Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Vaccine ; 38(46): 7246-7257, 2020 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-33012605

RESUMO

BACKGROUND: BK-SE36 is blood-stage malaria vaccine candidate that is undergoing clinical trials. Here, the safety and immunogenicity of BK-SE36 with a novel adjuvant, CpG-ODN(K3) (thus, BK-SE36/CpG) was assessed in a phase 1a trial in Japan. METHODS: An investigator-initiated, randomised, single-blind, placebo-controlled, dose-escalation study was conducted at Osaka University Hospital with 26 healthy malaria naïve Japanese male adults. The trial was conducted in two stages: Stage/Group 1, half-dose (n = 7 for BK-SE36/CpG and n = 3 for control) and Stage/Group 2, full-dose (n = 11 for BK-SE36/CpG and n = 5 for control). There were two intramuscular vaccinations 21 days apart for both half-dose (0.5 ml: 50 µg SE36 + 500 µg aluminum + 500 µg K3) and full-dose (1.0 ml: 100 µg SE36 + 1000 µg aluminum + 1000 µg K3). A one-year follow-up was done to monitor changes in autoimmune markers and vaccine-induced antibody response. RESULTS: BK-SE36/CpG was well tolerated. Vaccination site reactions were similar to those observed with BK-SE36. During the trial and follow-up period, no subject had clinical evidence of autoimmune disease. The full-dose group had significantly higher titres than the half-dose group (Student's t-test, p = 0.002) at 21 days post-second vaccination. Antibody titres remained above baseline values during 12 months of follow-up. The vaccine induced antibody was mostly composed of IgG1 and IgM, and recognised epitopes close to the polyserine region located in the middle of SE36. CONCLUSIONS: BK-SE36/CpG has an acceptable safety profile. Use of CpG-ODN(K3) greatly enhanced immunogenicity in malaria naïve Japanese adults when compared to BK-SE36 alone. The utility of BK-SE36/CpG is currently under evaluation in a malaria endemic setting in West Africa. TRIAL REGISTRATION: JMACCT Clinical Trial Registry JMA-IIA00109.


Assuntos
Vacinas Antimaláricas , Malária Falciparum , Adulto , África Ocidental , Antígenos de Protozoários , Método Duplo-Cego , Seguimentos , Humanos , Japão , Vacinas Antimaláricas/efeitos adversos , Malária Falciparum/prevenção & controle , Masculino , Plasmodium falciparum , Método Simples-Cego
2.
Proc Natl Acad Sci U S A ; 111(8): 3086-91, 2014 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-24516163

RESUMO

CpG DNA, a ligand for Toll-like receptor 9 (TLR9), has been one of the most promising immunotherapeutic agents. Although there are several types of potent humanized CpG oligodeoxynucleotide (ODN), developing "all-in-one" CpG ODNs activating both B cells and plasmacytoid dendritic cells forming a stable nanoparticle without aggregation has not been successful. In this study, we generated a novel nanoparticulate K CpG ODN (K3) wrapped by the nonagonistic Dectin-1 ligand schizophyllan (SPG), K3-SPG. In sharp contrast to K3 alone, K3-SPG stimulates human peripheral blood mononuclear cells to produce a large amount of both type I and type II IFN, targeting the same endosome where IFN-inducing D CpG ODN resides without losing its K-type activity. K3-SPG thus became a potent adjuvant for induction of both humoral and cellular immune responses, particularly CTL induction, to coadministered protein antigens without conjugation. Such potent adjuvant activity of K3-SPG is attributed to its nature of being a nanoparticle rather than targeting Dectin-1 by SPG, accumulating and activating antigen-bearing macrophages and dendritic cells in the draining lymph node. K3-SPG acting as an influenza vaccine adjuvant was demonstrated in vivo in both murine and nonhuman primate models. Taken together, K3-SPG may be useful for immunotherapeutic applications that require type I and type II IFN as well as CTL induction.


