1
|
Singh H, Kirar M, Yadav M, Dahiya N, Janjoter S, Sehrawat N. Molecular characterization and bioinformatic analysis of SGU protein in Anopheles culicifacies as target for transmission blocking activity. Immunol Res 2025; 73:34. [PMID: 39810016 DOI: 10.1007/s12026-024-09561-x] [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: 07/24/2024] [Accepted: 11/13/2024] [Indexed: 01/16/2025]
Abstract
In tropical countries, malaria transmission is the major health issue. To eradicate malaria, health communities depend on the control measure that affects economy and environment of the countries. To overcome these burdens, there is a great need to develop vaccine against malaria, but there is no vaccine to control malaria effectively. Transmission blocking vaccine (TBV) is the best alternative approach to control malaria, which inhibits the malaria transmission. Mosquito-based TBVs, blocks the transmission of Plasmodium developmental stages in mosquito. There are some potential candidate antigens for mosquito-based TBV, e.g., AgCPB1, AnAPN1, AgFREP1, etc. AcSGU is one of the most potential candidates for TBVs. AcSGU protein is glycol-anchored protein in Anopheles culicifacies which is highly expressed after blood feeding. In the present study acsgu gene was amplified from genomic DNA, sequenced (GenBank id: MN402758) and characterised. The sequence of acsgu (gene and protein) was analysed by different immuno-informatics tools to confirm its potentiality as a candidate antigen. The target protein was cloned, isolated and immunised for immunogenic response analysis. The acsgu gene is single exonic which encodes for AcSGU protein with single functional MBF2 domain. It is conserved in most of the Anopheles species. Bioinformatics analysis confirmed the stability and immunogenic nature of the protein. Protein-Protein interaction revealed effective interaction of AcSGU with Pf47 and TLR4 molecules. AcSGU protein was expressed in E. coli BL21 (DE3) by using expression vector pLATE51. The immunogenic response in AcSGU protein was remarkable in the rabbit. This study confirmed that AcSGU protein is the potential candidate for transmission blocking vaccine to generate anti-midgut immunity against plasmodium. It can be used as a candidate for the development of multistage targeting vaccines against malaria.
Collapse
Affiliation(s)
- Hitesh Singh
- Department of Life Science, PDM University, Bahadurgarh, Haryana, India
| | - Manisha Kirar
- School of Basic and Applied Sciences, Sanskaram University, Jhajjar, Haryana, India
| | - Mahima Yadav
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Nisha Dahiya
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Sangeeta Janjoter
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Neelam Sehrawat
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, India.
| |
Collapse
|
2
|
Dolan B, Correa Gaviria T, Dong Y, Cresswell P, Dimopoulos G, Chuang YM, Fikrig E. mosGILT antibodies interfere with Plasmodium sporogony in Anopheles gambiae. Nat Commun 2025; 16:592. [PMID: 39799117 PMCID: PMC11724845 DOI: 10.1038/s41467-025-55902-1] [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: 07/03/2024] [Accepted: 01/03/2025] [Indexed: 01/15/2025] Open
Abstract
Plasmodium, the causative agents of malaria, are obtained by mosquitoes from an infected human. Following Plasmodium acquisition by Anopheles gambiae, mosquito gamma-interferon-inducible lysosomal thiol reductase (mosGILT) plays a critical role in its subsequent sporogony in the mosquito. A critical location for this development is the midgut, a tissue we show expresses mosGILT. Using membrane-feeding and murine infection models, we demonstrate that antibodies against mosGILT reduce the number of P. falciparum and P. berghei oocysts in the midgut and the infection prevalence of both species in the mosquito. mosGILT antibodies act in the mosquito midgut, specifically impacting the Plasmodium oocyst stage. Targeting mosGILT can therefore interfere with the Plasmodium life cycle in the mosquito and potentially serve as a transmission-blocking vaccine.
Collapse
Affiliation(s)
- Brady Dolan
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Tomás Correa Gaviria
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Yuemei Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Peter Cresswell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Yu-Min Chuang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
3
|
Pourhashem Z, Nourani L, Pirahmadi S, Yousefi H, J. Sani J, Raz A, Zakeri S, Dinparast Djadid N, Abouie Mehrizi A. Malaria transmission blocking activity of Anopheles stephensi alanyl aminopeptidase N antigen formulated with MPL, CpG, and QS21 adjuvants. PLoS One 2024; 19:e0306664. [PMID: 38968270 PMCID: PMC11226095 DOI: 10.1371/journal.pone.0306664] [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: 10/24/2023] [Accepted: 02/29/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUNDS Malaria, a preventive and treatable disease, is still responsible for annual deaths reported in most tropical regions, principally in sub-Saharan Africa. Subunit recombinant transmission-blocking vaccines (TBVs) have been proposed as promising vaccines to succeed in malaria elimination and eradication. Here, a provisional study was designed to assess the immunogenicity and functional activity of alanyl aminopeptidase N (APN1) of Anopheles stephensi, as a TBV candidate, administered with MPL, CpG, and QS21 adjuvants in the murine model. METHODOLOGY/PRINCIPAL FINDINGS The mouse groups were immunized with recombinant APN1 (rAPN1) alone or formulated with CpG, MPL, QS-21, or a combination of adjuvants (CMQ), and the elicited immune responses were evaluated after the third immunization. The standard membrane feeding assay (SMFA) measured the functional activity of antibodies against bacterial-expressed APN1 protein in adjuvanted vaccine groups on transmission of P. falciparum (NF54) to An. stephensi mosquitoes. Evaluation of mice vaccinated with rAPN1 formulated with distinct adjuvants manifested a significant increase in the high-avidity level of anti-APN1 IgG and IgG subclasses; however, rAPN1 induced the highest level of high-avidity anti-APN1 IgG1, IgG2a, and IgG2b antibodies in the immunized vaccine group 5 (APN1/CMQ). In addition, vaccine group 5 (receiving APN1/CMQ), had still the highest level of anti-APN1 IgG antibodies relative to other immunized groups after six months, on day 180. The SMFA data indicates a trend towards higher transmission-reducing activity in groups 2 and 5, which received the antigen formulated with CpG or a combination of three adjuvants. CONCLUSIONS/SIGNIFICANCE The results have shown the capability of admixture to stimulate high-affinity and long-lasting antibodies against the target antigen to hinder Plasmodium parasite development in the mid-gut of An. stephensi. The attained results authenticated APN1/CMQ and APN1/CpG as a potent APN1-based TBV formulation which will be helpful in designing a vaccine in the future.
Collapse
Affiliation(s)
- Zeinab Pourhashem
- Pasteur Institute of Iran, Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Tehran, Iran
| | - Leila Nourani
- Pasteur Institute of Iran, Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Tehran, Iran
| | - Sakineh Pirahmadi
- Pasteur Institute of Iran, Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Tehran, Iran
| | - Hemn Yousefi
- Pasteur Institute of Iran, Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Tehran, Iran
| | - Jafar J. Sani
- Pasteur Institute of Iran, Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Tehran, Iran
| | - Abbasali Raz
- Pasteur Institute of Iran, Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Tehran, Iran
| | - Sedigheh Zakeri
- Pasteur Institute of Iran, Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Tehran, Iran
| | - Navid Dinparast Djadid
- Pasteur Institute of Iran, Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Tehran, Iran
| | - Akram Abouie Mehrizi
- Pasteur Institute of Iran, Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Tehran, Iran
| |
Collapse
|
4
|
Kefi M, Cardoso-Jaime V, Saab SA, Dimopoulos G. Curing mosquitoes with genetic approaches for malaria control. Trends Parasitol 2024; 40:487-499. [PMID: 38760256 DOI: 10.1016/j.pt.2024.04.010] [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: 03/12/2024] [Revised: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 05/19/2024]
Abstract
Malaria remains a persistent global public health challenge because of the limitations of current prevention tools. The use of transgenic mosquitoes incapable of transmitting malaria, in conjunction with existing methods, holds promise for achieving elimination of malaria and preventing its reintroduction. In this context, population modification involves the spread of engineered genetic elements through mosquito populations that render them incapable of malaria transmission. Significant progress has been made in this field over the past decade in revealing promising targets, optimizing genetic tools, and facilitating the transition from the laboratory to successful field deployments, which are subject to regulatory scrutiny. This review summarizes recent advances and ongoing challenges in 'curing' Anopheles vectors of the malaria parasite.
Collapse
Affiliation(s)
- Mary Kefi
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Victor Cardoso-Jaime
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Sally A Saab
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - George Dimopoulos
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
5
|
Pang W, Bai J, Zhu L, Liu F, Wu Y, Yang F, Zheng L, Liu P, Zhang Y, Wang M, Li J, Zhu X, Cui L, Cao Y. Functional characterization of a conserved membrane protein, Pbs54, involved in gamete fertilization in Plasmodium berghei. Int J Parasitol 2024; 54:99-107. [PMID: 37774810 PMCID: PMC11283859 DOI: 10.1016/j.ijpara.2023.08.003] [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: 07/18/2023] [Accepted: 08/25/2023] [Indexed: 10/01/2023]
Abstract
The successful completion of gamete fertilization is essential for malaria parasite transmission, and this process can be targeted by intervention strategies. In this study, we identified a conserved gene (PBANKA_0813300) in the rodent malaria parasite Plasmodium berghei, which encodes a protein of 54 kDa (designated as Pbs54). Localization studies indicated that Pbs54 is associated with the plasma membranes of gametes and ookinetes. Functional studies by gene disruption showed that the Δpbs54 parasites had no defect in asexual proliferation, gametocyte development, or gametogenesis. However, the interactions between male and female gametes were significantly decreased compared with wild-type parasites. The Δpbs54 lines did not show a further reduction in zygote and ookinete numbers during in vitro culture, indicating that the defects were probably restricted to gamete fertilization. Consistent with this finding, mosquitoes fed on Δpbs54-infected mice showed a 30.1% reduction in infection prevalence and a 74.7% reduction in oocyst intensity. Cross-fertilization assay indicated that both male and female gametes were impaired in the Δpbs54 parasites. To evaluate its transmission-blocking potential, we obtained polyclonal antibodies from mice immunized with the recombinant Pbs54 (rPbs54) protein. In vitro assays showed that anti-rPbs54 sera inhibited ookinete formation by 42.7%. Our experiments identified Pbs54 as a fertility factor required for mosquito transmission and a novel candidate for a malaria transmission-blocking vaccine.
Collapse
Affiliation(s)
- Wei Pang
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Jie Bai
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Liying Zhu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Fei Liu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yudi Wu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Fan Yang
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Li Zheng
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Pengbo Liu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yaowen Zhang
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Meilian Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Jun Li
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA
| | - Xiaotong Zhu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China.
| | - Liwang Cui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China.
| |
Collapse
|
6
|
Pourhashem Z, Nourani L, Sani JJ, Yousefi H, Pirahmadi S, Sabouri M, Raz A, Djadid ND, Zakeri S, Mehrizi AA. Evaluation of a new fusion antigen, cd loop and HAP2-GCS1 domain (cd-HAP) of Plasmodium falciparum Generative Cell Specific 1 antigen formulated with various adjuvants, as a transmission blocking vaccine. Malar J 2023; 22:374. [PMID: 38071314 PMCID: PMC10710725 DOI: 10.1186/s12936-023-04798-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Malaria is a major global health challenge, and for the elimination and eradication of this disease, transmission-blocking vaccines (TBVs) are a priority. Plasmodium falciparum Generative Cell Specific 1 (PfGCS1), a promising TBV candidate, is essential for gamete fertilization. The HAP2-GCS1 domain of this antigen as well as its cd loop could induce antibodies that partially inhibit transmission of P. falciparum. METHODS In the current study, a new synthetic fusion antigen containing cd loop and HAP2-GCS1 domain (cd-HAP) of PfGCS1 was evaluated as a transmission blocking vaccine candidate. Initially, the profile of naturally acquired IgG antibodies to the cd-HAP antigen was analysed in Iranian individuals infected with P. falciparum, to confirm that this new fusion protein has the appropriate structure containing common epitopes with the native form of PfGCS1. Then, the immunogenicity of cd-HAP was evaluated in BALB/c mice, using different adjuvant systems such as CpG, MPL, QS-21, and a combination of them (CMQ). Furthermore, the blocking efficacy of polyclonal antibodies induced against these formulations was also assessed by oocyst intensity and infection prevalence in the Standard Membrane Feeding Assay (SMFA). RESULTS The naturally acquired antibodies (dominantly IgG1 and IgG3 subclasses) induced in P. falciparum-infected individuals could recognize the cd-HAP antigen which implies that the new fusion protein has a proper conformation that mimics the native structure of PfGCS1. Concerning the immunogenicity of cd-HAP antigen, the highest IgG levels and titers, by a Th1-type immune profile, and elevated antibody avidity were induced in mice immunized with the cd-HAP antigen formulated with a combination of adjuvants (P < 0.0001). Additionally, cytokine profiling of the immunized mice displayed that a high level of IFN-γ response, a Th1-type immune response, was produced by splenocytes from immunized mice that received cd-HAP antigen in combination with CMQ adjuvants (P < 0.0001). This formulation of cd-HAP antigen with CMQ adjuvants could reduce oocyst intensity and infection prevalence by 82%, evidenced by the SMFA and hold significant implications for future malaria vaccine development. CONCLUSION Altogether, the results showed that cd-HAP antigen formulated with a combination of the adjuvants (CMQ), could be a promising formulation to develop a PfGCS1-based transmission-blocking vaccine.
