1
|
Kumari D, Mahajan S, Kour P, Singh K. Virulence factors of Leishmania parasite: Their paramount importance in unraveling novel vaccine candidates and therapeutic targets. Life Sci 2022; 306:120829. [PMID: 35872004 DOI: 10.1016/j.lfs.2022.120829] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/15/2022] [Accepted: 07/17/2022] [Indexed: 12/30/2022]
Abstract
Leishmaniasis is a neglected tropical disease and remains a global concern for healthcare. It is caused by an opportunistic protozoan parasite belonging to the genus Leishmania and affects millions worldwide. This disease is mainly prevalent in tropical and subtropical regions and is associated with a high risk of public morbidity and mortality if left untreated. Transmission of this deadly disease is aggravated by the bite of female sand-fly vectors (Phlebotomus and Lutzomyia). With time, significant advancement in leishmaniasis-related research has been carried out to cope with the disease burden. Still, the Leishmania parasite has also co-evolved with its host and adapted successfully within the host's lethal milieu/environment. Thus, understanding and knowledge of various leishmanial virulence factors responsible for the parasitic infection are essential for exploring drug targets and vaccine candidates. The present review elucidates the importance of virulence factors in pathogenesis and summarizes the major leishmanial virulence molecules contributing to the parasitic infection during host-pathogen interaction. Furthermore, we have also elaborated on the potential contribution of leishmanial virulence proteins in developing vaccine candidates and exploring novel therapeutics against this parasitic disease. We aim to represent a clearer picture of parasite pathogenesis within the human host that can further aid in unraveling new strategies to fight against the deadly infection of leishmaniasis.
Collapse
Affiliation(s)
- Diksha Kumari
- Infectious Diseases Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Shavi Mahajan
- Infectious Diseases Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Parampreet Kour
- Infectious Diseases Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Kuljit Singh
- Infectious Diseases Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
2
|
Lage DP, Vale DL, Linhares FP, Freitas CS, Machado AS, Cardoso JMO, de Oliveira D, Galvani NC, de Oliveira MP, Oliveira-da-Silva JA, Ramos FF, Tavares GSV, Ludolf F, Bandeira RS, Pereira IAG, Chávez-Fumagalli MA, Roatt BM, Machado-de-Ávila RA, Christodoulides M, Coelho EAF, Martins VT. A Recombinant Chimeric Protein-Based Vaccine Containing T-Cell Epitopes from Amastigote Proteins and Combined with Distinct Adjuvants, Induces Immunogenicity and Protection against Leishmania infantum Infection. Vaccines (Basel) 2022; 10:vaccines10071146. [PMID: 35891310 PMCID: PMC9317424 DOI: 10.3390/vaccines10071146] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/13/2022] [Accepted: 07/16/2022] [Indexed: 02/06/2023] Open
Abstract
Currently, there is no licensed vaccine to protect against human visceral leishmaniasis (VL), a potentially fatal disease caused by infection with Leishmania parasites. In the current study, a recombinant chimeric protein ChimT was developed based on T-cell epitopes identified from the immunogenic Leishmania amastigote proteins LiHyp1, LiHyV, LiHyC and LiHyG. ChimT was associated with the adjuvants saponin (Sap) or monophosphoryl lipid A (MPLA) and used to immunize mice, and their immunogenicity and protective efficacy were evaluated. Both ChimT/Sap and ChimT/MPLA induced the development of a specific Th1-type immune response, with significantly high levels of IFN-γ, IL-2, IL-12, TNF-α and GM-CSF cytokines produced by CD4+ and CD8+ T cell subtypes (p < 0.05), with correspondingly low production of anti-leishmanial IL-4 and IL-10 cytokines. Significantly increased (p < 0.05) levels of nitrite, a proxy for nitric oxide, and IFN-γ expression (p < 0.05) were detected in stimulated spleen cell cultures from immunized and infected mice, as was significant production of parasite-specific IgG2a isotype antibodies. Significant reductions in the parasite load in the internal organs of the immunized and infected mice (p < 0.05) were quantified with a limiting dilution technique and quantitative PCR and correlated with the immunological findings. ChimT/MPLA showed marginally superior immunogenicity than ChimT/Sap, and although this was not statistically significant (p > 0.05), ChimT/MPLA was preferred since ChimT/Sap induced transient edema in the inoculation site. ChimT also induced high IFN-γ and low IL-10 levels from human PBMCs isolated from healthy individuals and from VL-treated patients. In conclusion, the experimental T-cell multi-epitope amastigote stage Leishmania vaccine administered with adjuvants appears to be a promising vaccine candidate to protect against VL.
Collapse
Affiliation(s)
- Daniela P. Lage
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, Belo Horizonte 30130-100, MG, Brazil; (D.P.L.); (D.L.V.); (F.P.L.); (C.S.F.); (A.S.M.); (N.C.G.); (M.P.d.O.); (J.A.O.-d.-S.); (F.F.R.); (G.S.V.T.); (F.L.); (R.S.B.); (I.A.G.P.); (E.A.F.C.); (V.T.M.)
| | - Danniele L. Vale
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, Belo Horizonte 30130-100, MG, Brazil; (D.P.L.); (D.L.V.); (F.P.L.); (C.S.F.); (A.S.M.); (N.C.G.); (M.P.d.O.); (J.A.O.-d.-S.); (F.F.R.); (G.S.V.T.); (F.L.); (R.S.B.); (I.A.G.P.); (E.A.F.C.); (V.T.M.)
| | - Flávia P. Linhares
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, Belo Horizonte 30130-100, MG, Brazil; (D.P.L.); (D.L.V.); (F.P.L.); (C.S.F.); (A.S.M.); (N.C.G.); (M.P.d.O.); (J.A.O.-d.-S.); (F.F.R.); (G.S.V.T.); (F.L.); (R.S.B.); (I.A.G.P.); (E.A.F.C.); (V.T.M.)
| | - Camila S. Freitas
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, Belo Horizonte 30130-100, MG, Brazil; (D.P.L.); (D.L.V.); (F.P.L.); (C.S.F.); (A.S.M.); (N.C.G.); (M.P.d.O.); (J.A.O.-d.-S.); (F.F.R.); (G.S.V.T.); (F.L.); (R.S.B.); (I.A.G.P.); (E.A.F.C.); (V.T.M.)
| | - Amanda S. Machado
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, Belo Horizonte 30130-100, MG, Brazil; (D.P.L.); (D.L.V.); (F.P.L.); (C.S.F.); (A.S.M.); (N.C.G.); (M.P.d.O.); (J.A.O.-d.-S.); (F.F.R.); (G.S.V.T.); (F.L.); (R.S.B.); (I.A.G.P.); (E.A.F.C.); (V.T.M.)
| | - Jamille M. O. Cardoso
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas (NUPEB), Departamento de Ciências Biológicas, Insituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto CEP 35400-000, MG, Brazil; (J.M.O.C.); (B.M.R.)
| | - Daysiane de Oliveira
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma 88806-000, SC, Brazil; (D.d.O.); (R.A.M.-d.-Á.)
| | - Nathália C. Galvani
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, Belo Horizonte 30130-100, MG, Brazil; (D.P.L.); (D.L.V.); (F.P.L.); (C.S.F.); (A.S.M.); (N.C.G.); (M.P.d.O.); (J.A.O.-d.-S.); (F.F.R.); (G.S.V.T.); (F.L.); (R.S.B.); (I.A.G.P.); (E.A.F.C.); (V.T.M.)
| | - Marcelo P. de Oliveira
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, Belo Horizonte 30130-100, MG, Brazil; (D.P.L.); (D.L.V.); (F.P.L.); (C.S.F.); (A.S.M.); (N.C.G.); (M.P.d.O.); (J.A.O.-d.-S.); (F.F.R.); (G.S.V.T.); (F.L.); (R.S.B.); (I.A.G.P.); (E.A.F.C.); (V.T.M.)
