1
|
Eom GD, Chu KB, Yoon KW, Mao J, Kim SS, Quan FS. Immunizing Mice with Influenza Virus-like Particles Expressing the Leishmania amazonensis Promastigote Surface Antigen Alleviates Inflammation in Footpad. Vaccines (Basel) 2024; 12:793. [PMID: 39066431 PMCID: PMC11281337 DOI: 10.3390/vaccines12070793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Cutaneous leishmaniasis (CL) is a tropical disease endemic in many parts of the world. Characteristic clinical manifestations of CL include the formation of ulcerative skin lesions that can inflict life-long disability if left untreated. Although drugs are available, they are unaffordable and out of reach for individuals who need them the most. Developing a highly cost-efficient CL vaccine could address this problem but such a vaccine remains unavailable. Here, we developed a chimeric influenza virus-like particle expressing the Leishmania amazonensis promastigote surface antigen (LaPSA-VLP). LaPSA-VLPs were self-assembled in Spodoptera frugiperda insect cell lines using the baculovirus expression system. After characterizing the vaccines and confirming successful VLP assembly, BALB/c mice were immunized with these vaccines for efficacy assessment. Sera acquired from mice upon subcutaneous immunization with the LaPSA-VLP specifically interacted with the L. amazonensis soluble total antigens. LaPSA-VLP-immunized mice elicited significantly greater quantities of parasite-specific IgG from the spleens, popliteal lymph nodes, and footpads than unimmunized mice. LaPSA-VLP immunization also enhanced the proliferation of B cell populations in the spleens of mice and significantly lessened the CL symptoms, notably the footpad swelling and IFN-γ-mediated inflammatory response. Overall, immunizing mice with the LaPSA-VLPs prevented mice from developing severe CL symptoms, signifying their developmental potential.
Collapse
Affiliation(s)
- Gi-Deok Eom
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (G.-D.E.); (K.-W.Y.); (J.M.)
| | - Ki Back Chu
- Department of Parasitology, Inje University College of Medicine, Busan 47392, Republic of Korea;
- Department of Infectious Disease and Malaria, Paik Institute of Clinical Research, Inje University, Busan 47392, Republic of Korea
| | - Keon-Woong Yoon
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (G.-D.E.); (K.-W.Y.); (J.M.)
| | - Jie Mao
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (G.-D.E.); (K.-W.Y.); (J.M.)
| | - Sung Soo Kim
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Core Research Institute (CRI), Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Fu-Shi Quan
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Core Research Institute (CRI), Kyung Hee University, Seoul 02447, Republic of Korea;
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
2
|
Costa Souza BL, Pinto EF, Bezerra IP, Gomes DC, Martinez AMB, Ré MI, de Matos Guedes HL, Rossi-Bergmann B. Crosslinked chitosan microparticles as a safe and efficient DNA carrier for intranasal vaccination against cutaneous leishmaniasis. Vaccine X 2023; 15:100403. [PMID: 38026045 PMCID: PMC10665653 DOI: 10.1016/j.jvacx.2023.100403] [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: 01/11/2022] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023] Open
Abstract
Intranasal (i.n.) vaccination with adjuvant-free plasmid DNA encoding the leishmanial antigen LACK (LACK DNA) has shown to induce protective immunity against both cutaneous and visceral leishmaniasis in rodents. In the present work, we sought to evaluate the safety and effectiveness of d,l-glyceraldehyde cross-linked chitosan microparticles (CCM) as a LACK DNA non-intumescent mucoadhesive delivery system. CCM with 5 μm of diameter was prepared and adsorbed with a maximum of 2.4 % (w/w) of DNA with no volume alteration. Histological analysis of mouse nostrils instilled with LACK DNA / CCM showed microparticles to be not only mucoadherent but also mucopenetrant, inducing no local inflammation. Systemic safeness was confirmed by the observation that two nasal instillations one week apart did not alter the numbers of bronchoalveolar cells or blood eosinophils; did not alter ALT, AST and creatinine serum levels; and did not induce cutaneous hypersensitivity. When challenged in the footpad with Leishmania amazonensis, mice developed significantly lower parasite loads as compared with animals given naked LACK DNA or CCM alone. That was accompanied by increased stimulation of Th1-biased responses, as seen by the higher T-bet / GATA-3 ratio and IFN-γ levels. Together, these results demonstrate that CCM is a safe and effective mucopenetrating carrier that can increase the efficacy of i.n. LACK DNA vaccination against cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Beatriz L.S. Costa Souza
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Eduardo F. Pinto
- Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Izabella P.S. Bezerra
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Daniel C.O. Gomes
- Núcleo de Doenças Infecciosas/Núcleo de Biotecnologia, Universidade Federal do Espírito Santo, Brazil
| | - Ana Maria B. Martinez
- Laboratório de Neurodegeneração e Reparo, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil
| | - Maria Inês Ré
- Mines Albi, UMR-CNRS 5302, Centre RAPSODEE, Université de Toulouse, Campus Jarlard, Albi, France
| | - Herbert L. de Matos Guedes
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Bartira Rossi-Bergmann
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
3
|
Gupta AK, Das S, Kamran M, Ejazi SA, Ali N. The Pathogenicity and Virulence of Leishmania - interplay of virulence factors with host defenses. Virulence 2022; 13:903-935. [PMID: 35531875 PMCID: PMC9154802 DOI: 10.1080/21505594.2022.2074130] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Leishmaniasis is a group of disease caused by the intracellular protozoan parasite of the genus Leishmania. Infection by different species of Leishmania results in various host immune responses, which usually lead to parasite clearance and may also contribute to pathogenesis and, hence, increasing the complexity of the disease. Interestingly, the parasite tends to reside within the unfriendly environment of the macrophages and has evolved various survival strategies to evade or modulate host immune defense. This can be attributed to the array of virulence factors of the vicious parasite, which target important host functioning and machineries. This review encompasses a holistic overview of leishmanial virulence factors, their role in assisting parasite-mediated evasion of host defense weaponries, and modulating epigenetic landscapes of host immune regulatory genes. Furthermore, the review also discusses the diagnostic potential of various leishmanial virulence factors and the advent of immunomodulators as futuristic antileishmanial drug therapy.
Collapse
Affiliation(s)
- Anand Kumar Gupta
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Sonali Das
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Mohd Kamran
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Sarfaraz Ahmad Ejazi
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Nahid Ali
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| |
Collapse
|
4
|
Jafarzadeh A, Kumar S, Bodhale N, Jafarzadeh S, Nemati M, Sharifi I, Sarkar A, Saha B. The expression of PD-1 and its ligands increases in Leishmania infection and its blockade reduces the parasite burden. Cytokine 2022; 153:155839. [PMID: 35276636 DOI: 10.1016/j.cyto.2022.155839] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/16/2021] [Accepted: 02/21/2022] [Indexed: 11/03/2022]
Abstract
The expression of programmed cell death protein-1 (PD-1) and its ligands- PD-L1 and PD-L2- on T cells and macrophages', respectively, increases in Leishmania infection. The PD-1/PD-L1 interaction induces T cell anergy, T cell apoptosis and exhaustion, diversion of T cells toward TH2 and T-reg cells but inhibits M1 macrophage activities by suppression of nitric oxide (NO) and reactive oxygen species (ROS) production. These changes exacerbate Leishmania infection. As PD-L1-deficient, but not PD-L2-deficient, mice were protected againstL. mexicanainfection, differential roles have been proposed for PD-L1 and PD-L2 in mouse models of leishmaniasis. Blockade of PD-1/PD-L1 interaction in various in vitro and Leishmania-infected mouse, hamster and dog models enhanced IFN-γ and NO production, reduced IL-10 and TGF-β generation, promoted T cell proliferation and reduced parasite burden. Therefore, PD-1/PD-L1 blockade is being considered as a potential therapeutic strategy to restore protective immunity during leishmaniasis, particularly, in drug-resistant cases.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Sunil Kumar
- National Centre For Cell Science, Pune 411007, India
| | | | - Sara Jafarzadeh
- Student Research Committee, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Nemati
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Haematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Iraj Sharifi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Arup Sarkar
- Trident Academy of Creative Technology, Bhubaneswar, India
| | - Bhaskar Saha
- National Centre For Cell Science, Pune 411007, India; Student Research Committee, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Trident Academy of Creative Technology, Bhubaneswar, India.
| |
Collapse
|
5
|
Oliveira-Maciel D, dos-Santos JS, Oliveira-Silva G, de Mello MF, da Fonseca-Martins AM, Carneiro MPD, Ramos TD, Firmino-Cruz L, Gomes DCO, Rossi-Bergmann B, de Matos Guedes HL. MPLA and AddaVax ® Adjuvants Fail to Promote Intramuscular LaAg Vaccine Protectiveness against Experimental Cutaneous Leishmaniasis. Microorganisms 2021; 9:microorganisms9061272. [PMID: 34207948 PMCID: PMC8230739 DOI: 10.3390/microorganisms9061272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/04/2021] [Indexed: 11/23/2022] Open
Abstract
There is so far no vaccine approved for human leishmaniasis, mainly because of the lack of appropriate adjuvants. This study aimed to evaluate in mice the capacity of a mixture of monophosphoryl lipid A (MPLA) and AddaVax® adjuvants in enhancing the efficacy of a Leishvacin®-like vaccine comprised of Leishmania amazonensis whole antigens (LaAg). For that, mice were immunized with LaAg plus MPLA/AddaVax® by the intramuscular route (i.m.) prior to challenge with 2 × 105 and 2 × 106 living parasites. Immunization with LaAg alone reduced the lesion growth of the 2 × 105-challenged mice only in the peak of infection, but that was not accompanied by reduced parasite load, and thus not considered protective. Mice given a 2 × 106 -challenge were not protected by LaAg. The association of LaAg with MPLA/AddaVax® was able to enhance the cutaneous hypersensitivity response compared with LaAg alone. Despite this, there was no difference in proliferative cell response to antigen ex vivo. Moreover, regardless of the parasite challenge, association of LaAg with MPL/AddaVax® did not significantly enhance protection in comparison with LaAg alone. This work demonstrated that MPL/AddaVax® is not effective in improving the efficacy of i.m. LaAg vaccine against cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Diogo Oliveira-Maciel
- Laboratório de Imunofarmacologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.O.-M.); (J.S.d.-S.); (G.O.-S.); (A.M.d.F.-M.); (M.P.D.C.); (T.D.R.); (L.F.-C.); (B.R.-B.)