Collapse
Affiliation(s)
- Zeinab Pourhashem
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Leila Nourani
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Jafar J Sani
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Hemn Yousefi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Sakineh Pirahmadi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Mobina Sabouri
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Abbasali Raz
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Navid Dinparast Djadid
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Sedigheh Zakeri
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.
| | - Akram Abouie Mehrizi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
7
|
Leite JC, Gonçalves AAM, de Oliveira DS, Resende LA, Boas DFV, Ribeiro HS, Pereira DFS, da Silva AV, Mariano RMDS, Reis PCC, Nakasone EN, França-Silva JC, Galdino AS, Paes PRDO, Melo MM, Dias ES, Chávez-Fumagalli MA, da Silveira-Lemos D, Dutra WO, Giunchetti RC. Transmission-Blocking Vaccines for Canine Visceral Leishmaniasis: New Progress and Yet New Challenges. Vaccines (Basel) 2023; 11:1565. [PMID: 37896969 PMCID: PMC10610753 DOI: 10.3390/vaccines11101565] [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: 08/09/2023] [Revised: 09/17/2023] [Accepted: 09/19/2023] [Indexed: 10/29/2023] Open
Abstract
Dogs with visceral leishmaniasis play a key role in the transmission cycle of Leishmania infantum to humans in the urban environment. There is a consensus regarding the importance of developing a vaccine to control this disease. Despite many efforts to develop a protective vaccine against CVL, the ones currently available, Leish-tec® and LetiFend®, have limited effectiveness. This is due, in part, to the complexity of the immune response of the naturally infected dogs against the parasite and the complexity of the parasite transmission cycle. Thus, strategies, such as the development of a transmission-blocking vaccines (TBVs) already being applied to other vector-borne diseases like malaria and dengue, would be an attractive alternative to control leishmaniasis. TBVs induce the production of antibodies in the vertebrate host, which can inhibit parasite development in the vector and/or interfere with aspects of vector biology, leading to an interruption of parasite transmission. To date, there are few TBV studies for CVL and other leishmaniasis forms. However, the few studies that exist show promising results, thus justifying the further development of this approach.
Collapse
Affiliation(s)
- Jaqueline Costa Leite
- Laboratory of Biology of Cell Interactions, Department of Morphology, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil; (J.C.L.); (A.A.M.G.); (D.S.d.O.); (L.A.R.); (D.F.V.B.); (H.S.R.); (D.F.S.P.); (A.V.d.S.); (R.M.d.S.M.); (P.C.C.R.); (E.N.N.); (J.C.F.-S.); (D.d.S.-L.); (W.O.D.)
| | - Ana Alice Maia Gonçalves
- Laboratory of Biology of Cell Interactions, Department of Morphology, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil; (J.C.L.); (A.A.M.G.); (D.S.d.O.); (L.A.R.); (D.F.V.B.); (H.S.R.); (D.F.S.P.); (A.V.d.S.); (R.M.d.S.M.); (P.C.C.R.); (E.N.N.); (J.C.F.-S.); (D.d.S.-L.); (W.O.D.)
| | - Diana Souza de Oliveira
- Laboratory of Biology of Cell Interactions, Department of Morphology, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil; (J.C.L.); (A.A.M.G.); (D.S.d.O.); (L.A.R.); (D.F.V.B.); (H.S.R.); (D.F.S.P.); (A.V.d.S.); (R.M.d.S.M.); (P.C.C.R.); (E.N.N.); (J.C.F.-S.); (D.d.S.-L.); (W.O.D.)
| | - Lucilene Aparecida Resende
- Laboratory of Biology of Cell Interactions, Department of Morphology, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil; (J.C.L.); (A.A.M.G.); (D.S.d.O.); (L.A.R.); (D.F.V.B.); (H.S.R.); (D.F.S.P.); (A.V.d.S.); (R.M.d.S.M.); (P.C.C.R.); (E.N.N.); (J.C.F.-S.); (D.d.S.-L.); (W.O.D.)
| | - Diego Fernandes Vilas Boas
- Laboratory of Biology of Cell Interactions, Department of Morphology, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil; (J.C.L.); (A.A.M.G.); (D.S.d.O.); (L.A.R.); (D.F.V.B.); (H.S.R.); (D.F.S.P.); (A.V.d.S.); (R.M.d.S.M.); (P.C.C.R.); (E.N.N.); (J.C.F.-S.); (D.d.S.-L.); (W.O.D.)
| | - Helen Silva Ribeiro
- Laboratory of Biology of Cell Interactions, Department of Morphology, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil; (J.C.L.); (A.A.M.G.); (D.S.d.O.); (L.A.R.); (D.F.V.B.); (H.S.R.); (D.F.S.P.); (A.V.d.S.); (R.M.d.S.M.); (P.C.C.R.); (E.N.N.); (J.C.F.-S.); (D.d.S.-L.); (W.O.D.)
| | - Diogo Fonseca Soares Pereira
- Laboratory of Biology of Cell Interactions, Department of Morphology, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil; (J.C.L.); (A.A.M.G.); (D.S.d.O.); (L.A.R.); (D.F.V.B.); (H.S.R.); (D.F.S.P.); (A.V.d.S.); (R.M.d.S.M.); (P.C.C.R.); (E.N.N.); (J.C.F.-S.); (D.d.S.-L.); (W.O.D.)
| | - Augusto Ventura da Silva
- Laboratory of Biology of Cell Interactions, Department of Morphology, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil; (J.C.L.); (A.A.M.G.); (D.S.d.O.); (L.A.R.); (D.F.V.B.); (H.S.R.); (D.F.S.P.); (A.V.d.S.); (R.M.d.S.M.); (P.C.C.R.); (E.N.N.); (J.C.F.-S.); (D.d.S.-L.); (W.O.D.)
| | - Reysla Maria da Silveira Mariano
- Laboratory of Biology of Cell Interactions, Department of Morphology, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil; (J.C.L.); (A.A.M.G.); (D.S.d.O.); (L.A.R.); (D.F.V.B.); (H.S.R.); (D.F.S.P.); (A.V.d.S.); (R.M.d.S.M.); (P.C.C.R.); (E.N.N.); (J.C.F.-S.); (D.d.S.-L.); (W.O.D.)
| | - Pedro Campos Carvalhaes Reis
- Laboratory of Biology of Cell Interactions, Department of Morphology, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil; (J.C.L.); (A.A.M.G.); (D.S.d.O.); (L.A.R.); (D.F.V.B.); (H.S.R.); (D.F.S.P.); (A.V.d.S.); (R.M.d.S.M.); (P.C.C.R.); (E.N.N.); (J.C.F.-S.); (D.d.S.-L.); (W.O.D.)
| | - Eiji Nakasone Nakasone
- Laboratory of Biology of Cell Interactions, Department of Morphology, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil; (J.C.L.); (A.A.M.G.); (D.S.d.O.); (L.A.R.); (D.F.V.B.); (H.S.R.); (D.F.S.P.); (A.V.d.S.); (R.M.d.S.M.); (P.C.C.R.); (E.N.N.); (J.C.F.-S.); (D.d.S.-L.); (W.O.D.)
| | - João Carlos França-Silva
- Laboratory of Biology of Cell Interactions, Department of Morphology, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil; (J.C.L.); (A.A.M.G.); (D.S.d.O.); (L.A.R.); (D.F.V.B.); (H.S.R.); (D.F.S.P.); (A.V.d.S.); (R.M.d.S.M.); (P.C.C.R.); (E.N.N.); (J.C.F.-S.); (D.d.S.-L.); (W.O.D.)
| | - Alexsandro Sobreira Galdino
- Microorganism Biotechnology Laboratory, Federal University of São João Del-Rei (UFSJ), Midwest Campus, Divinópolis 35501-296, MG, Brazil;
| | - Paulo Ricardo de Oliveira Paes
- Department of Veterinary Clinic and Surgery, School of Veterinary, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil; (P.R.d.O.P.); (M.M.M.)
| | - Marília Martins Melo
- Department of Veterinary Clinic and Surgery, School of Veterinary, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil; (P.R.d.O.P.); (M.M.M.)
| | - Edelberto Santos Dias
- René Rachou Research Center, Oswaldo Cruz Foundation, Belo Horizonte 30190-002, MG, Brazil;
| | - Miguel Angel Chávez-Fumagalli
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Arequipa 04000, Peru;
| | - Denise da Silveira-Lemos
- Laboratory of Biology of Cell Interactions, Department of Morphology, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil; (J.C.L.); (A.A.M.G.); (D.S.d.O.); (L.A.R.); (D.F.V.B.); (H.S.R.); (D.F.S.P.); (A.V.d.S.); (R.M.d.S.M.); (P.C.C.R.); (E.N.N.); (J.C.F.-S.); (D.d.S.-L.); (W.O.D.)
| | - Walderez Ornelas Dutra
- Laboratory of Biology of Cell Interactions, Department of Morphology, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil; (J.C.L.); (A.A.M.G.); (D.S.d.O.); (L.A.R.); (D.F.V.B.); (H.S.R.); (D.F.S.P.); (A.V.d.S.); (R.M.d.S.M.); (P.C.C.R.); (E.N.N.); (J.C.F.-S.); (D.d.S.-L.); (W.O.D.)
| | - Rodolfo Cordeiro Giunchetti
- Laboratory of Biology of Cell Interactions, Department of Morphology, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil; (J.C.L.); (A.A.M.G.); (D.S.d.O.); (L.A.R.); (D.F.V.B.); (H.S.R.); (D.F.S.P.); (A.V.d.S.); (R.M.d.S.M.); (P.C.C.R.); (E.N.N.); (J.C.F.-S.); (D.d.S.-L.); (W.O.D.)
| |
Collapse
|
8
|
Ouologuem DT, Dara A, Kone A, Ouattara A, Djimde AA. Plasmodium falciparum Development from Gametocyte to Oocyst: Insight from Functional Studies. Microorganisms 2023; 11:1966. [PMID: 37630530 PMCID: PMC10460021 DOI: 10.3390/microorganisms11081966] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/06/2023] [Accepted: 07/14/2023] [Indexed: 08/27/2023] Open
Abstract
Malaria elimination may never succeed without the implementation of transmission-blocking strategies. The transmission of Plasmodium spp. parasites from the human host to the mosquito vector depends on circulating gametocytes in the peripheral blood of the vertebrate host. Once ingested by the mosquito during blood meals, these sexual forms undergo a series of radical morphological and metabolic changes to survive and progress from the gut to the salivary glands, where they will be waiting to be injected into the vertebrate host. The design of effective transmission-blocking strategies requires a thorough understanding of all the mechanisms that drive the development of gametocytes, gametes, sexual reproduction, and subsequent differentiation within the mosquito. The drastic changes in Plasmodium falciparum shape and function throughout its life cycle rely on the tight regulation of stage-specific gene expression. This review outlines the mechanisms involved in Plasmodium falciparum sexual stage development in both the human and mosquito vector, and zygote to oocyst differentiation. Functional studies unravel mechanisms employed by P. falciparum to orchestrate the expression of stage-specific functional products required to succeed in its complex life cycle, thus providing us with potential targets for developing new therapeutics. These mechanisms are based on studies conducted with various Plasmodium species, including predominantly P. falciparum and the rodent malaria parasites P. berghei. However, the great potential of epigenetics, genomics, transcriptomics, proteomics, and functional genetic studies to improve the understanding of malaria as a disease remains partly untapped because of limitations in studies using human malaria parasites and field isolates.
Collapse
Affiliation(s)
- Dinkorma T. Ouologuem
- Malaria Research and Training Center, Faculty of Pharmacy, Faculty of Medicine and Dentistry, University of Sciences, Techniques, and Technologies of Bamako, Bamako 1805, Mali
| | - Antoine Dara
- Malaria Research and Training Center, Faculty of Pharmacy, Faculty of Medicine and Dentistry, University of Sciences, Techniques, and Technologies of Bamako, Bamako 1805, Mali
| | - Aminatou Kone
- Malaria Research and Training Center, Faculty of Pharmacy, Faculty of Medicine and Dentistry, University of Sciences, Techniques, and Technologies of Bamako, Bamako 1805, Mali
| | - Amed Ouattara
- Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Abdoulaye A. Djimde
- Malaria Research and Training Center, Faculty of Pharmacy, Faculty of Medicine and Dentistry, University of Sciences, Techniques, and Technologies of Bamako, Bamako 1805, Mali
| |
Collapse
|
9
|
Mizushima D, Yamamoto DS, Tabbabi A, Arai M, Kato H. A rare sugar, allose, inhibits the development of Plasmodium parasites in the Anopheles mosquito independently of midgut microbiota. Front Cell Infect Microbiol 2023; 13:1162918. [PMID: 37545855 PMCID: PMC10400720 DOI: 10.3389/fcimb.2023.1162918] [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: 02/21/2023] [Accepted: 06/28/2023] [Indexed: 08/08/2023] Open
Abstract
A rare sugar, allose, was reported to inhibit the development of Plasmodium parasites in Anopheles mosquitoes; however, the mechanism remains unknown. The present study addressed the inhibitory mechanism of allose on the development of the Plasmodium parasite by connecting it with bacteria involvement in the midgut. In addition, further inhibitory sugars against Plasmodium infection in mosquitoes were explored. Antibiotic-treated and antibiotic-untreated Anopheles stephensi were fed fructose with or without allose. The mosquitoes were infected with luciferase-expressing Plasmodium berghei, and parasite development was evaluated by luciferase activity. Bacterial composition analysis in gut of their mosquitoes was performed with comprehensive 16S ribosomal RNA sequencing. As the result, allose inhibited the development of oocysts in mosquitoes regardless of prior antibiotic treatment. Microbiome analysis showed that the midgut bacterial composition in mosquitoes before and after blood feeding was not affected by allose. Although allose inhibited transient growth of the midgut microbiota of mosquitoes after blood feeding, neither toxic nor inhibitory effects of allose on the dominant midgut bacteria were observed. Ookinete development in the mosquito midgut was also not affected by allose feeding. Additional 15 sugars including six monosaccharides, four polyols, and five polysaccharides were tested; however, no inhibitory effect against Plasmodium development in mosquitoes was observed. These results indicated that allose inhibits parasite development in midgut stage of the mosquito independently of midgut microbiota. Although further studies are needed, our results suggest that allose may be a useful material for the vector control of malaria as a "transmission-blocking sugar."
Collapse
Affiliation(s)
- Daiki Mizushima
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Daisuke S. Yamamoto
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Ahmed Tabbabi
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Meiji Arai
- Department of International Medical Zoology, Faculty of Medicine, Kagawa University, Kita-gun, Kagawa, Japan
| | - Hirotomo Kato
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
10
|
Parres-Mercader M, Pance A, Gómez-Díaz E. Novel systems to study vector-pathogen interactions in malaria. Front Cell Infect Microbiol 2023; 13:1146030. [PMID: 37305421 PMCID: PMC10253182 DOI: 10.3389/fcimb.2023.1146030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/04/2023] [Indexed: 06/13/2023] Open
Abstract
Some parasitic diseases, such as malaria, require two hosts to complete their lifecycle: a human and an insect vector. Although most malaria research has focused on parasite development in the human host, the life cycle within the vector is critical for the propagation of the disease. The mosquito stage of the Plasmodium lifecycle represents a major demographic bottleneck, crucial for transmission blocking strategies. Furthermore, it is in the vector, where sexual recombination occurs generating "de novo" genetic diversity, which can favor the spread of drug resistance and hinder effective vaccine development. However, understanding of vector-parasite interactions is hampered by the lack of experimental systems that mimic the natural environment while allowing to control and standardize the complexity of the interactions. The breakthrough in stem cell technologies has provided new insights into human-pathogen interactions, but these advances have not been translated into insect models. Here, we review in vivo and in vitro systems that have been used so far to study malaria in the mosquito. We also highlight the relevance of single-cell technologies to progress understanding of these interactions with higher resolution and depth. Finally, we emphasize the necessity to develop robust and accessible ex vivo systems (tissues and organs) to enable investigation of the molecular mechanisms of parasite-vector interactions providing new targets for malaria control.
Collapse
Affiliation(s)
- Marina Parres-Mercader
- Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas (IPBLN, CSIC), Granada, Spain
| | - Alena Pance
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield, United Kingdom
| | - Elena Gómez-Díaz
- Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas (IPBLN, CSIC), Granada, Spain
| |
Collapse
|
11
|
Izquierdo L. The glycobiology of plasmodium falciparum: New approaches and recent advances. Biotechnol Adv 2023; 66:108178. [PMID: 37216996 DOI: 10.1016/j.biotechadv.2023.108178] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 04/22/2023] [Accepted: 05/18/2023] [Indexed: 05/24/2023]
Abstract
Like any other microorganism, pathogenic protozoan parasites rely heavily on glycoconjugates and glycan binding proteins to protect themselves from the environment and to interact with their diverse hosts. A thorough comprehension of how glycobiology contributes to the survival and virulence of these organisms may reveal unknown aspects of their biology and may open much needed avenues for the design of new strategies against them. In the case of Plasmodium falciparum, which causes the vast majority of malaria cases and deaths, the restricted variety and the simplicity of its glycans seemed to confer limited significance to the role played by glycoconjugates in the parasite. Nonetheless, the last 10 to 15 years of research are revealing a clearer and more defined picture. Thus, the use of new experimental techniques and the results obtained provide new avenues for understanding the biology of the parasite, as well as opportunities for the development of much required new tools against malaria.
Collapse
Affiliation(s)
- Luis Izquierdo
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain; CIBER de Enfermedades Infecciosas, Madrid, Spain.
| |
Collapse
|
12
|
Simons LM, Ferrer P, Gombakomba N, Underwood K, Herrera R, Narum DL, Canepa G, Acquah F, Amoah L, Duffy PE, Barillas-Mury C, Long C, Lee SM, Locke E, Miura K, Williamson KC. Extending the range of Plasmodium falciparum transmission blocking antibodies. Vaccine 2023; 41:3367-3379. [PMID: 37100721 PMCID: PMC10334573 DOI: 10.1016/j.vaccine.2023.04.042] [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: 12/01/2022] [Revised: 04/01/2023] [Accepted: 04/14/2023] [Indexed: 04/28/2023]
Abstract
Recent work demonstrating that asymptomatic carriers of P. falciparum parasites make up a large part of the infectious reservoir highlights the need for an effective malaria vaccine. Given the historical challenges of vaccine development, multiple parasite stages have been targeted, including the sexual stages required for transmission. Using flow cytometry to efficiently screen for P. falciparum gamete/zygote surface reactivity, we identified 82 antibodies that bound live P. falciparum gametes/zygotes. Ten antibodies had significant transmission-reducing activity (TRA) in a standard membrane feeding assay and were subcloned along with 9 nonTRA antibodies as comparators. After subcloning, only eight of the monoclonals obtained have significant TRA. These eight TRA mAbs do not recognize epitopes present in any of the current recombinant transmission-blocking vaccine candidates, Pfs230D1M, Pfs48/45.6C, Pf47 D2 and rPfs25. One TRA mAb immunoprecipitates two surface antigens, Pfs47 and Pfs230, that are expressed by both gametocytes and gametes/zygotes. These two proteins have not previously been reported to associate and the recognition of both by a single TRA mAb suggests the Pfs47/Pfs230 complex is a new vaccine target. In total, Pfs230 was the dominant target antigen, with five of the eight TRA mAbs and 8 of 11 nonTRA gamete/zygote surface reactive mAbs interacting with Pfs230. Of the three remaining TRA mAbs, two recognized non-reduced, parasite-produced Pfs25 and one bound non-reduced, parasite-produced Pfs48/45. None of the TRA mAbs bound protein on an immunoblot of reduced gamete/zygote extract and two TRA mAbs were immunoblot negative, indicating none of the new TRA epitopes are linear. The identification of eight new TRA mAbs that bind epitopes not included in any of the constructs currently under advancement as transmission-blocking vaccine candidates may provide new targets worthy of further study.
Collapse
Affiliation(s)
- Lacy M Simons
- Department of Biology, Loyola University Chicago, 1032 W Sheridan Rd, Chicago, IL 60660, USA
| | - Patricia Ferrer
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd., Bethesda, MD, 20814, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Suite 100, Bethesda, USA
| | - Nita Gombakomba
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd., Bethesda, MD, 20814, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Suite 100, Bethesda, USA
| | - Knashka Underwood
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd., Bethesda, MD, 20814, USA
| | - Raul Herrera
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - David L Narum
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Gaspar Canepa
- Mosquito Immunity and Vector Competence Section Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, MD 20852, USA
| | - Festus Acquah
- Immunology Department, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Linda Amoah
- Immunology Department, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Patrick E Duffy
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Carolina Barillas-Mury
- Mosquito Immunity and Vector Competence Section Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, MD 20852, USA
| | - Carole Long
- Malaria Immunology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, MD 20852, USA
| | - Shwu-Maan Lee
- Center for Vaccine Innovation and Access, PATH, 455 Massachusetts Ave, NW, Suite 1000, Washington, DC, 20001, USA
| | - Emily Locke
- Center for Vaccine Innovation and Access, PATH, 455 Massachusetts Ave, NW, Suite 1000, Washington, DC, 20001, USA
| | - Kazutoyo Miura
- Malaria Immunology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, MD 20852, USA
| | - Kim C Williamson
- Department of Biology, Loyola University Chicago, 1032 W Sheridan Rd, Chicago, IL 60660, USA; Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd., Bethesda, MD, 20814, USA.
| |
Collapse
|
13
|
Ramelow J, Keleta Y, Niu G, Wang X, Li J. Plasmodium parasitophorous vacuole membrane protein Pfs16 promotes malaria transmission by silencing mosquito immunity. J Biol Chem 2023:104824. [PMID: 37196765 DOI: 10.1016/j.jbc.2023.104824] [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: 12/26/2022] [Revised: 04/07/2023] [Accepted: 05/08/2023] [Indexed: 05/19/2023] Open
Abstract
With rising cases for the first time in years, malaria remains a significant public health burden. The sexual stage of the malaria parasite infects mosquitoes to transmit malaria from host to host. Hence, an infected mosquito plays an essential role in malaria transmission. Plasmodium falciparum is the most dominant and dangerous malaria pathogen. Previous studies identified a sexual stage-specific protein 16 (Pfs16) localized to the parasitophorous vacuole membrane (PVM). Here we elucidate the function of Pfs16 during malaria transmission. Our structural analysis identified Pfs16 as an alpha-helical integral membrane protein with one transmembrane domain connecting to two regions across PVM. ELISA assays showed that insect cell-expressed recombinant Pfs16 (rPfs16) interacted with An. gambiae midguts, and microscopy found that rPfs16 bound to midgut epithelial cells. Transmission-blocking assays demonstrated that polyclonal antibodies against Pfs16 significantly reduced the number of oocysts in mosquito midguts. However, on the contrary, feeding rPfs16 increased the number of oocysts. Further analysis revealed that Pfs16 reduced the activity of mosquito midgut caspase 3/7, a key enzyme in the mosquito Jun-N-terminal kinase (JNK) immune pathway. We conclude that Pfs16 facilitates parasites to invade mosquito midguts by actively silencing the mosquito's innate immunity through its interaction with the midgut epithelial cells. Therefore, Pfs16 is a potential target to control malaria transmission.
Collapse
Affiliation(s)
- Julian Ramelow
- Biomedical Sciences Graduate Program, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Yacob Keleta
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA
| | - Guodong Niu
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA
| | - Xiaohong Wang
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA
| | - Jun Li
- Biomedical Sciences Graduate Program, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; Department of Biological Sciences, Florida International University, Miami, FL 33199, USA; Biomolecular Sciences Institute, Florida International University, Miami, FL 33199, USA.
| |
Collapse
|
14
|
Rashidi S, Mansouri R, Ali-Hassanzadeh M, Muro A, Nguewa P, Manzano-Román R. The most prominent modulated annexins during parasitic infections. Acta Trop 2023; 243:106942. [PMID: 37172709 DOI: 10.1016/j.actatropica.2023.106942] [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: 02/27/2023] [Revised: 05/05/2023] [Accepted: 05/07/2023] [Indexed: 05/15/2023]
Abstract
Annexins (ANXs) exert different functions in cell biological and pathological processes and are thus known as double or multi-faceted proteins. These sophisticated proteins might express on both parasite structure and secretion and in parasite-infected host cells. In addition to the characterization of these pivotal proteins, describing their mechanism of action can be also fruitful in recognizing their roles in the pathogenesis of parasitic infections. Accordingly, this study presents the most prominent ANXs thus far identified and their relevant functions in parasites and infected host cells during pathogenesis, especially in the most important intracellular protozoan parasitic infections including leishmaniasis, toxoplasmosis, malaria and trypanosomiasis. The data provided in this study demonstrate that the helminth parasites most probably express and secret ANXs to develop pathogenesis while the modulation of the host-ANXs could be employed as a crucial strategy by intracellular protozoan parasites. Moreover, such data highlight that the use of analogs of both parasite and host ANX peptides (which mimic or regulate ANXs physiological functions through various strategies) might suggest novel therapeutic insights into the treatment of parasitic infections. Furthermore, due to the prominent immunoregulatory activities of ANXs during most parasitic infections and the expression levels of these proteins in some parasitic infected tissues, such multifunctional proteins might be also potentially relevant as vaccine and diagnostic biomarkers. We also suggest some prospects and insights that could be useful and applicable to form the basis of future experimental studies.
Collapse
Affiliation(s)
- Sajad Rashidi
- Molecular and Medicine Research Center, Khomein University of Medical Sciences, Khomein, Iran; Department of Medical Laboratory Sciences, Khomein University of Medical Sciences, Khomein, Iran
| | - Reza Mansouri
- Department of Immunology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Mohammad Ali-Hassanzadeh
- Department of Immunology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Antonio Muro
- Infectious and Tropical Diseases Group (e-INTRO), Institute of Biomedical Research of Salamanca-Research Center for Tropical Diseases at the University of Salamanca (IBSAL-CIETUS), Faculty of Pharmacy, University of Salamanca, 37008 Salamanca, Spain
| | - Paul Nguewa
- University of Navarra, ISTUN Institute of Tropical Health, Department of Microbiology and Parasitology. IdiSNA (Navarra Institute for Health Research), c/ Irunlarrea 1, 31008 Pamplona, Spain.
| | - Raúl Manzano-Román
- Infectious and Tropical Diseases Group (e-INTRO), Institute of Biomedical Research of Salamanca-Research Center for Tropical Diseases at the University of Salamanca (IBSAL-CIETUS), Faculty of Pharmacy, University of Salamanca, 37008 Salamanca, Spain.
| |
Collapse
|
15
|
Kaur H, Garber L, Murphy JW, Vinetz JM. Structure-function analysis of cysteine residues in the plasmodium falciparum chitinase, PfCHT1. Protein Sci 2022; 31:e4289. [PMID: 35481637 PMCID: PMC8994504 DOI: 10.1002/pro.4289] [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: 12/20/2021] [Revised: 02/13/2022] [Accepted: 02/16/2022] [Indexed: 11/11/2022]
Abstract
The Plasmodium ookinete uses chitinase activity to penetrate the acellular, chitin-containing peritrophic matrix to invade the mosquito vector. Plasmodium ookinetes from different parasite clades secrete two structurally distinct forms of chitinase, one, a short form lacking a C-terminal putative chitin-binding domain (CBD), the other, a long form with both proenzyme and C-terminal putative chitin-binding domains. Here, we structurally and functionally characterize the three cysteines in the short chitinase of the human-infecting malaria parasite, P. falciparum testing the hypothesis that one unpaired cysteine would not contribute to chitinase-specific enzymatic activity which would identify this residue as potentially involved in intermolecular disulfide bonding and heteromultimeric invasion complex formation as previously described. To test this hypothesis, we produced and characterized recombinant wild-type and cysteine-mutation PfCHT1 proteins in E. coli and used biophysical and enzymatic approaches to examine their enzymatic activities and chitin-binding affinities. The cysteine-203 PfCHT1 mutation had no effect on chitinolytic and chitin-binding functions. The cysteine-220 and cysteine-230 mutants were enzymatically inactive and did not bind to chitin. The artificial intelligence-based protein prediction algorithm, AlphaFold, correctly identified the involvement of cys-220 and cys-230 in the intramolecular disulfide linkages key to maintaining properly folded chitinase structural integrity. AlphaFold predicted that cys-203 cysteine is surface exposed and thus involved in intermolecular protein-protein interaction. Production of the cys-to-ser 203 PfCHT1 mutant facilitated recombinant protein production. Future cellular and biochemical studies are needed to further understand details of Plasmodium ookinete mosquito midgut invasion.
Collapse
Affiliation(s)
- Hargobinder Kaur
- Section of Infectious Diseases, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
| | - Laine Garber
- Section of Infectious Diseases, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
| | - James W. Murphy
- Department of PharmacologyYale University School of MedicineNew HavenConnecticutUSA
| | - Joseph M. Vinetz
- Section of Infectious Diseases, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
| |
Collapse
|
16
|
Aleshnick M, Florez-Cuadros M, Martinson T, Wilder BK. Monoclonal antibodies for malaria prevention. Mol Ther 2022; 30:1810-1821. [PMID: 35395399 PMCID: PMC8979832 DOI: 10.1016/j.ymthe.2022.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/04/2022] [Accepted: 04/01/2022] [Indexed: 11/29/2022] Open
Abstract
Monoclonal antibodies are highly specific proteins that are cloned from a single B cell and bind to a single epitope on a pathogen. These laboratory-made molecules can serve as prophylactics or therapeutics for infectious diseases and have an impressive capacity to modulate the progression of disease, as demonstrated for the first time on a large scale during the COVID-19 pandemic. The high specificity and natural starting point of monoclonal antibodies afford an encouraging safety profile, yet the high cost of production remains a major limitation to their widespread use. While a monoclonal antibody approach to abrogating malaria infection is not yet available, the unique life cycle of the malaria parasite affords many opportunities for such proteins to act, and preliminary research into the efficacy of monoclonal antibodies in preventing malaria infection, disease, and transmission is encouraging. This review examines the current status and future outlook for monoclonal antibodies against malaria in the context of the complex life cycle and varied antigenic targets expressed in the human and mosquito hosts, and provides insight into the strengths and limitations of this approach to curtailing one of humanity’s oldest and deadliest diseases.
Collapse
Affiliation(s)
- Maya Aleshnick
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | | | - Thomas Martinson
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Brandon K Wilder
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA; Department of Parasitology, U.S. Naval Medical Research 6 (NAMRU-6), Lima, Peru
| |
Collapse
|
17
|
Tavares CS, Mishra R, Ghobrial PN, Bonning BC. Composition and abundance of midgut surface proteins in the Asian citrus psyllid, Diaphorina citri. J Proteomics 2022; 261:104580. [DOI: 10.1016/j.jprot.2022.104580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/04/2022] [Accepted: 04/04/2022] [Indexed: 10/18/2022]
|
18
|
Yu S, Wang J, Luo X, Zheng H, Wang L, Yang X, Wang Y. Transmission-Blocking Strategies Against Malaria Parasites During Their Mosquito Stages. Front Cell Infect Microbiol 2022; 12:820650. [PMID: 35252033 PMCID: PMC8889032 DOI: 10.3389/fcimb.2022.820650] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/31/2022] [Indexed: 11/24/2022] Open
Abstract
Malaria is still the most widespread parasitic disease and causes the most infections globally. Owing to improvements in sanitary conditions and various intervention measures, including the use of antimalarial drugs, the malaria epidemic in many regions of the world has improved significantly in the past 10 years. However, people living in certain underdeveloped areas are still under threat. Even in some well-controlled areas, the decline in malaria infection rates has stagnated or the rates have rebounded because of the emergence and spread of drug-resistant malaria parasites. Thus, new malaria control methods must be developed. As the spread of the Plasmodium parasite is dependent on the part of its life cycle that occurs in mosquitoes, to eliminate the possibility of malaria infections, transmission-blocking strategies against the mosquito stage should be the first choice. In fact, after the gametocyte enters the mosquito body, it undergoes a series of transformation processes over a short period, thus providing numerous potential blocking targets. Many research groups have carried out studies based on targeting the blocking of transmission during the mosquito phase and have achieved excellent results. Meanwhile, the direct killing of mosquitoes could also significantly reduce the probability of malaria infections. Microorganisms that display complex interactions with Plasmodium, such as Wolbachia and gut flora, have shown observable transmission-blocking potential. These could be used as a biological control strategy and play an important part in blocking the transmission of malaria.
Collapse
Affiliation(s)
- Shasha Yu
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, China
| | - Jing Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, China
| | - Xue Luo
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, China
| | - Hong Zheng
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Luhan Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, China
| | - Xuesen Yang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, China
| | - Ying Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, China
- *Correspondence: Ying Wang,
| |
Collapse
|
19
|
Tachibana M, Takashima E, Morita M, Sattabongkot J, Ishino T, Culleton R, Torii M, Tsuboi T. Plasmodium vivax transmission-blocking vaccines: Progress, challenges and innovation. Parasitol Int 2021; 87:102525. [PMID: 34896614 DOI: 10.1016/j.parint.2021.102525] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 12/04/2021] [Indexed: 12/12/2022]
Abstract
Existing control measures have significantly reduced malaria morbidity and mortality in the last two decades, although these reductions are now stalling. Significant efforts have been undertaken to develop malaria vaccines. Recently, extensive progress in malaria vaccine development has been made for Plasmodium falciparum. To date, only the RTS,S/AS01 vaccine has been tested in Phase 3 clinical trials and is now under implementation, despite modest efficacy. Therefore, the development of a malaria transmission-blocking vaccine (TBV) will be essential for malaria elimination. Only a limited number of TBVs have reached pre-clinical or clinical development with several major challenges impeding their development, including low immunogenicity in humans. TBV development efforts against P. vivax, the second major cause of malaria morbidity, lag far behind those for P. falciparum. In this review we summarize the latest progress, challenges and innovations in P. vivax TBV research and discuss how to accelerate its development.
Collapse
Affiliation(s)
- Mayumi Tachibana
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime 791-0295, Japan.
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan.
| | - Masayuki Morita
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan.
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand.
| | - Tomoko Ishino
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime 791-0295, Japan.
| | - Richard Culleton
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime 791-0295, Japan.
| | - Motomi Torii
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime 791-0295, Japan; Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan.
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan.
| |
Collapse
|
20
|
Keleta Y, Ramelow J, Cui L, Li J. Molecular interactions between parasite and mosquito during midgut invasion as targets to block malaria transmission. NPJ Vaccines 2021; 6:140. [PMID: 34845210 PMCID: PMC8630063 DOI: 10.1038/s41541-021-00401-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 11/01/2021] [Indexed: 11/21/2022] Open
Abstract
Despite considerable effort, malaria remains a major public health burden. Malaria is caused by five Plasmodium species and is transmitted to humans via the female Anopheles mosquito. The development of malaria vaccines against the liver and blood stages has been challenging. Therefore, malaria elimination strategies advocate integrated measures, including transmission-blocking approaches. Designing an effective transmission-blocking strategy relies on a sophisticated understanding of the molecular mechanisms governing the interactions between the mosquito midgut molecules and the malaria parasite. Here we review recent advances in the biology of malaria transmission, focusing on molecular interactions between Plasmodium and Anopheles mosquito midgut proteins. We provide an overview of parasite and mosquito proteins that are either targets for drugs currently in clinical trials or candidates of promising transmission-blocking vaccines.
Collapse
Affiliation(s)
- Yacob Keleta
- Department of Biological Sciences, Florida International University, Miami, FL, 33199, USA
| | - Julian Ramelow
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Liwang Cui
- College of Public Health, University of South Florida, Tampa, FL, 33612, USA
| | - Jun Li
- Department of Biological Sciences, Florida International University, Miami, FL, 33199, USA.
- Biomolecular Science Institute, Florida International University, Miami, FL, 33199, USA.
| |
Collapse
|
21
|
Howard GP, Bender NG, Khare P, López-Gutiérrez B, Nyasembe V, Weiss WJ, Simecka JW, Hamerly T, Mao HQ, Dinglasan RR. Immunopotentiation by Lymph-Node Targeting of a Malaria Transmission-Blocking Nanovaccine. Front Immunol 2021; 12:729086. [PMID: 34512663 PMCID: PMC8432939 DOI: 10.3389/fimmu.2021.729086] [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: 06/22/2021] [Accepted: 08/06/2021] [Indexed: 11/13/2022] Open
Abstract
A successful malaria transmission blocking vaccine (TBV) requires the induction of a high antibody titer that leads to abrogation of parasite traversal of the mosquito midgut following ingestion of an infectious bloodmeal, thereby blocking the cascade of secondary human infections. Previously, we developed an optimized construct UF6b that elicits an antigen-specific antibody response to a neutralizing epitope of Anopheline alanyl aminopeptidase N (AnAPN1), an evolutionarily conserved pan-malaria mosquito midgut-based TBV target, as well as established a size-controlled lymph node targeting biodegradable nanoparticle delivery system that leads to efficient and durable antigen-specific antibody responses using the model antigen ovalbumin. Herein, we demonstrate that co-delivery of UF6b with the adjuvant CpG oligodeoxynucleotide immunostimulatory sequence (ODN ISS) 1018 using this biodegradable nanoparticle vaccine delivery system generates an AnAPN1-specific immune response that blocks parasite transmission in a standard membrane feeding assay. Importantly, this platform allows for antigen dose-sparing, wherein lower antigen payloads elicit higher-quality antibodies, therefore less antigen-specific IgG is needed for potent transmission-reducing activity. By targeting lymph nodes directly, the resulting immunopotentiation of AnAPN1 suggests that the de facto assumption that high antibody titers are needed for a TBV to be successful needs to be re-examined. This nanovaccine formulation is stable at -20°C storage for at least 3 months, an important consideration for vaccine transport and distribution in regions with poor healthcare infrastructure. Together, these data support further development of this nanovaccine platform for malaria TBVs.
Collapse
Affiliation(s)
- Gregory P Howard
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, United States
| | - Nicole G Bender
- Emerging Pathogens Institute, Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Prachi Khare
- Emerging Pathogens Institute, Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Borja López-Gutiérrez
- Emerging Pathogens Institute, Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Vincent Nyasembe
- Emerging Pathogens Institute, Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - William J Weiss
- Department of Pharmaceutical Sciences and UNTHSC Preclinical Services, University of North Texas System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Jerry W Simecka
- Department of Pharmaceutical Sciences and UNTHSC Preclinical Services, University of North Texas System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Timothy Hamerly
- Emerging Pathogens Institute, Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Hai-Quan Mao
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, United States.,Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, United States.,Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore MD, United States
| | - Rhoel R Dinglasan
- Emerging Pathogens Institute, Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
22
|
Adelman ZN, Kojin BB. Malaria-Resistant Mosquitoes (Diptera: Culicidae); The Principle is Proven, But Will the Effectors Be Effective? JOURNAL OF MEDICAL ENTOMOLOGY 2021; 58:1997-2005. [PMID: 34018548 DOI: 10.1093/jme/tjab090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Indexed: 06/12/2023]
Abstract
Over the last few decades, a substantial number of anti-malarial effector genes have been evaluated for their ability to block parasite infection in the mosquito vector. While many of these approaches have yielded significant effects on either parasite intensity or prevalence of infection, just a few have been able to completely block transmission. Additionally, many approaches, while effective against the parasite, also disrupt or alter important aspects of mosquito physiology, leading to corresponding changes in lifespan, reproduction, and immunity. As the most promising approaches move towards field-based evaluation, questions of effector gene robustness and durability move to the forefront. In this forum piece, we critically evaluate past effector gene approaches with an eye towards developing a deeper pipeline to augment the current best candidates.
Collapse
Affiliation(s)
- Zach N Adelman
- Department of Entomology and AgriLife Research, Texas A&M University, College Station, TX, USA
| | - Bianca B Kojin
- Department of Entomology and AgriLife Research, Texas A&M University, College Station, TX, USA
| |
Collapse
|
23
|
Niu G, Cui Y, Wang X, Keleta Y, Li J. Studies of the Parasite-Midgut Interaction Reveal Plasmodium Proteins Important for Malaria Transmission to Mosquitoes. Front Cell Infect Microbiol 2021; 11:654216. [PMID: 34262880 PMCID: PMC8274421 DOI: 10.3389/fcimb.2021.654216] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 05/10/2021] [Indexed: 11/16/2022] Open
Abstract
Malaria transmission relies on parasite-mosquito midgut interaction. The interactive proteins are hypothesized to be ideal targets to block malaria transmission to mosquitoes. We chose 76 genes that contain signal peptide-coding regions and are upregulated and highly abundant at sexual stages. Forty-six of these candidate genes (60%) were cloned and expressed using the baculovirus expression system in insect cells. Six of them, e.g., PF3D7_0303900, PF3D7_0406200 (Pfs16), PF3D7_1204400 (Pfs37), PF3D7_1214800, PF3D7_1239400, and PF3D7_1472800 were discovered to interact with blood-fed mosquito midgut lysate. Previous works showed that among these interactive proteins, knockout the orthologs of Pfs37 or Pfs16 in P. berghei reduced oocysts in mosquitoes. Here we further found that anti-Pfs16 polyclonal antibody significantly inhibited P. falciparum transmission to Anopheles gambiae. Investigating these candidate proteins will improve our understanding of malaria transmission and discover new targets to break malaria transmission.
Collapse
Affiliation(s)
| | | | | | | | - Jun Li
- Department of Biological Sciences, Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
| |
Collapse
|
24
|
Singh H, Sehrawat N. Molecular characterization and in-silico analysis of AsSGU (Secreted Glycocojugate of Unknown function) in malaria vector Anopheles stephensi for transmission blocking. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
25
|
Jakhar R, Gakhar SK. Identification and Characterization of Aminopeptidase N 1 Gene of the Indian Malaria Vector Anopheles culicifacies (Diptera: Culicidae). JOURNAL OF MEDICAL ENTOMOLOGY 2021; 58:1476-1481. [PMID: 33686425 DOI: 10.1093/jme/tjab011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Indexed: 06/12/2023]
Abstract
Aminopeptidase N1 (APN) is one of the important enzymes involved in blood digestion and is up-regulated along with several other enzymes in response to bloodmeal ingestion. APN is a zinc metalloprotease that cleaves one amino acid residue at a time from the amino terminus of the protein. The APN1 gene of the Indian malaria vector Anopheles culicifacies Giles was cloned and characterized. The An. culicifacies APN1 (AcAPN1) gene has an Open Reading Frame of 3084 basepairs which encodes a putative protein of 1027 amino acids. The coding region of the gene shares 81% and 78% similarity to the APN1 genes found in An. stephensi (Diptera: Culicidae) and An. gambiae (Diptera: Culicidae), respectively. The organization of the APN1 gene was studied in available mosquito genomes and a three-dimensional structure of AcAPN1 modeled using homology structure modeling. The enzymatic active site was predicted to consist of HEYAH and GAMEN amino acid residues, and a comparison of the protein sequences among different genera revealed the conservation of zinc-binding residues. The expression pattern of AcAPN1 showed that the gene was expressed rapidly in response to the ingestion of the bloodmeal and therefore this gene may be used to exploit its promoter region as an antiparasite candidate molecule.
Collapse
Affiliation(s)
- Renu Jakhar
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak-124001, Haryana, India
| | - Surendra Kumar Gakhar
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak-124001, Haryana, India
| |
Collapse
|
26
|
McCoy KD, Weldon CT, Ansumana R, Lamin JM, Stenger DA, Ryan SJ, Bardosh K, Jacobsen KH, Dinglasan RR. Are malaria transmission-blocking vaccines acceptable to high burden communities? Results from a mixed methods study in Bo, Sierra Leone. Malar J 2021; 20:183. [PMID: 33849572 PMCID: PMC8045381 DOI: 10.1186/s12936-021-03723-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 04/04/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Malaria transmission-blocking vaccines (TBVs) could help break the cycle of malaria transmission by conferring community rather than individual protection. When introducing new intervention strategies, uptake is dependent on acceptability, not just efficacy. In this exploratory study on acceptability of TBVs in Sierra Leone, it was hypothesized that TBVs would be largely acceptable to adults and health workers in areas with relatively few ongoing malaria interventions, and that (i) knowledge of malaria and vaccines, (ii) health behaviours associated with malaria and vaccines, and (iii) attitudes towards different vaccines types could lead to greater TBV acceptability. METHODS This study used a mixed methods approach in Bo, Sierra Leone, to understand community knowledge, attitudes, and practices related to malaria and vaccination in general. This included: (i) a population-based cross-sectional survey (n=615 adults), (ii) 6 focus group discussions with parents, and (iii) 20 key informant interviews. The concept of a TBV was explained to participants before they were asked about their willingness to accept this vaccine modality as part of an integrated malaria elimination programme. RESULTS This study found that most adults would be willing to receive a TBV vaccine. Respondents noted mostly positive past experiences with adult and childhood vaccinations for other infectious diseases and high levels of engagement in other malaria prevention behaviors such as bed nets. Perceived barriers to TBV acceptance were largely focused on general community-level distribution of a vaccine, including personal fears of vaccination and possible costs. After an explanation of the TBV mechanism, nearly all focus group and interview participants believed that community members would accept the vaccine as part of an integrated malaria control approach. Both parents and health workers offered insight on how to successfully roll-out a future TBV vaccination programme. CONCLUSIONS The willingness of community members in Bo, Sierra Leone to accept a TBV as part of an integrated anti-malarial strategy suggests that the atypical mechanism of TBV action might not be an obstacle to future clinical trials. This study's findings suggests that perceived general barriers to vaccination implementation, such as perceived personal fears and vaccine cost, must be addressed in future clinical and implementation research studies.
Collapse
Affiliation(s)
- Kaci D McCoy
- CDC Southeastern Center of Excellence in Vector Borne Diseases, Gainesville, FL, USA
- Emerging Pathogens Institute, Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Caroline T Weldon
- CDC Southeastern Center of Excellence in Vector Borne Diseases, Gainesville, FL, USA
| | - Rashid Ansumana
- Mercy Hospital Research Laboratory, Bo, Sierra Leone
- School of Community Health Sciences, Njala University, Bo, Sierra Leone
| | | | - David A Stenger
- Center for Bio/Molecular Science and Engineering, Naval Research Laboratory, Washington, DC, USA
| | - Sadie J Ryan
- CDC Southeastern Center of Excellence in Vector Borne Diseases, Gainesville, FL, USA
- Emerging Pathogens Institute, Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
- Department of Geography, College of Liberal Arts & Sciences, University of Florida, Gainesville, FL, USA
| | - Kevin Bardosh
- Center for One Health Research, School of Public Health, University of Washington, Seattle, WA, USA
| | - Kathryn H Jacobsen
- Department of Global and Community Health, College of Health and Human Services, George Mason University, Fairfax, VA, USA
| | - Rhoel R Dinglasan
- CDC Southeastern Center of Excellence in Vector Borne Diseases, Gainesville, FL, USA.
- Emerging Pathogens Institute, Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
27
|
Immunofocusing humoral immunity potentiates the functional efficacy of the AnAPN1 malaria transmission-blocking vaccine antigen. NPJ Vaccines 2021; 6:49. [PMID: 33824336 PMCID: PMC8024329 DOI: 10.1038/s41541-021-00309-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 02/24/2021] [Indexed: 11/08/2022] Open
Abstract
Malaria transmission-blocking vaccines (TBVs) prevent the completion of the developmental lifecycle of malarial parasites within the mosquito vector, effectively blocking subsequent infections. The mosquito midgut protein Anopheline alanyl aminopeptidase N (AnAPN1) is the leading, mosquito-based TBV antigen. Structure-function studies identified two Class II epitopes that can induce potent transmission-blocking (T-B) antibodies, informing the design of the next-generation AnAPN1. Here, we functionally screened new immunogens and down-selected to the UF6b construct that has two glycine-linked copies of the T-B epitopes. We then established a process for manufacturing UF6b and evaluated in outbred female CD1 mice the immunogenicity of the preclinical product with the human-safe adjuvant Glucopyranosyl Lipid Adjuvant in a liposomal formulation with saponin QS21 (GLA-LSQ). UF6b:GLA-LSQ effectively immunofocused the humoral response to one of the key T-B epitopes resulting in potent T-B activity, underscoring UF6b as a prime TBV candidate to aid in malaria elimination and eradication efforts.
Collapse
|
28
|
Couto J, Seixas G, Stutzer C, Olivier NA, Maritz-Olivier C, Antunes S, Domingos A. Probing the Rhipicephalusbursa Sialomes in Potential Anti-Tick Vaccine Candidates: A Reverse Vaccinology Approach. Biomedicines 2021; 9:363. [PMID: 33807386 PMCID: PMC8067113 DOI: 10.3390/biomedicines9040363] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/26/2021] [Accepted: 03/28/2021] [Indexed: 11/16/2022] Open
Abstract
In the wake of the 'omics' explosion of data, reverse vaccinology approaches are being applied more readily as an alternative for the discovery of candidates for next generation diagnostics and vaccines. Promising protective antigens for the control of ticks and tick-borne diseases can be discovered by mining available omics data for immunogenic epitopes. The present study aims to explore the previously obtained Rhipicephalus bursa sialotranscriptome during both feeding and Babesia infection, to select antigenic targets that are either membrane-associated or a secreted protein, as well as unique to the ectoparasite and not present in the mammalian host. Further, they should be capable of stimulating T and B cells for a potential robust immune response, and be non-allergenic or toxic to the host. From the R. bursa transcriptome, 5706 and 3025 proteins were identified as belonging to the surfaceome and secretome, respectively. Following a reverse genetics immunoinformatics pipeline, nine preferred candidates, consisting of one transmembrane-related and eight secreted proteins, were identified. These candidates showed a higher predicted antigenicity than the Bm86 antigen, with no homology to mammalian hosts and exposed regions. Only four were functionally annotated and selected for further in silico analysis, which examined their protein structure, surface accessibility, flexibility, hydrophobicity, and putative linear B and T-cell epitopes. Regions with overlapping coincident epitopes groups (CEGs) were evaluated to select peptides that were further analyzed for their physicochemical characteristics, potential allergenicity, toxicity, solubility, and potential propensity for crystallization. Following these procedures, a set of three peptides from the three R. bursa proteins were selected. In silico results indicate that the designed epitopes could stimulate a protective and long-lasting immune response against those tick proteins, reflecting its potential as anti-tick vaccines. The immunogenicity of these peptides was evaluated in a pilot immunization study followed by tick feeding to evaluate its impact on tick behavior and pathogen transmission. Combining in silico methods with in vivo immunogenicity evaluation enabled the screening of vaccine candidates prior to expensive infestation studies on the definitive ovine host animals.
Collapse
Affiliation(s)
- Joana Couto
- Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira, 100, 1349-008 Lisboa, Portugal; (G.S.); (A.D.)
- Global Health and Tropical Medicine, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa (GHTM-IHMT-UNL), Rua da Junqueira, 100, 1349-008 Lisboa, Portugal
| | - Gonçalo Seixas
- Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira, 100, 1349-008 Lisboa, Portugal; (G.S.); (A.D.)
- Global Health and Tropical Medicine, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa (GHTM-IHMT-UNL), Rua da Junqueira, 100, 1349-008 Lisboa, Portugal
| | - Christian Stutzer
- Division of Genetics, Department of Biochemistry, Genetics and Microbiology, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria 0002, South Africa; (C.S.); (C.M.-O.)
| | - Nicholas A. Olivier
- Department of Plant and Soil Sciences, University of Pretoria, Pretoria 0002, South Africa;
- Forestry and Agricultural Biotechnology Institute (FABI), University of Pretoria, Pretoria 0002, South Africa
| | - Christine Maritz-Olivier
- Division of Genetics, Department of Biochemistry, Genetics and Microbiology, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria 0002, South Africa; (C.S.); (C.M.-O.)
| | - Sandra Antunes
- Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira, 100, 1349-008 Lisboa, Portugal; (G.S.); (A.D.)
- Global Health and Tropical Medicine, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa (GHTM-IHMT-UNL), Rua da Junqueira, 100, 1349-008 Lisboa, Portugal
| | - Ana Domingos
- Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira, 100, 1349-008 Lisboa, Portugal; (G.S.); (A.D.)
- Global Health and Tropical Medicine, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa (GHTM-IHMT-UNL), Rua da Junqueira, 100, 1349-008 Lisboa, Portugal
| |
Collapse
|
29
|
Chawla J, Oberstaller J, Adams JH. Targeting Gametocytes of the Malaria Parasite Plasmodium falciparum in a Functional Genomics Era: Next Steps. Pathogens 2021; 10:346. [PMID: 33809464 PMCID: PMC7999360 DOI: 10.3390/pathogens10030346] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/25/2021] [Accepted: 03/03/2021] [Indexed: 02/04/2023] Open
Abstract
Mosquito transmission of the deadly malaria parasite Plasmodium falciparum is mediated by mature sexual forms (gametocytes). Circulating in the vertebrate host, relatively few intraerythrocytic gametocytes are picked up during a bloodmeal to continue sexual development in the mosquito vector. Human-to-vector transmission thus represents an infection bottleneck in the parasite's life cycle for therapeutic interventions to prevent malaria. Even though recent progress has been made in the identification of genetic factors linked to gametocytogenesis, a plethora of genes essential for sexual-stage development are yet to be unraveled. In this review, we revisit P. falciparum transmission biology by discussing targetable features of gametocytes and provide a perspective on a forward-genetic approach for identification of novel transmission-blocking candidates in the future.
Collapse
Affiliation(s)
- Jyotsna Chawla
- Molecular Medicine, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, MDC 7, Tampa, FL 33612, USA;
| | - Jenna Oberstaller
- Center for Global Health and Infectious Diseases Research and USF Genomics Program, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Suite 404, Tampa, FL 33612, USA;
| | - John H. Adams
- Center for Global Health and Infectious Diseases Research and USF Genomics Program, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Suite 404, Tampa, FL 33612, USA;
| |
Collapse
|
30
|
De SL, Ntumngia FB, Nicholas J, Adams JH. Progress towards the development of a P. vivax vaccine. Expert Rev Vaccines 2021; 20:97-112. [PMID: 33481638 PMCID: PMC7994195 DOI: 10.1080/14760584.2021.1880898] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 01/21/2021] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Plasmodium vivax causes significant public health problems in endemic regions. A vaccine to prevent disease is critical, considering the rapid spread of drug-resistant parasite strains, and the development of hypnozoites in the liver with potential for relapse. A minimally effective vaccine should prevent disease and transmission while an ideal vaccine provides sterile immunity. AREAS COVERED Despite decades of research, the complex life cycle, technical challenges and a lack of funding have hampered progress of P. vivax vaccine development. Here, we review the progress of potential P. vivax vaccine candidates from different stages of the parasite life cycle. We also highlight the challenges and important strategies for rational vaccine design. These factors can significantly increase immune effector mechanisms and improve the protective efficacy of these candidates in clinical trials to generate sustained protection over longer periods of time. EXPERT OPINION A vaccine that presents functionally-conserved epitopes from multiple antigens from various stages of the parasite life cycle is key to induce broadly neutralizing strain-transcending protective immunity to effectively disrupt parasite development and transmission.