| | - João A. Oliveira-da-Silva
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, Belo Horizonte 30130-100, MG, Brazil; (D.P.L.); (D.L.V.); (F.P.L.); (C.S.F.); (A.S.M.); (N.C.G.); (M.P.d.O.); (J.A.O.-d.-S.); (F.F.R.); (G.S.V.T.); (F.L.); (R.S.B.); (I.A.G.P.); (E.A.F.C.); (V.T.M.)
| | - Fernanda F. Ramos
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, Belo Horizonte 30130-100, MG, Brazil; (D.P.L.); (D.L.V.); (F.P.L.); (C.S.F.); (A.S.M.); (N.C.G.); (M.P.d.O.); (J.A.O.-d.-S.); (F.F.R.); (G.S.V.T.); (F.L.); (R.S.B.); (I.A.G.P.); (E.A.F.C.); (V.T.M.)
| | - Grasiele S. V. Tavares
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, Belo Horizonte 30130-100, MG, Brazil; (D.P.L.); (D.L.V.); (F.P.L.); (C.S.F.); (A.S.M.); (N.C.G.); (M.P.d.O.); (J.A.O.-d.-S.); (F.F.R.); (G.S.V.T.); (F.L.); (R.S.B.); (I.A.G.P.); (E.A.F.C.); (V.T.M.)
| | - Fernanda Ludolf
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, Belo Horizonte 30130-100, MG, Brazil; (D.P.L.); (D.L.V.); (F.P.L.); (C.S.F.); (A.S.M.); (N.C.G.); (M.P.d.O.); (J.A.O.-d.-S.); (F.F.R.); (G.S.V.T.); (F.L.); (R.S.B.); (I.A.G.P.); (E.A.F.C.); (V.T.M.)
| | - Raquel S. Bandeira
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, Belo Horizonte 30130-100, MG, Brazil; (D.P.L.); (D.L.V.); (F.P.L.); (C.S.F.); (A.S.M.); (N.C.G.); (M.P.d.O.); (J.A.O.-d.-S.); (F.F.R.); (G.S.V.T.); (F.L.); (R.S.B.); (I.A.G.P.); (E.A.F.C.); (V.T.M.)
| | - Isabela A. G. Pereira
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, Belo Horizonte 30130-100, MG, Brazil; (D.P.L.); (D.L.V.); (F.P.L.); (C.S.F.); (A.S.M.); (N.C.G.); (M.P.d.O.); (J.A.O.-d.-S.); (F.F.R.); (G.S.V.T.); (F.L.); (R.S.B.); (I.A.G.P.); (E.A.F.C.); (V.T.M.)
| | - Miguel A. Chávez-Fumagalli
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Urb. San José S/N, Umacollo, Arequipa 04000, Peru;
| | - Bruno M. Roatt
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas (NUPEB), Departamento de Ciências Biológicas, Insituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto CEP 35400-000, MG, Brazil; (J.M.O.C.); (B.M.R.)
| | - Ricardo A. Machado-de-Ávila
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma 88806-000, SC, Brazil; (D.d.O.); (R.A.M.-d.-Á.)
| | - Myron Christodoulides
- Neisseria Research Group, Molecular Microbiology, Faculty of Medicine, School of Clinical and Experimental Sciences, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
- Correspondence: ; Tel.: +44-02381-205120
| | - Eduardo A. F. Coelho
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, Belo Horizonte 30130-100, MG, Brazil; (D.P.L.); (D.L.V.); (F.P.L.); (C.S.F.); (A.S.M.); (N.C.G.); (M.P.d.O.); (J.A.O.-d.-S.); (F.F.R.); (G.S.V.T.); (F.L.); (R.S.B.); (I.A.G.P.); (E.A.F.C.); (V.T.M.)
- Departamento de Patologia Clínica, Colégio Técnico (COLTEC), Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil
| | - Vívian T. Martins
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, Belo Horizonte 30130-100, MG, Brazil; (D.P.L.); (D.L.V.); (F.P.L.); (C.S.F.); (A.S.M.); (N.C.G.); (M.P.d.O.); (J.A.O.-d.-S.); (F.F.R.); (G.S.V.T.); (F.L.); (R.S.B.); (I.A.G.P.); (E.A.F.C.); (V.T.M.)
| |
Collapse
|
3
|
Kamdem BP, Elizabeth FI. The Role of Nitro (NO 2-), Chloro (Cl), and Fluoro (F) Substitution in the Design of Antileishmanial and Antichagasic Compounds. Curr Drug Targets 2021; 22:379-398. [PMID: 33371845 DOI: 10.2174/1389450121666201228122239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/22/2020] [Accepted: 11/11/2020] [Indexed: 11/22/2022]
Abstract
Neglected tropical diseases (NTDs) are responsible for over 500,000 deaths annually and are characterized by multiple disabilities. Leishmaniasis and Chagas diseases are among the most severe NTDs, and are caused by the Leishmania sp and Trypanosoma cruzi, respectively. Glucantime, pentamidine, and miltefosine are commonly used to treat leishmaniasis, whereas nifurtimox, benznidazole are current treatments for Chagas disease. However, these treatments are associated with drug resistance and severe side effects. Hence, the development of synthetic products, especially those containing N02, F, or Cl, are known to improve biological activity. The present work summarizes the information on the antileishmanial and antitrypanosomal activity of nitro-, chloro-, and fluorosynthetic derivatives. Scientific publications referring to halogenated derivatives in relation to antileishmanial and antitrypanosomal activities were hand-searched in databases such as SciFinder, Wiley, Science Direct, PubMed, ACS, Springer, Scielo, and so on. According to the literature information, more than 90 compounds were predicted as lead molecules with reference to their IC50/EC50 values in in vitro studies. It is worth mentioning that only active compounds with known cytotoxic effects against mammalian cells were considered in the present study. The observed activity was attributed to the presence of nitro-, fluoro-, and chloro-groups in the compound backbone. All in all, nitro and halogenated derivatives are active antileishmanial and antitrypanosomal compounds and can serve as the baseline for the development of new drugs against leishmaniasis and Chagas disease. However, efforts in in vitro and in vivo toxicity studies of the active synthetic compounds is still needed. Pharmacokinetic studies and the mechanism of action of the promising compounds need to be explored. The use of new catalysts and chemical transformation can afford unexplored halogenated compounds with improved antileishmanial and antitrypanosomal activity.
Collapse
Affiliation(s)
- Boniface P Kamdem
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Ferreira I Elizabeth
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
4
|
Elmahallawy EK, Alkhaldi AAM. Insights into Leishmania Molecules and Their Potential Contribution to the Virulence of the Parasite. Vet Sci 2021; 8:vetsci8020033. [PMID: 33672776 PMCID: PMC7924612 DOI: 10.3390/vetsci8020033] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/11/2021] [Accepted: 02/14/2021] [Indexed: 12/12/2022] Open
Abstract
Neglected parasitic diseases affect millions of people worldwide, resulting in high morbidity and mortality. Among other parasitic diseases, leishmaniasis remains an important public health problem caused by the protozoa of the genus Leishmania, transmitted by the bite of the female sand fly. The disease has also been linked to tropical and subtropical regions, in addition to being an endemic disease in many areas around the world, including the Mediterranean basin and South America. Although recent years have witnessed marked advances in Leishmania-related research in various directions, many issues have yet to be elucidated. The intention of the present review is to give an overview of the major virulence factors contributing to the pathogenicity of the parasite. We aimed to provide a concise picture of the factors influencing the reaction of the parasite in its host that might help to develop novel chemotherapeutic and vaccine strategies.