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil
- Grupo de Imunologia e Vacinologia, Instituto de Microbiologia Paulo de Goés, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Júlio Souza dos-Santos
- Laboratório de Imunofarmacologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.O.-M.); (J.S.d.-S.); (G.O.-S.); (A.M.d.F.-M.); (M.P.D.C.); (T.D.R.); (L.F.-C.); (B.R.-B.)
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil
- Grupo de Imunologia e Vacinologia, Instituto de Microbiologia Paulo de Goés, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Gabriel Oliveira-Silva
- Laboratório de Imunofarmacologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.O.-M.); (J.S.d.-S.); (G.O.-S.); (A.M.d.F.-M.); (M.P.D.C.); (T.D.R.); (L.F.-C.); (B.R.-B.)
- Grupo de Imunologia e Vacinologia, Instituto de Microbiologia Paulo de Goés, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Mirian França de Mello
- Grupo de Imunologia e Vacinologia, Instituto de Microbiologia Paulo de Goés, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Alessandra Marcia da Fonseca-Martins
- Laboratório de Imunofarmacologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.O.-M.); (J.S.d.-S.); (G.O.-S.); (A.M.d.F.-M.); (M.P.D.C.); (T.D.R.); (L.F.-C.); (B.R.-B.)
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil
- Grupo de Imunologia e Vacinologia, Instituto de Microbiologia Paulo de Goés, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Monique Pacheco Duarte Carneiro
- Laboratório de Imunofarmacologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.O.-M.); (J.S.d.-S.); (G.O.-S.); (A.M.d.F.-M.); (M.P.D.C.); (T.D.R.); (L.F.-C.); (B.R.-B.)
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil
- Grupo de Imunologia e Vacinologia, Instituto de Microbiologia Paulo de Goés, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Tadeu Diniz Ramos
- Laboratório de Imunofarmacologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.O.-M.); (J.S.d.-S.); (G.O.-S.); (A.M.d.F.-M.); (M.P.D.C.); (T.D.R.); (L.F.-C.); (B.R.-B.)
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil
- Grupo de Imunologia e Vacinologia, Instituto de Microbiologia Paulo de Goés, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Luan Firmino-Cruz
- Laboratório de Imunofarmacologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.O.-M.); (J.S.d.-S.); (G.O.-S.); (A.M.d.F.-M.); (M.P.D.C.); (T.D.R.); (L.F.-C.); (B.R.-B.)
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil
- Grupo de Imunologia e Vacinologia, Instituto de Microbiologia Paulo de Goés, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Daniel Claudio Oliveira Gomes
- Núcleo de Doenças Infecciosas/Núcleo de Biotecnologia-Universidade Federal do Espírito Santo, Vitória 29075-910, Brazil;
| | - Bartira Rossi-Bergmann
- Laboratório de Imunofarmacologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.O.-M.); (J.S.d.-S.); (G.O.-S.); (A.M.d.F.-M.); (M.P.D.C.); (T.D.R.); (L.F.-C.); (B.R.-B.)
| | - Herbert Leonel de Matos Guedes
- Laboratório de Imunofarmacologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.O.-M.); (J.S.d.-S.); (G.O.-S.); (A.M.d.F.-M.); (M.P.D.C.); (T.D.R.); (L.F.-C.); (B.R.-B.)
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil
- Grupo de Imunologia e Vacinologia, Instituto de Microbiologia Paulo de Goés, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
- Correspondence: or or ; Tel.: +55-213-98-6571; Fax: +55-212-280-8193
| |
Collapse
|
6
|
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
|
7
|
da Fonseca-Martins AM, Ramos TD, Pratti JES, Firmino-Cruz L, Gomes DCO, Soong L, Saraiva EM, de Matos Guedes HL. Immunotherapy using anti-PD-1 and anti-PD-L1 in Leishmania amazonensis-infected BALB/c mice reduce parasite load. Sci Rep 2019; 9:20275. [PMID: 31889072 PMCID: PMC6937231 DOI: 10.1038/s41598-019-56336-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 12/04/2019] [Indexed: 01/07/2023] Open
Abstract
Leishmaniasis is a neglected disease, for which current treatment presents numerous issues. Leishmania amazonensis is the etiological agent of cutaneous and diffuse cutaneous leishmaniasis. The roles of the programmed death-1 (PD-1) receptor on lymphocytes and its ligand (PD-L1) on antigen-presenting cells have been well studied in tumor and other infection models; but little is known about their roles in non-healing cutaneous leishmaniasis. In this study, we observed that L. amazonensis induced PD-1 expression on both CD4+ and CD8+ T cells and PD-L1 on dendritic cells on BALB/c mice. We tested the therapeutic potential of anti-PD-1 and anti-PD-L1 monoclonal antibodies (MoAbs) against a non-healing L. amazonensis infection in BALB/c mice, and that anti-PD-1 and anti-PD-L1 treatment significantly increased IFN-γ-producing CD4+ and CD8+ T cells, respectively. Compared with infection controls, mice treated with anti-PD-1 and anti-PD-L1, but not anti-PD-L2, displayed bigger lesions with significantly lower parasite loads. Treatment did not affect anti-Leishmania antibody (IgM, IgG, IgG1 and IgG2a) or IL-10 production, but anti-PD-1 treatment reduced both IL-4 and TGF-β production. Together, our results highlight the therapeutic potential of an anti-PD-1-based treatment in promoting the reinvigoration of T cells for the control of parasite burden.
Collapse
Affiliation(s)
- Alessandra M da Fonseca-Martins
- Instituto de Biofísica Carlos Chagas Filho, Laboratório de Imunofarmacologia, Grupo de Imunologia e Vacinologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.,Paulo de Góes Microbiology Institute, Immunology Department, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Tadeu D Ramos
- Instituto de Biofísica Carlos Chagas Filho, Laboratório de Imunofarmacologia, Grupo de Imunologia e Vacinologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Juliana E S Pratti
- Instituto de Biofísica Carlos Chagas Filho, Laboratório de Imunofarmacologia, Grupo de Imunologia e Vacinologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Luan Firmino-Cruz
- Instituto de Biofísica Carlos Chagas Filho, Laboratório de Imunofarmacologia, Grupo de Imunologia e Vacinologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Lynn Soong
- Department of Microbiology and Immunology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Elvira M Saraiva
- Paulo de Góes Microbiology Institute, Immunology Department, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Herbert L de Matos Guedes
- Instituto de Biofísica Carlos Chagas Filho, Laboratório de Imunofarmacologia, Grupo de Imunologia e Vacinologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil. .,Universidade Federal do Rio de Janeiro - Campus Duque de Caxias Professor Geraldo Cidade, Rio de Janeiro, RJ, Brazil. .,Laboratório Interdisciplinar de pesquisas Médicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.
| |
Collapse
|
8
|
Bezerra IPS, Costa-Souza BLS, Carneiro G, Ferreira LAM, de Matos Guedes HL, Rossi-Bergmann B. Nanoencapsulated retinoic acid as a safe tolerogenic adjuvant for intranasal vaccination against cutaneous leishmaniasis. Vaccine 2019; 37:3660-3667. [PMID: 31133469 DOI: 10.1016/j.vaccine.2019.05.043] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 04/18/2019] [Accepted: 05/13/2019] [Indexed: 12/15/2022]
Abstract
Mucosal, but not peripheral, vaccination with whole Leishmania amazonensis antigen (LaAg) effectively protects mice against leishmaniasis, likely through a tolerogenic mechanism. Given the crucial role of retinoic acid (RA) in CD4+ Foxp3+ regulatory T cell (Treg) differentiation and mucosal tolerance, here we evaluated the capacity of RA to improve intranasal (i.n.) vaccination with LaAg. To prevent degradation and possible mucosa irritation, RA was encapsulated in solid lipid nanoparticles (RA-SLN). Thus, BALB/c mice were given two i.n. doses of LaAg alone or in association with RA-SLN (LaAg/RA-SLN) prior to challenge with L. amazonensis. No histological sign of irritation or inflammation was produced in the nasal mucosa after RA-SLN administration. LaAg/RA-SLN vaccine was more effective in delaying lesion growth and reducing parasite burdens than LaAg alone (96% and 61% reduction, respectively). At two months after challenge, both vaccinated groups displayed similar T helper (Th) 1-skewed in situ cytokine responses, different from early infection where both Th1 and Th2 responses were suppressed, except for transforming growth factor (TGF)-β mRNA, that was higher in mice given RA-SLN. At the mucosa, RA-SLN promoted enhanced expression of interleukin (IL)-10 and CD4+ Foxp3+ Treg population. In sum, these data show that RA-SLN is an effective and safe tolerogenic adjuvant for i.n. vaccination against leishmaniasis.
Collapse
Affiliation(s)
- Izabella P S Bezerra
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Beatriz L S Costa-Souza
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Guilherme Carneiro
- Departamento de Farmácia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil
| | | | | | - Bartira Rossi-Bergmann
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
9
|
Abstract
The investigation of the glycan repertoire of several organisms has revealed a wide variation in terms of structures and abundance of glycan moieties. Among the parasites, it is possible to observe different sets of glycoconjugates across taxa and developmental stages within a species. The presence of distinct glycoconjugates throughout the life cycle of a parasite could relate to the ability of that organism to adapt and survive in different hosts and environments. Carbohydrates on the surface, and in excretory-secretory products of parasites, play essential roles in host-parasite interactions. Carbohydrate portions of complex molecules of parasites stimulate and modulate host immune responses, mainly through interactions with specific receptors on the surface of dendritic cells, leading to the generation of a pattern of response that may benefit parasite survival. Available data reviewed here also show the frequent aspect of parasite immunomodulation of mammalian responses through specific glycan interactions, which ultimately makes these molecules promising in the fields of diagnostics and vaccinology.
Collapse
|
10
|
Pratti JES, da Fonseca Martins AM, da Silva JP, Ramos TD, Pereira JC, Firmino-Cruz L, Oliveira-Maciel D, Vieira TSDS, Lacerda LL, Vale AM, Freire-de-Lima CG, Gomes DCO, Saraiva EM, Rossi-Bergmann B, de Matos Guedes HL. The role of TLR9 on Leishmania amazonensis infection and its influence on intranasal LaAg vaccine efficacy. PLoS Negl Trop Dis 2019; 13:e0007146. [PMID: 30802247 PMCID: PMC6405171 DOI: 10.1371/journal.pntd.0007146] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 03/07/2019] [Accepted: 01/08/2019] [Indexed: 12/13/2022] Open
Abstract
Leishmania (L.) amazonensis is one of the etiological agents of cutaneous leishmaniasis (CL) in Brazil. Currently, there is no vaccine approved for human use against leishmaniasis, although several vaccine preparations are in experimental stages. One of them is Leishvacin, or LaAg, a first-generation vaccine composed of total L. amazonensis antigens that has consistently shown an increase of mouse resistance against CL when administered intranasally (i.n.). Since Toll-like receptor 9 (TLR9) is highly expressed in the nasal mucosa and LaAg is composed of TLR9-binding DNA CpG motifs, in this study we proposed to investigate the role of TLR9 in both L. amazonensis infection and in LaAg vaccine efficacy in C57BL/6 (WT) mice and TLR9-/- mice. First, we evaluated, the infection of macrophages by L. amazonensis in vitro, showing no significant difference between macrophages from WT and TLR9-/- mice in terms of both infection percentage and total number of intracellular amastigotes, as well as NO production. In addition, neutrophils from WT and TLR9-/- mice had similar capacity to produce neutrophil extracellular traps (NETs) in response to L. amazonensis. L. amazonensis did not activate dendritic cells from WT and TLR9-/- mice, analysed by MHCII and CD86 expression. However, in vivo, TLR9-/- mice were slightly more susceptible to L. amazonensis infection than WT mice, presenting a larger lesion and an increased parasite load at the peak of infection and in the chronic phase. The increased TLR9-/- mice susceptibility was accompanied by an increased IgG and IgG1 production; a decrease of IFN-γ in infected tissue, but not IL-4 and IL-10; and a decreased number of IFN-γ producing CD8+ T cells, but not CD4+ T cells in the lesion-draining lymph nodes. Also, TLR9-/- mice could not control parasite growth following i.n. LaAg vaccination unlike the WT mice. This protection failure was associated with a reduction of the hypersensitivity response induced by immunization. The TLR9-/- vaccinated mice failed to respond to antigen stimulation and to produce IFN-γ by lymph node cells. Together, these results suggest that TLR9 contributes to C57BL/6 mouse resistance against L. amazonensis, and that the TLR9-binding LaAg comprising CpG motifs may be important for intranasal vaccine efficacy against CL.
Collapse
Affiliation(s)
| | - Alessandra Marcia da Fonseca Martins
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Department of Immunology, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Juliana Paiva da Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Tadeu Diniz Ramos
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Joyce Carvalho Pereira
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Luan Firmino-Cruz
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Diogo Oliveira-Maciel
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Thiago Soares de Souza Vieira
- Department of Immunology, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Leandra Linhares Lacerda
- Department of Immunology, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Andre Macedo Vale
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Celio G. Freire-de-Lima
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Daniel C. Oliveira Gomes
- Laboratório de Imunobiologia, Núcleo de Doenças Infecciosas/ Núcleo de Biotecnologia, Universidade Federal do Espírito Santo, ES, Brazil
| | - Elvira M. Saraiva
- Department of Immunology, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Bartira Rossi-Bergmann
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Herbert Leonel de Matos Guedes
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Núcleo Multidisciplinar de Pesquisa UFRJ–Xerém em Biologia (NUMPEX-BIO), Campus Duque de Caxias Professor Geraldo Cidade (Polo Avançado de Xerém), Universidade Federal do Rio de Janeiro, Duque de Caxias, RJ, Brazil
| |
Collapse
|
11
|
dos Santos IB, da Silva DAM, Paz FACR, Garcia DM, Carmona AK, Teixeira D, Longo-Maugéri IM, Katz S, Barbiéri CL. Leishmanicidal and Immunomodulatory Activities of the Palladacycle Complex DPPE 1.1, a Potential Candidate for Treatment of Cutaneous Leishmaniasis. Front Microbiol 2018; 9:1427. [PMID: 30018604 PMCID: PMC6038773 DOI: 10.3389/fmicb.2018.01427] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 06/11/2018] [Indexed: 01/18/2023] Open
Abstract
The present study focused on the activity of the palladacycle complex DPPE 1.1 on Leishmania (Leishmania) amazonensis. Promastigotes of L. (L.) amazonensis were destroyed in vitro by nanomolar concentrations of DPPE 1.1, whereas intracellular amastigotes were killed at drug concentrations fivefold less toxic than those harmful to macrophages. L. (L.) amazonensis-infected BALB/c mice were treated by intralesional injection of DPPE 1.1. Animals treated with 3.5 and 7.0 mg/kg of DPPE 1.1 showed a significant decrease of foot lesion sizes and a parasite load reduction of 93 and 99%, respectively, when compared to untreated controls. Furthermore, DPPE 1.1 was non-toxic to treated animals. The cathepsin B activity of L. (L.) amazonensis amastigotes was inhibited by DPPE 1.1 as demonstrated spectrofluorometrically by use of a specific fluorogenic substrate. Analysis of T-cells populations in mice treated with DPPE 1.1 and untreated controls was performed by fluorescence-activated cell sorter (FACS). IFN-γ was measured in supernatants of lymphocytes from popliteal and inguinal lymph nodes isolated from treated and untreated mice and stimulated with L. (L.) amazonensis amastigotes extract and active TGF-β was evaluated in supernatants of foot lesions; both dosages were carried out by means of a double-sandwich ELISA assay. A significant increase of TCD4+ and TCD8+ lymphocytes and IFN-γ secretion was displayed in mice treated with DPPE 1.1 compared to untreated animals, whereas a significant reduction of active TGF-β was observed in treated mice. These findings open perspectives for further investment in DPPE 1.1 as an alternative option for the chemotherapy of cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Isabela B. dos Santos
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Danielle A. M. da Silva
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Fabiana A. C. R. Paz
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Daniel M. Garcia
- Departamento de Farmacologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Adriana K. Carmona
- Departamento de Biofísica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Daniela Teixeira
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ieda M. Longo-Maugéri
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Simone Katz
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Clara L. Barbiéri
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
12
|
Pereira Silva Bezerra I, Amaral Abib M, Rossi-Bergmann B. Intranasal but not subcutaneous vaccination with LaAg allows rapid expansion of protective immunity against cutaneous leishmaniasis. Vaccine 2018; 36:2480-2486. [DOI: 10.1016/j.vaccine.2018.03.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 02/14/2018] [Accepted: 03/08/2018] [Indexed: 12/22/2022]
|
13
|
Caffeic acid combined with autoclaved Leishmania major boosted the protection of infected BALB/c mice by enhancing IgG2 production, IFN-γ/TGF-β and iNO synthase/arginase1 ratios, and the death of infected phagocytes. Inflammopharmacology 2017; 26:621-634. [DOI: 10.1007/s10787-017-0399-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 09/12/2017] [Indexed: 12/19/2022]
|
14
|
Paladi CS, da Silva DAM, Motta PD, Garcia DM, Teixeira D, Longo-Maugéri IM, Katz S, Barbiéri CL. Treatment of Leishmania (Leishmania) Amazonensis-Infected Mice with a Combination of a Palladacycle Complex and Heat-Killed Propionibacterium acnes Triggers Protective Cellular Immune Responses. Front Microbiol 2017; 8:333. [PMID: 28321209 PMCID: PMC5337482 DOI: 10.3389/fmicb.2017.00333] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 02/17/2017] [Indexed: 12/12/2022] Open
Abstract
Palladacycle complex DPPE 1.2 was previously reported to inhibit the in vitro and in vivo infection by Leishmania (Leishmania) amazonensis. The aim of the present study was to compare the effect of DPPE 1.2, in association with heat-killed Propionibacterium acnes, on L. (L.) amazonensis infection in two mouse strains, BALB/c and C57BL/6, and to evaluate the immune responses of the treated animals. Foot lesions of L. (L.) amazonensis-infected mice were injected with DPPE 1.2 alone, or associated with P. acnes as an adjuvant. Analysis of T-cell populations in the treated mice and in untreated controls was performed by FACS. Detection of IFN-γ-secreting lymphocytes was carried out by an ELISPOT assay and active TGF-β was measured by means of a double-sandwich ELISA test. The treatment with DPPE 1.2 resulted in a significant reduction of foot lesion sizes and parasite burdens in both mouse strains, and the lowest parasite burden was found in mice treated with DPPE 1.2 plus P. acnes. Mice treated with DPPE 1.2 alone displayed a significant increase of TCD4+ and TCD8+ lymphocytes and IFN-γ secretion which were significantly higher in animals treated with DPPE 1.2 plus P. acnes. A significant reduction of active TGF-β was observed in mice treated with DPPE 1.2 alone or associated with P. acnes. Moreover, DPPE 1.2 associated to P. acnes was non-toxic to treated animals. The destruction of L. (L.) amazonensis by DPPE 1.2 was followed by host inflammatory responses which were exacerbated when the palladacycle complex was associated with P. acnes.