Collapse
Affiliation(s)
- Sai Lata De
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa – 33612, FL
| | - Francis B. Ntumngia
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa – 33612, FL
| | - Justin Nicholas
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa – 33612, FL
| | - John H. Adams
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa – 33612, FL
| |
Collapse
|
31
|
Patra KP, Kaur H, Kolli SK, Wozniak JM, Prieto JH, Yates JR, Gonzalez DJ, Janse CJ, Vinetz JM. A Hetero-Multimeric Chitinase-Containing Plasmodium falciparum and Plasmodium gallinaceum Ookinete-Secreted Protein Complex Involved in Mosquito Midgut Invasion. Front Cell Infect Microbiol 2021; 10:615343. [PMID: 33489941 PMCID: PMC7821095 DOI: 10.3389/fcimb.2020.615343] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 11/30/2020] [Indexed: 12/14/2022] Open
Abstract
Malaria parasites are transmitted by Anopheles mosquitoes. During its life cycle in the mosquito vector the Plasmodium ookinete escapes the proteolytic milieu of the post-blood meal midgut by traversing the midgut wall. This process requires penetration of the chitin-containing peritrophic matrix lining the midgut epithelium, which depends in part on ookinete-secreted chitinases. Plasmodium falciparum ookinetes have one chitinase (PfCHT1), whereas ookinetes of the avian-infecting parasite, P. gallinaceum, have two, a long and a short form, PgCHT1 and PgCHT2, respectively. Published data indicates that PgCHT2 forms a high molecular weight (HMW) reduction-sensitive complex; and one binding partner is the ookinete-produced von Willebrand A-domain-containing protein, WARP. Size exclusion chromatography data reported here show that P. gallinaceum PgCHT2 and its ortholog, P. falciparum PfCHT1 are covalently-linked components of a HMW chitinase-containing complex (> 1,300 kDa). Mass spectrometry of ookinete-secreted proteins isolated using a new chitin bead pull-down method identified chitinase-associated proteins in P. falciparum and P. gallinaceum ookinete-conditioned culture media. Mass spectrometry of this complex showed the presence of several micronemal proteins including von Willebrand factor A domain-related protein (WARP), ookinete surface enolase, and secreted ookinete adhesive protein (SOAP). To test the hypothesis that ookinete-produced PfCHT1 can form a high molecular homo-multimer or, alternatively, interacts with P. berghei ookinete-produced proteins to produce an HMW hetero-multimer, we created chimeric P. berghei parasites expressing PfCHT1 to replace PbCHT1, enabling the production of large numbers of PfCHT1-expressing ookinetes. We show that chimeric P. berghei ookinetes express monomeric PfCHT1, but a HMW complex containing PfCHT1 is not present. A better understanding of the chitinase-containing HMW complex may enhance development of next-generation vaccines or drugs that target malaria transmission stages.
Collapse
Affiliation(s)
- Kailash P Patra
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Hargobinder Kaur
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Surendra Kumar Kolli
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Jacob M Wozniak
- Department of Pharmacology and the Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, United States
| | - Judith Helena Prieto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States.,Department of Chemistry, Western Connecticut State University, Danbury, CT, United States
| | - John R Yates
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - David J Gonzalez
- Department of Pharmacology and the Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, United States
| | - Chris J Janse
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Joseph M Vinetz
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
32
|
Perner J, Helm D, Haberkant P, Hatalova T, Kropackova S, Ribeiro JM, Kopacek P. The Central Role of Salivary Metalloproteases in Host Acquired Resistance to Tick Feeding. Front Cell Infect Microbiol 2020; 10:563349. [PMID: 33312963 PMCID: PMC7708348 DOI: 10.3389/fcimb.2020.563349] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 10/20/2020] [Indexed: 01/07/2023] Open
Abstract
During feeding on vertebrate hosts, ticks secrete saliva composed of a rich cocktail of bioactive molecules modulating host immune responses. Although most of the proteinaceous fraction of tick saliva is of little immunogenicity, repeated feeding of ticks on mammalian hosts may lead to impairment of tick feeding, preventing full engorgement. Here, we challenged rabbits with repeated feeding of both Ixodes ricinus nymphs and adults and observed the formation of specific antibodies against several tick salivary proteins. Repeated feeding of both I. ricinus stages led to a gradual decrease in engorged weights. To identify the salivary antigens, isolated immunoglobulins from repeatedly infested rabbits were utilized for a protein pull-down from the saliva of pilocarpine-treated ticks. Eluted antigens were first identified by peptide mass fingerprinting with the aid of available I. ricinus salivary gland transcriptomes originating from early phases of tick feeding. To increase the authenticity of immunogens identified, we also performed, for the first time, de novo assembly of the sialome from I. ricinus females fed for six days, a timepoint used for pilocarpine-salivation. The most dominant I. ricinus salivary immunogens identified in our study were zinc-dependent metalloproteases of three different families. To corroborate the role of metalloproteases at the tick/host interface, we fed ticks micro-injected with a zinc metalloprotease inhibitor, phosphoramidon, on a rabbit. These ticks clearly failed to initiate feeding and to engorge. However, neither feeding to ticks immune blood of repeatedly infested rabbits, nor phosphoramidon injection into ticks, prevented their engorgement when fed in vitro on an artificial membrane system. These data show that Zn metalloproteases play a decisive role in the success of tick feeding, mediated by complex molecular interactions between the host immune, inflammatory, and hemostatic processes, which are absent in in vitro feeding. This basic concept warrants further investigation and reconsideration of the current strategies towards the development of an effective “anti-tick” vaccine.
Collapse
Affiliation(s)
- Jan Perner
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czechia
| | - Dominic Helm
- Proteomics Core Facility, The European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Per Haberkant
- Proteomics Core Facility, The European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Tereza Hatalova
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czechia
| | - Sara Kropackova
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czechia
| | - Jose M Ribeiro
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Petr Kopacek
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czechia
| |
Collapse
|
33
|
Analysis of blood-induced Anopheles gambiae midgut proteins and sexual stage Plasmodium falciparum interaction reveals mosquito genes important for malaria transmission. Sci Rep 2020; 10:14316. [PMID: 32868841 PMCID: PMC7459308 DOI: 10.1038/s41598-020-71186-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 08/11/2020] [Indexed: 12/12/2022] Open
Abstract
Plasmodium invasion of mosquito midguts is a mandatory step for malaria transmission. The roles of mosquito midgut proteins and parasite interaction during malaria transmission are not clear. This study aims to identify mosquito midgut proteins that interact with and affect P. falciparum invasion. Based on gene expression profiles and protein sequences, 76 mosquito secretory proteins that are highly expressed in midguts and up-regulated by blood meals were chosen for analysis. About 61 candidate genes were successfully cloned from Anopheles gambiae and expressed in insect cells. ELISA analysis showed that 25 of the insect cell-expressed recombinant mosquito proteins interacted with the P. falciparum-infected cell lysates. Indirect immunofluorescence assays confirmed 17 of them interacted with sexual stage parasites significantly stronger than asexual stage parasites. Knockdown assays found that seven candidate genes significantly changed mosquitoes' susceptibility to P. falciparum. Four of them (AGAP006268, AGAP002848, AGAP006972, and AGAP002851) played a protective function against parasite invasion, and the other three (AGAP008138, FREP1, and HPX15) facilitated P. falciparum transmission to mosquitoes. Notably, AGAP008138 is a unique gene that only exists in Anopheline mosquitoes. These gene products are ideal targets to block malaria transmission.
Collapse
|
34
|
Qiu Y, Zhao Y, Liu F, Ye B, Zhao Z, Thongpoon S, Roobsoong W, Sattabongkot J, Cui L, Fan Q, Cao Y. Evaluation of Plasmodium vivax HAP2 as a transmission-blocking vaccine candidate. Vaccine 2020; 38:2841-2848. [PMID: 32093983 DOI: 10.1016/j.vaccine.2020.02.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 01/31/2020] [Accepted: 02/05/2020] [Indexed: 12/23/2022]
Abstract
Transmission-blocking vaccine (TBV) is a promising strategy to interfere with the transmission of malaria. To date, only limited TBV candidate antigens have been identified for Plasmodium vivax. HAP2 is a gamete membrane fusion protein, with homology to the class II viral fusion proteins. Herein we reported the characterization of the PvHAP2 for its potential as a TBV candidate for P. vivax. The HAP2/GCS1 domain of PvHAP2 was expressed in the baculovirus expression system and the recombinant protein was used to raise antibodies in rabbits. Indirect immunofluorescence assays showed that anti-PvHAP2 antibodies reacted only with the male gametocytes on blood smears. Direct membrane feeding assays were conducted using four field P. vivax isolates in Anopheles dirus. At a mean infection intensity of 72.4, 70.7, 51.3, and 15.6 oocysts/midgut with the control antibodies, anti-PvHAP2 antibodies significantly reduced the midgut oocyst intensity by 40.3, 44.4, 61.9, and 89.7%. Whereas the anti-PvHAP2 antibodies were not effective in reducing the infection prevalence at higher parasite exposure (51.3-72.4 oocysts/midgut in the control group), the anti-PvHAP2 antibodies reduced infection prevalence by 50% at a low challenge (15.6 oocysts/midgut). Multiple sequence alignment showed 100% identity among these Thai P. vivax isolates, suggesting that polymorphism may not be an impediment for the utilization of PvHAP2 as a TBV antigen. In conclusion, our results suggest that PvHAP2 could serve as a TBV candidate for P. vivax, and further optimization and evaluation are warranted.
Collapse
Affiliation(s)
- Yue Qiu
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning 110122, China
| | - Yan Zhao
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning 110122, China
| | - Fei Liu
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning 110122, China
| | - Bo Ye
- Dalian Institute of Biotechnology, Dalian, Liaoning, China
| | - Zhenjun Zhao
- Dalian Institute of Biotechnology, Dalian, Liaoning, China
| | - Sataporn Thongpoon
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Thailand
| | - Wanlapa Roobsoong
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Thailand
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Thailand
| | - Liwang Cui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Suite 304, Tampa, FL 33612, USA
| | - Qi Fan
- Dalian Institute of Biotechnology, Dalian, Liaoning, China
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
35
|
Dadgar Pakdel J, Zakeri S, Raz A, Dinparast Djadid N. Identification, molecular characterization and expression of aminopeptidase N-1 (APN-1) from Anopheles stephensi in SF9 cell line as a candidate molecule for developing a vaccine that interrupt malaria transmission. Malar J 2020; 19:79. [PMID: 32075635 PMCID: PMC7029531 DOI: 10.1186/s12936-020-03154-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 02/09/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND According to the World Health Organization reports, billions of people around the world are at risk for malaria disease and it is important to consider the preventive strategies for protecting the people that are living in high risk areas. One of the main reasons of disease survival is diversity of vectors and parasites in different malaria regions that have their specific features, behaviour and biology. Therefore, specific regional strategies are necessary for successful control of malaria. One of the tools that needs to be developed for elimination and prevention of reintroduction of malaria is a vaccine that interrupt malaria transmission (VIMTs). VIMT is a broad concept that should be adjusted to the biological characteristics of the disease in each region. One type of VIMT is a vector-based vaccine that affects the sexual stage of Plasmodium life cycle. According to recent studies, the aminopeptidase N-1 of Anopheles gambiae (AgAPN-1) is as a potent vector-based VIMT with considerable inhibition activity against the sexual stage of Plasmodium parasite. METHODS Systems for rapid amplification of cDNA ends (3'-RACE) and genome walking methods were used for sequence determination of apn-1 gene from Anopheles stephensi and distinct bioinformatics software were used for structural analysis. AsAPN-1 was expressed in Spodoptera frugiperda (Sf9) insect cell line using the baculovirus expression system. Recombinant AsAPN-1 was purified under the hybrid condition and its biological activity was assayed. RESULTS Asapn-1 gene and its coded protein from An. stephensi were characterized for the first time in this study. Subsequently, the structural features and immunological properties of its coded protein were evaluated by in silico approaches. Enzymatic activity of the recombinant AsAPN-1, which was expressed in Sf9 insect cell line, was equal to 6 unit/μl. CONCLUSIONS Results of this study revealed that AsAPN-1 is very similar to its counterpart in An. gambiae. In silico evaluation and fundamental data which are necessary for its evaluation as a VIMT-based vaccine in the next steps were acquired in this study and those could be useful for research groups that study on malaria vaccine for countries that An. stephensi is the main malaria vector there.
Collapse
Affiliation(s)
- Javad Dadgar Pakdel
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran (PII), Pasteur Avenue, P.O. Box 1316943551, Tehran, Iran.,Trauma Research Center, Sina Hospital, Tehran University of Medical Sciences, Hassan Abad Square, Imam Khomeini Avenue, PO BOX: 1136746911, Tehran, Iran
| | - Sedigheh Zakeri
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran (PII), Pasteur Avenue, P.O. Box 1316943551, Tehran, Iran
| | - Abbasali Raz
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran (PII), Pasteur Avenue, P.O. Box 1316943551, Tehran, Iran.
| | - Navid Dinparast Djadid
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran (PII), Pasteur Avenue, P.O. Box 1316943551, Tehran, Iran.
| |
Collapse
|
36
|
Narula AK, Azad CS, Nainwal LM. New dimensions in the field of antimalarial research against malaria resurgence. Eur J Med Chem 2019; 181:111353. [DOI: 10.1016/j.ejmech.2019.05.043] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 04/16/2019] [Accepted: 05/15/2019] [Indexed: 12/20/2022]
|
37
|
Miura K, Tachibana M, Takashima E, Morita M, Kanoi BN, Nagaoka H, Baba M, Torii M, Ishino T, Tsuboi T. Malaria transmission-blocking vaccines: wheat germ cell-free technology can accelerate vaccine development. Expert Rev Vaccines 2019; 18:1017-1027. [PMID: 31566026 PMCID: PMC11000147 DOI: 10.1080/14760584.2019.1674145] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 09/25/2019] [Indexed: 12/18/2022]
Abstract
Introduction: Highly effective malaria vaccines are essential component toward malaria elimination. Although the leading malaria vaccine, RTS,S/AS01, with modest efficacy is being evaluated in a pilot feasibility trial, development of a malaria transmission-blocking vaccine (TBV) could make a major contribution toward malaria elimination. Only a few TBV antigens have reached pre-clinical or clinical development but with several challenges including difficulties in the expression of malaria recombinant proteins and low immunogenicity in humans. Therefore, novel approaches to accelerate TBV research to preclinical development are critical to generate an efficacious TBV.Areas covered: PubMed was searched to review the progress and future prospects of malaria TBV research and development. We also reviewed registered trials at ClinicalTrials.gov as well as post-genome TBV candidate discovery research including our efforts.Expert opinion: Wheat germ cell-free protein synthesis technology can accelerate TBV development by overcoming some current challenges of TBV research.
Collapse
Affiliation(s)
- Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Mayumi Tachibana
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Japan
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Masayuki Morita
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Bernard N Kanoi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Hikaru Nagaoka
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Minami Baba
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Japan
| | - Motomi Torii
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Japan
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Tomoko Ishino
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Japan
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| |
Collapse
|
38
|
Abstract
In the progression of the life cycle of Plasmodium falciparum, a small proportion of asexual parasites differentiate into male or female sexual forms called gametocytes. Just like their asexual counterparts, gametocytes are contained within the infected host's erythrocytes (RBCs). However, unlike their asexual partners, they do not exit the RBC until they are taken up in a blood meal by a mosquito. In the mosquito midgut, they are stimulated to emerge from the RBC, undergo fertilization, and ultimately produce tens of thousands of sporozoites that are infectious to humans. This transmission cycle can be blocked by antibodies targeting proteins exposed on the parasite surface in the mosquito midgut, a process that has led to the development of candidate transmission-blocking vaccines (TBV), including some that are in clinical trials. Here we review the leading TBV antigens and highlight the ongoing search for additional gametocyte/gamete surface antigens, as well as antigens on the surfaces of gametocyte-infected erythrocytes, which can potentially become a new group of TBV candidates.
Collapse
|
39
|
Kengne-Ouafo JA, Sutherland CJ, Binka FN, Awandare GA, Urban BC, Dinko B. Immune Responses to the Sexual Stages of Plasmodium falciparum Parasites. Front Immunol 2019; 10:136. [PMID: 30804940 PMCID: PMC6378314 DOI: 10.3389/fimmu.2019.00136] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 01/16/2019] [Indexed: 11/13/2022] Open
Abstract
Malaria infections remain a serious global health problem in the world, particularly among children and pregnant women in Sub-Saharan Africa. Moreover, malaria control and elimination is hampered by rapid development of resistance by the parasite and the vector to commonly used antimalarial drugs and insecticides, respectively. Therefore, vaccine-based strategies are sorely needed, including those designed to interrupt disease transmission. However, a prerequisite for such a vaccine strategy is the understanding of both the human and vector immune responses to parasite developmental stages involved in parasite transmission in both man and mosquito. Here, we review the naturally acquired humoral and cellular responses to sexual stages of the parasite while in the human host and the Anopheles vector. In addition, updates on current anti-gametocyte, anti-gamete, and anti-mosquito transmission blocking vaccines are given. We conclude with our views on some important future directions of research into P. falciparum sexual stage immunity relevant to the search for the most appropriate transmission-blocking vaccine.
Collapse
Affiliation(s)
- Jonas A Kengne-Ouafo
- West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Accra, Ghana
| | - Colin J Sutherland
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Fred N Binka
- Department of Epidemiology and Biostatistics, School of Public Health, University of Health and Allied Sciences, Ho, Ghana
| | - Gordon A Awandare
- West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Accra, Ghana
| | - Britta C Urban
- Faculty of Biological Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Bismarck Dinko
- Department of Biomedical Sciences, School of Basic and Biomedical Sciences, University of Health and Allied Sciences, Ho, Ghana
| |
Collapse
|
40
|
Wilson KL, Flanagan KL, Prakash MD, Plebanski M. Malaria vaccines in the eradication era: current status and future perspectives. Expert Rev Vaccines 2019; 18:133-151. [PMID: 30601095 DOI: 10.1080/14760584.2019.1561289] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The challenge to eradicate malaria is an enormous task that will not be achieved by current control measures, thus an efficacious and long-lasting malaria vaccine is required. The licensing of RTS, S/AS01 is a step forward in providing some protection, but a malaria vaccine that protects across multiple transmission seasons is still needed. To achieve this, inducing beneficial immune responses while minimising deleterious non-targeted effects will be essential. AREAS COVERED This article discusses the current challenges and advances in malaria vaccine development and reviews recent human clinical trials for each stage of infection. Pubmed and ScienceDirect were searched, focusing on cell mediated immunity and how T cell subsets might be targeted in future vaccines using novel adjuvants and emerging vaccine technologies. EXPERT COMMENTARY Despite decades of research there is no highly effective licensed malaria vaccine. However, there is cause for optimism as new adjuvants and vaccine systems emerge, and our understanding of correlates of protection increases, especially regarding cellular immunity. The new field of heterologous (non-specific) effects of vaccines also highlights the broader consequences of immunization. Importantly, the WHO led Malaria Vaccine Technology Roadmap illustrates that there is a political will among the global health community to make it happen.
Collapse
Affiliation(s)
- K L Wilson
- a Department of Immunology and Pathology, Faculty of Medicine, Nursing and Health Sciences , Monash University , Melbourne , Australia.,b School of Health and Biomedical Sciences , RMIT University , Bundoora , Australia
| | - K L Flanagan
- a Department of Immunology and Pathology, Faculty of Medicine, Nursing and Health Sciences , Monash University , Melbourne , Australia.,b School of Health and Biomedical Sciences , RMIT University , Bundoora , Australia.,c School of Medicine, Faculty of Health Sciences , University of Tasmania , Launceston , Australia
| | - M D Prakash
- b School of Health and Biomedical Sciences , RMIT University , Bundoora , Australia
| | - M Plebanski
- b School of Health and Biomedical Sciences , RMIT University , Bundoora , Australia
| |
Collapse
|
41
|
Khamsehnejad MI, Djadid ND, Raz A. Identification, Molecular Characterization, and In Silico Structural Analysis of Carboxypeptidase B2 of Anopheles stephensi. JOURNAL OF MEDICAL ENTOMOLOGY 2019; 56:72-85. [PMID: 30124910 DOI: 10.1093/jme/tjy127] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Indexed: 06/08/2023]
Abstract
Malaria is a vector-borne infectious disease that is considered a priority of the World Health Organization due to its enormous impacts on global health. Plasmodium spp. (Haemosporida: Plasmodiidae), Anopheles spp. (Diptera: Culicidae), and a suitable host are the key elements for malaria transmission. To disrupt the parasitic life cycle of malaria or prevent its transmission, these three key elements should be targeted by effective control strategies. Development of vaccines that interrupt malaria transmission is one of the solutions that has been recommended to the countries that aim to eliminate malaria. With respect to the important role of Anopheles stephensi in malaria transmission and involvement of Anopheles carboxypeptidase B1 in sexual parasite development, we characterized the second member of cpb gene family (cpbAs2) of An. Stephensi to provide some basic information and evaluate significance of cpbAs2's role in complementing sexual plasmodium development role of cpbAs1. The cpbAs2 mRNA sequence was characterized by 3' and 5' RACE and the structural features of its coded protein were studied by in silico modeling. The coding sequence and gene structure of cpbAs2 were determined empirically and compared with the in silico predictions from the An. stephensi genome sequencing project. Furthermore, homology modeling revealed that its structure is very similar to the structurally important domains of procarboxypeptidase B2 in humans. This study provides basic molecular and structural information about another member of the cpb gene family of An. stephensi. The reported results are informative and necessary for evaluation of the role of this gene in sexual parasite development by future studies.
Collapse
Affiliation(s)
- Mahdokht Ilbeigi Khamsehnejad
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran (PII), Pasteur Avenue, Tehran, Iran
| | - Navid Dinparast Djadid
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran (PII), Pasteur Avenue, Tehran, Iran
| | - Abbasali Raz
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran (PII), Pasteur Avenue, Tehran, Iran
| |
Collapse
|
42
|
Hugo RLE, Birrell GW. Proteomics of Anopheles Vectors of Malaria. Trends Parasitol 2018; 34:961-981. [DOI: 10.1016/j.pt.2018.08.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/08/2018] [Accepted: 08/10/2018] [Indexed: 12/12/2022]
|
43
|
Structural delineation of potent transmission-blocking epitope I on malaria antigen Pfs48/45. Nat Commun 2018; 9:4458. [PMID: 30367064 PMCID: PMC6203815 DOI: 10.1038/s41467-018-06742-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 09/17/2018] [Indexed: 12/28/2022] Open
Abstract
Interventions that can block the transmission of malaria-causing Plasmodium falciparum (Pf) between the human host and Anopheles vector have the potential to reduce the incidence of malaria. Pfs48/45 is a gametocyte surface protein critical for parasite development and transmission, and its targeting by monoclonal antibody (mAb) 85RF45.1 leads to the potent reduction of parasite transmission. Here, we reveal how the Pfs48/45 6C domain adopts a (SAG1)-related-sequence (SRS) fold. We structurally delineate potent epitope I and show how mAb 85RF45.1 recognizes an electronegative surface with nanomolar affinity. Analysis of Pfs48/45 sequences reveals that polymorphisms are rare for residues involved at the binding interface. Humanization of rat-derived mAb 85RF45.1 conserved the mode of recognition and activity of the parental antibody, while also improving its thermostability. Our work has implications for the development of transmission-blocking interventions, both through improving vaccine designs and the testing of passive delivery of mAbs in humans. Malaria protein Pfs48/45 is a promising transmission-blocking antigen targeted by antibodies. Here, the authors determine the structure of its transmission-blocking epitope I, and generate a humanized monoclonal antibody that binds Pfs48/45 with high affinity.
Collapse
|
44
|
Characterization of Plasmodium berghei Pbg37 as Both a Pre- and Postfertilization Antigen with Transmission-Blocking Potential. Infect Immun 2018; 86:IAI.00785-17. [PMID: 29866905 DOI: 10.1128/iai.00785-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 05/26/2018] [Indexed: 01/29/2023] Open
Abstract
Transmission-blocking vaccines (TBVs) interrupting malaria transmission are an integrated tool for malaria eradication. We characterized a sexual-stage-specific gene (PBANKA_060330) from Plasmodium berghei and studied its potential for use as a TBV. This gene, referred to as pbg37, encodes a protein of 37 kDa with a signal peptide and multiple transmembrane domains and is preferentially expressed in gametocytes. A recombinant Pbg37 (rPbg37) protein targeting the N-terminal 63 amino acids (amino acids 26 to 88) expressed in bacteria elicited strong antibody responses in mice. Western blotting demonstrated Pbg37 expression in gametocytes, zygotes, and, to a lesser extent, ookinetes and its predominant association with the membranes of gametocytes. Indirect immunofluorescence assay showed an abundant surface localization of Pbg37 on gametes and zygotes but reduced amounts on retorts and ookinetes. Knockout of pbg37 (Δpbg37) led to a considerable reduction in gametocytemia, which translated into a ~92.1% decrease in the oocyst number in mosquitoes. Deletion of pbg37 had a more substantial influence on the development and maturation of microgametocytes. As a result, the Δpbg37 lines exhibited a higher female/male gametocyte ratio, fewer mature male gametocytes, and defects in the exflagellation of mature microgametocytes. To test the transmission-blocking potential of Pbg37, an in vitro ookinete assay showed that the major inhibitory effects of anti-Pbg37 antiserum were on the exflagellation and fertilization processes. Direct feeding of mosquitoes on mice immunized with rPbg37 or a control protein showed that rPbg37-immunized and P. berghei-infected mice had a significant reduction (49.1%) in oocyst density compared to the controls. The conservation of this gene in Plasmodium warrants further investigations in human malaria parasites.
Collapse
|
45
|
Stutzer C, Richards SA, Ferreira M, Baron S, Maritz-Olivier C. Metazoan Parasite Vaccines: Present Status and Future Prospects. Front Cell Infect Microbiol 2018; 8:67. [PMID: 29594064 PMCID: PMC5859119 DOI: 10.3389/fcimb.2018.00067] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 02/26/2018] [Indexed: 12/21/2022] Open
Abstract
Eukaryotic parasites and pathogens continue to cause some of the most detrimental and difficult to treat diseases (or disease states) in both humans and animals, while also continuously expanding into non-endemic countries. Combined with the ever growing number of reports on drug-resistance and the lack of effective treatment programs for many metazoan diseases, the impact that these organisms will have on quality of life remain a global challenge. Vaccination as an effective prophylactic treatment has been demonstrated for well over 200 years for bacterial and viral diseases. From the earliest variolation procedures to the cutting edge technologies employed today, many protective preparations have been successfully developed for use in both medical and veterinary applications. In spite of the successes of these applications in the discovery of subunit vaccines against prokaryotic pathogens, not many targets have been successfully developed into vaccines directed against metazoan parasites. With the current increase in -omics technologies and metadata for eukaryotic parasites, target discovery for vaccine development can be expedited. However, a good understanding of the host/vector/pathogen interface is needed to understand the underlying biological, biochemical and immunological components that will confer a protective response in the host animal. Therefore, systems biology is rapidly coming of age in the pursuit of effective parasite vaccines. Despite the difficulties, a number of approaches have been developed and applied to parasitic helminths and arthropods. This review will focus on key aspects of vaccine development that require attention in the battle against these metazoan parasites, as well as successes in the field of vaccine development for helminthiases and ectoparasites. Lastly, we propose future direction of applying successes in pursuit of next generation vaccines.