Collapse
Affiliation(s)
- Ehab Kotb Elmahallawy
- Department of Zoonoses, Faculty of Veterinary Medicine, Sohag University, Sohag 82524, Egypt
- Correspondence: (E.K.E.); (A.A.M.A.)
| | - Abdulsalam A. M. Alkhaldi
- Biology Department, College of Science, Jouf University, Sakaka, Aljouf 2014, Saudi Arabia
- Correspondence: (E.K.E.); (A.A.M.A.)
| |
Collapse
|
5
|
Salari S, Sharifi I, Bamorovat M, Ghasemi Nejad Almani P. The immunity of the recombinant prokaryotic and eukaryotic subunit vaccines against cutaneous leishmaniasis. Microb Pathog 2021; 153:104807. [PMID: 33609648 DOI: 10.1016/j.micpath.2021.104807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 01/15/2021] [Accepted: 02/07/2021] [Indexed: 10/22/2022]
Abstract
Leishmaniasis counts as one of the most neglected tropical diseases. Despite the amount of research perceived in this field, no fully effective and approved vaccine against this disease is yet available in humans. In this study, LACK and KMP11 antigens were constructed simultaneously by recombinant methods in prokaryotic and eukaryotic expression systems and were compared and assessed along with the CpG adjuvant in BALB/c mice. In the prokaryotic method, LACK and KMP11 protein gene sequences were synthesized in pET28a-TEV vector. In order to extract these two proteins after expression, His-tag and S-tag sequences were added to the constructs, respectively for LACK and KMP11. The pET28a-TEV-LACK/KMP11 construct was transformed into Escherichia coli, and the inserts were verified by Colony PCR. Pure proteins were verified by western blot, and groups of BALB/c mice were injected with the created prokaryotic recombinant proteins along with an ODN CpG adjuvant. In the eukaryotic method, antigen sequences were constructed in the pLEXSY-neo 2.1 vector, E.coli Top10 strain was cloned in the bacteria, and after being linearized were transfected into Leishmania tarentolae genome. After recombinant strains were selected, they were verified by molecular methods. After the extraction and purification of the protein using the method above, groups of mice were injected with the recombinant antigens and ODN CpG adjuvant. Eukaryotic subunit vaccines showed more effective immunization compared with prokaryotic vaccines and caused an immune system shift towards Th1 and protection. Protein expression in L. tarentolae by the constructs created in this host contains Post-Translational Modifications. The constructed protein will be significantly similar to eukaryotic proteins, considering that they are identical epitopes. More comprehensive studies aiming to improve the effectiveness of this vaccine are being conducted to improve immune profiles and immunological memory stimulation in future designs.
Collapse
Affiliation(s)
- Samira Salari
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman, Iran; Department of Medical Parasitology and Mycology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Iraj Sharifi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| | - Mehdi Bamorovat
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | | |
Collapse
|
6
|
Understanding the immune responses involved in mediating protection or immunopathology during leishmaniasis. Biochem Soc Trans 2021; 49:297-311. [PMID: 33449103 DOI: 10.1042/bst20200606] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/18/2020] [Accepted: 12/23/2020] [Indexed: 01/21/2023]
Abstract
Leishmaniasis is a vector-borne Neglected Tropical Disease (NTD) transmitted by the sand fly and is a major public health problem worldwide. Infections caused by Leishmania clinically manifest as a wide range of diseases, such as cutaneous (CL), diffuse cutaneous (DCL), mucosal (MCL) and visceral leishmaniasis (VL). The host innate and adaptative immune responses play critical roles in the defense against leishmaniasis. However, Leishmania parasites also manipulate the host immune response for their survival and replication. In addition, other factors such as sand fly salivary proteins and microbiota also promote disease susceptibility and parasite spread by modulating local immune response. Thus, a complex interplay between parasite, sand fly and the host immunity governs disease severity and outcome. In this review, we discuss the host immune response during Leishmania infection and highlight the factors associated with resistance or susceptibility.
Collapse
|
7
|
Ratnapriya S, Keerti, Sahasrabuddhe AA, Dube A. Visceral leishmaniasis: An overview of vaccine adjuvants and their applications. Vaccine 2019; 37:3505-3519. [PMID: 31103364 DOI: 10.1016/j.vaccine.2019.04.092] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 03/08/2019] [Accepted: 04/30/2019] [Indexed: 11/25/2022]
Abstract
Although there has been an extensive research on vaccine development over the last decade and some vaccines have been commercialized for canine visceral leishmaniasis (CVL), but as yet no effective vaccine is available for anthroponotic VL which may partly be due to the absence of an appropriate adjuvant system. Vaccines alone yield poor immunity hence requiring an adjuvant which can boost the immunosuppressed state of VL infected individuals by eliciting adaptive immune responses to achieve required immunological enhancement. Recent studies have documented the continuous efforts that are being made in the field of adjuvants research in an attempt to render vaccines more effective. This review article focuses on adjuvants, particularly particulate and non-particulate ones, which have been assessed with VL vaccine candidates in several preclinical and clinical trials outlining the induction of immune responses obtained from these studies. Moreover, we have emphasized the applicability of multiple adjuvants combination for an improvement in the potential of a VL vaccine.
Collapse
Affiliation(s)
- Sneha Ratnapriya
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Keerti
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Amogh A Sahasrabuddhe
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Anuradha Dube
- Division of Parasitology, CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India.
| |
Collapse
|
8
|
Volpedo G, Costa L, Ryan N, Halsey G, Satoskar A, Oghumu S. Nanoparticulate drug delivery systems for the treatment of neglected tropical protozoan diseases. J Venom Anim Toxins Incl Trop Dis 2019; 25:e144118. [PMID: 31130996 PMCID: PMC6483407 DOI: 10.1590/1678-9199-jvatitd-1441-18] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 11/06/2018] [Indexed: 12/13/2022] Open
Abstract
Neglected Tropical Diseases (NTDs) comprise of a group of seventeen infectious
conditions endemic in many developing countries. Among these diseases are three
of protozoan origin, namely leishmaniasis, Chagas disease, and African
trypanosomiasis, caused by the parasites Leishmania spp.,
Trypanosoma cruzi, and Trypanosoma brucei
respectively. These diseases have their own unique challenges which are
associated with the development of effective prevention and treatment methods.
Collectively, these parasitic diseases cause more deaths worldwide than all
other NTDs combined. Moreover, many current therapies for these diseases are
limited in their efficacy, possessing harmful or potentially fatal side effects
at therapeutic doses. It is therefore imperative that new treatment strategies
for these parasitic diseases are developed. Nanoparticulate drug delivery
systems have emerged as a promising area of research in the therapy and
prevention of NTDs. These delivery systems provide novel mechanisms for targeted
drug delivery within the host, maximizing therapeutic effects while minimizing
systemic side effects. Currently approved drugs may also be repackaged using
these delivery systems, allowing for their potential use in NTDs of protozoan
origin. Current research on these novel delivery systems has provided insight
into possible indications, with evidence demonstrating their improved ability to
specifically target pathogens, penetrate barriers within the host, and reduce
toxicity with lower dose regimens. In this review, we will examine current
research on these delivery systems, focusing on applications in the treatment of
leishmaniasis, Chagas disease, and African trypanosomiasis. Nanoparticulate
systems present a unique therapeutic alternative through the repositioning of
existing medications and directed drug delivery.
Collapse
Affiliation(s)
- Greta Volpedo
- Ohio State University Wexner Medical Center, Department of Pathology, Columbus, OH, 43210, USA.,Ohio State University, Department of Microbiology, Columbus, OH, 43210, USA
| | - Lourena Costa
- Ohio State University Wexner Medical Center, Department of Pathology, Columbus, OH, 43210, USA.,Universidade Federal de Minas Gerais, Faculdade de Medicina, Departamento de Infectologia e Medicina Tropical, Belo Horizonte, MG, Brasil
| | - Nathan Ryan
- Ohio State University Wexner Medical Center, Department of Pathology, Columbus, OH, 43210, USA
| | - Gregory Halsey
- Ohio State University Wexner Medical Center, Department of Pathology, Columbus, OH, 43210, USA
| | - Abhay Satoskar
- Ohio State University Wexner Medical Center, Department of Pathology, Columbus, OH, 43210, USA.,Ohio State University, Department of Microbiology, Columbus, OH, 43210, USA
| | - Steve Oghumu
- Ohio State University Wexner Medical Center, Department of Pathology, Columbus, OH, 43210, USA
| |
Collapse
|
9
|
Leishmania treatment and prevention: Natural and synthesized drugs. Eur J Med Chem 2018; 160:229-244. [DOI: 10.1016/j.ejmech.2018.10.022] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 12/31/2022]
|
10
|
He J, Huang F, Zhang J, Chen H, Chen Q, Zhang J, Li J, Zheng Z, Chen D, Chen J. DNA prime-protein boost vaccine encoding HLA-A2, HLA-A24 and HLA-DR1 restricted epitopes of CaNA2 against visceral leishmaniasis. Immunology 2018; 156:94-108. [PMID: 30285279 DOI: 10.1111/imm.13007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 09/09/2018] [Accepted: 09/26/2018] [Indexed: 12/21/2022] Open
Abstract
Visceral leishmaniasis is a tropical and neglected disease with an estimated 200 000-400 000 cases and 60 000 deaths worldwide each year. Currently, no clinically valid vaccine is available for this disease. In this study, we formulated DNA and protein vaccines encoding HLA-A2, HLA-A24 and HLA-DR1 restricted epitopes of CaNA2 against visceral leishmaniasis. We predicted the secondary and tertiary structures, surface properties, subcellular localization, potential binding sites and HLA-A2, HLA-A24 and HLA-DR1 restricted epitopes of CaNA2. The best candidate CpG ODN (2395, M362, D-SL03 or 685) was screened out as a DNA vaccine adjuvant. We also prepared Kmp-11 and Kmp-11/CaNA2 DNA and protein vaccines, respectively, for comparison. BALB/c mice were immunized with a DNA prime-protein boost immunization strategy and challenged with a newly isolated Leishmania strain from an individual with visceral leishmaniasis. The IgG antibody titers showed that our vaccine had strong immunogenicity with a long duration, especially cellular immunity. The spleen parasite burden of each group demonstrated that the CaNA2 vaccine had a certain immune protective effect on visceral leishmaniasis in BALB/c mice, and the amastigote reduction rate reached 76%. Preliminary safety tests confirmed the safety of the vaccine. Our work demonstrates that the HLA-A2, HLA-A24 and HLA-DR1 restricted epitope CaNA2 DNA prime-protein boost vaccine may be a safe and effective epitope vaccine candidate against visceral leishmaniasis.
Collapse
Affiliation(s)
- Jinlei He
- Department of Parasitology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Fan Huang
- Surgical Department, Chengdu Shuangliu Hospital of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Jianhui Zhang
- Department of Parasitology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Han Chen
- Department of Parasitology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Qiwei Chen
- Department of Parasitology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Junrong Zhang
- Department of Parasitology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Jiao Li
- Department of Parasitology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Zhiwan Zheng
- Department of Parasitology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Dali Chen
- Department of Parasitology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Jianping Chen
- Department of Parasitology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China.,Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
11
|
Mortazavidehkordi N, Fallah A, Abdollahi A, Kia V, Khanahmad H, Najafabadi ZG, Hashemi N, Estiri B, Roudbari Z, Najafi A, Farjadfar A, Hejazi SH. A lentiviral vaccine expressing KMP11-HASPB fusion protein increases immune response to Leishmania major in BALB/C. Parasitol Res 2018; 117:2265-2273. [PMID: 29845415 DOI: 10.1007/s00436-018-5915-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 05/09/2018] [Indexed: 11/28/2022]
Abstract
Hydrophilic acylated surface protein B (HASPB) is an immunogenic Leishmania-specific protein that antibodies are produced against it in the sera of Leishmania-infected individuals. Kinetoplastid membrane protein 11 (KMP11) is another Leishmania antigen and considered as the suitable candidate for vaccine development Leishmaniasis. It is a highly conserved surface protein expressed in both promastigotes and amastigotes. In this study, KMP11 and HASPB coding sequences were cloned into a pCDH-cGFP lentiviral vector as a fusion protein to be used as a DNA vaccine against L. major. The KMP11-HASPB fusion protein was successfully expressed as evidenced by RT-PCR and Western blot assays. The effect of the vaccine was determined by evaluating the level of IFN-γ, IL-10, IgG1, and IgG2a performed using ELISA as well as determining the parasite load after challenge with L. major in vaccinated mice. The results revealed that IFN-γ, IL-10, IgG1, and IgG2a significantly increased after vaccination using KMP11-HASPB-expressing lentiviruses in BALB/c mice. It is noteworthy that the level of IFN-γ and IgG2a was higher than that of IL-10 and IgG1, respectively, which indicates the activation Th1 cells, macrophages, and cellular immunity. Moreover, the parasite load in the spleen and lymph node of vaccinated mice after challenge was significantly lower than that of controls.
Collapse
Affiliation(s)
| | - Ali Fallah
- Systems and Synthetic Biology Group, Mede Bioeconomy Company, Tehran, Iran
- BioViva USA Inc, Bainbridge Island, WA, USA
| | - Abbas Abdollahi
- Department of Medical Microbiology, Fasa University of Medical Sciences, Fars, Iran
| | - Vahid Kia
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hossein Khanahmad
- Department of Genetics, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Nooshin Hashemi
- Vector-borne Diseases Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Bahareh Estiri
- Iranian Institute of Cell and Gene Therapy, Tehran, Iran
| | - Zahra Roudbari
- Department of Animal Science, Faculty of Agriculture, University of Jiroft, P.O. Box 364, Jiroft, Iran
| | - Ali Najafi
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Akbar Farjadfar
- Department of Medical Biotechnology, Fasa University of Medical Sciences, Fasa, Iran.
| | - Seyed Hossein Hejazi
- Skin Diseases and Leishmaniasis Research Center, Department of Parasitology and Mycology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
12
|
Abkar M, Fasihi-Ramandi M, Kooshki H, Lotfi AS. Intraperitoneal immunization with Urease loaded N -trimethyl Chitosan nanoparticles elicits high protection against Brucella melitensis and Brucella abortus infections. Immunol Lett 2018; 199:53-60. [DOI: 10.1016/j.imlet.2018.03.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 03/02/2018] [Accepted: 03/10/2018] [Indexed: 01/21/2023]
|
13
|
Kumar A, Dikhit MR, Amit A, Zaidi A, Pandey RK, Singh AK, Suman SS, Ali V, Das VNR, Pandey K, kumar V, Singh SK, Narayan S, Chourasia HK, Das P, Bimal S. Immunomodulation induced through ornithine decarboxylase DNA immunization in Balb/c mice infected with Leishmania donovani. Mol Immunol 2018; 97:33-44. [DOI: 10.1016/j.molimm.2018.03.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 03/05/2018] [Accepted: 03/07/2018] [Indexed: 12/21/2022]
|
14
|
Ghorbani M, Farhoudi R. Leishmaniasis in humans: drug or vaccine therapy? DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 12:25-40. [PMID: 29317800 PMCID: PMC5743117 DOI: 10.2147/dddt.s146521] [Citation(s) in RCA: 214] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Leishmania is an obligate intracellular pathogen that invades phagocytic host cells. Approximately 30 different species of Phlebotomine sand flies can transmit this parasite either anthroponotically or zoonotically through their bites. Leishmaniasis affects poor people living around the Mediterranean Basin, East Africa, the Americas, and Southeast Asia. Affected regions are often remote and unstable, with limited resources for treating this disease. Leishmaniasis has been reported as one of the most dangerous neglected tropical diseases, second only to malaria in parasitic causes of death. People can carry some species of Leishmania for long periods without becoming ill, and symptoms depend on the form of the disease. There are many drugs and candidate vaccines available to treat leishmaniasis. For instance, antiparasitic drugs, such as amphotericin B (AmBisome), are a treatment of choice for leishmaniasis depending on the type of the disease. Despite the availability of different treatment approaches to treat leishmaniasis, therapeutic tools are not adequate to eradicate this infection. In the meantime, drug therapy has been limited because of adverse side effects and unsuccessful vaccine preparation. However, it can immediately make infections inactive. According to other studies, vaccination cannot eradicate leishmaniasis. There is no perfect vaccine or suitable drug to eradicate leishmaniasis completely. So far, no vaccine or drug has been provided to induce long-term protection and ensure effective immunity against leishmaniasis. Therefore, it is necessary that intensive research should be performed in drug and vaccine fields to achieve certain results.