Assuntos
Ilhas de CpG/genética , Imunoterapia/métodos , Lectinas Tipo C/metabolismo , Nanopartículas/metabolismo , Oligodesoxirribonucleotídeos/farmacologia , Sizofirano/metabolismo , Receptor Toll-Like 9/agonistas , Adjuvantes Imunológicos/farmacologia , Animais , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Humanos , Indutores de Interferon/farmacologia , Lectinas Tipo C/genética , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica , Oligodesoxirribonucleotídeos/genética , Oligodesoxirribonucleotídeos/metabolismo
3.
PLoS One ; 8(5): e64073, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23724021

RESUMO

BACKGROUND: Up to now a malaria vaccine remains elusive. The Plasmodium falciparum serine repeat antigen-5 formulated with aluminum hydroxyl gel (BK-SE36) is a blood-stage malaria vaccine candidate that has undergone phase 1a trial in malaria-naive Japanese adults. We have now assessed the safety and immunogenicity of BK-SE36 in a malaria endemic area in Northern Uganda. METHODS: We performed a two-stage, randomized, single-blinded, placebo-controlled phase 1b trial (Current Controlled trials ISRCTN71619711). A computer-generated sequence randomized healthy subjects for 2 subcutaneous injections at 21-day intervals in Stage1 (21-40 year-olds) to 1-mL BK-SE36 (BKSE1.0) (n = 36) or saline (n = 20) and in Stage2 (6-20 year-olds) to BKSE1.0 (n = 33), 0.5-mL BK-SE36 (BKSE0.5) (n = 33), or saline (n = 18). Subjects and laboratory personnel were blinded. Safety and antibody responses 21-days post-second vaccination (Day42) were assessed. Post-trial, to compare the risk of malaria episodes 130-365 days post-second vaccination, Stage2 subjects were age-matched to 50 control individuals. RESULTS: Nearly all subjects who received BK-SE36 had induration (Stage1, n = 33, 92%; Stage2, n = 63, 96%) as a local adverse event. No serious adverse event related to BK-SE36 was reported. Pre-existing anti-SE36 antibody titers negatively correlated with vaccination-induced antibody response. At Day42, change in antibody titers was significant for seronegative adults (1.95-fold higher than baseline [95% CI, 1.56-2.43], p = 0.004) and 6-10 year-olds (5.71-fold [95% CI, 2.38-13.72], p = 0.002) vaccinated with BKSE1.0. Immunogenicity response to BKSE0.5 was low and not significant (1.55-fold [95% CI, 1.24-1.94], p = 0.75). In the ancillary analysis, cumulative incidence of first malaria episodes with ≥5000 parasites/µL was 7 cases/33 subjects in BKSE1.0 and 10 cases/33 subjects in BKSE0.5 vs. 29 cases/66 subjects in the control group. Risk ratio for BKSE1.0 was 0.48 (95% CI, 0.24-0.98; p = 0.04). CONCLUSION: BK-SE36 is safe and immunogenic. The promising potential of BK-SE36, observed in the follow-up study, warrants a double-blind phase 1/2b trial in children under 5 years. TRIAL REGISTRATION: Controlled-Trials.com ISRCTN71619711.


Assuntos
Antígenos de Protozoários/imunologia , Estágios do Ciclo de Vida , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Malária Falciparum/prevenção & controle , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/imunologia , Adulto , Animais , Anticorpos Antiprotozoários/imunologia , Seguimentos , Humanos , Estimativa de Kaplan-Meier , Vacinas Antimaláricas/efeitos adversos , Resultado do Tratamento , Uganda , Vacinação , Adulto Jovem
4.
Vaccine ; 30(52): 7658-61, 2012 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-23084848

RESUMO

The effectiveness of influenza vaccines is still controversial, and the role of adjuvants in such vaccines is briefly reviewed in this paper. Inactivated whole virus vaccines may include components that function as adjuvants, meaning that additive adjuvants are often not required. MF59 and AS03 showed higher adjuvanticity than aluminum salts in several clinical studies. Recent research has suggested that immune cell recruitment is the main mechanism underlying adjuvant actions in general, and that aluminum salts induce this recruitment via inflammation at the injected site. The aspect of how oil-based adjuvants, such as MF59 and AS03, recruit immune cells remains to be clarified.


Assuntos
Adjuvantes Imunológicos/farmacologia , Vacinas contra Influenza/imunologia , Compostos de Alúmen/farmacologia , Combinação de Medicamentos , Humanos , Polissorbatos/farmacologia , Esqualeno/farmacologia , Vacinas de Produtos Inativados/imunologia , alfa-Tocoferol/farmacologia
5.
Eur J Immunol ; 41(5): 1365-75, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21469097

RESUMO

Both iron-deficient anemia (IDA) and malaria remain a threat to children in developing countries. Children with IDA are resistant to malaria, but the reasons for this are unknown. In this study, we addressed the mechanisms underlying the protection against malaria observed in IDA individuals using a rodent malaria parasite, Plasmodium yoelii (Py). We showed that the intra-erythrocytic proliferation and amplification of Py parasites were not suppressed in IDA erythrocytes and immune responses specific for Py parasites were not enhanced in IDA mice. We also found that parasitized IDA cells were more susceptible to engulfment by phagocytes in vitro than control cells, resulting in rapid clearance of parasitized cells and that protection of IDA mice from malaria was abrogated by inhibiting phagocytosis. One possible reason for this rapid clearance might be increased exposure of phosphatidylserine at the outer leaflet of parasitized IDA erythrocytes. The results of this study suggest that parasitized IDA erythrocytes are eliminated by phagocytic cells, which sense alterations in the membrane structure of parasitized IDA erythrocytes.