Collapse
Affiliation(s)
- Christian Stutzer
- Tick Vaccine Group, Department of Genetics, University of Pretoria, Pretoria, South Africa
| | | | | | | | | |
Collapse
|
46
|
Dong Y, Simões ML, Marois E, Dimopoulos G. CRISPR/Cas9 -mediated gene knockout of Anopheles gambiae FREP1 suppresses malaria parasite infection. PLoS Pathog 2018. [PMID: 29518156 PMCID: PMC5843335 DOI: 10.1371/journal.ppat.1006898] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Plasmodium relies on numerous agonists during its journey through the mosquito vector, and these agonists represent potent targets for transmission-blocking by either inhibiting or interfering with them pre- or post-transcriptionally. The recently developed CRISPR/Cas9-based genome editing tools for Anopheles mosquitoes provide new and promising opportunities for the study of agonist function and for developing malaria control strategies through gene deletion to achieve complete agonist inactivation. Here we have established a modified CRISPR/Cas9 gene editing procedure for the malaria vector Anopheles gambiae, and studied the effect of inactivating the fibrinogen-related protein 1 (FREP1) gene on the mosquito’s susceptibility to Plasmodium and on mosquito fitness. FREP1 knockout mutants developed into adult mosquitoes that showed profound suppression of infection with both human and rodent malaria parasites at the oocyst and sporozoite stages. FREP1 inactivation, however, resulted in fitness costs including a significantly lower blood-feeding propensity, fecundity and egg hatching rate, a retarded pupation time, and reduced longevity after a blood meal. The causative agent of malaria, Plasmodium, has to complete a complex infection cycle in the Anopheles gambiae mosquito vector in order to reach the salivary gland from where it can be transmitted to a human host. The parasite’s development in the mosquito relies on numerous host factors (agonists), and their inhibition or inactivation can thereby result in suppression of infection and consequently malaria transmission. The recently developed CRISPR/Cas9-based genome editing tools for Anopheles mosquitoes provide new and promising opportunities to delete (inactivate) Plasmodium agonists to better understand their function and for blocking malaria transmission. Here we have established a modified CRISPR/Cas9 genome editing technique for malaria vector A. gambiae mosquitoes. Through this approach we have inactivated the fibrinogen-related protein 1 (FREP1) gene, via CRISPR/Cas9 genome editing, and the impact of this manipulation on the mosquito’s susceptibility to Plasmodium and on mosquito fitness. FREP1 knockout mutants showed a profound suppression of infection with both human and rodent malaria parasites, while it also resulted in fitness costs: a significantly lower blood-feeding propensity, fecundity and egg hatching rate, and a retarded larval development and pupation time, and reduced longevity after a blood meal.
Collapse
Affiliation(s)
- Yuemei Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Malaria Research Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Maria L. Simões
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Malaria Research Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Eric Marois
- Inserm, CNRS, Université de Strasbourg, Strasbourg, France
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Malaria Research Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
47
|
Eldering M, Bompard A, Miura K, Stone W, Morlais I, Cohuet A, van Gemert GJ, Brock PM, Rijpma SR, van de Vegte-Bolmer M, Graumans W, Siebelink-Stoter R, Da DF, Long CA, Morin MJ, Sauerwein RW, Churcher TS, Bousema T. Comparative assessment of An. gambiae and An. stephensi mosquitoes to determine transmission-reducing activity of antibodies against P. falciparum sexual stage antigens. Parasit Vectors 2017; 10:489. [PMID: 29041962 PMCID: PMC5646129 DOI: 10.1186/s13071-017-2414-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 10/01/2017] [Indexed: 01/08/2023] Open
Abstract
Background With the increasing interest in vaccines to interrupt malaria transmission, there is a demand for harmonization of current methods to assess Plasmodium transmission in laboratory settings. Potential vaccine candidates are currently tested in the standard membrane feeding assay (SMFA) that commonly relies on Anopheles stephensi mosquitoes. Other mosquito species including Anopheles gambiae are the dominant malaria vectors for Plasmodium falciparum in sub-Saharan Africa. Methods Using human serum and monoclonal pre-fertilization (anti-Pfs48/45) and post-fertilization (anti-Pfs25) antibodies known to effectively inhibit sporogony, we directly compared SMFA based estimates of transmission-reducing activity (TRA) for An. stephensi and An. gambiae mosquitoes. Results In the absence of transmission-reducing antibodies, average numbers of oocysts were similar between An. gambiae and An. stephensi. Antibody-mediated TRA was strongly correlated between both mosquito species, and absolute TRA estimates for pre-fertilisation monoclonal antibodies (mAb) showed no significant difference between the two species. TRA estimates for IgG of naturally exposed individuals and partially effective concentrations of anti-Pfs25 mAb were higher for An. stephensi than for An. gambiae. Conclusion Our findings support the use of An. stephensi in the SMFA for target prioritization. As a vaccine moves through product development, better estimates of TRA and transmission-blocking activity (TBA) may need to be obtained in epidemiologically relevant parasite-species combination. Electronic supplementary material The online version of this article (10.1186/s13071-017-2414-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Maarten Eldering
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anaïs Bompard
- MRC Centre for Outbreak Analysis and Modelling, Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - Kazutoyo Miura
- National Institute of Allergy and Infectious Diseases, Laboratory of Malaria and Vector Research, National Institutes of Health, Rockville, MD, USA
| | - Will Stone
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Isabelle Morlais
- Institut de Recherche pour le Développement, UMR MIVEGEC UM-CNRS 5290-IRD 224, Montpellier, France
| | - Anna Cohuet
- Institut de Recherche pour le Développement, UMR MIVEGEC UM-CNRS 5290-IRD 224, Montpellier, France
| | - Geert-Jan van Gemert
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Patrick M Brock
- MRC Centre for Outbreak Analysis and Modelling, Department of Infectious Disease Epidemiology, Imperial College London, London, UK.,Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Sanna R Rijpma
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Wouter Graumans
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rianne Siebelink-Stoter
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Dari F Da
- Institut de Recherche en Sciences de la Santé, Bobo Dioulasso, Burkina Faso
| | - Carole A Long
- National Institute of Allergy and Infectious Diseases, Laboratory of Malaria and Vector Research, National Institutes of Health, Rockville, MD, USA
| | | | - Robert W Sauerwein
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Thomas S Churcher
- MRC Centre for Outbreak Analysis and Modelling, Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - Teun Bousema
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands. .,Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK.
| |
Collapse
|
48
|
Dong S, Balaraman V, Kantor AM, Lin J, Grant DG, Held NL, Franz AWE. Chikungunya virus dissemination from the midgut of Aedes aegypti is associated with temporal basal lamina degradation during bloodmeal digestion. PLoS Negl Trop Dis 2017; 11:e0005976. [PMID: 28961239 PMCID: PMC5636170 DOI: 10.1371/journal.pntd.0005976] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 10/11/2017] [Accepted: 09/19/2017] [Indexed: 11/21/2022] Open
Abstract
In the mosquito, the midgut epithelium is the initial tissue to become infected with an arthropod-borne virus (arbovirus) that has been acquired from a vertebrate host along with a viremic bloodmeal. Following its replication in midgut epithelial cells, the virus needs to exit the midgut and infect secondary tissues including the salivary glands before it can be transmitted to another vertebrate host. The viral exit mechanism from the midgut, the midgut escape barrier (MEB), is poorly understood although it is an important determinant of mosquito vector competence for arboviruses. Using chikungunya virus (CHIKV) as a model in Aedes aegypti, we demonstrate that the basal lamina (BL) of the extracellular matrix (ECM) surrounding the midgut constitutes a potential barrier for the virus. The BL, predominantly consisting of collagen IV and laminin, becomes permissive during bloodmeal digestion in the midgut lumen. Bloodmeal digestion, BL permissiveness, and CHIKV dissemination are coincident with increased collagenase activity, diminished collagen IV abundance, and BL shredding in the midgut between 24–32 h post-bloodmeal. This indicates that there may be a window-of-opportunity during which the MEB in Ae. aegypti becomes permissive for CHIKV. Matrix metalloproteinases (MMPs) are the principal extracellular endopeptidases responsible for the degradation/remodeling of the ECM including the BL. We focused on Ae. aegypti (Ae)MMP1, which is expressed in midgut epithelial cells, is inducible upon bloodfeeding, and shows collagenase (gelatinase) activity. However, attempts to inhibit AeMMP activity in general or specifically that of AeMMP1 did not seem to affect its function nor produce an altered midgut escape phenotype. As an alternative, we silenced and overexpressed the Ae. aegyptitissue inhibitor of metalloproteinases (AeTIMP) in the mosquito midgut. AeTIMP was highly upregulated in the midgut during bloodmeal digestion and was able to inhibit MMP activity in vitro. Bloodmeal-inducible, midgut-specific overexpression of AeTIMP or its expression via a recombinant CHIKV significantly increased midgut dissemination rates of the virus. Possibly, AeTIMP overexpression affected BL degradation and/or restoration thereby increasing the midgut dissemination efficiency of the virus. The biological nature of the midgut escape barrier in insects for arthropod-borne viruses has been a mystery for decades. Here we show that the basal lamina (BL) surrounding the mosquito midgut acts as a barrier for chikungunya virus, an alphavirus, which has emerged in the New World hemisphere around three years ago. The barrier became permissive for the virus during digestion of a viremic bloodmeal inside the midgut lumen. Concurrent with BL permissiveness, we observed that collagen IV, a major component of the BL became temporally degraded while the BL was visibly damaged. Based on previous findings, we hypothesized that matrix metalloproteinases such as Ae. aegypti (Ae)MMP1 may be involved in BL degradation. We confirmed that recombinant AeMMP1 exhibited strong gelatinase activity, which was profoundly reduced when recombinant AeMMP1 interacted in vitro with the recombinant Ae. aegypti tissue inhibitor of metalloproteinases (AeTIMP). When transgenically overexpressing AeTIMP in an attempt to temporally inhibit general MMP activity in the mosquito midgut, we observed that the dissemination efficiency of chikungunya virus became significantly increased, while its midgut infection was not affected. It is possible that AeTIMP overexpression affected BL degradation/restoration permitting increased quantities of virus to escape from the midgut.
Collapse
Affiliation(s)
- Shengzhang Dong
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
| | - Velmurugan Balaraman
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
| | - Asher M. Kantor
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
| | - Jingyi Lin
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
| | - DeAna G. Grant
- Electron Microscopy Core Facility, University of Missouri, Columbia, Missouri, United States of America
| | - Nicole L. Held
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
| | - Alexander W. E. Franz
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
- * E-mail:
| |
Collapse
|
49
|
Chaturvedi N, Bharti PK, Tiwari A, Singh N. Strategies & recent development of transmission-blocking vaccines against Plasmodium falciparum. Indian J Med Res 2017; 143:696-711. [PMID: 27748294 PMCID: PMC5094109 DOI: 10.4103/0971-5916.191927] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Transmission blocking malaria vaccines are aimed to block the development and maturity of sexual stages of parasite within mosquitoes. The vaccine candidate antigens (Pfs25, Pfs48/45, Pfs230) that have shown transmission blocking immunity in model systems are in different stages of development. These antigens are immunogenic with limited genetic diversity. Pfs25 is a leading candidate and currently in phase I clinical trial. Efforts are now focused on the cost-effective production of potent antigens using safe adjuvants and optimization of vaccine delivery system that are capable of inducing strong immune responses. This review addresses the potential usefulness, development strategies, challenges, clinical trials and current status of Plasmodium falciparum sexual stage malaria vaccine candidate antigens for the development of transmission-blocking vaccines.
Collapse
Affiliation(s)
- Neha Chaturvedi
- National Institute for Research in Tribal Health (ICMR), Jabalpur, School of Biotechnology, Rajiv Gandhi Proudyogiki Vishwavidyalaya (State Technological University of Madhya Pradesh), Bhopal, India
| | - Praveen K Bharti
- National Institute for Research in Tribal Health (ICMR), Jabalpur, India
| | - Archana Tiwari
- School of Biotechnology, Rajiv Gandhi Proudyogiki Vishwavidyalaya (State Technological University of Madhya Pradesh), Bhopal, India
| | - Neeru Singh
- National Institute for Research in Tribal Health (ICMR), Jabalpur, India
| |
Collapse
|
50
|
Niu G, Franc A C, Zhang G, Roobsoong W, Nguitragool W, Wang X, Prachumsri J, Butler NS, Li J. The fibrinogen-like domain of FREP1 protein is a broad-spectrum malaria transmission-blocking vaccine antigen. J Biol Chem 2017; 292:11960-11969. [PMID: 28533429 DOI: 10.1074/jbc.m116.773564] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 05/15/2017] [Indexed: 01/01/2023] Open
Abstract
FREP1 in mosquito midguts facilitates Plasmodium falciparum parasite transmission. The fibrinogen-like (FBG) domain of FREP1 is highly conserved (>90% identical) among Anopheles species from different continents, suggesting that anti-FBG antibodies may block malaria transmission to all anopheline mosquitoes. Using standard membrane-feeding assays, anti-FREP1 polyclonal antibodies significantly blocked transmission of Plasmodium berghei and Plasmodium vivax to Anopheles gambiae and Anopheles dirus, respectively. Furthermore, in vivo studies of mice immunized with FBG achieved >75% blocking efficacy of P. berghei to A. gambiae without triggering immunopathology. Anti-FBG serum also reduced >81% of P. falciparum infection to A. gambiae Finally, we showed that FBG interacts with Plasmodium gametocytes and ookinetes, revealing the molecular mechanism of its antibody transmission-blocking activity. Collectively, our data support that FREP1-mediated Plasmodium transmission to mosquitoes is a conserved pathway and that targeting the FBG domain of FREP1 will limit the transmission of multiple Plasmodium species to multiple Anopheles species.
Collapse
Affiliation(s)
- Guodong Niu
- Department of Biological Sciences, Florida International University, Miami, Florida 33199
| | - Caio Franc A
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019
| | - Genwei Zhang
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019
| | - Wanlapa Roobsoong
- Mahidol Vivax Research Center, Mahidol University Faculty of Tropical Medicine, Bangkok 10400, Thailand
| | - Wang Nguitragool
- Mahidol Vivax Research Center, Mahidol University Faculty of Tropical Medicine, Bangkok 10400, Thailand
| | - Xiaohong Wang
- Department of Biological Sciences, Florida International University, Miami, Florida 33199
| | - Jetsumon Prachumsri
- Mahidol Vivax Research Center, Mahidol University Faculty of Tropical Medicine, Bangkok 10400, Thailand
| | - Noah S Butler
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa 52242
| | - Jun Li
- Department of Biological Sciences, Florida International University, Miami, Florida 33199; Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019.
| |
Collapse
|