Collapse
Affiliation(s)
- Masoud Ghorbani
- Department of Viral Vaccine Production, Pasteur Institute of Iran, Research and Production Complex, Karaj, Iran
| | - Ramin Farhoudi
- Department of Viral Vaccine Production, Pasteur Institute of Iran, Research and Production Complex, Karaj, Iran
| |
Collapse
|
15
|
Cecílio P, Pérez-Cabezas B, Fernández L, Moreno J, Carrillo E, Requena JM, Fichera E, Reed SG, Coler RN, Kamhawi S, Oliveira F, Valenzuela JG, Gradoni L, Glueck R, Gupta G, Cordeiro-da-Silva A. Pre-clinical antigenicity studies of an innovative multivalent vaccine for human visceral leishmaniasis. PLoS Negl Trop Dis 2017; 11:e0005951. [PMID: 29176865 PMCID: PMC5720812 DOI: 10.1371/journal.pntd.0005951] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 12/07/2017] [Accepted: 09/11/2017] [Indexed: 01/18/2023] Open
Abstract
The notion that previous infection by Leishmania spp. in endemic areas leads to robust anti-Leishmania immunity, supports vaccination as a potentially effective approach to prevent disease development. Nevertheless, to date there is no vaccine available for human leishmaniasis. We optimized and assessed in vivo the safety and immunogenicity of an innovative vaccine candidate against human visceral leishmaniasis (VL), consisting of Virus-Like Particles (VLP) loaded with three different recombinant proteins (LJL143 from Lutzomyia longipalpis saliva as the vector-derived (VD) component, and KMP11 and LeishF3+, as parasite-derived (PD) antigens) and adjuvanted with GLA-SE, a TLR4 agonist. No apparent adverse reactions were observed during the experimental time-frame, which together with the normal hematological parameters detected seems to point to the safety of the formulation. Furthermore, measurements of antigen-specific cellular and humoral responses, generally higher in immunized versus control groups, confirmed the immunogenicity of the vaccine formulation. Interestingly, the immune responses against the VD protein were reproducibly more robust than those elicited against leishmanial antigens, and were apparently not caused by immunodominance of the VD antigen. Remarkably, priming with the VD protein alone and boosting with the complete vaccine candidate contributed towards an increase of the immune responses to the PD antigens, assessed in the form of increased ex vivo CD4+ and CD8+ T cell proliferation against both the PD antigens and total Leishmania antigen (TLA). Overall, our immunogenicity data indicate that this innovative vaccine formulation represents a promising anti-Leishmania vaccine whose efficacy deserves to be tested in the context of the "natural infection".
Collapse
Affiliation(s)
- Pedro Cecílio
- Parasite Disease group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- IBMC—Instituto de Biologia Celular e Molecular, Universidade do Porto, Porto, Portugal
- Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto, Porto, Portugal
| | - Begoña Pérez-Cabezas
- Parasite Disease group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- IBMC—Instituto de Biologia Celular e Molecular, Universidade do Porto, Porto, Portugal
| | - Laura Fernández
- WHO Collaborating Centre for Leishmaniasis, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Moreno
- WHO Collaborating Centre for Leishmaniasis, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - Eugenia Carrillo
- WHO Collaborating Centre for Leishmaniasis, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - José M. Requena
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Epifanio Fichera
- Etna Biotech S.R.L, via Vincenzo Lancia, 57—Zona Industriale Blocco Palma 1, Catania, Italy
| | - Steven G. Reed
- Infectious Disease Research Institute (IDRI), Seattle, WA, United States of America
| | - Rhea N. Coler
- Infectious Disease Research Institute (IDRI), Seattle, WA, United States of America
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, NIAID, NIH, Rockville, MD, United States of America
| | - Fabiano Oliveira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, NIAID, NIH, Rockville, MD, United States of America
| | - Jesus G. Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, NIAID, NIH, Rockville, MD, United States of America
| | - Luigi Gradoni
- Unit of Vector-borne Diseases and International Health, Istituto Superiore di Sanità, Rome, Italy
| | - Reinhard Glueck
- Etna Biotech S.R.L, via Vincenzo Lancia, 57—Zona Industriale Blocco Palma 1, Catania, Italy
| | - Gaurav Gupta
- Etna Biotech S.R.L, via Vincenzo Lancia, 57—Zona Industriale Blocco Palma 1, Catania, Italy
| | - Anabela Cordeiro-da-Silva
- Parasite Disease group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- IBMC—Instituto de Biologia Celular e Molecular, Universidade do Porto, Porto, Portugal
- Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto, Porto, Portugal
| |
Collapse
|
16
|
Skin vaccination using microneedles coated with a plasmid DNA cocktail encoding nucleosomal histones of Leishmania spp. Int J Pharm 2017; 533:236-244. [DOI: 10.1016/j.ijpharm.2017.09.055] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 09/18/2017] [Accepted: 09/19/2017] [Indexed: 02/08/2023]
|
17
|
Probing the efficacy of a heterologous Leishmania/L. Viannia braziliensis recombinant enolase as a candidate vaccine to restrict the development of L. infantum in BALB/c mice. Acta Trop 2017; 171:8-16. [PMID: 28288798 DOI: 10.1016/j.actatropica.2017.03.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 02/11/2017] [Accepted: 03/09/2017] [Indexed: 11/23/2022]
Abstract
In the present study, the Leishmania braziliensis enolase protein was evaluated as a vaccine candidate against visceral leishmaniasis (VL). The DNA sequence was cloned and the recombinant protein (rEnolase) was evaluated as a vaccine, associated with saponin, as an immune adjuvant. The protective efficacy of the rEnolase plus saponin combination was investigated in BALB/c mice against Leishmania infantum infection. The results revealed that the vaccine induced higher levels of IFN-γ, IL-12, and GM-CSF when a capture ELISA and flow cytometry were performed, as well as an antileishmanial nitrite production after using in vitro stimulation with rEnolase and an antigenic Leishmania preparation. The vaccinated animals, when compared to the control groups, showed a lower parasite burden in the liver, spleen, bone marrow, and paws' draining lymph nodes when both a limiting dilution technique and RT-PCR assay were performed. In addition, these mice showed low levels of antileishmanial IL-4, IL-10, and anti-Leishmania IgG1 isotype antibodies. Partial protection was associated with IFN-γ production, which was mainly mediated by CD4+ T cells. In conclusion, the present study's data showed that the L. braziliensis enolase protein could be considered a vaccine candidate that offers heterologous protection against VL.
Collapse
|
18
|
Optimizing Immunization Strategies for the Induction of Antigen-Specific CD4 and CD8 T Cell Responses for Protection against Intracellular Parasites. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2016; 23:785-94. [PMID: 27466350 PMCID: PMC5014921 DOI: 10.1128/cvi.00251-16] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 07/18/2016] [Indexed: 11/20/2022]
Abstract
Immunization strategies that generate either CD4 or CD8 T cell responses are relatively well described, but less is known with regard to optimizing regimens to induce both CD4 and CD8 memory T cells. Considering the importance of both CD4 and CD8 T cells in the control of intracellular pathogens such as Leishmania donovani, we wanted to identify vaccines that could raise both CD4 and CD8 T cell responses and determine how to configure immunization strategies to generate the best combined protective T cell response. We examined responses generated against the Leishmania vaccine antigen F3 following its administration in either recombinant form with the Toll-like receptor 4 (TLR4) agonist-containing adjuvant formulation GLA-SE (F3+GLA-SE) or as a gene product delivered in an adenoviral vector (Ad5-F3). Homologous immunization strategies using only F3+GLA-SE or Ad5-F3 preferentially generated either CD4 or CD8 T cells, respectively. In contrast, heterologous strategies generated both antigen-specific CD4 and CD8 T cells. Administration of F3+GLA-SE before Ad5-F3 generated the greatest combined CD4 and CD8 responses. Cytotoxic CD8 T cell responses were highest when Th1 cells were generated prior to their induction by Ad5-F3. Finally, a single immunization with a combination of F3+GLA-SE mixed with Ad5-F3 was found to be sufficient to provide protection against experimental L. donovani infection. Taken together, our data delineate immunization regimens that induce antigen-specific CD4 and CD8 T cell memory responses, and identify a single immunization strategy that could be used to rapidly provide protection against intracellular pathogens in regions where access to health care is limited or sporadic.
Collapse
|
19
|
Duthie MS, Favila M, Hofmeyer KA, Tutterrow YL, Reed SJ, Laurance JD, Picone A, Guderian J, Bailor HR, Vallur AC, Liang H, Mohamath R, Vergara J, Howard RF, Coler RN, Reed SG. Strategic evaluation of vaccine candidate antigens for the prevention of Visceral Leishmaniasis. Vaccine 2016; 34:2779-86. [PMID: 27142329 PMCID: PMC4889780 DOI: 10.1016/j.vaccine.2016.04.067] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 04/18/2016] [Accepted: 04/23/2016] [Indexed: 11/03/2022]
Abstract
Infection with Leishmania parasites results in a range of clinical manifestations and outcomes, the most severe of which is visceral leishmaniasis (VL). Vaccination will likely provide the most effective long-term control strategy, as the large number of vectors and potential infectious reservoirs renders sustained interruption of Leishmania parasite transmission extremely difficult. Selection of the best vaccine is complicated because, although several vaccine antigen candidates have been proposed, they have emerged following production in different platforms. To consolidate the information that has been generated into a single vaccine platform, we expressed seven candidates as recombinant proteins in E. coli. After verifying that each recombinant protein could be recognized by VL patients, we evaluated their protective efficacy against experimental L. donovani infection of mice. Administration in formulation with the Th1-potentiating adjuvant GLA-SE indicated that each antigen could elicit antigen-specific Th1 responses that were protective. Considering the ability to reduce parasite burden along with additional factors such as sequence identity across Leishmania species, we then generated a chimeric fusion protein comprising a combination of the 8E, p21 and SMT proteins. This E. coli –expressed fusion protein was also demonstrated to protect against L. donovani infection. These data indicate a novel recombinant vaccine antigen with the potential for use in VL control programs.
Collapse
Affiliation(s)
- Malcolm S Duthie
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA 98102, USA.
| | - Michelle Favila
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA 98102, USA
| | - Kimberley A Hofmeyer
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA 98102, USA
| | - Yeung L Tutterrow
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA 98102, USA
| | - Steven J Reed
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA 98102, USA
| | - John D Laurance
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA 98102, USA
| | - Alessandro Picone
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA 98102, USA
| | - Jeffrey Guderian
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA 98102, USA
| | - H Remy Bailor
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA 98102, USA
| | - Aarthy C Vallur
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA 98102, USA
| | - Hong Liang
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA 98102, USA
| | - Raodoh Mohamath
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA 98102, USA
| | - Julie Vergara
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA 98102, USA
| | - Randall F Howard
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA 98102, USA
| | - Rhea N Coler
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA 98102, USA
| | - Steven G Reed
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA 98102, USA
| |
Collapse
|
20
|
Kumar A, Samant M. DNA vaccine against visceral leishmaniasis: a promising approach for prevention and control. Parasite Immunol 2016; 38:273-81. [DOI: 10.1111/pim.12315] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 03/21/2016] [Indexed: 01/23/2023]
Affiliation(s)
- A. Kumar
- Department of Biotechnology; National Institute of Technology; Raipur Chhattisgarh India
| | - M. Samant
- Cell and Molecular biology laboratory; Department of Zoology; Kumaun University SSJ Campus; Almora Uttarakhand India
| |
Collapse
|
21
|
De Luca PM, Macedo ABB. Cutaneous Leishmaniasis Vaccination: A Matter of Quality. Front Immunol 2016; 7:151. [PMID: 27148270 PMCID: PMC4838622 DOI: 10.3389/fimmu.2016.00151] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 04/07/2016] [Indexed: 11/25/2022] Open
Abstract
There have been exhaustive efforts to develop an efficient vaccine against leishmaniasis. Factors like host and parasite genetic characteristics, virulence, epidemiological scenarios, and, mainly, diverse immune responses triggered by Leishmania species make the achievement of this aim a complex task. It is already clear that the induction of a Th1, pro-inflammatory response, is important in the protection against Leishmania infection. However, many questions must still be answered to fully understand Leishmania immunopathology, especially regarding Leishmania-specific Th1 response induction, regulation, and persistence. A large number of Leishmania antigens able to induce pro-inflammatory response have been selected so far, but none of them demonstrated efficiency in protection assays. A possible explanation is that CD4 T cells display marked heterogeneity at a single-cell level especially regarding the production of Th1-defining cytokines and multifunctionality. It has been established in the literature that Th1 cells undergo a differentiation process, which can generate cells with diverse phenotypes and survival capabilities. Despite that, only a few studies evaluate this heterogenic response and the amount of multifunctional CD4 T cells induced by Leishmania vaccine candidates, missing what can be a crucial point in defining a correlate of protection after vaccination. Moreover, most of the knowledge involving the development of cutaneous leishmaniasis (CL) vaccines comes from the mouse model of infection with Leishmania major, which cannot be fully applied to New World Leishmaniasis. For this reason, the immune response triggered by infection with New World Leishmania species, as well as vaccine candidates, need further studies. In this review, we will reinforce the importance of evaluating the quality of immune response against Leishmania, using a multiparametric analysis in order to understand better this complex host-parasite interaction, discussing the differences in the responses triggered by different New World Leishmania species, as well as the impact on the development of an effective vaccine against CL.
Collapse
Affiliation(s)
- Paula Mello De Luca
- Laboratório de Imunoparasitologia, Instituto Oswaldo Cruz, FIOCRUZ , Rio de Janeiro , Brazil
| | - Amanda Beatriz Barreto Macedo
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine , Salt Lake City, UT , USA
| |
Collapse
|
22
|
Gholami E, Zahedifard F, Rafati S. Delivery systems for Leishmania vaccine development. Expert Rev Vaccines 2016; 15:879-95. [DOI: 10.1586/14760584.2016.1157478] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Elham Gholami
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran (the Islamic Republic of)
- School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farnaz Zahedifard
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran (the Islamic Republic of)
| | - Sima Rafati
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran (the Islamic Republic of)
| |
Collapse
|
23
|
Domínguez-Bernal G, Horcajo P, Orden JA, Ruiz-Santa-Quiteria JA, De La Fuente R, Ordóñez-Gutiérrez L, Martínez-Rodrigo A, Mas A, Carrión J. HisAK70: progress towards a vaccine against different forms of leishmaniosis. Parasit Vectors 2015; 8:629. [PMID: 26653170 PMCID: PMC4675018 DOI: 10.1186/s13071-015-1246-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 12/03/2015] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Leishmania major and Leishmania infantum are among the main species that are responsible for cutaneous leishmaniosis (CL) and visceral leishmaniosis (VL), respectively. The leishmanioses represent the second-largest parasitic killer in the world after malaria. Recently, we succeeded in generating a plasmid DNA (pCMV-HISA70m2A) and demonstrated that immunized mice were protected against L. major challenge. The efficacy of the DNA-vaccine was further enhanced by the inclusion of KMP-11 antigen into the antibiotic-free plasmid pVAX1-asd. METHODS Here, we describe the use of a HisAK70 DNA-vaccine encoding seven Leishmania genes (H2A, H2B, H3, H4, A2, KMP11 and HSP70) for vaccination of mice to assess the induction of a resistant phenotype against VL and CL. RESULTS HisAK70 was successful in vaccinated mice, resulting in a high amount of efficient sterile hepatic granulomas associated with a hepatic parasite burden fully resolved in the VL model; and resulting in 100% inhibition of parasite visceralization in the CL model. CONCLUSIONS The results suggest that immunization with the HisAK70 DNA-vaccine may provide a rapid, suitable, and efficient vaccination strategy to confer cross-protective immunity against VL and CL.
Collapse
Affiliation(s)
- Gustavo Domínguez-Bernal
- INMIVET, Department of Animal Health, Faculty of Veterinary Science, Complutense University of Madrid, Madrid, 28040, Spain.
| | - Pilar Horcajo
- SALUVET, Department of Animal Health, Faculty of Veterinary Science, Complutense University of Madrid, Madrid, 28040, Spain.
| | - José A Orden
- INMIVET, Department of Animal Health, Faculty of Veterinary Science, Complutense University of Madrid, Madrid, 28040, Spain.
| | - José A Ruiz-Santa-Quiteria
- INMIVET, Department of Animal Health, Faculty of Veterinary Science, Complutense University of Madrid, Madrid, 28040, Spain.
| | - Ricardo De La Fuente
- INMIVET, Department of Animal Health, Faculty of Veterinary Science, Complutense University of Madrid, Madrid, 28040, Spain.
| | | | - Abel Martínez-Rodrigo
- INMIVET, Department of Animal Health, Faculty of Veterinary Science, Complutense University of Madrid, Madrid, 28040, Spain.
| | - Alicia Mas
- INMIVET, Department of Animal Health, Faculty of Veterinary Science, Complutense University of Madrid, Madrid, 28040, Spain.
| | - Javier Carrión
- INMIVET, Department of Animal Health, Faculty of Veterinary Science, Complutense University of Madrid, Madrid, 28040, Spain.
| |
Collapse
|
24
|
de Mendonça SCF, Cysne-Finkelstein L, Matos DCDS. Kinetoplastid Membrane Protein-11 as a Vaccine Candidate and a Virulence Factor in Leishmania. Front Immunol 2015; 6:524. [PMID: 26528287 PMCID: PMC4602152 DOI: 10.3389/fimmu.2015.00524] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 09/25/2015] [Indexed: 01/02/2023] Open
Abstract
Kinetoplastid membrane protein-11 (KMP-11), a protein present in all kinetoplastid protozoa, is considered a potential candidate for a leishmaniasis vaccine. In Leishmania amazonensis, KMP-11 is expressed in promastigotes and amastigotes. In both stages, the protein was found in association with membrane structures at the cell surface, flagellar pocket, and intracellular vesicles. More importantly, its surface expression is higher in amastigotes than in promastigotes and increases during metacyclogenesis. The increased expression of KMP-11 in metacyclic promastigotes, and especially in amastigotes, indicates a role for this molecule in the parasite relationship with the mammalian host. In this connection, we have shown that addition of KMP-11 exacerbates L. amazonensis infection in peritoneal macrophages from BALB/c mice by increasing interleukin (IL)-10 secretion and arginase activity while reducing nitric oxide production. The doses of KMP-11, the IL-10 levels, and the intracellular amastigote loads were strongly, positively, and significantly correlated. The increase in parasite load induced by KMP-11 was inhibited by anti-KMP-11 or anti-IL-10-neutralizing antibodies, but not by isotype controls. The neutralizing antibodies, but not the isotype controls, were also able to significantly decrease the parasite load in macrophages cultured without the addition of KMP-11, demonstrating that KMP-11-induced exacerbation of the infection is not dependent on the addition of exogenous KMP-11 and that the protein naturally expressed by the parasite is able to promote it. All these data indicate that KMP-11 acts as a virulence factor in L. amazonensis infection.
Collapse
Affiliation(s)
| | - Léa Cysne-Finkelstein
- Laboratório de Imunoparasitologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz , Rio de Janeiro , Brazil
| | - Denise Cristina de Souza Matos
- Laboratório de Tecnologia Imunológica, Instituto de Tecnologia em Imunobiológicos, Fundação Oswaldo Cruz , Rio de Janeiro , Brazil
| |
Collapse
|
25
|
Abstract
Replication of Trypanosoma cruzi, the etiological agent of Chagas disease, displays peculiar features, such as absence of chromosome condensation and closed mitosis. Although previous proteome and subproteome analyses of T. cruzi have been carried out, the nuclear subproteome of this protozoan has not been described. Here, we report, for the first time to the best of our knowledge, the isolation and proteome analysis of T. cruzi nuclear fraction. For that, T. cruzi epimastigote cells were lysed and subjected to cell fractionation using two steps of sucrose density gradient centrifugation. The purity of the nuclear fraction was confirmed by phase contrast and fluorescence microscopy. Liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) allowed the identification of 864 proteins. Among those, 272 proteins were annotated as putative uncharacterized, and 275 had not been previously reported on global T. cruzi proteome analysis. Additionally, to support our enrichment method, bioinformatics analysis in DAVID was carried out. It grouped the nuclear proteins in 65 gene clusters, wherein the clusters with the highest enrichment scores harbor members with chromatin organization and DNA binding functions.
Collapse
|
26
|
Jain K, Jain NK. Vaccines for visceral leishmaniasis: A review. J Immunol Methods 2015; 422:1-12. [PMID: 25858230 DOI: 10.1016/j.jim.2015.03.017] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Revised: 02/21/2015] [Accepted: 03/28/2015] [Indexed: 01/09/2023]
Abstract
Visceral leishmaniasis, which is also known as Kala-Azar, is one of the most severely neglected tropical diseases recognized by the World Health Organization (WHO). The threat of this debilitating disease continues due to unavailability of promising drug therapy or human vaccine. An extensive research is undergoing to develop a promising vaccine to prevent this devastating disease. In this review we compiled the findings of recent research with a view to facilitate knowledge on experimental vaccinology for visceral leishmaniasis. Various killed or attenuated parasite based first generation vaccines, second generation vaccines based on antigenic protein or recombinant protein, and third generation vaccines derived from antigen-encoding DNA plasmids including heterologous prime-boost Leishmania vaccine have been examined for control and prevention of visceral leishmaniasis. Vaccines based on recombinant protein and antigen-encoding DNA plasmids have given promising results and few vaccines including Leishmune®, Leishtec, and CaniLeish® have been licensed for canine visceral leishmaniasis. A systematic investigation of these vaccine candidates can lead to development of promising vaccine for human visceral leishmaniasis, most probably in the near future.
Collapse
Affiliation(s)
- Keerti Jain
- Pharmaceutical Nanotechnology Research Laboratory, ISF College of Pharmacy, Moga, Punjab 142001, India.
| | - N K Jain
- Pharmaceutical Nanotechnology Research Laboratory, ISF College of Pharmacy, Moga, Punjab 142001, India.
| |
Collapse
|
27
|
Abstract
Leishmaniasis is a neglected tropical disease spread by an arthropod vector. It remains a significant health problem with an incidence of 0.2–0.4 million visceral leishmaniasis and 0.7–1.2 million cutaneous leishmaniasis cases each year. There are limitations associated with the current therapeutic regimens for leishmaniasis and the fact that after recovery from infection the host becomes immune to subsequent infection therefore, these factors force the feasibility of a vaccine for leishmaniasis. Publication of the genome sequence of Leishmania has paved a new way to understand the pathogenesis and host immunological status therefore providing a deep insight in the field of vaccine research. This review is an effort to study the antigenic targets in Leishmania to develop an anti-leishmanial vaccine.
Collapse
|
28
|
Guha R, Das S, Ghosh J, Sundar S, Dujardin JC, Roy S. Antimony resistant Leishmania donovani but not sensitive ones drives greater frequency of potent T-regulatory cells upon interaction with human PBMCs: role of IL-10 and TGF-β in early immune response. PLoS Negl Trop Dis 2014; 8:e2995. [PMID: 25032977 PMCID: PMC4102415 DOI: 10.1371/journal.pntd.0002995] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Accepted: 05/21/2014] [Indexed: 12/16/2022] Open
Abstract
In India the sand fly, Phlebotomus argentipes, transmitted parasitic disease termed kala-azar is caused by Leishmania donovani (LD) in humans. These immune-evading parasites have increasingly developed resistance to the drug sodium antimony gluconate in endemic regions. Lack of early diagnosis methods for the disease limits the information available regarding the early interactions of this parasite with either human tissues or cell lineages. We reasoned that peripheral blood mononuclear cells (PBMCs) from healthy human beings could help compare some of their immune signatures once they were exposed for up to 8 days, to either pentavalent antimony sensitive (SbS-LD) or resistant (SbR-LD) Leishmania donovani isolates. At day 2, PBMC cultures exposed to SbS-LD and SbR-LD stationary phase promastigotes had four and seven fold higher frequency of IL-10 secreting monocyte-macrophage respectively, compared to cultures unexposed to parasites. Contrasting with the CD4+CD25−CD127− type-1 T-regulatory (Tr1) cell population that displayed similar features whatever the culture conditions, there was a pronounced increase in the IL-10 producing CD4+CD25+CD127low/− inducible T-regulatory cells (iTregs) in the PBMC cultures sampled at day 8 post addition of SbR-LD. Sorted iTregs from different cultures on day 8 were added to anti-CD3/CD28 induced naïve PBMCs to assess their suppressive ability. We observed that iTregs from SbR-LD exposed PBMCs had more pronounced suppressive ability compared to SbS-LD counterpart on a per cell basis and is dependent on both IL-10 and TGF-β, whereas IL-10 being the major factor contributing to the suppressive ability of iTregs sorted from PBMC cultures exposed to SbS–LD. Of note, iTreg population frequency value remained at the basal level after addition of genetically modified SbR-LD lacking unique terminal sugar in surface glycan. Even with limitations of this artificial in vitro model of L. donovani-human PBMC interactions, the present findings suggest that SbR-LD have higher immunomodulatory capacity which may favour aggressive pathology. The disease Kala-azar is caused by Leishmania donovani (LD). The disease is characterized by the depression of cellular immune response. In the Indian subcontinent LD parasites are mostly resistant to commonly used antileishmanial drug, like sodium antimony gluconate (SAG). It is known that infection with pentavalent antimony (Sb)-resistant parasites induces aggressive pathology- the cause is still not known. Sb-resistant parasites endowed with unique glycan which may also play an important role in the pathogenesis as following removal of terminal sugar of glycan these parasites behave like sensitive parasites. The diagnosis of the disease is possible after the disease sets in and therefore limited information is available on the host-parasite interaction at the onset of disease. As a surrogate of in vivo scenario we studied the interaction between normal human PBMC with Sb-sensitive and Sb-resistant parasites. The Sb-resistant parasites upon interaction with human peripheral blood mononuclear cells (PBMC) in vitro produced two distinct inhibitory cytokines, IL-10 and TGF-β. Similar experiment with Sb-sensitive LD induced much less amount of above cytokines. Thus aggressive pathology induced by Sb-resistant LD, may be, in part attributed to production of dual inhibitory cytokines where surface glycan of the parasite may play a decisive role.
Collapse
Affiliation(s)
- Rajan Guha
- Division of Infectious diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Shantanabha Das
- Division of Infectious diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - June Ghosh
- Division of Infectious diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Shyam Sundar
- Institute of Medical Sciences, Banaras Hindu University, Banaras, India
| | | | - Syamal Roy
- Division of Infectious diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
- * E-mail:
| |
Collapse
|
29
|
Guha R, Gupta D, Rastogi R, Vikram R, Krishnamurthy G, Bimal S, Roy S, Mukhopadhyay A. Vaccination with leishmania hemoglobin receptor-encoding DNA protects against visceral leishmaniasis. Sci Transl Med 2014; 5:202ra121. [PMID: 24027025 DOI: 10.1126/scitranslmed.3006406] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Leishmaniasis is a severe infectious disease. Drugs used for leishmaniasis are very toxic, and no vaccine is available. We found that the hemoglobin receptor (HbR) of Leishmania was conserved across various strains of Leishmania, and anti-HbR antibody could be detected in kala-azar patients' sera. Our results showed that immunization with HbR-DNA induces complete protection against virulent Leishmania donovani infection in both BALB/c mice and hamsters. Moreover, HbR-DNA immunization stimulated the production of protective cytokines like interferon-γ (IFN-γ), interleukin-12 (IL-12), and tumor necrosis factor-α (TNF-α) with concomitant down-regulation of disease-promoting cytokines like IL-10 and IL-4. HbR-DNA vaccination also induced a protective response by generating multifunctional CD4(+) and CD8(+) T cells. All HbR-DNA-vaccinated hamsters showed sterile protection and survived during an experimental period of 8 months. These findings demonstrate the potential of HbR as a vaccine candidate against visceral leishmaniasis.
Collapse
Affiliation(s)
- Rajan Guha
- Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700 032, India
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Agallou M, Athanasiou E, Koutsoni O, Dotsika E, Karagouni E. Experimental Validation of Multi-Epitope Peptides Including Promising MHC Class I- and II-Restricted Epitopes of Four Known Leishmania infantum Proteins. Front Immunol 2014; 5:268. [PMID: 24959167 PMCID: PMC4051127 DOI: 10.3389/fimmu.2014.00268] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 05/25/2014] [Indexed: 12/25/2022] Open
Abstract
Leishmaniasis is a significant worldwide health problem for which no vaccine exists. Activation of CD4(+) and CD8(+) T cells is crucial for the generation of protective immunity against parasite. Recent trend in vaccine design has been shifted to epitope-based vaccines that are more specific, safe, and easy to produce. In the present study, four known antigenic Leishmania infantum proteins, cysteine peptidase A (CPA), histone H1, KMP-11, and Leishmania eukaryotic initiation factor (LeIF) were analyzed for the prediction of binding epitopes to H2(d) MHC class I and II molecules, using online available algorithms. Based on in silico analysis, eight peptides including highly scored MHC class I- and II-restricted epitopes were synthesized. Peptide immunogenicity was validated in MHC compatible BALB/c mice immunized with each synthetic peptide emulsified in complete Freund's adjuvant/incomplete Freund's adjuvant. CPA_p2, CPA_p3, H1_p1, and LeIF_p6 induced strong spleen cell proliferation upon in vitro peptide re-stimulation. In addition, the majority of the peptides, except of LeIF_p1 and KMP-11_p1, induced IFN-γ secretion, while KMP-11_p1 indicated a suppressive effect on IL-10 production. CPA_p2, CPA_p3, LeIF_p3, and LeIF_p6 induced IFN-γ-producing CD4(+) T cells indicating a TH1-type response. In addition, CPA_p2, CPA_p3, and H1_p1 induced also the induction of CD8(+) T cells. The induction of peptide-specific IgG in immunized mice designated also the existence of B cell epitopes in peptide sequences. Combining immunoinformatic tools and experimental validation, we demonstrated that CPA_p2, CPA_p3, H1_p1, H1_p3, CPA_p2, LeIF_p3, and LeIF_p6 are likely to include potential epitopes for the induction of protective cytotoxic and/or TH1-type immune responses supporting the feasibility of peptide-based vaccine development for leishmaniasis.
Collapse
Affiliation(s)
- Maria Agallou
- Laboratory of Cellular Immunology, Department of Microbiology, Hellenic Pasteur Institute , Athens , Greece
| | - Evita Athanasiou
- Laboratory of Cellular Immunology, Department of Microbiology, Hellenic Pasteur Institute , Athens , Greece
| | - Olga Koutsoni
- Laboratory of Cellular Immunology, Department of Microbiology, Hellenic Pasteur Institute , Athens , Greece
| | - Eleni Dotsika
- Laboratory of Cellular Immunology, Department of Microbiology, Hellenic Pasteur Institute , Athens , Greece
| | - Evdokia Karagouni
- Laboratory of Cellular Immunology, Department of Microbiology, Hellenic Pasteur Institute , Athens , Greece
| |
Collapse
|
31
|
Vaccines to prevent leishmaniasis. Clin Transl Immunology 2014; 3:e13. [PMID: 25505961 PMCID: PMC4232054 DOI: 10.1038/cti.2014.4] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 02/11/2014] [Accepted: 02/12/2014] [Indexed: 12/19/2022] Open
Abstract
Leishmaniasis is a parasitic disease that encompasses a range of clinical manifestations affecting people in tropical and subtropical regions of the world. Epidemiological and experimental data indicate that protection from disease can be achieved in most people. In addition, we know how the host immune system must respond to infection in order to control parasite growth. However, there is still no vaccine for use in humans. Here, we review our understanding of host immunity following Leishmania infection and also discuss recent advances in the development of vaccines to prevent leishmaniasis, highlighting a new promising approach that targets the parasite hemoglobin receptor.
Collapse
|