Assuntos
Anemia Ferropriva/imunologia , Eritrócitos/imunologia , Malária/imunologia , Fagocitose/imunologia , Plasmodium yoelii/imunologia , Imunidade Adaptativa , Animais , Linfócitos T CD4-Positivos/imunologia , Cálcio/metabolismo , Ensaio de Imunoadsorção Enzimática , Eritrócitos/química , Eritrócitos/parasitologia , Citometria de Fluxo , Imunidade Inata , Subunidade alfa de Receptor de Interleucina-2/imunologia , Malária/parasitologia , Malária/prevenção & controle , Membranas/química , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Fagócitos/imunologia , Fosfatidilserinas/imunologia , Plasmodium yoelii/crescimento & desenvolvimento , Plasmodium yoelii/patogenicidade
6.
Eur J Immunol ; 40(4): 1053-61, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20101613

RESUMO

When developing malaria vaccines, the most crucial step is to elucidate the mechanisms involved in protective immunity against the parasites. We found that CD8(+) T cells contribute to protective immunity against infection with blood-stage parasites of Plasmodium yoelii. Infection of C57BL/6 mice with P. yoelii 17XL was lethal, while all mice infected with a low-virulence strain of the parasite 17XNL acquired complete resistance against re-infection with P. yoelii 17XL. However, the host mice transferred with CD8(+) T cells from mice primed only with P. yoelii 17XNL failed to acquire protective immunity. On the other hand, the irradiated host mice were completely resistant to P. yoelii 17XL infection, showing no grade of parasitemia when adoptively transferred with CD8(+) T cells from immune mice that survived infection with both P. yoelii XNL and, subsequently, P. yoelii 17XL. These protective CD8(+) T cells from immune WT mice had the potential to generate IFN-gamma, perforin (PFN) and granzyme B. When mice deficient in IFN-gamma were used as donor mice for CD8(+) T cells, protective immunity in the host mice was fully abrogated, and the immunity was profoundly attenuated in PFN-deficient mice. Thus, CD8(+) T cells producing IFN-gamma and PFN appear to be involved in protective immunity against infection with blood-stage malaria.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Malária/imunologia , Parasitemia/imunologia , Plasmodium yoelii/imunologia , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/transplante , Convalescença , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Feminino , Granzimas/biossíntese , Interferon gama/biossíntese , Interferon gama/deficiência , Interferon gama/genética , Macrófagos/imunologia , Malária/prevenção & controle , Vacinas Antimaláricas/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Parasitemia/prevenção & controle , Plasmodium yoelii/patogenicidade , Proteínas Citotóxicas Formadoras de Poros/biossíntese , Proteínas Citotóxicas Formadoras de Poros/deficiência , Proteínas Citotóxicas Formadoras de Poros/genética , Quimera por Radiação , Virulência
7.
Eur J Immunol ; 39(12): 3385-94, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19830724

RESUMO

Proteasome-mediated proteolysis is responsible for the generation of immunogenic epitopes presented by MHC class I molecules, which activate antigen-specific CD8+ T cells. Immunoproteasomes, defined by the presence of the three catalytic subunits LMP2, MECL-1, and LMP7, have been hypothesized to optimize MHC class I antigen processing. In this study, we demonstrate that the infection of mice with a protozoan parasite, Toxoplasma gondii, induced the expression of LMP7 mRNA in APC and increased the capacity of APC to induce the production of IFN-gamma by antigen-specific CD8+ T cells. In vitro infection of a DC cell line with T. gondii also induced the expression of LMP7 and resulted in enhanced proteasome proteolytic activity. Finally, mice lacking LMP7 were highly susceptible to infection with T. gondii and showed a reduced number of functional CD8+ T cells. These results demonstrate that proteasomes containing LMP7 play an indispensable role in the survival of mice infected with T. gondii, presumably due to the efficient generation of CTL epitopes required for the functional development of CD8+ T cells.


