Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Infect Immun ; 88(10)2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32719159

RESUMEN

The circumsporozoite protein (CSP) builds up the surface coat of sporozoites and is the leading malaria pre-erythrocytic-stage vaccine candidate. CSP has been shown to induce robust CD8+ T cell responses that are capable of eliminating developing parasites in hepatocytes, resulting in protective immunity. In this study, we characterized the importance of the immunodominant CSP-derived epitope SYIPSAEKI of Plasmodium berghei in both sporozoite- and vaccine-induced protection in murine infection models. In BALB/c mice, where SYIPSAEKI is efficiently presented in the context of the major histocompatibility complex class I (MHC-I) molecule H-2-Kd, we established that epitope-specific CD8+ T cell responses contribute to parasite killing following sporozoite immunization. Yet, sterile protection was achieved in the absence of this epitope, substantiating the concept that other antigens can be sufficient for parasite-induced protective immunity. Furthermore, we demonstrated that SYIPSAEKI-specific CD8+ T cell responses elicited by viral-vectored CSP-expressing vaccines effectively targeted parasites in hepatocytes. The resulting sterile protection strictly relied on the expression of SYIPSAEKI. In C57BL/6 mice, which are unable to present the immunodominant epitope, CSP-based vaccines did not confer complete protection, despite the induction of high levels of CSP-specific antibodies. These findings underscore the significance of CSP in protection against malaria pre-erythrocytic stages and demonstrate that a significant proportion of the protection against the parasite is mediated by CD8+ T cells specific for the immunodominant CSP-derived epitope.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Vacunas contra la Malaria/inmunología , Malaria/prevención & control , Plasmodium berghei/inmunología , Proteínas Protozoarias/inmunología , Animales , Anticuerpos Antiprotozoarios/sangre , Anticuerpos Antiprotozoarios/inmunología , Presentación de Antígeno , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Modelos Animales de Enfermedad , Epítopos de Linfocito T/química , Inmunización , Malaria/inmunología , Malaria/parasitología , Vacunas contra la Malaria/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fragmentos de Péptidos , Proteínas Protozoarias/química , Especificidad de la Especie , Esporozoítos/inmunología
2.
Infect Immun ; 86(9)2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29986894

RESUMEN

Vivax malaria remains one of the most serious and neglected tropical diseases, with 132 to 391 million clinical cases per year and 2.5 billion people at risk of infection. A vaccine against Plasmodium vivax could have more impact than any other intervention, and the use of a vaccine targeting multiple antigens may result in higher efficacy against sporozoite infection than targeting a single antigen. Here, two leading P. vivax preerythrocytic vaccine candidate antigens, the P. vivax circumsporozoite protein (PvCSP) and the thrombospondin-related adhesion protein (PvTRAP) were delivered as a combined vaccine. This strategy provided a dose-sparing effect, with 100% sterile protection in mice using doses that individually conferred low or no protection, as with the unadjuvanted antigens PvTRAP (0%) and PvCSP (50%), and reached protection similar to that of adjuvanted components. Efficacy against malaria infection was assessed using a new mouse challenge model consisting of a double-transgenic Plasmodium berghei parasite simultaneously expressing PvCSP and PvTRAP used in mice immunized with the virus-like particle (VLP) Rv21 previously reported to induce high efficacy in mice using Matrix-M adjuvant, while PvTRAP was concomitantly administered in chimpanzee adenovirus and modified vaccinia virus Ankara (MVA) vectors (viral-vectored TRAP, or vvTRAP) to support effective induction of T cells. We examined immunity elicited by these vaccines in the context of two adjuvants approved for human use (AddaVax and Matrix-M). Matrix-M supported the highest anti-PvCSP antibody titers when combined with Rv21, and, interestingly, mixing PvCSP Rv21 and PvTRAP viral vectors enhanced immunity to malaria over levels provided by single vaccines.


Asunto(s)
Vacunas contra la Malaria/inmunología , Malaria Vivax/prevención & control , Plasmodium vivax/inmunología , Proteínas Protozoarias/inmunología , Adenoviridae/genética , Adyuvantes Inmunológicos , Animales , Anticuerpos Antiprotozoarios/sangre , Antígenos de Protozoos/inmunología , Femenino , Vectores Genéticos , Malaria Vivax/inmunología , Ratones , Ratones Endogámicos BALB C , Nanopartículas/administración & dosificación , Plasmodium berghei/genética , Plasmodium berghei/inmunología , Polisorbatos/administración & dosificación , Proteínas Protozoarias/administración & dosificación , Saponinas/administración & dosificación , Escualeno/administración & dosificación , Vacunas de Partículas Similares a Virus/administración & dosificación , Vacunas de Partículas Similares a Virus/genética , Vacunas de Partículas Similares a Virus/inmunología , Virus Vaccinia/genética
3.
Curr Hypertens Rep ; 18(4): 33, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27059041

RESUMEN

Persistently raised blood pressure is one of the major risk factors for diseases such as myocardial infarction and stroke. Uncontrolled hypertension is also associated with high rates of mortality, particularly in middle and high-income countries. Lifestyle factors such as poor diet, obesity, physical inactivity and smoking are all thought to contribute to the development of hypertension. As a result, the management of hypertension should begin with modifying these lifestyle factors. Beyond this, drug interventions are used as the predominant form of management. However, adherence to medications can be highly variable, medication side effects are common, and may require regular monitoring or, in some individuals may be ineffective. Therefore, additional non-pharmacologic interventions that lower blood pressure may be advantageous when combined with lifestyle modifications. Such interventions may include relaxation therapies such as slow breathing exercises, which can be initiated by means of specific devices. The technique of device-guided breathing (DGB) has been considered by guideline developers in the management of hypertension. One specific device, the Resperate, has received US FDA and UK NHS approval over the last few years. In this review, we summarise the evidence base on efficacy and find that although some clinical trials exist that demonstrate a BP-lowering effect, others do not. There is currently insufficient evidence from pooled data to recommend the routine use of device-guided breathing in hypertensive patients.


Asunto(s)
Hipertensión/fisiopatología , Respiración , Presión Sanguínea/fisiología , Ejercicios Respiratorios , Humanos , Factores de Riesgo
4.
Infect Immun ; 84(3): 622-34, 2015 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-26667840

RESUMEN

The leading malaria vaccine candidate, RTS,S, based on the Plasmodium falciparum circumsporozoite protein (CSP), will likely be the first publicly adopted malaria vaccine. However, this and other subunit vaccines, such as virus-vectored thrombospondin-related adhesive protein (TRAP), provide only intermediate to low levels of protection. In this study, the Plasmodium berghei homologues of antigens CSP and TRAP are combined. TRAP is delivered using adenovirus- and vaccinia virus-based vectors in a prime-boost regime. Initially, CSP is also delivered using these viral vectors; however, a reduction of anti-CSP antibodies is seen when combined with virus-vectored TRAP, and the combination is no more protective than either subunit vaccine alone. Using an adenovirus-CSP prime, protein-CSP boost regime, however, increases anti-CSP antibody titers by an order of magnitude, which is maintained when combined with virus-vectored TRAP. This combination regime using protein CSP provided 100% protection in C57BL/6 mice compared to no protection using virus-vectored TRAP alone and 40% protection using adenovirus-CSP prime and protein-CSP boost alone. This suggests that a combination of CSP and TRAP subunit vaccines could enhance protection against malaria.


Asunto(s)
Eritrocitos/parasitología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/inmunología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Animales , Anticuerpos Antiprotozoarios/inmunología , Eritrocitos/inmunología , Femenino , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Vacunas contra la Malaria/genética , Malaria Falciparum/parasitología , Malaria Falciparum/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Virus Vaccinia/genética , Virus Vaccinia/metabolismo
5.
PLoS Pathog ; 9(5): e1003303, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23675294

RESUMEN

CD8⁺ T cells mediate immunity against Plasmodium liver stages. However, the paucity of parasite-specific epitopes of CD8⁺ T cells has limited our current understanding of the mechanisms influencing the generation, maintenance and efficiency of these responses. To identify antigenic epitopes in a stringent murine malaria immunisation model, we performed a systematic profiling of H(2b)-restricted peptides predicted from genome-wide analysis. We describe the identification of Plasmodium berghei (Pb) sporozoite-specific gene 20 (S20)- and thrombospondin-related adhesive protein (TRAP)-derived peptides, termed PbS20318 and PbTRAP130 respectively, as targets of CD8⁺ T cells from C57BL/6 mice vaccinated by whole parasite strategies known to protect against sporozoite challenge. While both PbS20318 and PbTRAP130 elicit effector and effector memory phenotypes in both the spleens and livers of immunised mice, only PbTRAP130-specific CD8⁺ T cells exhibit in vivo cytotoxicity. Moreover, PbTRAP130-specific, but not PbS20318-specific, CD8⁺ T cells significantly contribute to inhibition of parasite development. Prime/boost vaccination with PbTRAP demonstrates CD8⁺ T cell-dependent efficacy against sporozoite challenge. We conclude that PbTRAP is an immunodominant antigen during liver-stage infection. Together, our results underscore the presence of CD8⁺ T cells with divergent potencies against distinct Plasmodium liver-stage epitopes. Our identification of antigen-specific CD8⁺ T cells will allow interrogation of the development of immune responses against malaria liver stages.


Asunto(s)
Antígenos de Protozoos/inmunología , Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Malaria/inmunología , Secuencia de Aminoácidos , Animales , Antígenos de Protozoos/aislamiento & purificación , Epítopos de Linfocito T/aislamiento & purificación , Estudio de Asociación del Genoma Completo , Hígado/parasitología , Ratones , Ratones Endogámicos C57BL , Plasmodium berghei , Proteínas Protozoarias/inmunología , Proteínas Protozoarias/aislamiento & purificación , Esporozoítos/inmunología
6.
Infect Immun ; 82(3): 1277-86, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24379295

RESUMEN

Plasmodium vivax is the world's most widely distributed malaria parasite and a potential cause of morbidity and mortality for approximately 2.85 billion people living mainly in Southeast Asia and Latin America. Despite this dramatic burden, very few vaccines have been assessed in humans. The clinically relevant vectors modified vaccinia virus Ankara (MVA) and the chimpanzee adenovirus ChAd63 are promising delivery systems for malaria vaccines due to their safety profiles and proven ability to induce protective immune responses against Plasmodium falciparum thrombospondin-related anonymous protein (TRAP) in clinical trials. Here, we describe the development of new recombinant ChAd63 and MVA vectors expressing P. vivax TRAP (PvTRAP) and show their ability to induce high antibody titers and T cell responses in mice. In addition, we report a novel way of assessing the efficacy of new candidate vaccines against P. vivax using a fully infectious transgenic Plasmodium berghei parasite expressing P. vivax TRAP to allow studies of vaccine efficacy and protective mechanisms in rodents. Using this model, we found that both CD8+ T cells and antibodies mediated protection against malaria using virus-vectored vaccines. Our data indicate that ChAd63 and MVA expressing PvTRAP are good preerythrocytic-stage vaccine candidates with potential for future clinical application.


Asunto(s)
Adenoviridae/inmunología , Vacunas contra la Malaria/inmunología , Malaria Vivax/inmunología , Plasmodium berghei/inmunología , Plasmodium vivax/inmunología , Proteínas Protozoarias/genética , Virus Vaccinia/inmunología , Adenoviridae/genética , Animales , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Linfocitos T CD8-positivos/inmunología , Culicidae/inmunología , Femenino , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Vacunas contra la Malaria/genética , Malaria Vivax/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Pan troglodytes/inmunología , Pan troglodytes/virología , Plasmodium berghei/genética , Proteínas Protozoarias/inmunología , Vaccinia/genética , Vaccinia/inmunología , Virus Vaccinia/genética
7.
Artículo en Inglés | MEDLINE | ID: mdl-38452888

RESUMEN

OBJECTIVES: To identify preoperative predictors of postcardiotomy cardiogenic shock in patients with ischemic and nonischemic cardiomyopathy and evaluate trajectory of postoperative ventricular function. METHODS: From January 2017 to January 2020, 238 patients with ejection fraction <30% (206/238) or 30% to 34% with at least moderately severe mitral regurgitation (32/238) underwent conventional cardiac surgery at Cleveland Clinic, 125 with ischemic and 113 with nonischemic cardiomyopathy. Preoperative ejection fraction was 25 ± 4.5%. The primary outcome was postcardiotomy cardiogenic shock, defined as need for microaxial temporary left ventricular assist device, extracorporeal membrane oxygenation, or vasoactive-inotropic score >25. RandomForestSRC was used to identify its predictors. RESULTS: Postcardiotomy cardiogenic shock occurred in 27% (65/238). Pulmonary artery pulsatility index <3.5 and pulmonary capillary wedge pressure >19 mm Hg were the most important factors predictive of postcardiotomy cardiogenic shock in ischemic cardiomyopathy. Cardiac index <2.2 L·min-1 m-2 and pulmonary capillary wedge pressure >21 mm Hg were the most important predictive factors in nonischemic cardiomyopathy. Operative mortality was 1.7%. Ejection fraction at 12 months after surgery increased to 39% (confidence interval, 35-40%) in the ischemic group and 37% (confidence interval, 35-38%) in the nonischemic cardiomyopathy group. CONCLUSIONS: Predictors of postcardiotomy cardiogenic shock were different in ischemic and nonischemic cardiomyopathy. Right heart dysfunction, indicated by low pulmonary artery pulsatility index, was the most important predictor in ischemic cardiomyopathy, whereas greater degree of cardiac decompensation was the most important in nonischemic cardiomyopathy. Therefore, preoperative right heart catheterization will help identify patients with low ejection fraction who are at greater risk of postcardiotomy cardiogenic shock.

8.
J Immunol ; 187(3): 1347-57, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21715686

RESUMEN

Identification of correlates of protection for infectious diseases including malaria is a major challenge and has become one of the main obstacles in developing effective vaccines. We investigated protection against liver-stage malaria conferred by vaccination with adenoviral (Ad) and modified vaccinia Ankara (MVA) vectors expressing pre-erythrocytic malaria Ags. By classifying CD8(+) T cells into effector, effector memory (T(EM)), and central memory subsets using CD62L and CD127 markers, we found striking differences in T cell memory generation. Although MVA induced accelerated central memory T cell generation, which could be efficiently boosted by subsequent Ad administration, it failed to protect against malaria. In contrast, Ad vectors, which permit persistent Ag delivery, elicit a prolonged effector T cell and T(EM) response that requires long intervals for an efficient boost. A preferential T(EM) phenotype was maintained in liver, blood, and spleen after Ad/MVA prime-boost regimens, and animals were protected against malaria sporozoite challenge. Blood CD8(+) T(EM) cells correlated with protection against malaria liver-stage infection, assessed by estimation of number of parasites emerging from the liver into the blood. The protective ability of Ag-specific T(EM) cells was confirmed by transfer experiments into naive recipient mice. Thus, we identify persistent CD8 T(EM) populations as essential for vaccine-induced pre-erythrocytic protection against malaria, a finding that has important implications for vaccine design.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/parasitología , Memoria Inmunológica , Parasitosis Hepáticas/inmunología , Parasitosis Hepáticas/prevención & control , Malaria/inmunología , Malaria/prevención & control , Adenoviridae/genética , Adenoviridae/inmunología , Animales , Linfocitos T CD8-positivos/patología , Epítopos de Linfocito T/administración & dosificación , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/inmunología , Vectores Genéticos/uso terapéutico , Antígenos H-2/administración & dosificación , Antígenos H-2/genética , Antígenos H-2/inmunología , Humanos , Memoria Inmunológica/genética , Parasitosis Hepáticas/patología , Malaria/patología , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/genética , Vacunas contra la Malaria/inmunología , Ratones , Ratones Endogámicos BALB C , Plasmodium berghei/genética , Plasmodium berghei/inmunología , Proteínas Protozoarias/administración & dosificación , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología , Transgenes/inmunología , Vaccinia/genética , Vaccinia/inmunología
9.
Mol Ther ; 20(8): 1633-47, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22354374

RESUMEN

Substantial protection can be provided against the pre-erythrocytic stages of malaria by vaccination first with an adenoviral and then with an modified vaccinia virus Ankara (MVA) poxviral vector encoding the same ME.TRAP transgene. We investigated whether the two vaccine components adenovirus (Ad) and MVA could be coinjected as a mixture to enhance protection against malaria. A single-shot mixture at specific ratios of Ad and MVA (Ad+MVA) enhanced CD8(+) T cell-dependant protection of mice against challenge with Plasmodium berghei. Moreover, the degree of protection could be enhanced after homologous boosting with the same Ad+MVA mixture to levels comparable with classic heterologous Ad prime-MVA boost regimes. The mixture increased transgene-specific responses while decreasing the CD8(+) T cell antivector immunity compared to each vector used alone, particularly against the MVA backbone. Mixed vector immunization led to increased early circulating interferon-γ (IFN-γ) response levels and altered transcriptional microarray profiles. Furthermore, we found that sequential immunizations with the Ad+MVA mixture led to consistent boosting of the transgene-specific CD8(+) response for up to three mixture immunizations, whereas each vector used alone elicited progressively lower responses. Our findings offer the possibility of simplifying the deployment of viral vectors as a single mixture product rather than in heterologous prime-boost regimens.


