scispace - formally typeset
Search or ask a question
Author

Raul Arjona-Sabido

Bio: Raul Arjona-Sabido is an academic researcher from Universidad Autónoma de Yucatán. The author has contributed to research in topics: Polyacrylamide gel electrophoresis & Antigen. The author has an hindex of 1, co-authored 1 publications receiving 10 citations.

Papers
More filters
Journal ArticleDOI
TL;DR: Mice immunized with the LdNH36 antigen in combination with the GLA-SE adjuvant and challenged with Leishmania mexicana showed significant reductions in parasite burden, confirming the protective efficacy of this vaccine candidate.

11 citations


Cited by
More filters
Journal ArticleDOI
TL;DR: The epitope presentation in a recombinant chimera optimizes immunogenicity and efficacy above the levels induced by the independent or admixed F1 and F3 domains and advanced in the design of a NH36 polytope vaccine capable of inducing cross-protection to cutaneous leishmaniasis.
Abstract: The Leishmania donovani-nucleoside hydrolase (NH36) and NH A34480 of L. amazonensis share 93% of sequence identity. In mice, the NH36 induced protection against visceral leishmaniasis is mediated by a CD4+ T cell response against its C-terminal domain (F3). Besides this CD4+ Th1 response, prevention and cure of L. amazonensis infection require also additional CD8+ and regulatory T-cell responses to the NH36 N-terminal (F1 domain). We investigated if mice vaccination with F1 and F3 domains cloned in tandem, in a recombinant chimera, with saponin, optimizes the vaccine efficacy against L. amazonensis infection above the levels promoted by the two admixed domains or by each domain independently. The chimera induced the highest IgA, IgG and IgG2a anti-NH36 antibody, IDR, IFN-γ and IL-10 responses, while TNF-α was more secreted by mice vaccinated with F3 or all F3-contaning vaccines. Additionally, the chimera and the F1 vaccine also induced the highest proportions of CD4+ and CD8+ T cells secreting IL-2, TNF-α or IFN-γ alone, TNF-α in combination with IL-2 or IFN-γ and of CD4+ multifunctional cells secreting IL-2, TNF-α and IFN-γ. Correlating with the immunological results the strongest reductions of skin lesions sizes were determined by the admixed domains (80 %) and by the chimera (84 %), which also promoted the most pronounced and significant reduction of the parasite load (99.8 %). Thus, the epitope presentation in a recombinant chimera optimizes immunogenicity and efficacy above the levels induced by the independent or admixed F1 and F3 domains. The multiparameter analysis disclosed that the Th1-CD4+ T helper response induced by the chimera is mainly directed against its FRYPRPKHCHTQVA epitope. Additionally, the YPPEFKTKL epitope of F1 induced the second most important CD4+ T cell response, and, followed by the DVAGIVGVPVAAGCT, FMLQILDFYTKVYE and ELLAITTVVGNQ sequences, also the most potent CD8+ T cell responses and IL-10 secretion. Remarkably, the YPPEFKTKL epitope shows high amino acid identity with a multipotent PADRE sequence and stimulates simultaneously the CD4+, CD8+ T cell and a probable T regulatory response. With this approach we advanced in the design of a nucleoside hydrolase NH36 polytope vaccine capable of inducing cross-protection to cutaneous leishmaniasis.

30 citations

Journal ArticleDOI
TL;DR: It is clear that the vaccine platform of choice can have a significant impact upon the level of protection induced by particular antigens, and some examples for which the vaccine system used has impacted the protective efficacy imparted are provided.
Abstract: From experimental models and the analyses of patients, it is well documented that antigen-specific T cells are critical for protection against Leishmania infection Effective vaccines require both targeting to the pathogen and an immune stimulant to induce maturation of appropriate immune responses While a great number of antigens have been examined as vaccine candidates against various Leishmania species, few have advanced to human or canine clinical trials With emphasis on antigen expression, in this minireview we discuss some of the vaccine platforms that are currently being explored for the development of Leishmania vaccines It is clear that the vaccine platform of choice can have a significant impact upon the level of protection induced by particular antigens, and we provide and highlight some examples for which the vaccine system used has impacted the protective efficacy imparted

19 citations

Journal ArticleDOI
TL;DR: Over the last decade, there has been additional excitement over the potential development and introduction of new vaccines to prevent highly lethal respiratory virus infections, as well as tuberculosis, malaria, HIV/AIDS and several neglected tropical diseases.

11 citations

Journal ArticleDOI
TL;DR: The aim of this study is to discuss the research and development of new agents for the treatment of Leishmania, and to stimulate the formulation of new NH inhibitors, to eliminate leishmaniasis disease in infected individuals.

10 citations

Journal ArticleDOI
TL;DR: The FML-saponin vaccine became the first licensed veterinary vaccine against leishmaniasis (Leishmune®) which reduced the incidence of human and canine VL in endemic areas and identified the most immunogenic NH36 domains and epitopes for PBMC of active human VL, cured or asymptomatic and DTH+ patients.
Abstract: NH36 is a vital enzyme of the DNA metabolism and a specific target for anti-Leishmania chemotherapy. We developed second-generation vaccines composed of the FML complex or its main native antigen, the NH36 nucleoside hydrolase of Leishmania (L.) donovani and saponin, and a DNA vaccine containing the NH36 gene. All these vaccines were effective in prophylaxis and treatment of mice and dog visceral leishmaniasis (VL). The FML-saponin vaccine became the first licensed veterinary vaccine against leishmaniasis (Leishmune®) which reduced the incidence of human and canine VL in endemic areas. The NH36, DNA or recombinant protein vaccines induced a Th1 CD4+IFN-γ+ mediated protection in mice. Efficacy against VL was mediated by a CD4+TNF-α T lymphocyte response against the NH36-F3 domain, while against tegumentary leishmaniasis (TL) a CD8+ T lymphocyte response to F1 was also required. These domains were 36-41 % more protective than NH36, and a recombinant F1F3 chimera was 21% stronger than the domains, promoting a 99.8% reduction of the parasite load. We also identified the most immunogenic NH36 domains and epitopes for PBMC of active human VL, cured or asymptomatic and DTH+ patients. Currently, the NH36 subunit recombinant vaccine is turning into a multi-epitope T cell synthetic vaccine against VL and TL.

9 citations