Assuntos
Complexos Multienzimáticos/genética , Toxoplasma/imunologia , Toxoplasmose/genética , Toxoplasmose/imunologia , Animais , Western Blotting , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/parasitologia , Células Cultivadas , Cisteína Endopeptidases/biossíntese , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/parasitologia , Indução Enzimática , Expressão Gênica , Predisposição Genética para Doença , Interações Hospedeiro-Patógeno , Imunidade Inata/genética , Imunidade Inata/imunologia , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexos Multienzimáticos/biossíntese , Complexos Multienzimáticos/metabolismo , Complexo de Endopeptidases do Proteassoma , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Toxoplasma/fisiologia , Toxoplasmose/parasitologia
8.
Eur J Immunol ; 39(10): 2822-30, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19728313

RESUMO

Malaria and intestinal nematode infection are widespread and co-infection frequently occurs. We investigated whether co-infected intestinal nematodes modulate immunity against co-existing malaria parasites. Infection of C57BL/6 mice with Plasmodium yoelii 17XNL (Py) was transient and self-limiting, but preceding infection with Heligmosomoides polygyrus (Hp), a mouse intestinal nematode, exacerbated malaria resulting in higher parasite burdens and poor survival of the mice. Co-infection with Hp led to reduced Py-responsive proliferation and IFN-gamma production of spleen cells, and higher activation of CD4(+)CD25(+)Foxp3(+) Treg. In vivo depletion of Treg recovered anti-Py immunity and rescued co-infected mice from exacerbated malaria. However, we did not observe any obvious ex vivo activation of Treg by either Hp products or living worms. Our results suggest that intestinal nematodes moderate host immune responses during acute malaria infection by aggressive activation of Treg. Elucidation of the mechanisms of Treg activation in situ is a target for future analyses.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Malária/imunologia , Nematospiroides dubius/imunologia , Plasmodium yoelii/imunologia , Infecções por Strongylida/imunologia , Linfócitos T Reguladores/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Antígenos de Helmintos/imunologia , Contagem de Células , Concanavalina A/farmacologia , Células Dendríticas/imunologia , Eritrócitos/imunologia , Eritrócitos/parasitologia , Tolerância Imunológica/imunologia , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Interferon gama/metabolismo , Subunidade alfa de Receptor de Interleucina-2/imunologia , Ativação Linfocitária/imunologia , Depleção Linfocítica , Malária/complicações , Malária/parasitologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Parasitemia , Baço/citologia , Baço/imunologia , Infecções por Strongylida/complicações , Análise de Sobrevida , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/metabolismo
9.
Vaccine ; 27(44): 6154-9, 2009 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-19712768

RESUMO

Chagas' disease, caused by infection with the protozoan parasite Trypanosoma cruzi (T. cruzi), is intractable showing a high mortality rate, and the development of effective vaccines is much desired. To examine the efficacy of a new mode of recombinant viral vaccine, we constructed two non-transmissible Sendai viruses (rSeV/dF) encoding the full-length parasite antigen amastigote surface protein-2 (ASP2) or ASP2 fused with a mono-ubiquitin on its N-terminus (UASP2). C57BL/6 mice immunized intranasally with rSeV/dF expressing either ASP2 or UASP2 showed significantly suppressed parasitemia and could be protected from lethal T. cruzi challenge. Depletion of CD8(+) T cells around the time of infection with T. cruzi completely abolished this protection, confirming that acquired immunity against the infection of T. cruzi is dependent on CD8(+) T cells. We also demonstrated that the protective immunity correlated with higher secretion of interferon-gamma (IFN-gamma) by spleen cells on in vitro-specific or non-specific stimulation. Increased CTL activity was also confirmed by degranulation or CTL assays. Interestingly, the control virus, rSeV/dF-GFP, induced even a higher IFN-gamma production from spleen cells following non-specific but not specific stimulation in vitro, suggesting that SeV may also be a good adjuvant when used as a vaccine vehicle. Taking together, the current findings indicate that recombinant Sendai virus expressing the ASP2 or UASP2 antigens of T. cruzi are interesting candidates for the development of a new mode of recombinant viral vaccine against Chagas' disease.


Assuntos
Doença de Chagas/prevenção & controle , Neuraminidase/imunologia , Vacinas Protozoárias/imunologia , Trypanosoma cruzi/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Doença de Chagas/imunologia , Feminino , Interferon gama/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Vacinas Protozoárias/genética , Vírus Sendai/imunologia , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
10.
Am J Trop Med Hyg ; 79(6): 819-22, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19052285

RESUMO

We investigated whether concurrent infection with Heligmosomoides polygyrus, an intestinal nematode, modulated anti-malaria parasite immunity and development of experimental cerebral malaria (ECM) in mice. The C57BL/6 mice infected with Plasmodium berghei ANKA showed typical symptoms of ECM. Interestingly, preceding H. polygyrus infection did not alter ECM development, despite accelerated P. berghei growth in vivo. Our observation provides a new insight that ECM can be induced in a fashion independent of the immune responses affected by concurrent H. polygyrus. Differentiation between protective immunity and infection-associated host-damaging inflammatory response is urgently required for understanding the pathogenesis of cerebral malaria.


Assuntos
Malária Cerebral/complicações , Malária Cerebral/imunologia , Nematospiroides dubius , Plasmodium berghei , Infecções por Strongylida/complicações , Animais , Fezes/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Contagem de Ovos de Parasitas , Parasitemia , Fatores de Tempo
11.
J Immunol ; 180(4): 2496-503, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18250459

RESUMO

Malaria is still a life-threatening infectious disease that continues to produce 2 million deaths annually. Malaria parasites have acquired immune escape mechanisms and prevent the development of sterile immunity. Regulatory T cells (Tregs) have been reported to contribute to immune evasion during malaria in mice and humans, suggesting that activating Tregs is one of the mechanisms by which malaria parasites subvert host immune systems. However, little is known about how these parasites activate Tregs. We herein show that TLR9 signaling to dendritic cells (DCs) is crucial for activation of Tregs. Infection of mice with the rodent malaria parasite Plasmodium yoelii activates Tregs, leading to enhancement of their suppressive function. In vitro activation of Tregs requires the interaction of DCs with parasites in a TLR9-dependent manner. Furthermore, TLR9(-/-) mice are partially resistant to lethal infection, and this is associated with impaired activation of Tregs and subsequent development of effector T cells. Thus, malaria parasites require TLR9 to activate Tregs for immune escape.


Assuntos
Interações Hospedeiro-Parasita/imunologia , Ativação Linfocitária/imunologia , Malária/imunologia , Plasmodium yoelii/imunologia , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/parasitologia , Receptor Toll-Like 9/fisiologia , Animais , Células Cultivadas , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/parasitologia , Malária/metabolismo , Malária/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Plasmodium yoelii/patogenicidade , Transdução de Sinais/genética , Linfócitos T Reguladores/metabolismo , Receptor Toll-Like 9/deficiência , Receptor Toll-Like 9/genética
12.
Eur J Immunol ; 35(12): 3516-24, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16304635

RESUMO

CD4+ T cells are the major effector T cells against blood-stage Plasmodium yoelii infection. On the other hand, the lethal strain of P. yoelii (PyL) has acquired an escape mechanism from host T cell immunity by activating CD4+CD25+ regulatory T cells (Treg). Although the activation of Treg during PyL infection precludes the clearance of PyL from mice, it remains unclear whether activation of Treg is attributable to a specific response against PyL infection. Thus, we examined here whether Treg proliferate in an antigen-dependent manner during PyL infection. We also investigated the effector and regulatory mechanisms of Treg. Infection with PyL increased the number of CD4+CD25+ T cells, in which expression of Foxp3 mRNA is up-regulated. The Treg that were transferred into mice infected with PyL, but not with a non-lethal strain of P. yoelii (PyNL), proliferated during the initial 5 days following infection. The Treg from PyL-infected mice showed strong suppression compared with those from naive or PyNL-infected mice, and could suppress T cell activation by recognizing PyL- but not PyNL-derived antigens. Furthermore, the suppressive function of Treg activated in PyL-infected but not in naive mice could not be inhibited by treatment with an anti-glucocorticoid-induced TNFR family-related protein (GITR) mAb. These findings indicate that PyL infection specifically activates Treg that are specific for PyL-derived antigens. The infection also induces resistance for Treg to GITR signaling, and this eventually contributes to the escape of parasites from host T cell immunity.


Assuntos
Malária/imunologia , Plasmodium yoelii/imunologia , Receptores de Fator de Crescimento Neural/fisiologia , Receptores do Fator de Necrose Tumoral/fisiologia , Tolerância a Antígenos Próprios , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Antiprotozoários/farmacologia , Feminino , Proteína Relacionada a TNFR Induzida por Glucocorticoide , Imunidade Inata , Malária/metabolismo , Malária/mortalidade , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium yoelii/patogenicidade , Receptores de Fator de Crescimento Neural/imunologia , Receptores do Fator de Necrose Tumoral/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...