Collapse
Affiliation(s)
- Carolina S Paladi
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo São Paulo, Brazil
| | - Danielle A M da Silva
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo São Paulo, Brazil
| | - Priscila D Motta
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo São Paulo, Brazil
| | - Daniel M Garcia
- Departamento de Farmacologia, Escola Paulista de Medicina, Universidade Federal de São Paulo São Paulo, Brazil
| | - Daniela Teixeira
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo São Paulo, Brazil
| | - Ieda M Longo-Maugéri
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo São Paulo, Brazil
| | - Simone Katz
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo São Paulo, Brazil
| | - Clara L Barbiéri
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo São Paulo, Brazil
| |
Collapse
|
15
|
Pratti JES, Ramos TD, Pereira JC, da Fonseca-Martins AM, Maciel-Oliveira D, Oliveira-Silva G, de Mello MF, Chaves SP, Gomes DCO, Diaz BL, Rossi-Bergmann B, de Matos Guedes HL. Efficacy of intranasal LaAg vaccine against Leishmania amazonensis infection in partially resistant C57Bl/6 mice. Parasit Vectors 2016; 9:534. [PMID: 27716449 PMCID: PMC5052793 DOI: 10.1186/s13071-016-1822-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 09/29/2016] [Indexed: 12/12/2022] Open
Abstract
Background We have previously demonstrated that intranasal vaccination of highly susceptible BALB/c mice with whole Leishmania amazonensis antigens (LaAg) leads to protection against murine cutaneous leishmaniasis. Here, we evaluate the response of partially resistant C57BL/6 mice to vaccination as a more representative experimental model of human cutaneous leishmaniasis. Methods C57BL/6 mice from different animal facilities were infected with L. amazonensis (Josefa strain) to establish the profile of infection. Intranasal vaccination was performed before the infection challenge with two doses of 10 μg of LaAg alone or associated with the adjuvant ADDAVAX® by instillation in the nostrils. The lesion progression was measured with a dial caliper and the parasite load by limited dilution assay in the acute and chronic phases of infection. Cytokines were quantified by ELISA in the homogenates of infected footpads. Results C57BL/6 mice from different animal facilities presented the same L. amazonensis infection profile, displaying a progressive acute phase followed by a controlled chronic phase. Parasites cultured in M199 and Schneider’s media were equally infective. Intranasal vaccination with LaAg led to milder acute and chronic phases of the disease. The mechanism of protection was associated with increased production of IFN-gamma in the infected tissue as measured in the acute phase. Association with the ADDAVAX® adjuvant did not improve the efficacy of intranasal LaAg vaccination. Rather, ADDAVAX® reduced vaccination efficacy. Conclusion This study demonstrates that the efficacy of adjuvant-free intranasal vaccination with LaAg is extendable to the more resistant C57Bl/6 mouse model of infection with L. amazonensis, and is thus not exclusive to the susceptible BALB/c model. These results imply that mucosal immunomodulation by LaAg leads to peripheral protection irrespective of the genetic background of the host. Electronic supplementary material The online version of this article (doi:10.1186/s13071-016-1822-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Juliana Elena Silveira Pratti
- Laboratório de Inflamação, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Tadeu Diniz Ramos
- Laboratório de Inflamação, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Joyce Carvalho Pereira
- Laboratório de Inflamação, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Diogo Maciel-Oliveira
- Laboratório de Inflamação, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Gabriel Oliveira-Silva
- Laboratório de Inflamação, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Mirian França de Mello
- Laboratório de Inflamação, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Suzana Passos Chaves
- Laboratório Integrado de Imunoparasitologia, Campus Macaé-Universidade Federal do Rio de Janeiro, Macaé, Brazil
| | - Daniel Claudio Oliveira Gomes
- Laboratório de Imunobiologia, Núcleo de Doenças Infecciosas/Núcleo de Biotecnologia, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | - Bruno Lourenço Diaz
- Laboratório de Inflamação, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Bartira Rossi-Bergmann
- Laboratório de Imunofarmacologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Herbert Leonel de Matos Guedes
- Laboratório de Inflamação, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil. .,Núcleo Multidisciplinar de Pesquisa UFRJ-Xerém em Biologia (NUMPEX-BIO), Polo Avançado de Xerém-Universidade Federal do Rio de Janeiro, Duque de Caxias, Rio de Janeiro, Brazil.
| |
Collapse
|
16
|
Almeida-Souza F, Cardoso FDO, Souza BVDC, do Valle TZ, de Sá JC, Oliveira IDSDS, de Souza CDSF, Moragas Tellis CJ, Chagas MDSDS, Behrens MD, Abreu-Silva AL, Calabrese KDS. Morinda citrifolia Linn. Reduces Parasite Load and Modulates Cytokines and Extracellular Matrix Proteins in C57BL/6 Mice Infected with Leishmania (Leishmania) amazonensis. PLoS Negl Trop Dis 2016; 10:e0004900. [PMID: 27579922 PMCID: PMC5006983 DOI: 10.1371/journal.pntd.0004900] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 07/13/2016] [Indexed: 12/12/2022] Open
Abstract
The absence of an effective vaccine and the debilitating chemotherapy for Leishmaniasis demonstrate the need for developing alternative treatments. Several studies conducted with Morinda citrifolia have shown various biological activities, including antileishmanial activity, however its mechanisms of action are unknown. This study aimed to analyze the in vivo activity of M. citrifolia fruit juice (Noni) against Leishmania (Leishmania) amazonensis in C57BL/6 mice. M. citrifolia fruit juice from the Brazilian Amazon has shown the same constitution of other juices produced around the world and liquid chromatography–mass spectrometry analysis identified five compounds: deacetylasperulosidic acid, asperulosidic acid, rutin, nonioside B and nonioside C. Daily intragastric treatment with Noni was carried out after 55 days of L. (L.) amazonensis infection in C57BL/6 mice. Parasitic loads, cytokine and extracellular protein matrix expressions of the lesion site were analyzed by qPCR. Histopathology of the lesion site, lymph nodes and liver were performed to evaluate the inflammatory processes. Cytokines and biochemical parameters of toxicity from sera were also evaluated. The Noni treatment at 500 mg.kg-1.day-1 for 60 days decreased the lesion size and parasitic load in the footpad infected with L. (L.) amazonensis. The site of infection also showed decreased inflammatory infiltrates and decreased cytokine expressions for IL-12, TNF-α, TGF-β and IL-10. On the other hand, Noni treatment enhanced the extracellular matrix protein expressions of collagen IV, fibronectin and laminin in the infected footpad as well collagen I and II, fibronectin and laminin in the mock-infected footpads. No toxicity was observed at the end of treatment. These data show the efficacy of Noni treatment. Leishmaniasis is a complex of diseases caused by parasites of the Leishmania genus, which affects thousands of people around the world. The parasite lives within the cells and the disease manifests itself in different ways, one of them is wound-like lesions on the skin that do not heal. The treatment, with a medicament discovered in 1912, causes several side effects, its intramuscular administration is painful and it is given daily over a long period of time. These characteristics show the need for new alternatives for leishmaniasis treatment justifying works like this. The Morinda citrifolia is a plant native to the Polynesian islands and has a fruit commonly known as Noni. Noni has been analyzed for various targets such as anticancer, anti-inflammatory and antimicrobial effects; however, the antileishmanial has not yet been fully evaluated. This work proves that Noni treatment can promote a fast clinical cure in mice with leishmaniasis by decreasing parasite number, acting on the immune system and repairing skin components.
Collapse
Affiliation(s)
- Fernando Almeida-Souza
- Laboratório de Imunomodulação e Protozoologia, Instituto Oswaldo Cruz-Fiocruz, Rio de Janeiro, Brazil
- Departamento de Patologia, Universidade Estadual do Maranhão, São Luís, Brazil
| | | | | | - Tânia Zaverucha do Valle
- Laboratório de Imunomodulação e Protozoologia, Instituto Oswaldo Cruz-Fiocruz, Rio de Janeiro, Brazil
| | - Joicy Cortez de Sá
- Departamento de Patologia, Universidade Estadual do Maranhão, São Luís, Brazil
| | | | | | | | | | - Maria Dutra Behrens
- Departamento de Produtos Naturais, Farmanguinhos-Fiocruz, Rio de Janeiro, Brazil
| | - Ana Lúcia Abreu-Silva
- Departamento de Patologia, Universidade Estadual do Maranhão, São Luís, Brazil
- * E-mail:
| | - Kátia da Silva Calabrese
- Laboratório de Imunomodulação e Protozoologia, Instituto Oswaldo Cruz-Fiocruz, Rio de Janeiro, Brazil
| |
Collapse
|
17
|
Intranasal vaccination with killed Leishmania amazonensis promastigotes antigen (LaAg) associated with CAF01 adjuvant induces partial protection in BALB/c mice challenged with Leishmania (infantum) chagasi. Parasitology 2015; 142:1640-6. [DOI: 10.1017/s0031182015001250] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
SUMMARYThe CAF01 adjuvant has previously been shown to be safe for human use and to be a potent adjuvant for several vaccine antigens. In the present work, we sought to optimize the Leishmania amazonensis antigens (LaAg) intranasal vaccine in an attempt to enhance the protective immune responses against Leishmania (infantum) chagasi by using the CAF01 association. LaAg/CAF01 vaccinated mice that were challenged 15 days after booster dose with L. (infantum) chagasi showed a significant reduction in their parasite burden in both the spleen and liver, which is associated with an increase in specific production of IFN-γ and nitrite, and a decrease in IL-4 production. In addition, LaAg/CAF01 intranasal delivery was able to increase lymphoproliferative immune responses after parasite antigen recall. These results suggest the feasibility of using the intranasal route for the delivery of crude antigens and of a human-compatible adjuvant against visceral leishmaniasis.
Collapse
|
18
|
Forestier CL, Gao Q, Boons GJ. Leishmania lipophosphoglycan: how to establish structure-activity relationships for this highly complex and multifunctional glycoconjugate? Front Cell Infect Microbiol 2015; 4:193. [PMID: 25653924 PMCID: PMC4301024 DOI: 10.3389/fcimb.2014.00193] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 12/22/2014] [Indexed: 01/09/2023] Open
Abstract
A key feature of many pathogenic microorganisms is the presence of a dense glycocalyx at their surface, composed of lipid-anchored glycoproteins and non-protein-bound polysaccharides. These surface glycolipids are important virulence factors for bacterial, fungal and protozoan pathogens. The highly complex glycoconjugate lipophosphoglycan (LPG) is one of the dominant surface macromolecules of the promastigote stage of all Leishmania parasitic species. LPG plays critical pleiotropic roles in parasite survival and infectivity in both the sandfly vector and the mammalian host. Here, we review the composition of the Leishmania glycocalyx, the chemical structure of LPG and what is currently known about its effects in the mammalian host, specifically. We will then discuss the current approaches employed to elucidate LPG functions. Finally, we will provide a viewpoint on future directions that this area of investigation could take to unravel in detail the biological activity of the specific molecular elements composing the structurally complex LPG.
Collapse
Affiliation(s)
| | - Qi Gao
- Complex Carbohydrate Research Center, Department of Chemistry, University of Georgia Athens, GA, USA
| | - Geert-Jan Boons
- Complex Carbohydrate Research Center, Department of Chemistry, University of Georgia Athens, GA, USA
| |
Collapse
|
19
|
Intranasal vaccination with leishmanial antigens protects golden hamsters (Mesocricetus auratus) against Leishmania (Viannia) Braziliensis infection. PLoS Negl Trop Dis 2015; 9:e3439. [PMID: 25569338 PMCID: PMC4287559 DOI: 10.1371/journal.pntd.0003439] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 11/24/2014] [Indexed: 11/19/2022] Open
Abstract
Background Previous results have shown that oral and intranasal administration of particulate Leishmania (Leishmania) amazonensis antigens (LaAg) partially protects mice against L. amazonensis infection. However, vaccination studies on species of the subgenus Viannia, the main causative agent of cutaneous and mucosal leishmaniasis in the Americas, have been hampered by the lack of easy-to-handle bio-models that accurately mimic the human disease. Recently, we demonstrated that the golden hamster is an appropriate model for studying the immunopathogenesis of cutaneous leishmaniasis caused by L. (Viannia) braziliensis. Using the golden hamster model, our current study investigated whether the protective effect of intranasal immunisation with LaAg can be extended to L. braziliensis infection. Methodology/Principal Findings Golden hamsters vaccinated with either two intranasal (IN) doses of LaAg (10 µg) or two intramuscular doses of LaAg (20 µg) were challenged 2 weeks post-vaccination with L. braziliensis. The results showed that IN immunisation with LaAg significantly reduced lesion growth and parasitic load as well as serum IgG and IgG2 levels. At the experimental endpoint on day 114 post-infection, IN-immunised hamsters that were considered protected expressed IFN-γ and IL10 mRNA levels that returned to uninfected skin levels. In contrast to the nasal route, intramuscular (IM) immunisation failed to provide protection. Conclusions/Significance These results demonstrate for the first time that the nasal route of immunisation can induce cross protection against L. braziliensis infection. Leishmaniasis is a disease that is common in most tropical countries. In Brazil, the cutaneous form of the disease is highly prevalent, with approximately 28,000 new cases reported annually. L. (Viannia) braziliensis is the main causative agent of cutaneous leishmaniasis; however, vaccine studies against protozoans of the subgenus Viannia have been largely neglected, mainly due to the high resistance of most mouse strains to the infection. Here, the authors used the golden hamster, which is highly susceptible to dermotropic Leishmania spp infection. It was previously shown that oral and intranasal vaccination with whole L. (Leishmania) amazonensis antigens (LaAg) protected mice against L. amazonensis infection. In the present study, the authors investigated whether the protective effect of intranasal immunisation with LaAg can be extended to L. braziliensis infection using the golden hamster model. The results showed that intranasal immunisation with LaAg significantly reduced lesion growth and parasitic load as well as IgG and IgG2 serum levels. At the endpoint of the experiment, intranasally immunised hamsters that were considered protected expressed IFN-γ and IL10 mRNA at levels similar to those in uninfected skin. These data show that the use of a proper animal model and/or different vaccination strategies may facilitate the development of an effective vaccine against L. braziliensis.
Collapse
|
20
|
de Matos Guedes HL, da Silva Costa BL, Chaves SP, de Oliveira Gomes DC, Nosanchuk JD, De Simone SG, Rossi-Bergmann B. Intranasal vaccination with extracellular serine proteases of Leishmania amazonensis confers protective immunity to BALB/c mice against infection. Parasit Vectors 2014; 7:448. [PMID: 25239157 PMCID: PMC4261548 DOI: 10.1186/1756-3305-7-448] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 09/01/2014] [Indexed: 12/12/2022] Open
Abstract
Background Previously, we demonstrated that unlike subcutaneous or intramuscular vaccination, intranasal vaccination of BALB/c mice with whole Leishmania amazonensis antigens leads to protection against cutaneous leishmaniasis. Here, the role of parasite serine proteases in the protective immunity was investigated. Findings Serine Proteases were partially purified from both soluble (LaSP-Sol) and extracellular (LaSP-Ex) Leishmania amazonensis promastigote extracts by aprotinin-agarose chromatography. BALB/c mice were intranasally immunized with LaSP-Sol and LaSP-Ex prior to infection with L. amazonensis. LaSP-Ex but not LaSP-Sol vaccination led to significantly smaller lesions and parasite burdens as compared with non-vaccinated controls. Protection was accompanied by systemic Th1 polarization with increased IFN-γ and decreased IL-4 and IL-10 splenic production. Likewise, increased production of IFN-γ, IL-12 and IL-4 concomitant with decreased TGF-β and TNF-α was locally observed in the infected footpad. Conclusion This study indicates that extracellular serine proteases of L. amazonensis are strong candidates for a more defined intranasal vaccine against cutaneous leishmaniasis. Electronic supplementary material The online version of this article (doi:10.1186/1756-3305-7-448) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Herbert Leonel de Matos Guedes
- Laboratório de Imunofarmacologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil.
| | | | | | | | | | | | | |
Collapse
|
21
|
de Araújo CF, Silva VMG, Cronemberger-Andrade A, Aragão-França LS, Rocha VCJ, Santos PSL, Pontes-de-Carvalho L. Leishmania braziliensis and Leishmania amazonensis amastigote extracts differ in their enhancement effect on Leishmania infection when injected intradermally. BMC Res Notes 2014; 7:70. [PMID: 24484604 PMCID: PMC3922628 DOI: 10.1186/1756-0500-7-70] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 01/27/2014] [Indexed: 11/29/2022] Open
Abstract
Background It has been reported that repeated intravenous injections of a relatively large amount of Leishmania amazonensis amastigote extract (LaE) in BALB/c mice exacerbates the infection of these mice by Leishmania braziliensis. The identification of the extract active principle(s) through physicochemical purification often involves dilution and losses of protein in the course of successive purification procedures. The large amount of the extract required to induce the phenomenon, therefore, hinders the carrying out of experiments aimed at identifying the active molecule(s) through extract purification. In the present work, a dose–response experiment was done to find out if smaller amounts of LaE than that necessary to be used by the intravenous route would reproduce the phenomenon when injected by the intradermal route. In addition, it was also investigated whether a Leishmania braziliensis amastigote extract (LbE) would exert the same effect and whether the effect would occur in C57BL/6 mice. Results It was found that a single injection of either LaE or LbE containing 5 μg of protein was capable of enhancing the infection in BALB/c but not in C57BL/6 mice. In addition, it was observed that the largest tested doses of LbE (containing 30 and 180 μg of protein) failed to enhance the infection by L. braziliensis, whereas all doses of LaE enhanced equally that infection. Conclusions Those results indicate the possible existence in LbE, and not in LaE, of molecules that interfere with the extract infection-enhancing activity when it is injected in large amounts, and that the inoculation of Leishmania extracts through the intravenous and intradermal routes potentiate the infection by L. braziliensis through the same mechanism.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lain Pontes-de-Carvalho
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz, Rua Waldemar Falcão 121, Salvador, Brazil.
| |
Collapse
|
22
|
Martínez Salazar MB, Delgado Domínguez J, Silva Estrada J, González Bonilla C, Becker I. Vaccination with Leishmania mexicana LPG induces PD-1 in CD8⁺ and PD-L2 in macrophages thereby suppressing the immune response: a model to assess vaccine efficacy. Vaccine 2014; 32:1259-65. [PMID: 24462405 DOI: 10.1016/j.vaccine.2014.01.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 12/20/2013] [Accepted: 01/10/2014] [Indexed: 11/16/2022]
Abstract
Leishmania lipophosphoglycan (LPG) is a molecule that has been used as a vaccine candidate, with contradictory results. Since unsuccessful protection could be related to suppressed T cell responses, we analyzed the expression of inhibitory receptor PD-1 in CD8(+) and CD4(+) lymphocytes and it is ligand PD-L2 in macrophages of BALB/c mice immunized with various doses of Leishmania mexicana LPG and re-stimulated in vitro with different concentrations of LPG. Vaccination with LPG enhanced the expression of PD-1 in CD8(+) cells. Activation molecules CD137 were reduced in CD8(+) cells from vaccinated mice. In vitro re-stimulation enhanced PD-L2 expression in macrophages of healthy mice in a dose-dependent fashion. The expression of PD-1, PD-L2 and CD137 is modulated according to the amount of LPG used during immunization and in vitro re-stimulation. We analyzed the expression of these molecules in mice infected with 1×10(4) or 1×10(5)L. mexicana promastigotes and re-stimulated in vitro with LPG. Infection with 1×10(5) parasites increased the PD-1 expression in CD8(+) and diminished PD-L2 in macrophages. When these CD8(+) cells were re-stimulated in vitro with LPG, simulating a second exposure to parasite antigens, PD-1 expression increased significantly more, in a dose dependent fashion. We conclude that CD8(+) T lymphocytes and macrophages express inhibition molecules according to the concentrations of Leishmania LPG and to the parasite load. Vaccination with increased amounts of LPG or infections with higher parasite numbers induces enhanced expression of PD-1 and functional inactivation of CD8(+) cells, which can have critical consequences in leishmaniasis, since these cells are crucial for disease control. These results call for pre-vaccination evaluations of potential immunogens, specifically where CD8 cells are required, since inhibiting molecules can be induced after certain thresholds of antigen concentrations. We propose that the analysis of PD-1 and PD-L2 are useful tools to monitor the optimal dose for vaccination candidates.
Collapse
Affiliation(s)
- M B Martínez Salazar
- Departamento de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Dr. Balmis 148, Colonia Doctores, 06726, Mexico D.F., Mexico
| | - J Delgado Domínguez
- Departamento de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Dr. Balmis 148, Colonia Doctores, 06726, Mexico D.F., Mexico
| | - J Silva Estrada
- Departamento de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Dr. Balmis 148, Colonia Doctores, 06726, Mexico D.F., Mexico
| | - C González Bonilla
- Laboratorios de Vigilancia e Investigación Epidemiológica, Coordinación de Vigilancia Epidemiológica, Instituto Mexicano del Seguro Social, México D.F., Mexico
| | - I Becker
- Departamento de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Dr. Balmis 148, Colonia Doctores, 06726, Mexico D.F., Mexico.
| |
Collapse
|
23
|
Soares DC, Calegari-Silva TC, Lopes UG, Teixeira VL, de Palmer Paixão ICN, Cirne-Santos C, Bou-Habib DC, Saraiva EM. Dolabelladienetriol, a compound from Dictyota pfaffii algae, inhibits the infection by Leishmania amazonensis. PLoS Negl Trop Dis 2012; 6:e1787. [PMID: 22970332 PMCID: PMC3435235 DOI: 10.1371/journal.pntd.0001787] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Accepted: 07/06/2012] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Chemotherapy for leishmaniasis, a disease caused by Leishmania parasites, is expensive and causes side effects. Furthermore, parasite resistance constitutes an increasing problem, and new drugs against this disease are needed. In this study, we examine the effect of the compound 8,10,18-trihydroxy-2,6-dolabelladiene (Dolabelladienetriol), on Leishmania growth in macrophages. The ability of this compound to modulate macrophage function is also described. METHODOLOGY/PRINCIPAL FINDINGS Leishmania-infected macrophages were treated with Dolabelladienetriol, and parasite growth was measured using an infectivity index. Nitric oxide (NO), TNF-α and TGF-β production were assayed in macrophages using specific assays. NF-kB nuclear translocation was analyzed by western blot. Dolabelladienetriol inhibited Leishmania in a dose-dependent manner; the IC(50) was 44 µM. Dolabelladienetriol diminished NO, TNF-α and TGF-β production in uninfected and Leishmania-infected macrophages and reduced NF-kB nuclear translocation. Dolabelladienetriol inhibited Leishmania infection even when the parasite growth was exacerbated by either IL-10 or TGF-β. In addition, Dolabelladienetriol inhibited Leishmania growth in HIV-1-co-infected human macrophages. CONCLUSION Our results indicate that Dolabelladienetriol significantly inhibits Leishmania in macrophages even in the presence of factors that exacerbate parasite growth, such as IL-10, TGF-β and HIV-1 co-infection. Our results suggest that Dolabelladienetriol is a promising candidate for future studies regarding treatment of leishmaniasis, associated or not with HIV-1 infection.
Collapse
Affiliation(s)
- Deivid Costa Soares
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Teresa C. Calegari-Silva
- Laboratório de Parasitologia Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ulisses G. Lopes
- Laboratório de Parasitologia Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Valéria L. Teixeira
- Departamento de Biologia Marinha, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| | - Izabel C. N. de Palmer Paixão
- Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| | - Claudio Cirne-Santos
- Laboratório de Imunologia Clínica, Instituto Oswaldo Cruz/FIOCRUZ, Rio de Janeiro, Brazil
| | | | - Elvira M. Saraiva
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
24
|
Silva-Almeida M, Pereira BAS, Ribeiro-Guimarães ML, Alves CR. Proteinases as virulence factors in Leishmania spp. infection in mammals. Parasit Vectors 2012; 5:160. [PMID: 22871236 PMCID: PMC3436776 DOI: 10.1186/1756-3305-5-160] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 07/12/2012] [Indexed: 11/10/2022] Open
Abstract
Leishmania parasites cause human tegumentary and visceral infections that are commonly referred to as leishmaniasis. Despite the high incidence and prevalence of cases, leishmaniasis has been a neglected disease because it mainly affects developing countries. The data obtained from the analysis of patients' biological samples and from assays with animal models confirm the involvement of an array of the parasite's components in its survival inside the mammalian host. These components are classified as virulence factors. In this review, we focus on studies that have explored the role of proteinases as virulence factors that promote parasite survival and immune modulation in the mammalian host. Additionally, the direct involvement of proteinases from the host in lesion evolution is analyzed. The gathered data shows that both parasite and host proteinases are involved in the clinical manifestation of leishmaniasis. It is interesting to note that although the majority of the classes of proteinases are present in Leishmania spp., only cysteine-proteinases, metalloproteinases and, to a lesser scale, serine-proteinases have been adequately studied. Members from these classes have been implicated in tissue invasion, survival in macrophages and immune modulation by parasites. This review reinforces the importance of the parasite proteinases, which are interesting candidates for new chemo or immunotherapies, in the clinical manifestations of leishmaniasis.
Collapse
Affiliation(s)
- Mariana Silva-Almeida
- Laboratório de Biologia Molecular e Doenças Endêmicas, IOC, Fiocruz, Avenida Brasil, 4365 Manguinhos Pavilhão Leônidas Deane-Sala 209, CEP: 21040-900, Rio de Janeiro, RJ, Brasil
| | | | | | | |
Collapse
|
25
|
Paladi CDS, Pimentel IAS, Katz S, Cunha RLOR, Judice WADS, Caires ACF, Barbiéri CL. In vitro and in vivo activity of a palladacycle complex on Leishmania (Leishmania) amazonensis. PLoS Negl Trop Dis 2012; 6:e1626. [PMID: 22616018 PMCID: PMC3352823 DOI: 10.1371/journal.pntd.0001626] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 03/08/2012] [Indexed: 11/18/2022] Open
Abstract
Background Antitumor cyclopalladated complexes with low toxicity to laboratory animals have shown leishmanicidal effect. These findings stimulated us to test the leishmanicidal property of one palladacycle compound called DPPE 1.2 on Leishmania (Leishmania) amazonensis, an agent of simple and diffuse forms of cutaneous leishmaniasis in the Amazon region, Brazil. Methodology/Principal Findings Promastigotes of L. (L.) amazonensis and infected bone marrow-derived macrophages were treated with different concentrations of DPPE 1.2. In in vivo assays foot lesions of L. (L.) amazonensis-infected BALB/c mice were injected subcutaneously with DPPE 1.2 and control animals received either Glucantime or PBS. The effect of DPPE 1.2 on cathepsin B activity of L. (L.) amazonensis amastigotes was assayed spectrofluorometrically by use of fluorogenic substrates. The main findings were: 1) axenic L. (L.) amazonensis promastigotes were destroyed by nanomolar concentrations of DPPE 1.2 (IC50 = 2.13 nM); 2) intracellular parasites were killed by DPPE 1.2 (IC50 = 128.35 nM), and the drug displayed 10-fold less toxicity to macrophages (CC50 = 1,267 nM); 3) one month after intralesional injection of DPPE 1.2 infected BALB/c mice showed a significant decrease of foot lesion size and a reduction of 97% of parasite burdens when compared to controls that received PBS; 4) DPPE 1.2 inhibited the cysteine protease activity of L. (L.) amazonensis amastigotes and more significantly the cathepsin B activity. Conclusions/Significance The present results demonstrated that DPPE 1.2 can destroy L. (L.) amazonensis in vitro and in vivo at concentrations that are non toxic to the host. We believe these findings support the potential use of DPPE 1.2 as an alternative choice for the chemotherapy of leishmaniasis. Leishmaniasis is an important public health problem with an estimated annual incidence of 1.5 million of new human cases of cutaneous leishmaniasis and 500,000 of visceral leishmaniasis. Treatment of the diseases is limited by toxicity and parasite resistance to the drugs currently in use, validating the need to develop new leishmanicidal compounds. We evaluated the killing by the palladacycle complex DPPE 1.2 of Leishmania (Leishmania) amazonensis, an agent of human cutaneous leishmaniasis in the Amazon region, Brazil. DPPE 1.2 destroyed promastigotes of L. (L.) amazonensis in vitro at nanomolar concentrations, whereas intracellular amastigotes were killed at drug concentrations 10-fold less toxic than those displayed to macrophages. L. (L.) amazonensis-infected BALB/c mice treated by intralesional injection of DPPE 1.2 exhibited a significant decrease of foot lesion sizes and a 97% reduction of parasite burdens when compared to untreated controls. Additional experiments indicated the inhibition of the cathepsin B activity of L. (L.) amazonensis amastigotes by DPPE 1.2. Further studies are needed to explore the potential of DPPE 1.2 as an additional option for the chemotherapy of leishmaniasis.
Collapse
Affiliation(s)
- Carolina de Siqueira Paladi
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | | | - Simone Katz
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Rodrigo L. O. R. Cunha
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André, São Paulo, Brazil
| | - Wagner Alves de Souza Judice
- Centro Interdisciplinar de Investigação Bioquímica, Universidade de Mogi das Cruzes, Mogi das Cruzes, São Paulo, Brazil
| | - Antonio C. F. Caires
- Centro Interdisciplinar de Investigação Bioquímica, Universidade de Mogi das Cruzes, Mogi das Cruzes, São Paulo, Brazil
| | - Clara Lúcia Barbiéri
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
26
|
Macedo ABB, Sánchez-Arcila JC, Schubach AO, Mendonça SCF, Marins-Dos-Santos A, de Fatima Madeira M, Gagini T, Pimentel MIF, De Luca PM. Multifunctional CD4⁺ T cells in patients with American cutaneous leishmaniasis. Clin Exp Immunol 2012; 167:505-13. [PMID: 22288594 DOI: 10.1111/j.1365-2249.2011.04536.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Leishmaniasis is a group of important parasitic diseases affecting millions worldwide. To understand more clearly the quality of T helper type 1 (Th1) response stimulated after Leishmania infection, we applied a multiparametric flow cytometry protocol to evaluate multifunctional T cells induced by crude antigen extracts obtained from promastigotes of Leishmania braziliensis (LbAg) and Leishmania amazonensis (LaAg) in peripheral blood mononuclear cells from healed cutaneous leishmaniasis patients. Although no significant difference was detected in the percentage of total interferon (IFN)-γ-producing CD4(+) T cells induced by both antigens, multiparametric flow cytometry analysis revealed clear differences in the quality of Th1 responses. LbAg induced an important proportion of multifunctional CD4(+) T cells (28% of the total Th1 response evaluated), whereas LaAg induced predominantly single-positive cells (68%), and 57% of those were IFN-γ single-positives. Multifunctional CD4(+) T cells showed the highest mean fluorescence intensity (MFI) for the three Th1 cytokines assessed and MFIs for IFN-γ and interleukin-2 from those cells stimulated with LbAg were significantly higher than those obtained after LaAg stimulation. These major differences observed in the generation of multifunctional CD4(+) T cells suggest that the quality of the Th1 response induced by L. amazonensis antigens can be involved in the mechanisms responsible for the high susceptibility observed in L. amazonensis-infected individuals. Ultimately, our results call attention to the importance of studying a Th1 response regarding its quality, not just its magnitude, and indicate that this kind of evaluation might help understanding of the complex and diverse immunopathogenesis of American tegumentary leishmaniasis.
Collapse
Affiliation(s)
- A B B Macedo
- Laboratório de Imunoparasitologia, Instituto Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Soong L. Subversion and Utilization of Host Innate Defense by Leishmania amazonensis. Front Immunol 2012; 3:58. [PMID: 22566939 PMCID: PMC3342205 DOI: 10.3389/fimmu.2012.00058] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Accepted: 03/06/2012] [Indexed: 01/07/2023] Open
Abstract
Infection with Leishmania amazonensis and other members of the Leishmania mexicana complex can lead to diverse clinical manifestations, some of which are relatively difficult to control, even with standard chemotherapy. Diffuse cutaneous leishmaniasis (CL) is a rare but severe form, and its clinical hallmark is excessive parasitic growth in infected cells accompanied by profound impairments in host immune responses to the parasites. Since these parasites also cause non-healing CL in most inbred strains of mice, these animals are valuable models for dissecting the mechanisms of persistent infection and disease pathogenesis. In comparison to other Leishmania species, L. amazonensis infections are most remarkable for their ability to repress the activation and effector functions of macrophages, dendritic cells, and CD4(+) T cells, implying discrete mechanisms at work. In addition to this multilateral suppression of host innate and adaptive immunity, the activation of types I and II interferon-mediated responses and autophagic/lipid metabolic pathways actually promotes rather than restrains L. amazonensis infection. These seemingly contradictory findings reflect the remarkable adaptation of the parasites to the ancient defense machinery of the host, as well as the complex parasite-host interactions at different stages of infection, which collectively contribute to non-healing leishmaniasis in the New World. This review article highlights new evidence that reveals the strategies utilized by L. amazonensis parasites to subvert or modulate host innate defense machinery in neutrophils and macrophages, as well as the regulatory roles of host innate responses in promoting parasite survival and replication within the huge parasitophorous vacuoles. A better understanding of unique features in host responses to these parasites at early and late stages of infection is important for the rational design of control strategies for non-healing leishmaniasis.
Collapse
Affiliation(s)
- Lynn Soong
- Center for Tropical Diseases, Sealy Center for Vaccine Development, Department of Microbiology and Immunology, The University of Texas Medical Branch Galveston, TX, USA
| |
Collapse
|
28
|
Corporeau C, Groisillier A, Jeudy A, Barbeyron T, Fleury E, Fabioux C, Czjzek M, Huvet A. A functional study of transforming growth factor-beta from the gonad of Pacific oyster Crassostrea gigas. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2011; 13:971-980. [PMID: 21271272 DOI: 10.1007/s10126-010-9361-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Accepted: 12/25/2010] [Indexed: 05/30/2023]
Abstract
The transforming growth factor (TGF)-β superfamily is a group of important growth factors involved in multiple processes such as differentiation, cell proliferation, apoptosis and cellular growth. In the Pacific oyster Crassostrea gigas, the oyster gonadal (og) TGF-β gene was recently characterized through genome-wide expression profiling of oyster lines selected to be resistant or susceptible to summer mortality. Og TGF-β appeared specifically expressed in the gonad to reach a maximum when gonads are fully mature, which singularly contrasts with the pleiotropic roles commonly ascribed to most TGF-β family members. The function of og TGF-β protein in oysters is unknown, and defining its role remains challenging. In this study, we develop a rapid bacterial production system to obtain recombinant og TGF-β protein, and we demonstrate that og TGF-β is processed by furin to a mature form of the protein. This mature form can be detected in vivo in the gonad. Functional inhibition of mature og TGF-β in the gonad was conducted by inactivation of the protein using injection of antibodies. We show that inhibition of og TGF-β function tends to reduce gonadic area. We conclude that mature og TGF-β probably functions as an activator of germ cells development in oyster.
Collapse
|
29
|
Enhancement of experimental cutaneous leishmaniasis by Leishmania molecules is dependent on interleukin-4, serine protease/esterase activity, and parasite and host genetic backgrounds. Infect Immun 2010; 79:1236-43. [PMID: 21173308 DOI: 10.1128/iai.00309-10] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Most inbred strains of mice, like the BALB/c strain, are susceptible to Leishmania amazonensis infections and resistant to Leishmania braziliensis infections. This parasite-related difference could result from the activity of an L. amazonensis-specific virulence factor. In agreement with this hypothesis, it is shown here that the intravenous injection of BALB/c mice with L. amazonensis amastigote extract (LaE) but not the L. braziliensis extract confers susceptibility to L. braziliensis infection. This effect was associated with high circulating levels of IgG1 anti-L. amazonensis antibodies and with an increase in interleukin-4 (IL-4) production and a decrease in gamma interferon production by draining lymph node cells. Moreover, the effect was absent in IL-4-knockout mice. The biological activity in the LaE was not mediated by amphiphilic molecules and was inhibited by pretreatment of the extract with irreversible serine protease inhibitors. These findings indicate that the LaE contains a virulence-related factor that (i) enhances the Leishmania infection by promoting Th2-type immune responses, (ii) is not one of the immunomodulatory Leishmania molecules described so far, and (iii) is either a serine protease or has an effect that depends on that protease activity. In addition to being Leishmania species specific, the infection-enhancing activity was also shown to depend on the host genetic makeup, as LaE injections did not affect the susceptibility of C57BL/6 mice to L. braziliensis infection. The identification of Leishmania molecules with infection-enhancing activity could be important for the development of a vaccine, since the up- or downmodulation of the immune response against a virulence factor could well contribute to controlling the infection.
Collapse
|
30
|
de Matos Guedes HL, Pinheiro RO, Chaves SP, De-Simone SG, Rossi-Bergmann B. Serine proteases of Leishmania amazonensis as immunomodulatory and disease-aggravating components of the crude LaAg vaccine. Vaccine 2010; 28:5491-6. [PMID: 20472030 DOI: 10.1016/j.vaccine.2010.04.109] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2010] [Revised: 04/07/2010] [Accepted: 04/30/2010] [Indexed: 10/19/2022]
Abstract
We previously demonstrated that intradermal and intramuscular vaccination with Leishmania amazonensis promastigote antigens (LaAg) increases the susceptibility of BALB/c mice to cutaneous leishmaniasis. In this study, we investigated the role played by serine and cysteine proteases as disease-promoting components of LaAg. Mice were immunized by the intramuscular route with LaAg that was pre-treated with a pool of serine or cysteine protease inhibitors (SPi and CPi, respectively) prior to infection with L. amazonensis. Neutralization of either enzyme type reversed the disease-promoting effect of LaAg, as seen by the slower lesion development. However, the parasite burden was only effectively controlled in mice receiving SPi-treated LaAg. Protection was associated with diminished production of TGF-beta and particularly IL-10 in response to parasite antigens by the lesion-draining lymph node cells of vaccinated mice relative to control. In vitro, soluble proteases isolated from LaAg (LaSP-Sol) directly activated IL-4, IL-10 and TGF-beta production by immune cells. Like native LaAg, vaccination with LaSP-Sol primed mice to respond to parasite challenge with a strong Jones-Mote cutaneous hypersensitivity reaction, and increased susceptibility to infection. Furthermore, neutralization of serine but not cysteine proteases blocked the capacity of LaAg to sensitize mice for Jones-Mote reaction. Together, these results indicate that soluble serine proteases are key components of LaAg responsible for its disease-promoting immunity.
Collapse
Affiliation(s)
- Herbert Leonel de Matos Guedes
- Laboratório de Imunofarmacologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil
| | | | | | | | | |
Collapse
|
31
|
Pinheiro RO, Nunes MP, Pinheiro CS, D'Avila H, Bozza PT, Takiya CM, Côrte-Real S, Freire-de-Lima CG, DosReis GA. Induction of autophagy correlates with increased parasite load of Leishmania amazonensis in BALB/c but not C57BL/6 macrophages. Microbes Infect 2008; 11:181-90. [PMID: 19070676 DOI: 10.1016/j.micinf.2008.11.006] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2008] [Revised: 11/03/2008] [Accepted: 11/11/2008] [Indexed: 12/12/2022]
Abstract
We investigated the role of autophagy in infection of macrophages by Leishmania amazonensis. Induction of autophagy by IFN-gamma or starvation increased intracellular parasite load and the percentages of infected macrophages from BALB/c but not from C57BL/6 mice. In contrast, starvation did not affect the replication of either Leishmania major or Trypanosoma cruzi in BALB/c macrophages. In BALB/c macrophages, starvation resulted in increased monodansylcadaverine staining and in the appearance of double-membrane and myelin-like vesicles characteristic of autophagosomes. Increased parasite load was associated with a reduction in NO levels and was attenuated by wortmannin, an inhibitor of autophagy. In infected macrophages from BALB/c, but not from C57BL/6 mice, starvation increased the number of lipid bodies and the amounts of PGE(2) produced. Exogenous PGE(2) increased parasite load in macrophages from BALB/c, but not C57BL/6 mice. The cyclooxygenase inhibitor indomethacin prevented the increase of parasite load in starved BALB/c macrophages, and actually induced parasite killing. These results suggest that autophagy regulates the outcome of L. amazonensis infection in macrophages in a host strain specific manner.
Collapse
|
32
|
Pinheiro RO, Rossi-Bergmann B. Interferon-gamma is required for the late but not early control of Leishmania amazonensis infection in C57Bl/6 mice. Mem Inst Oswaldo Cruz 2007; 102:79-82. [PMID: 17294004 DOI: 10.1590/s0074-02762007000100013] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2006] [Accepted: 01/12/2007] [Indexed: 05/13/2023] Open
Abstract
The critical role of interferon-gamma (IFN-gamma) in the resistance of C57Bl/6 mice to Leishmania major is widely established but its role in the relative resistance of these animals to L. amazonensis infection is still not clear. In this work we use C57Bl/6 mice congenitally deficient in the IFN-gamma gene (IFN-gamma KO) to address this issue. We found that IFN-gamma KO mice were as resistant as their wild-type (WT) counterparts at least during the first two months of infection. Afterwards, whereas WT mice maintained lesion growth under control, IFN-gamma KO mice developed devastating lesions. At day 97 of infection, their lesions were 9-fold larger than WT controls, concomitant with an increased parasite burden. At this stage, lesion-draining cells from IFN-gamma KO mice had impaired capacity to produce interleukin-12 (IL-12) and tumour necrosis factor-a in response to parasite antigens whereas IL-4 was slightly increased in comparison to infected WT mice. Together, these results show that IFN-gamma is not critical for the initial control of L. amazonensis infection in C57Bl/6 mice, but is essential for the development of a protective Th1 type immune response in the later stages.
Collapse
Affiliation(s)
- Roberta Olmo Pinheiro
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-900, Brasil
| | | |
Collapse
|
33
|
Pinheiro RO, Pinto EF, de Matos Guedes HL, Filho OAA, de Mattos KA, Saraiva EM, de Mendonça SCF, Rossi-Bergmann B. Protection against cutaneous leishmaniasis by intranasal vaccination with lipophosphoglycan. Vaccine 2006; 25:2716-22. [PMID: 16814903 DOI: 10.1016/j.vaccine.2006.05.093] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2006] [Revised: 05/24/2006] [Accepted: 05/29/2006] [Indexed: 11/21/2022]
Abstract
We previously showed the opposing effect of systemic and mucosal vaccination with whole Leishmania amazonensis antigen (LaAg). Here, the role played by lipophosphoglycan (LPG) as the key disease-promoting component of intramuscular (i.m.) LaAg and its usefulness as a defined intranasal vaccine was investigated in murine cutaneous leishmaniasis. BALB/c mice were twice vaccinated by the i.m. route with 25mug of intact LaAg or with LaAg that was pretreated with anti-LPG 3A1-La monoclonal antibody, prior to infection with L. amazonensis. LPG neutralization rendered the otherwise disease-promoting LaAg antigen protective, as observed by the smaller lesion sizes and reduced parasite burden. The increased resistance was accompanied by a markedly lower antigen-driven TGF-beta and IL-10 responses in the lesion-draining lymph nodes, concomitant with significantly higher IFN-gamma production. To test for intranasal efficacy, 10 microg of affinity-purified LPG and its parental LaAg were twice instilled in the nostrils prior to L. amazonensis infection. In both cases, similarly slower lesion growth and lower parasite burden were found that was associated with increased IFN-gamma and IL-10 responses in the lesion-draining lymph nodes. These results support a role for LPG in the dual route-related effect of LaAg and shows its strong potential as a defined needle-free and adjuvant-free vaccine for cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Roberta Olmo Pinheiro
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21949-900 Rio de Janeiro, RJ, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Calvopina M, Barroso PA, Marco JD, Korenaga M, Cooper PJ, Nonaka S, Hashiguchi Y. Efficacy of vaccination with a combination of Leishmania amastigote antigens and the lipid A-analogue ONO-4007 for immunoprophylaxis and immunotherapy against Leishmania amazonensis infection in a murine model of New World cutaneous leishmaniasis. Vaccine 2006; 24:5645-52. [PMID: 16621179 DOI: 10.1016/j.vaccine.2006.03.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2006] [Accepted: 03/02/2006] [Indexed: 10/24/2022]
Abstract
Activation of innate immunity using adjuvants that activate Toll-like receptor 4 pathways have great potential for improving the protection induced by parasite vaccines. We investigated protective and therapeutic effects of a vaccine against leishmaniasis containing a combination of an adjuvant synthetic lipid A-analogue, ONO-4007 and Leishmania amazonensis antigens. ONO-4007 was co-injected with soluble and membrane-enriched L. amazonensis-amastigote antigens into BALB/c mice that had either already been infected with 1 x 10(6) L. amazonensis promastigotes (immunotherapy study) or before challenge with the same infectious dose (immunoprophylaxis study). Sixty percent of mice vaccinated before infectious challenge controlled their Leishmania infections - defined by the absence of footpad-swelling and negative Leishmania cultures - compared to 0% of controls, and 40% of mice vaccinated after infection resolved their infections compared to 0% of controls. Protective immunity in both immunoprophylaxis and immunotherapy models was associated with increased protein production of IL-12 and IFN-gamma. These data suggest that vaccination with a combination of ONO-4007 and amastigote antigens of L. amazonensis may be useful for the prevention and treatment of leishmaniasis, and that the protective immunity induced is associated with the production of type-1 cytokines.
Collapse
Affiliation(s)
- Manuel Calvopina
- Department of Parasitology, Kochi Medical School, Kochi University, Kochi 783-8505, Japan.
| | | | | | | | | | | | | |
Collapse
|