Asunto(s)
Adenoviridae/inmunología , Inmunización/métodos , Virus Vaccinia/inmunología , Adenoviridae/genética , Animales , Linfocitos T CD8-positivos/inmunología , Femenino , Vectores Genéticos/genética , Malaria/inmunología , Malaria/prevención & control , Ratones , Ratones Endogámicos BALB C , Plasmodium berghei/inmunología , Virus Vaccinia/genética
10.
JACC Clin Electrophysiol ; 7(4): 494-501, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33358671

RESUMEN

OBJECTIVES: The objective of this study was to describe the risk of cardiac implantable electronic devices (CIEDs) complications in patients with left ventricular assist devices (LVADs). BACKGROUND: Patients with LVADs are predisposed to ventricular arrhythmias and frequently have CIEDs before receiving their LVAD. However, the role of CIED procedures such as generator changes (GC) are unclear in this population, given the potential complications of bleeding and infection. METHODS: This was a retrospective, multicenter study from January 1, 2012, to September 30, 2018. All patients with LVADs were screened and those who had a CIED GC, implantation, or revision were included in the study and followed until December 31, 2018. RESULTS: A total of 179 patients across 6 centers had a CIED procedure after LVAD implantation. The mean age was 59.5 ± 13.4, with the cohort comprising mostly men (78%), destination LVAD therapy (53.8%), and GC (66%). The 30-day primary composite endpoint of hematoma or device infection occurred in 34 (19%) patients. The secondary endpoints of rehospitalization within 30 days and appropriate device therapy during follow-up occurred in 40 (22%) and 42 (24%) patients respectively. Of the 126 patients without previous device therapy, 14.3% received appropriate therapy during follow-up. CONCLUSIONS: In this large, multicenter cohort, we report the incidence of complications for CIED procedures in the LVAD population; specifically, LVAD patients are at increased risk of pocket hematomas, without downstream risk of infection, and do experience a high rate of appropriate device therapies.


Asunto(s)
Insuficiencia Cardíaca , Corazón Auxiliar , Electrónica , Insuficiencia Cardíaca/epidemiología , Corazón Auxiliar/efectos adversos , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Estudios Retrospectivos
11.
PLoS One ; 14(1): e0209028, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30625136

RESUMEN

Malaria remains one the world's most deadly infectious diseases, with almost half a million deaths and over 150 million clinical cases each year. An effective vaccine would contribute enormously to malaria control and will almost certainly be required for eventual eradication of the disease. However, the leading malaria vaccine candidate, RTS,S, shows only 30-50% efficacy under field conditions, making it less cost-effective than long-lasting insecticide treated bed nets. Other subunit malaria vaccine candidates, including TRAP-based vaccines, show no better protective efficacy. This has led to increased interest in combining subunit malaria vaccines as a means of enhancing protective efficacy. Mathematical models of the effect of combining such vaccines on protective efficacy can help inform optimal vaccine strategies and decision-making at all stages of the clinical process. So far, however, no such model has been developed for pre-clinical murine studies, the stage at which all candidate antigens and combinations begin evaluation. To address this gap, this paper develops a mathematical model of vaccine combination adapted to murine malaria studies. The model is based on simple probabilistic assumptions which put the model on a firmer theoretical footing than previous clinical models, which rather than deriving a relationship between immune responses and protective efficacy posit the relationship to be either exponential or Hill curves. Data from pre-clinical murine malaria studies are used to derive values for unknowns in the model which in turn allows simulations of vaccine combination efficacy and suggests optimal strategies to pursue. Finally, the ability of the model to shed light on fundamental biological variables of murine malaria such as the blood stage growth rate and sporozoite infectivity is explored.


Asunto(s)
Malaria/prevención & control , Modelos Teóricos , Animales , Femenino , Vacunas contra la Malaria/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas Protozoarias/metabolismo
12.
Sci Rep ; 7: 46482, 2017 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-28417968

RESUMEN

Development of a protective and broadly-acting vaccine against the most widely distributed human malaria parasite, Plasmodium vivax, will be a major step towards malaria elimination. However, a P. vivax vaccine has remained elusive by the scarcity of pre-clinical models to test protective efficacy and support further clinical trials. In this study, we report the development of a highly protective CSP-based P. vivax vaccine, a virus-like particle (VLP) known as Rv21, able to provide 100% sterile protection against a stringent sporozoite challenge in rodent models to malaria, where IgG2a antibodies were associated with protection in absence of detectable PvCSP-specific T cell responses. Additionally, we generated two novel transgenic rodent P. berghei parasite lines, where the P. berghei csp gene coding sequence has been replaced with either full-length P. vivax VK210 or the allelic VK247 csp that additionally express GFP-Luciferase. Efficacy of Rv21 surpassed viral-vectored vaccination using ChAd63 and MVA. We show for the first time that a chimeric VK210/247 antigen can elicit high level cross-protection against parasites expressing either CSP allele, which provide accessible and affordable models suitable to support the development of P. vivax vaccines candidates. Rv21 is progressing to GMP production and has entered a path towards clinical evaluation.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Vacunas contra la Malaria , Malaria Vivax , Plasmodium vivax/inmunología , Proteínas Protozoarias , Vacunación , Animales , Vacunas contra la Malaria/genética , Vacunas contra la Malaria/inmunología , Vacunas contra la Malaria/farmacología , Malaria Vivax/genética , Malaria Vivax/inmunología , Malaria Vivax/patología , Malaria Vivax/prevención & control , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Ratones Transgénicos , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología , Proteínas Protozoarias/farmacología
13.
J Evid Based Med ; 8(2): 108-10, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25955430

RESUMEN

In post-Soviet countries, where medical practice largely relies on experience alone, the incorporation of the best research evidence in clinical practice is limited. In order to promote the awareness and utilization of evidence-based medicine (EBM) among Lithuanian doctors, we organized EBM conferences in each of the two Lithuanian medical schools. More than 500 medical professionals and students attended the conferences in Vilnius (2013) and Kaunas (2014) demonstrating that there is a high demand for formal EBM teaching. Building on the success of these seminal conferences, and to start addressing the lack of EBM practice in the country, the first Lithuanian Centre for Evidence-Based Medicine was established at Vilnius University Medical Faculty in 2014. The Centre will focus on the implementation of EBM teaching in medical school curriculum, formulating management guidelines, writing systematic reviews and supporting Lithuanian authors in doing so.


Asunto(s)
Actitud del Personal de Salud , Curriculum , Medicina Basada en la Evidencia/educación , Facultades de Medicina/normas , Enseñanza/normas , Humanos , Lituania
14.
PLoS One ; 10(3): e0119880, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25822951

RESUMEN

The development of an efficacious vaccine against the Plasmodium parasite remains a top priority. Previous research has demonstrated the ability of a prime-boost virally vectored sub-unit vaccination regimen, delivering the liver-stage expressed malaria antigen TRAP, to produce high levels of antigen-specific T cells. The liver-stage of malaria is the main target of T cell-mediated immunity, yet a major challenge in assessing new T cell inducing vaccines has been the lack of a suitable pre-clinical assay. We have developed a flow-cytometry based in vitro T cell killing assay using a mouse hepatoma cell line, Hepa1-6, and Plasmodium berghei GFP expressing sporozoites. Using this assay, P. berghei TRAP-specific CD8+ T cell enriched splenocytes were shown to inhibit liver-stage parasites in an effector-to-target ratio dependent manner. Further development of this assay using human hepatocytes and P. falciparum would provide a new method to pre-clinically screen vaccine candidates and to elucidate mechanisms of protection in vitro.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra la Malaria/inmunología , Plasmodium berghei/inmunología , Proteínas Protozoarias/inmunología , Animales , Antígenos de Protozoos/inmunología , Línea Celular , Femenino , Proteínas Fluorescentes Verdes/genética , Humanos , Técnicas In Vitro , Hígado/inmunología , Hígado/parasitología , Malaria/inmunología , Malaria/prevención & control , Ratones , Ratones Endogámicos C57BL , Plasmodium berghei/genética , Plasmodium berghei/patogenicidad , Bazo/citología , Bazo/inmunología , Esporozoítos/inmunología
15.
Sci Rep ; 5: 16756, 2015 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-26576856

RESUMEN

Replication defective adenoviruses are promising vectors for the delivery of vaccine antigens. However, the potential of a vector to elicit transgene-specific adaptive immune responses is largely dependent on the viral serotype used. HAdV-5 (Human adenovirus C) vectors are more immunogenic than chimpanzee adenovirus vectors from species Human adenovirus E (ChAdOx1 and AdC68) in mice, though the mechanisms responsible for these differences in immunogenicity remain poorly understood. In this study, superior immunogenicity was associated with markedly higher levels of transgene expression in vivo, particularly within draining lymph nodes. To investigate the viral factors contributing to these phenotypes, we generated recombinant ChAdOx1 vectors by exchanging components of the viral capsid reported to be principally involved in cell entry with the corresponding sequences from HAdV-5. Remarkably, pseudotyping with the HAdV-5 fiber and/or penton RGD loop had little to no effect on in vivo transgene expression or transgene-specific adaptive immune responses despite considerable species-specific sequence heterogeneity in these components. Our results suggest that mechanisms governing vector transduction after intramuscular administration in mice may be different from those described in vitro.


Asunto(s)
Adenoviridae/inmunología , Adenovirus Humanos/inmunología , Proteínas de la Cápside/inmunología , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Receptores de Orexina/genética , Adenoviridae/genética , Adenovirus Humanos/genética , Animales , Anticuerpos Antivirales/inmunología , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Antígenos Virales/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Proteínas de la Cápside/química , Expresión Génica , Vectores Genéticos/administración & dosificación , Humanos , Inmunidad , Inmunización , Inyecciones Intramusculares , Ratones , Pan troglodytes , Transducción Genética , Transgenes , Vacunas/genética , Vacunas/inmunología
16.
Sci Rep ; 3: 1706, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23609325

RESUMEN

Rodent malaria species Plasmodium yoelii and P. chabaudi have been widely used to validate vaccine approaches targeting blood-stage merozoite antigens. However, increasing data suggest the P. berghei rodent malaria may be able to circumvent vaccine-induced anti-merozoite responses. Here we confirm a failure to protect against P. berghei, despite successful antibody induction against leading merozoite antigens using protein-in-adjuvant or viral vectored vaccine delivery. No subunit vaccine approach showed efficacy in mice following immunization and challenge with the wild-type P. berghei strains ANKA or NK65, or against a chimeric parasite line encoding a merozoite antigen from P. falciparum. Protection was not improved in knockout mice lacking the inhibitory Fc receptor CD32b, nor against a Δsmac P. berghei parasite line with a non-sequestering phenotype. An improved understanding of the mechanisms responsible for protection, or failure of protection, against P. berghei merozoites could guide the development of an efficacious vaccine against P. falciparum.


Asunto(s)
Formación de Anticuerpos/inmunología , Antimaláricos/inmunología , Vacunas contra la Malaria/inmunología , Malaria/inmunología , Merozoítos/inmunología , Plasmodium berghei/inmunología , Animales , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/inmunología , Línea Celular , Femenino , Células HEK293 , Humanos , Inmunización/métodos , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Conejos , Receptores de IgG/inmunología , Roedores/inmunología
17.
Virology ; 421(2): 245-52, 2011 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-22033262

RESUMEN

Human monocytes are susceptible to dengue virus (DV) infection through an FcR-dependent pathway known as antibody-dependent enhancement (ADE). In this study, infection enhancement was observed when purified monocytes were infected with DV serotypes in the presence of serially diluted immune serum antibodies. Analyzing binding of the DV-antibody immune complexes to monocytes by quantifying the amount of viruses attached to monocytes, we found that binding did not correlate with the input amount of antibodies; rather, it peaked at suboptimal antibody concentrations, correlating with the observed infection enhancement. These results suggested that immune complexes are involved in hindering DV from binding to FcR-bearing cells; when such a protective feature is weakened, enhancement of viral attachment and ADE are observed. Further, increased cytokine production (TNF-alpha and IFN-alpha), and costimulatory marker expression (CD86 and CD40), were found to be associated with infection enhancement, suggesting a pathological role of ADE-affected monocytes in dengue hemorrhagic diseases.


Asunto(s)
Anticuerpos Antivirales/inmunología , Acrecentamiento Dependiente de Anticuerpo , Virus del Dengue/inmunología , Leucocitos Mononucleares/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/metabolismo , Anticuerpos Antivirales/metabolismo , Afinidad de Anticuerpos , Antígeno B7-2/biosíntesis , Antígenos CD40/biosíntesis , Chlorocebus aethiops , Virus del Dengue/metabolismo , Virus del Dengue/fisiología , Humanos , Interferón-alfa/biosíntesis , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/virología , Dengue Grave/inmunología , Dengue Grave/virología , Factor de Necrosis Tumoral alfa/biosíntesis , Células Vero , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA