Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Ir J Med Sci ; 192(1): 231-261, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35394635

RESUMO

Toxoplasma gondii is an obligate intracellular parasite that causes toxoplasmosis. It has been shown that the severity of symptoms depends on the functioning of the host immune system. Although T. gondii infection typically does not lead to severe disease in healthy people and after infection, it induces a stable immunity, but it can contribute to severe and even lethal Toxoplasmosis in immunocompromised individuals (AIDS, bone marrow transplant and neoplasia). The antigens that have been proposed to be used in vaccine candidate in various studies include surface antigens and secretory excretions that have been synthesized and evaluated in different studies. In some studies, secretory antigens play an important role in stimulating the host immune response. Various antigens such as SAG, GRA, ROP, ROM, and MAG have been from different strains of T. gondii have been synthesized and their protective effects have been evaluated in animal models in different vaccine platforms including recombinant antigens, nanoparticles, and DNA vaccine. Four bibliographic databases including Science Direct, PubMed Central (PMC), Scopus, and Google Scholar were searched for articles published up to 2020.The current review article focuses on recent studies on the use and usefulness of recombinant antigens, nanoparticles, and DNA vaccines.


Assuntos
Vacinas Protozoárias , Toxoplasma , Toxoplasmose , Vacinas de DNA , Animais , Humanos , Camundongos , Toxoplasma/genética , Antígenos de Protozoários/genética , Proteínas de Protozoários/genética , Vacinas Protozoárias/uso terapêutico , Vacinas Protozoárias/genética , Toxoplasmose/prevenção & controle , Toxoplasmose/parasitologia , Vacinas de DNA/uso terapêutico , Vacinas de DNA/genética , Camundongos Endogâmicos BALB C
2.
Front Immunol ; 12: 635513, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33953712

RESUMO

Schistosome infection is a major cause of global morbidity, particularly in sub-Saharan Africa. However, there is no effective vaccine for this major neglected tropical disease, and re-infection routinely occurs after chemotherapeutic treatment. Following invasion through the skin, larval schistosomula enter the circulatory system and migrate through the lung before maturing to adulthood in the mesenteric or urogenital vasculature. Eggs released from adult worms can become trapped in various tissues, with resultant inflammatory responses leading to hepato-splenic, intestinal, or urogenital disease - processes that have been extensively studied in recent years. In contrast, although lung pathology can occur in both the acute and chronic phases of schistosomiasis, the mechanisms underlying pulmonary disease are particularly poorly understood. In chronic infection, egg-mediated fibrosis and vascular destruction can lead to the formation of portosystemic shunts through which eggs can embolise to the lungs, where they can trigger granulomatous disease. Acute schistosomiasis, or Katayama syndrome, which is primarily evident in non-endemic individuals, occurs during pulmonary larval migration, maturation, and initial egg-production, often involving fever and a cough with an accompanying immune cell infiltrate into the lung. Importantly, lung migrating larvae are not just a cause of inflammation and pathology but are a key target for future vaccine design. However, vaccine efforts are hindered by a limited understanding of what constitutes a protective immune response to larvae. In this review, we explore the current understanding of pulmonary immune responses and inflammatory pathology in schistosomiasis, highlighting important unanswered questions and areas for future research.


Assuntos
Pneumopatias Parasitárias/parasitologia , Pulmão/parasitologia , Schistosoma/patogenicidade , Esquistossomose/parasitologia , Animais , Modelos Animais de Doenças , Interações Hospedeiro-Parasita , Humanos , Evasão da Resposta Imune , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pneumopatias Parasitárias/imunologia , Pneumopatias Parasitárias/prevenção & controle , Camundongos , Vacinas Protozoárias/uso terapêutico , Schistosoma/efeitos dos fármacos , Schistosoma/imunologia , Esquistossomose/imunologia , Esquistossomose/prevenção & controle , Esquistossomicidas/uso terapêutico
3.
Front Immunol ; 12: 635985, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33746974

RESUMO

Despite mass drug administration programmes with praziquantel, the prevalence of schistosomiasis remains high. A vaccine is urgently needed to control transmission of this debilitating disease. As some promising schistosomiasis vaccine candidates are moving through pre-clinical and clinical testing, we review the immunological challenges that these vaccine candidates may encounter in transitioning through the clinical trial phases in endemic settings. Prior exposure of the target population to schistosomes and other infections may impact vaccine response and efficacy and therefore requires considerable attention. Schistosomes are known for their potential to induce T-reg/IL-10 mediated immune suppression in populations which are chronically infected. Moreover, endemicity of schistosomiasis is focal whereby target and trial populations may exhibit several degrees of prior exposure as well as in utero exposure which may increase heterogeneity of vaccine responses. The age dependent distribution of exposure and development of acquired immunity, and general differences in the baseline immunological profile, adds to the complexity of selecting suitable trial populations. Similarly, prior or concurrent infections with other parasitic helminths, viral and bacterial infections, may alter immunological responses. Consequently, treatment of co-infections may benefit the immunogenicity of vaccines and may be considered despite logistical challenges. On the other hand, viral infections leave a life-long immunological imprint on the human host. Screening for serostatus may be needed to facilitate interpretation of vaccine responses. Co-delivery of schistosome vaccines with PZQ is attractive from a perspective of implementation but may complicate the immunogenicity of schistosomiasis vaccines. Several studies have reported PZQ treatment to induce both transient and long-term immuno-modulatory effects as a result of tegument destruction, worm killing and subsequent exposure of worm antigens to the host immune system. These in turn may augment or antagonize vaccine immunogenicity. Understanding the complex immunological interactions between vaccine, co-infections or prior exposure is essential in early stages of clinical development to facilitate phase 3 clinical trial design and implementation policies. Besides well-designed studies in different target populations using schistosome candidate vaccines or other vaccines as models, controlled human infections could also help identify markers of immune protection in populations with different disease and immunological backgrounds.


Assuntos
Desenvolvimento de Medicamentos , Doenças Endêmicas/prevenção & controle , Vacinas Protozoárias/uso terapêutico , Schistosoma/imunologia , Esquistossomose/prevenção & controle , Animais , Coinfecção , Desenho de Fármacos , Interações Hospedeiro-Parasita , Humanos , Imunogenicidade da Vacina , Praziquantel/uso terapêutico , Vacinas Protozoárias/efeitos adversos , Vacinas Protozoárias/imunologia , Schistosoma/patogenicidade , Esquistossomose/epidemiologia , Esquistossomose/imunologia , Esquistossomose/transmissão , Esquistossomicidas/uso terapêutico
4.
Front Immunol ; 10: 2738, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31824512

RESUMO

Parasites, including African trypanosomes, utilize several immune evasion strategies to ensure their survival and completion of their life cycles within their hosts. The defense factors activated by the host to resolve inflammation and restore homeostasis during active infection could be exploited and/or manipulated by the parasites in an attempt to ensure their survival and propagation. This often results in the parasites evading the host immune responses as well as the host sustaining some self-inflicted collateral tissue damage. During infection with African trypanosomes, both effector and suppressor cells are activated and the balance between these opposing arms of immunity determines susceptibility or resistance of infected host to the parasites. Immune evasion by the parasites could be directly related to parasite factors, (e.g., antigenic variation), or indirectly through the induction of suppressor cells following infection. Several cell types, including suppressive macrophages, myeloid-derived suppressor cells (MDSCs), and regulatory T cells have been shown to contribute to immunosuppression in African trypanosomiasis. In this review, we discuss the key factors that contribute to immunity and immunosuppression during T. congolense infection, and how these factors could aid immune evasion by African trypanosomes. Understanding the regulatory mechanisms that influence resistance and/or susceptibility during African trypanosomiasis could be beneficial in designing effective vaccination and therapeutic strategies against the disease.


Assuntos
Evasão da Resposta Imune , Macrófagos/imunologia , Células Supressoras Mieloides/imunologia , Linfócitos T Reguladores/imunologia , Trypanosoma congolense/imunologia , Tripanossomíase Africana/imunologia , Animais , Humanos , Vacinas Protozoárias/imunologia , Vacinas Protozoárias/uso terapêutico , Tripanossomíase Africana/prevenção & controle
5.
PLoS One ; 14(8): e0220865, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31465461

RESUMO

Although the efforts to develop vaccine against Toxoplasma gondii infection were made for decades, there is currently no licensed vaccine available for humans. Upon discovering a number of T or B cell epitope regions from T. gondii IMC, ROP18 and MIC8, multi-antigen VLPs or combination VLPs were generated. Mice immunized with multi-antigen VLPs or combination VLPs were challenge infected with T. gondii (ME49). T. gondii-specific IgG, IgG isotypes and IgA antibody responses, memory T and B cell responses and protection were evaluated. All the mice survived upon T. gondii challenge infection by multi-antigen VLPs vaccination. Vaccinated mice elicited higher levels of parasite-specific IgG and IgG2a antibody responses in sera, IgA antibody responses in feces, CD4+ and CD8+ T cell responses, and cytokines (IFN-γ, IL-10) responses compared to combination VLPs. In particular, the multi-antigen VLPs vaccination showed significantly higher levels of antibody secreting cell (ASC) responses, CD4+ and CD8+ effector memory T cells, and memory B cells than combination VLPs. Multi-antigen VLPs vaccination showed significant reduction of brain cyst counts and size, and all mice survived. Prediction and analysis of epitopes have indicated that IMC, ROP18 and MIC8 showed partially overlapping epitopes for T and B cells. Our results indicated that antibody responses, memory T and B cells induced by multi-antigen VLPs vaccination might contribute to the complete protection upon T. gondii (ME49) challenge infection.


Assuntos
Proteínas de Protozoários/uso terapêutico , Vacinas Protozoárias/uso terapêutico , Toxoplasma/imunologia , Toxoplasmose/prevenção & controle , Vacinas de Partículas Semelhantes a Vírus/uso terapêutico , Animais , Linfócitos B/imunologia , Feminino , Humanos , Memória Imunológica , Camundongos Endogâmicos BALB C , Proteínas de Protozoários/imunologia , Vacinas Protozoárias/imunologia , Linfócitos T/imunologia , Toxoplasmose/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia
6.
Vaccine ; 36(41): 6124-6132, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30181047

RESUMO

No effective human vaccine against Toxoplasma gondii (T. gondii) has yet been developed; however, a protective vaccine using immunogenic peptides in a safe delivery vehicle system offers promise. Here, we employed bioinformatics to design a multimeric recombinant T. gondii vaccine using predicted T and B cell epitopes of SAG1, AMA1, ROP2, and GRA4 proteins based on their binding capabilities to common major histocompatibility complex (MHC) molecules. Furthermore, we encapsulated the expressed protein in poly lactic-co-glycolic acid (PLGA) nanoparticles as a delivery vehicle and also used alum as an adjuvant to determine the vaccine potency of this multimeric antigen. BALB/c mice were vaccinated and then challenged with T. gondii RH strain, and the survival rate and cytokine profiles were studied. Mice vaccinated with the multi-epitope-based vaccine, both with and without PLGA, had greater Th1 immune responses, survival rates, specific antibody titers, and IFN-γ and IL-2 levels than controls, while the alum-adsorbed vaccine stimulated a Th2-type humoral immune response.


Assuntos
Antígenos de Protozoários/imunologia , Nanopartículas/química , Peptídeos/imunologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Vacinas Protozoárias/imunologia , Vacinas Protozoárias/uso terapêutico , Toxoplasma/imunologia , Toxoplasma/patogenicidade , Adjuvantes Imunológicos , Animais , Anticorpos Antiprotozoários/imunologia , Biologia Computacional , Feminino , Imunidade Humoral/fisiologia , Interferon gama/metabolismo , Interleucina-2/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Proteínas de Protozoários/imunologia
7.
Front Immunol ; 9: 861, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29743881

RESUMO

Infection with the intravascular diecious trematode Schistosoma spp. remains a serious tropical disease and public health problem in the developing world, affecting over 258 million people worldwide. During chronic Schistosoma mansoni infection, complex immune responses to tissue-entrapped parasite eggs provoke granulomatous inflammation which leads to serious damage of the liver and intestine. The suppression of protective host immune mechanisms by helminths promotes parasite survival and benefits the host by reducing tissue damage. However, immune-suppressive cytokines may reduce vaccine-induced immune responses. By combining a single-sex infection system with a murine air pouch model, we were able to demonstrate that male and female schistosomes play opposing roles in modulating the host's immune response. Female schistosomes suppress early innate immune responses to invading cercariae in the skin and upregulate anergy-associated genes. In contrast, male schistosomes trigger strong innate immune reactions which lead to a reduction in worm and egg burden in the liver. Our data suggest that the female worm is a neglected player in the dampening of the host's immune defense system and is therefore a promising target for new immune modulatory therapies.


Assuntos
Cercárias/imunologia , Interações Hospedeiro-Parasita/imunologia , Vacinas Protozoárias/uso terapêutico , Schistosoma mansoni/imunologia , Esquistossomose mansoni/imunologia , Animais , Anticorpos Anti-Helmínticos/imunologia , Biomphalaria/parasitologia , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Tolerância Imunológica/imunologia , Fígado/imunologia , Fígado/parasitologia , Masculino , Camundongos , Vacinas Protozoárias/imunologia , Esquistossomose mansoni/parasitologia , Esquistossomose mansoni/prevenção & controle , Fatores Sexuais
8.
Vet Res ; 47(1): 111, 2016 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-27825377

RESUMO

Coccidiosis is endemic in the commercial broiler industry capable of inflicting devastating economic losses to poultry operations. Vaccines are relatively effective in controlling the disease; their efficacy could potentially be improved with concurrent use of probiotics as evaluated in this study using an Eimeria challenge. Day of hatch 400 Cobb-500 male broilers were assigned to one of four treatment groups including control (CON), vaccine-only gel application (VNC), probiotic-only gel application (NPC), and vaccine-plus-probiotic gel application (VPC). Birds were placed in floor pens (6 replicate pens/treatment, 16-17 birds/pen). NPC and VPC birds received the probiotics in the water on days 2-4, 8, 14-20, 22, 29, and 34-36. On day 15, birds were mildly challenged with 0.5 mL of a mixed oral inoculum of Eimeria sp. prepared with the coccidiosis vaccine at 10× the vaccination dose. Performance measurements were recorded on first day and weekly afterwards, and lesion scores were evaluated 6 days post-challenge. Overall, the probiotics and coccidiosis vaccine resulted in an enhanced protective effect against the challenge, with VPC birds exhibiting lower lesion scores in the duodenum than VNC or NPC birds. Birds in the VPC treatment also demonstrated higher weight gains during days 1-15, days 7-15, and days 21-28 when compared to the VNC birds. These results suggest that the combination of probiotics and coccidiosis vaccines could enhance performance and provide an additional protective effect against a mixed Eimeria challenge.


Assuntos
Coccidiose/veterinária , Eimeria , Doenças das Aves Domésticas/prevenção & controle , Probióticos/uso terapêutico , Vacinas Protozoárias/uso terapêutico , Animais , Galinhas/parasitologia , Coccidiose/prevenção & controle , Eimeria/imunologia , Masculino , Vacinas Protozoárias/imunologia
9.
PLoS One ; 10(9): e0137683, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26367128

RESUMO

In the present study, two Leishmania infantum hypothetical proteins present in the amastigote stage, LiHyp1 and LiHyp6, were combined with a promastigote protein, IgE-dependent histamine-releasing factor (HRF); to compose a polyproteins vaccine to be evaluated against L. infantum infection. Also, the antigenicity of the three proteins was analyzed, and their use for the serodiagnosis of canine visceral leishmaniasis (CVL) was evaluated. The LiHyp1, LiHyp6, and HRF DNA coding sequences were cloned in prokaryotic expression vectors and the recombinant proteins were purified. When employed in ELISA assays, all proteins were recognized by sera from visceral leishmaniasis (VL) dogs, and presented no cross-reactivity with either sera from dogs vaccinated with a Brazilian commercial vaccine, or sera of Trypanosoma cruzi-infected or Ehrlichia canis-infected animals. In addition, the antigens were not recognized by antibodies from non-infected animals living in endemic or non-endemic areas for leishmaniasis. The immunogenicity and protective efficacy of the three proteins administered in the presence of saponin, individually or in combination (composing a polyproteins vaccine), were evaluated in a VL murine model: BALB/c mice infected with L. infantum. Spleen cells from mice inoculated with the individual proteins or with the polyproteins vaccine plus saponin showed a protein-specific production of IFN-γ, IL-12, and GM-CSF after an in vitro stimulation, which was maintained after infection. These animals presented significant reductions in the parasite burden in different evaluated organs, when compared to mice inoculated with saline or saponin. The decrease in parasite burden was associated with an IL-12-dependent production of IFN-γ against parasite total extracts (produced mainly by CD4+ T cells), correlated to the induction of parasite proteins-driven NO production. Mice inoculated with the recombinant protein-based vaccines showed also high levels of parasite-specific IgG2a antibodies. The polyproteins vaccine administration induced a more pronounced Th1 response before and after challenge infection than individual vaccines, which was correlated to a higher control of parasite dissemination to internal organs.


Assuntos
Antígenos de Protozoários/uso terapêutico , Leishmania infantum/imunologia , Leishmaniose Visceral/prevenção & controle , Proteínas de Protozoários/imunologia , Vacinas Protozoárias/uso terapêutico , Animais , Citocinas/metabolismo , Cães , Feminino , Imunidade Humoral , Leishmania infantum/crescimento & desenvolvimento , Camundongos Endogâmicos BALB C , Nitritos/metabolismo , Carga Parasitária
10.
Parasit Vectors ; 8: 121, 2015 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-25885641

RESUMO

Chagas disease is a zoonosis caused by Trypanosoma cruzi in which the most affected organ is the heart. Conventional chemotherapy has a very low effectiveness; despite recent efforts, there is currently no better or more effective treatment available. DNA vaccines provide a new alternative for both prevention and treatment of a variety of infectious disorders, including Chagas disease. Recombinant DNA technology has allowed some vaccines to be developed using recombinant proteins or virus-like particles capable of inducing both a humoral and cellular specific immune response. This type of immunization has been successfully used in preclinical studies and there are diverse models for viral, bacterial and/or parasitic diseases, allergies, tumors and other diseases. Therefore, several research groups have been given the task of designing a DNA vaccine against experimental infection with T. cruzi. In this review we explain what DNA vaccines are and the most recent studies that have been done to develop them with prophylactic or therapeutic purposes against Chagas disease.


Assuntos
Doença de Chagas/prevenção & controle , Doença de Chagas/terapia , Vacinas Protozoárias/imunologia , Vacinas Protozoárias/isolamento & purificação , Vacinas de DNA/imunologia , Vacinas de DNA/isolamento & purificação , Animais , Doença de Chagas/imunologia , Descoberta de Drogas/tendências , Humanos , Vacinas Protozoárias/uso terapêutico , Vacinação/métodos , Vacinas de DNA/uso terapêutico
11.
PLoS Pathog ; 11(1): e1004594, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25617628

RESUMO

Chagas disease (CD), caused by the protozoan Trypanosoma cruzi, is a prototypical neglected tropical disease. Specific immunity promotes acute phase survival. Nevertheless, one-third of CD patients develop chronic chagasic cardiomyopathy (CCC) associated with parasite persistence and immunological unbalance. Currently, the therapeutic management of patients only mitigates CCC symptoms. Therefore, a vaccine arises as an alternative to stimulate protective immunity and thereby prevent, delay progression and even reverse CCC. We examined this hypothesis by vaccinating mice with replication-defective human Type 5 recombinant adenoviruses (rAd) carrying sequences of amastigote surface protein-2 (rAdASP2) and trans-sialidase (rAdTS) T. cruzi antigens. For prophylactic vaccination, naïve C57BL/6 mice were immunized with rAdASP2+rAdTS (rAdVax) using a homologous prime/boost protocol before challenge with the Colombian strain. For therapeutic vaccination, rAdVax administration was initiated at 120 days post-infection (dpi), when mice were afflicted by CCC. Mice were analyzed for electrical abnormalities, immune response and cardiac parasitism and tissue damage. Prophylactic immunization with rAdVax induced antibodies and H-2Kb-restricted cytotoxic and interferon (IFN)γ-producing CD8+ T-cells, reduced acute heart parasitism and electrical abnormalities in the chronic phase. Therapeutic vaccination increased survival and reduced electrical abnormalities after the prime (analysis at 160 dpi) and the boost (analysis at 180 and 230 dpi). Post-therapy mice exhibited less heart injury and electrical abnormalities compared with pre-therapy mice. rAdVax therapeutic vaccination preserved specific IFNγ-mediated immunity but reduced the response to polyclonal stimuli (anti-CD3 plus anti-CD28), CD107a+ CD8+ T-cell frequency and plasma nitric oxide (NO) levels. Moreover, therapeutic rAdVax reshaped immunity in the heart tissue as reduced the number of perforin+ cells, preserved the number of IFNγ+ cells, increased the expression of IFNγ mRNA but reduced inducible NO synthase mRNA. Vaccine-based immunostimulation with rAd might offer a rational alternative for re-programming the immune response to preserve and, moreover, recover tissue injury in Chagas' heart disease.


Assuntos
Cardiomiopatia Chagásica/prevenção & controle , Doença de Chagas/imunologia , Doença de Chagas/terapia , Vacinas Protozoárias/uso terapêutico , Trypanosoma cruzi/imunologia , Adenoviridae/genética , Adenoviridae/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Doença Crônica , Feminino , Fenômenos do Sistema Imunitário , Camundongos , Camundongos Endogâmicos C57BL , Vacinação , Vacinas de DNA/genética , Vacinas de DNA/imunologia
12.
Trends Parasitol ; 29(9): 431-7, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23928100

RESUMO

Toxoplasma gondii is an intracellular parasite that has evolved to actively control its invaded host cells. Toxoplasma triggers then actively regulates host innate interleukin-12 (IL-12) and interferon-γ (IFN-γ) responses that elicit T cell control of infection. A live, nonreplicating avirulent uracil auxotroph vaccine strain (cps) of Toxoplasma triggers novel innate immune responses that stimulate amplified CD8(+) T cell responses and life-long immunity in vaccinated mice. Here, we review recent reports showing that intratumoral treatment with cps activated immune-mediated regression of established solid tumors in mice. We speculate that a better understanding of host-parasite interaction at the molecular level and applying improved genetic models based on Δku80 Toxoplasma strains will stimulate development of highly effective immunotherapeutic cancer vaccine strategies using engineered uracil auxotrophs.


Assuntos
Neoplasias/terapia , Vacinas Protozoárias/uso terapêutico , Toxoplasma/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/uso terapêutico , Imunidade Inata/imunologia , Camundongos , Vacinas Atenuadas/uso terapêutico
13.
PLoS Negl Trop Dis ; 7(4): e2174, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23638195

RESUMO

Visceral leishmaniasis (VL) is a vector-borne disease affecting humans and domestic animals that constitutes a serious public health problem in many countries. Although many antigens have been examined so far as protein- or DNA-based vaccines, none of them conferred complete long-term protection. The use of the lizard non-pathogenic to humans Leishmania (L.) tarentolae species as a live vaccine vector to deliver specific Leishmania antigens is a recent approach that needs to be explored further. In this study, we evaluated the effectiveness of live vaccination in protecting BALB/c mice against L. infantum infection using prime-boost regimens, namely Live/Live and DNA/Live. As a live vaccine, we used recombinant L. tarentolae expressing the L. donovani A2 antigen along with cysteine proteinases (CPA and CPB without its unusual C-terminal extension (CPB(-CTE))) as a tri-fusion gene. For DNA priming, the tri-fusion gene was encoded in pcDNA formulated with cationic solid lipid nanoparticles (cSLN) acting as an adjuvant. At different time points post-challenge, parasite burden and histopathological changes as well as humoral and cellular immune responses were assessed. Our results showed that immunization with both prime-boost A2-CPA-CPB(-CTE)-recombinant L. tarentolae protects BALB/c mice against L. infantum challenge. This protective immunity is associated with a Th1-type immune response due to high levels of IFN-γ production prior and after challenge and with lower levels of IL-10 production after challenge, leading to a significantly higher IFN-γ/IL-10 ratio compared to the control groups. Moreover, this immunization elicited high IgG1 and IgG2a humoral immune responses. Protection in mice was also correlated with a high nitric oxide production and low parasite burden. Altogether, these results indicate the promise of the A2-CPA-CPB(-CTE)-recombinant L. tarentolae as a safe live vaccine candidate against VL.


Assuntos
Leishmaniose Visceral/imunologia , Leishmaniose Visceral/prevenção & controle , Vacinas Protozoárias/uso terapêutico , Vacinas de DNA/uso terapêutico , Vacinas Sintéticas/uso terapêutico , Animais , Anticorpos Antiprotozoários/imunologia , Feminino , Fusão Gênica/genética , Imunidade Humoral/imunologia , Imunoglobulina G/metabolismo , Interferon gama/metabolismo , Interleucina-10/metabolismo , Camundongos , Camundongos Endogâmicos BALB C
14.
PLoS Negl Trop Dis ; 7(4): e2164, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23593527

RESUMO

BACKGROUND: Toll-like receptor (TLR) ligands have been explored as vaccine adjuvants for tumor and virus immunotherapy, but few TLR ligands affecting schistosoma vaccines have been characterized. Previously, we developed a partially protective DNA vaccine encoding the 26-kDa glutathione S-transferase of Schistosoma japonicum (pVAX1-Sj26GST). METHODOLOGY/PRINCIPAL FINDINGS: In this study, we evaluated a TLR7/8 ligand (R848) and a TLR9 ligand (CpG oligodeoxynucleotides, or CpG) as adjuvants for pVAX1-Sj26GST and assessed their effects on the immune system and protection against S. japonicum. We show that combining CpG and R848 with pVAX1-Sj26GST immunization significantly increases splenocyte proliferation and IgG and IgG2a levels, decreases CD4(+)CD25(+)Foxp3(+) regulatory T cells (Treg) frequency in vivo, and enhances protection against S. japonicum. CpG and R848 inhibited Treg-mediated immunosuppression, upregulated the production of interferon (IFN)-γ, tumor necrosis factor (TNF)-α, interleukin (IL)-4, IL-10, IL-2, and IL-6, and decreased Foxp3 expression in vitro, which may contribute to prevent Treg suppression and conversion during vaccination and allow expansion of antigen-specific T cells against pathogens. CONCLUSIONS: Our data shows that selective TLR ligands can increase the protective efficacy of DNA vaccines against schistosomiasis, potentially through combined antagonism of Treg-mediated immunosuppression and conversion.


Assuntos
Imidazóis/uso terapêutico , Oligodesoxirribonucleotídeos/uso terapêutico , Vacinas Protozoárias/uso terapêutico , Schistosoma japonicum/imunologia , Schistosoma japonicum/metabolismo , Esquistossomose/tratamento farmacológico , Esquistossomose/terapia , Receptor 7 Toll-Like/metabolismo , Receptor 8 Toll-Like/metabolismo , Vacinas de DNA/uso terapêutico , Animais , Feminino , Glutationa Transferase/metabolismo , Proteínas de Helminto/agonistas , Proteínas de Helminto/genética , Camundongos , Schistosoma japonicum/patogenicidade , Baço/citologia , Receptor 7 Toll-Like/agonistas , Receptor 8 Toll-Like/agonistas , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/metabolismo
15.
Fish Shellfish Immunol ; 34(3): 885-91, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23291105

RESUMO

In order to elucidate the immune-protective mechanisms of inactivated Cryptocaryon irritans vaccine, different doses of C. irritans theronts were used to immunize orange-spotted grouper (Epinephelus coioides). We measured serum immobilization titer, blood leukocyte respiratory burst activity, serum alternative complement activity, and serum lysozyme activity weekly. In addition, the expression levels of immune-related genes such as interleukin-1ß (IL-1ß), tumor necrosis factor-α (TNF-α), major histocompatibility complexes I and II (MHC I and II), and transforming growth factor-ß1 (TGF-ß1) were determined in spleen and gills. The results showed that the immobilization titer, respiratory burst activity, and alternative complement activity of immunized fish were significantly increased, and the levels of the last two immune parameters in the high-dose vaccine group were significantly higher than in the low-dose vaccine group. Serum lysozyme activity in the high-dose vaccine group was significantly higher than in the PBS control group. Vaccination also regulated host immune-related gene expression. For example, at 2- and 3- weeks post immunization, IL-1ß expression in the high-dose vaccine group spleen was significantly increased. At 4-weeks post immunization, the fish were challenged with a lethal dose of parasite, and the survival rates of high-dose vaccine group, low-dose vaccine group, PBS control group, and adjuvant control group were 80%, 40%, 0%, and 10% respectively. These results demonstrate that inactivated C. irritans vaccination improves specific and nonspecific immune responses in fish, enhancing their anti-parasite ability. These effects are vaccine antigen dose-dependent.


Assuntos
Bass , Infecções por Cilióforos/veterinária , Cilióforos/imunologia , Citocinas/genética , Doenças dos Peixes/imunologia , Vacinas Protozoárias/uso terapêutico , Transcriptoma , Animais , Infecções por Cilióforos/genética , Infecções por Cilióforos/imunologia , Infecções por Cilióforos/prevenção & controle , Citocinas/imunologia , Doenças dos Peixes/genética , Doenças dos Peixes/prevenção & controle , Perfilação da Expressão Gênica/veterinária , Testes Hematológicos/veterinária , Complexo Principal de Histocompatibilidade , Vacinas Protozoárias/administração & dosagem , Fatores de Tempo , Vacinas de Produtos Inativados/uso terapêutico
16.
Invest. clín ; 52(4): 365-375, dic. 2011. ilus
Artigo em Inglês | LILACS | ID: lil-659226

RESUMO

A patient with localized cutaneous leishmaniasis due to Leishmania (Leishmania) amazonensis infection was treated with an antigen containing heat-killed L. (L.) amazonensis promastigotes plus BCG. Expression of T-cell differentiation, memory and senescence receptors markers were analyzed on T cell subpopulations, in order to establish the correlation between the percentages of expression of these receptors and his clinical status, at different stages of his follow up. The following case reports on the achievement of a successful clinical outcome with complete resolution after receiving immunotherapy. A thorough clinical and immunological follow up supporting the healing process of this patient’s lesion is presented in detail.


Un paciente con leishmaniasis cutánea localizada producida por Leishmania (Leishmania) amazonensis fue tratado con un antígeno compuesto por promastigotes de L. (L.) amazonensis muertos por calor combinado con BCG. Se analizó la expresión de distintos receptores de diferenciación, de memoria y de senescencia en las subpoblaciones de células T, con el fin de establecer una relación entre los porcentajes de expresión de dichos receptores y la clínica del paciente en diferentes momentos del seguimiento. Se reporta en este caso un resultado exitoso, con resolución completa de la lesión después de recibir la inmunoterapia, y se presenta en detalle un seguimiento clínico e inmunológico completo durante el proceso de curación.


Assuntos
Adulto , Humanos , Masculino , Antígenos de Protozoários/uso terapêutico , Vacina BCG/uso terapêutico , Imunoterapia Ativa , Leishmania mexicana/imunologia , Leishmaniose Cutânea/terapia , Doenças Profissionais/terapia , Vacinas Protozoárias/uso terapêutico , Antígenos de Protozoários/administração & dosagem , Antígenos de Protozoários/imunologia , Argentina/epidemiologia , Vacina BCG/administração & dosagem , Pesqueiros , Imunidade Celular , Memória Imunológica , Injeções Intradérmicas , Úlcera da Perna/etiologia , Úlcera da Perna/parasitologia , Leishmania mexicana/crescimento & desenvolvimento , Leishmaniose Cutânea/epidemiologia , Leishmaniose Cutânea/imunologia , Leishmaniose Cutânea/parasitologia , Doenças Profissionais/imunologia , Doenças Profissionais/parasitologia , Vacinas Protozoárias/administração & dosagem , Vacinas Protozoárias/imunologia , Subpopulações de Linfócitos T/imunologia , Vacinas de Produtos Inativados
17.
Expert Rev Anti Infect Ther ; 9(11): 1077-86, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22029525

RESUMO

Cryptosporidium spp. that develop in intestinal epithelial cells are responsible for the diarrhoeal disease cryptosporidiosis, which is common in humans of all ages and in neonatal livestock. Following infection, parasite reproduction increases for a number of days before it is blunted and then impeded by innate and adaptive immune responses. Immunocompromised hosts often cannot establish strong immunity and develop chronic infections that can lead to death. Few drugs consistently inhibit parasite reproduction in the host, and chemotherapy might be ineffective in immunodeficient hosts. Future options for prevention or treatment of cryptosporidiosis might include vaccines or recombinant immunological molecules, but this will probably require a better understanding of both the mucosal immune system and intestinal immune responses to the parasite.


Assuntos
Imunidade Adaptativa , Criptosporidiose/tratamento farmacológico , Cryptosporidium parvum/efeitos dos fármacos , Diarreia/tratamento farmacológico , Imunidade Inata , Imunidade nas Mucosas , Animais , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/uso terapêutico , Anticorpos Antiprotozoários/biossíntese , Anticorpos Antiprotozoários/imunologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Linfócitos B/parasitologia , Criptosporidiose/complicações , Criptosporidiose/imunologia , Criptosporidiose/parasitologia , Criptosporidiose/prevenção & controle , Cryptosporidium parvum/imunologia , Citocinas/biossíntese , Diarreia/etiologia , Diarreia/imunologia , Diarreia/parasitologia , Diarreia/prevenção & controle , Humanos , Hospedeiro Imunocomprometido , Imunoterapia , Intestinos/efeitos dos fármacos , Intestinos/imunologia , Intestinos/parasitologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/parasitologia , Vacinas Protozoárias/administração & dosagem , Vacinas Protozoárias/uso terapêutico , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/parasitologia
18.
Immunol Lett ; 138(2): 187-96, 2011 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-21545808

RESUMO

Toxoplasma gondii is an obligate intracellular parasite that infects a variety of mammals and birds. T. gondii also causes human toxoplasmosis; although toxoplasmosis is generally a benign disease, ocular, congenital or reactivated disease is associated with high numbers of disabled people. Infection occurs orally through the ingestion of meat containing cysts or by the intake of food or water contaminated with oocysts. Although the immune system responds to acute infection and mediates the clearance of tachyzoites, parasite cysts persist for the lifetime of the host in tissues such as the eye, muscle, and CNS. However, T. gondii RH strain tachyzoites irradiated with 255Gy do not cause residual infection and induce the same immunity as a natural infection. To assess the humoral response in BALB/c and C57BL/6J mice immunized with irradiated tachyzoites either by oral gavage (p.o.) or intraperitoneal (i.p.) injection, we analyzed total and high-affinity IgG and IgA antibodies in the serum. High levels of antigen-specific IgG were detected in the serum of parenterally immunized mice, with lower levels in mice immunized via the oral route. However, most serum antibodies exhibited low affinity for antigen in both mice strain. We also found antigen specific IgA antibodies in the stools of the mice, especially in orally immunized BALB/c mice. Examination of bone marrow and spleen cells demonstrated that both groups of immunized mice clearly produced specific IgG, at levels comparable to chronic infection, suggesting the generation of IgG specific memory. Next, we challenged i.p. or p.o. immunized mice with cysts from ME49, VEG or P strains of T. gondii. Oral immunization resulted in partial protection as compared to challenged naive mice; these findings were more evident in highly pathogenic ME49 strain challenge. Additionally, we found that while mucosal IgA was important for protection against infection, antigen-specific IgG antibodies were involved with protection against disease and disease pathogenesis. Most antigen responsive cells in culture produced specific high-affinity IgG after immunization, diverse of the findings in serum IgG or from cells after infection, which produced low proportion of high-avidity IgG.


Assuntos
Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/sangue , Imunidade Humoral , Imunoglobulina G/imunologia , Vacinas Protozoárias , Toxoplasma/efeitos dos fármacos , Toxoplasmose Animal , Vacinas Atenuadas , Administração Oral , Animais , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/imunologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/imunologia , Cobalto , Raios gama , Humanos , Imunização/métodos , Imunoglobulina A/sangue , Imunoglobulina A/imunologia , Imunoglobulina G/sangue , Injeções Intravenosas , Estágios do Ciclo de Vida/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Vacinas Protozoárias/administração & dosagem , Vacinas Protozoárias/uso terapêutico , Baço/citologia , Baço/efeitos dos fármacos , Baço/imunologia , Toxoplasma/crescimento & desenvolvimento , Toxoplasma/patogenicidade , Toxoplasmose Animal/sangue , Toxoplasmose Animal/imunologia , Toxoplasmose Animal/prevenção & controle , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/uso terapêutico
19.
Arch Dermatol Res ; 303(6): 399-415, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21328087

RESUMO

A first generation vaccine (AS100-1) was manufactured with protein from four cultured Leishmania species, which proved to be effective in the treatment of psoriasis. A single blind trial on 3,132 psoriasis patients revealed 508 (16.2%) subjects with psoriatic arthritis (PsA) that received AS100-1 antigens. The study group was distributed according to percent psoriasis area and severity index (PASI) reduction from PASI 10 to PASI 100. All groups decreased in arthritis score (AS), tender joints counts and nail changes after treatment; the highest decreased in the PASI 100 group. Relapses of psoriasis and PsA had PASI and AS lower than initial values before treatment. Clinical remissions were at lower doses and less time, after the second course of treatment. Peripheral blood mononuclear cells (PBMC) lymphocyte subsets (LS) varied with PASI range (1-10, 11-20 and 21-72). Pre-treatment, absolute values of gated LS: CD4+, CD8+HLA-, CD8+HLA+, CD8+CD3-, CD8+CD3+ decreased in PBMC as PASI increased, suggesting migration from the blood to the skin. In contrary to the previous finding, the following LS: CD8+CD4-, CD3+CD8-, HLA+CD8-, CD19, CD8+CD4+ and membrane surface immunoglobulin IgA+, IgD+, IgM+, IgE+, and IgG+ increased in PBMC as PASI increased suggesting activation and proliferation by unknown antigens creating a homeostatic cycle between skin/joints and peripheral blood. After nine doses of AS100-1, the following LS: CD8+CD3+, CD8+HLA+, CD3+CD8-, CD4+CD8-, CD8+HLA-, HLA+CD8-, CD8+CD3-, CD19+, CD8+CD4-, CD8+CD4+, IgA+, IgD+, IgM+, IgE+, and IgG+ decreased significantly as compared with values before treatment. The LS decreased stops the vicious cycle between skin/joints and blood explaining clinical remission of lesions.


Assuntos
Artrite Psoriásica/tratamento farmacológico , Linfócitos T CD8-Positivos/metabolismo , Leishmania/imunologia , Subpopulações de Linfócitos/metabolismo , Vacinas Protozoárias/uso terapêutico , Adolescente , Adulto , Idoso , Antígenos CD/metabolismo , Antígenos de Protozoários/imunologia , Artrite Psoriásica/imunologia , Artrite Psoriásica/fisiopatologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Separação Celular , Criança , Pré-Escolar , Progressão da Doença , Feminino , Citometria de Fluxo , Humanos , Imunofenotipagem , Subpopulações de Linfócitos/efeitos dos fármacos , Subpopulações de Linfócitos/imunologia , Subpopulações de Linfócitos/patologia , Masculino , Pessoa de Meia-Idade , Indução de Remissão , Índice de Gravidade de Doença
20.
Vaccine ; 28(3): 597-603, 2010 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-19800443

RESUMO

Leishmune, the first licensed vaccine for prophylaxis against canine visceral leishmaniasis (CVL) and is also immunotherapeutic when used with double saponin adjuvant concentration. The Leishmune therapeutic vaccine was assessed for immunotherapy (IT) in 31 infected dogs and for immunochemotherapy (ICT) in combination with allopurinol or amphotericinB/allopurinol, in 35 dogs. Compared to infected untreated control dogs, at month 3, both treatments increased the proportion of dogs showing intradermal response to Leishmania antigen to a similar extent (from 8 to 67%, in the IT and to 76%, in the ICT groups), and conversely reduced from 100 to 38% (IT) and to 18% (ICT) the proportion of symptomatic cases, from 54 to 12% (IT) and to 15% (ICT) the proportion of parasite evidence in lymph nodes and from 48 to 19% (IT) and 12% (ICT) the proportion of deaths, indicating that the immunotherapy with enriched-Leishmune vaccine promotes the control of the clinical and parasitological signs of CVL rendering most dogs asymptomatic although PCR positive. By month 8, negative lymph node PCR results were obtained in 80% of the ICT-treated dogs, but only in 33% of the IT group (p=0.0253), suggesting that the combination of additional chemotherapy with Leishmune-enriched saponin vaccination abolished, not only the symptoms but also the latent infection condition, curing the dogs. The animals were followed up until 4.5 years after the beginning of the experiment and, compared to the untreated control group at month 3 (12/25 dogs; 48%), a decrease in the rate of CVL deaths was only seen after ICT treatment (7/35 dogs; 20%; 0.0273) but not after IT treatment (10/31 dogs; 32%; p=0.278), pointing out an additional advantage of the ICT treatment with the enriched-Leishmune in the control and cure of CVL.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Doenças do Cão/terapia , Tratamento Farmacológico/métodos , Imunoterapia/métodos , Leishmaniose Visceral/veterinária , Vacinas Protozoárias/uso terapêutico , Saponinas/uso terapêutico , Alopurinol/uso terapêutico , Anfotericina B/uso terapêutico , Animais , Antiprotozoários , Doenças do Cão/patologia , Cães , Quimioterapia Combinada , Seguimentos , Leishmaniose Visceral/patologia , Leishmaniose Visceral/terapia , Linfonodos/parasitologia , Análise de Sobrevida , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA