Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 171(1): 229-241.e15, 2017 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-28938115

RESUMEN

Zika virus (ZIKV), a mosquito-borne flavivirus, causes devastating congenital birth defects. We isolated a human monoclonal antibody (mAb), ZKA190, that potently cross-neutralizes multi-lineage ZIKV strains. ZKA190 is highly effective in vivo in preventing morbidity and mortality of ZIKV-infected mice. NMR and cryo-electron microscopy show its binding to an exposed epitope on DIII of the E protein. ZKA190 Fab binds all 180 E protein copies, altering the virus quaternary arrangement and surface curvature. However, ZIKV escape mutants emerged in vitro and in vivo in the presence of ZKA190, as well as of other neutralizing mAbs. To counter this problem, we developed a bispecific antibody (FIT-1) comprising ZKA190 and a second mAb specific for DII of E protein. In addition to retaining high in vitro and in vivo potencies, FIT-1 robustly prevented viral escape, warranting its development as a ZIKV immunotherapy.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/uso terapéutico , Infección por el Virus Zika/terapia , Virus Zika/química , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/química , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Neutralizantes/química , Anticuerpos Antivirales/administración & dosificación , Anticuerpos Antivirales/química , Microscopía por Crioelectrón , Epítopos , Humanos , Espectroscopía de Resonancia Magnética , Ratones , Modelos Moleculares , Alineación de Secuencia , Proteínas del Envoltorio Viral/química , Virus Zika/inmunología
2.
PLoS Pathog ; 15(7): e1007938, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31356638

RESUMEN

Arthropod-borne flaviviruses cause life-threatening diseases associated with endothelial hyperpermeability and vascular leak. We recently found that vascular leak can be triggered by dengue virus (DENV) non-structural protein 1 (NS1) via the disruption of the endothelial glycocalyx-like layer (EGL). However, the molecular determinants of NS1 required to trigger EGL disruption and the cellular pathway(s) involved remain unknown. Here we report that mutation of a single glycosylated residue of NS1 (N207Q) abolishes the ability of NS1 to trigger EGL disruption and induce endothelial hyperpermeability. Intriguingly, while this mutant bound to the surface of endothelial cells comparably to wild-type NS1, it was no longer internalized, suggesting that NS1 binding and internalization are distinct steps. Using endocytic pathway inhibitors and gene-specific siRNAs, we determined that NS1 was endocytosed into endothelial cells in a dynamin- and clathrin-dependent manner, which was required to trigger endothelial dysfunction in vitro and vascular leak in vivo. Finally, we found that the N207 glycosylation site is highly conserved among flaviviruses and is also essential for West Nile and Zika virus NS1 to trigger endothelial hyperpermeability via clathrin-mediated endocytosis. These data provide critical mechanistic insight into flavivirus NS1-induced pathogenesis, presenting novel therapeutic and vaccine targets for flaviviral diseases.


Asunto(s)
Virus del Dengue/patogenicidad , Proteínas no Estructurales Virales/fisiología , Sustitución de Aminoácidos , Sitios de Unión/genética , Permeabilidad Capilar , Línea Celular , Virus del Dengue/genética , Virus del Dengue/fisiología , Endocitosis/fisiología , Células Endoteliales/fisiología , Células Endoteliales/virología , Glicocálix/fisiología , Glicosilación , Células HEK293 , Humanos , Modelos Biológicos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Mutación , Estructura Cuaternaria de Proteína , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética
3.
J Immunol ; 202(4): 1153-1162, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30642979

RESUMEN

Endothelial dysfunction and vascular leak, pathogenic hallmarks of severe dengue disease, are directly triggered by dengue virus (DENV) nonstructural protein 1 (NS1). Previous studies have shown that immunization with NS1, as well as passive transfer of NS1-immune serum or anti-NS1 mAb, prevent NS1-mediated lethality in vivo. In this study, we evaluated the immunogenicity and protective capacity of recombinant DENV NS1 administered with cyclic dinucleotides (CDNs), potent activators of innate immune pathways and highly immunogenic adjuvants. Using both wild-type C57BL/6 mice and IFN-α/ß receptor-deficient mice, we show that NS1-CDN immunizations elicit serotype-specific and cross-reactive Ab and T cell responses. Furthermore, NS1-CDN vaccinations conferred significant homotypic and heterotypic protection from DENV2-induced morbidity and mortality. In addition, we demonstrate that high anti-NS1 Ab titers are associated with protection, supporting the role of humoral responses against DENV NS1 as correlates of protection. These findings highlight the potential of CDN-based adjuvants for inducing Ab and T cell responses and validate NS1 as an important candidate for dengue vaccine development.


Asunto(s)
Adyuvantes Inmunológicos , Anticuerpos Antivirales/inmunología , Virus del Dengue/inmunología , Nucleótidos Cíclicos/inmunología , Linfocitos T/inmunología , Proteínas no Estructurales Virales/inmunología , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
4.
Cardiol Young ; 30(12): 1866-1873, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32993838

RESUMEN

BACKGROUND: Acute respiratory infection is one of the main causes of morbidity in children. Some studies have suggested that pulmonary hypertension and congenital heart disease with haemodynamic repercussion increase the severity of respiratory infections, but there are few publications in developing countries. METHODS: This was a prospective cohort study evaluating the impact of pulmonary hypertension and congenital heart disease (CHD) with haemodynamic repercussion as predictors of severity in children under 5 years of age hospitalised for acute respiratory infection. RESULTS: Altogether, 217 children hospitalised for a respiratory infection who underwent an echocardiogram were evaluated; 62 children were diagnosed with CHD with haemodynamic repercussion or pulmonary hypertension. Independent predictors of admission to intensive care included: pulmonary hypertension (RR 2.14; 95% CI 1.06-4.35, p = 0.034), respiratory syncytial virus (RR 2.52; 95% CI 1.29-4.92, p = 0.006), and bacterial pneumonia (RR 3.09; 95% CI 1.65-5.81, p = 0.000). A significant difference was found in average length of hospital stay in children with the cardiovascular conditions studied (p = 0.000). CONCLUSIONS: Pulmonary hypertension and CHD with haemodynamic repercussion as well as respiratory syncytial virus and bacterial pneumonia were predictors of severity in children with respiratory infections in this study. Early recognition of cardiovascular risks in paediatric populations is necessary to lessen the impact on respiratory infections.


Asunto(s)
Cardiopatías Congénitas , Hipertensión Pulmonar , Infecciones por Virus Sincitial Respiratorio , Infecciones del Sistema Respiratorio , Niño , Preescolar , Colombia/epidemiología , Cardiopatías Congénitas/complicaciones , Cardiopatías Congénitas/epidemiología , Hemodinámica , Humanos , Hipertensión Pulmonar/epidemiología , Lactante , Estudios Prospectivos , Derivación y Consulta , Infecciones del Sistema Respiratorio/complicaciones , Infecciones del Sistema Respiratorio/epidemiología , América del Sur
5.
J Gen Virol ; 100(11): 1515-1522, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31526452

RESUMEN

Dengue virus (DENV) causes the most prevalent arboviral infection of humans, resulting in a spectrum of outcomes, ranging from asymptomatic infection to dengue fever to severe dengue characterized by vascular leakage and shock. Previously, we determined that DENV nonstructural protein 1 (NS1) induces endothelial hyperpermeability, disrupts the endothelial glycocalyx layer (EGL) in vitro and triggers shedding of structural components, including sialic acid (Sia) and heparan sulfate. Here, using a murine model of dengue disease disease, we found high levels of Sia and NS1 circulating in mice with DENV-induced morbidity and lethal DENV infection. Further, we developed a liquid chromatography/mass spectrometry-based method for quantifying free Sia in serum and determined that the levels of free N-glycolylneuraminic acid were significantly higher in DENV-infected mice than in uninfected controls. These data provide additional evidence that DENV infection disrupts EGL components in vivo and warrant further research assessing Sia as a biomarker of severe dengue disease.


Asunto(s)
Biomarcadores/sangre , Dengue/patología , Ácido N-Acetilneuramínico/sangre , Suero/química , Animales , Cromatografía Liquida , Modelos Animales de Enfermedad , Espectrometría de Masas , Ratones , Análisis de Supervivencia , Proteínas no Estructurales Virales/sangre
6.
PLoS Pathog ; 13(11): e1006673, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29121099

RESUMEN

Dengue virus (DENV) is the most prevalent, medically important mosquito-borne virus. Disease ranges from uncomplicated dengue to life-threatening disease, characterized by endothelial dysfunction and vascular leakage. Previously, we demonstrated that DENV nonstructural protein 1 (NS1) induces endothelial hyperpermeability in a systemic mouse model and human pulmonary endothelial cells, where NS1 disrupts the endothelial glycocalyx-like layer. NS1 also triggers release of inflammatory cytokines from PBMCs via TLR4. Here, we examined the relative contributions of inflammatory mediators and endothelial cell-intrinsic pathways. In vivo, we demonstrated that DENV NS1 but not the closely-related West Nile virus NS1 triggers localized vascular leak in the dorsal dermis of wild-type C57BL/6 mice. In vitro, we showed that human dermal endothelial cells exposed to DENV NS1 do not produce inflammatory cytokines (TNF-α, IL-6, IL-8) and that blocking these cytokines does not affect DENV NS1-induced endothelial hyperpermeability. Further, we demonstrated that DENV NS1 induces vascular leak in TLR4- or TNF-α receptor-deficient mice at similar levels to wild-type animals. Finally, we blocked DENV NS1-induced vascular leak in vivo using inhibitors targeting molecules involved in glycocalyx disruption. Taken together, these data indicate that DENV NS1-induced endothelial cell-intrinsic vascular leak is independent of inflammatory cytokines but dependent on endothelial glycocalyx components.


Asunto(s)
Virus del Dengue/metabolismo , Dengue/metabolismo , Endotelio Vascular/metabolismo , Glicocálix/metabolismo , Leucocitos Mononucleares/metabolismo , Proteínas no Estructurales Virales/metabolismo , Animales , Citocinas/genética , Citocinas/metabolismo , Dengue/genética , Virus del Dengue/genética , Endotelio Vascular/patología , Endotelio Vascular/virología , Glicocálix/genética , Humanos , Leucocitos Mononucleares/patología , Leucocitos Mononucleares/virología , Ratones , Ratones Noqueados , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Proteínas no Estructurales Virales/genética
7.
Infect Immun ; 85(2)2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27895131

RESUMEN

Recent studies have shown that immune responses against the cell-traversal protein for Plasmodium ookinetes and sporozoites (CelTOS) can inhibit parasite infection. While these studies provide important evidence toward the development of vaccines targeting this protein, it remains unknown whether these responses could engage the Plasmodium falciparum CelTOS in vivo Using a newly developed rodent malaria chimeric parasite expressing the P. falciparum CelTOS (PfCelTOS), we evaluated the protective effect of in vivo immune responses elicited by vaccination and assessed the neutralizing capacity of monoclonal antibodies specific against PfCelTOS. Mice immunized with recombinant P. falciparum CelTOS in combination with the glucopyranosyl lipid adjuvant-stable emulsion (GLA-SE) or glucopyranosyl lipid adjuvant-liposome-QS21 (GLA-LSQ) adjuvant system significantly inhibited sporozoite hepatocyte infection. Notably, monoclonal antibodies against PfCelTOS strongly inhibited oocyst development of P. falciparum and Plasmodium berghei expressing PfCelTOS in Anopheles gambiae mosquitoes. Taken together, our results demonstrate that anti-CelTOS responses elicited by vaccination or passive immunization can inhibit sporozoite and ookinete infection and impair vector transmission.


Asunto(s)
Antígenos de Protozoos/inmunología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Esporozoítos/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Anticuerpos Antiprotozoarios/sangre , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/genética , Modelos Animales de Enfermedad , Hepatocitos/efectos de los fármacos , Hepatocitos/parasitología , Inmunización , Inmunización Pasiva , Estadios del Ciclo de Vida , Malaria Falciparum/prevención & control , Malaria Falciparum/transmisión , Ratones , Plasmodium falciparum/crecimiento & desarrollo , Proteínas Protozoarias/genética , Proteínas Recombinantes
8.
PLoS Pathog ; 11(2): e1004637, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25658939

RESUMEN

Malaria infection begins when a female Anopheles mosquito injects Plasmodium sporozoites into the skin of its host during blood feeding. Skin-deposited sporozoites may enter the bloodstream and infect the liver, reside and develop in the skin, or migrate to the draining lymph nodes (DLNs). Importantly, the DLN is where protective CD8(+) T cell responses against malaria liver stages are induced after a dermal route of infection. However, the significance of parasites in the skin and DLN to CD8(+) T cell activation is largely unknown. In this study, we used genetically modified parasites, as well as antibody-mediated immobilization of sporozoites, to determine that active sporozoite migration to the DLNs is required for robust CD8(+) T cell responses. Through dynamic in vivo and static imaging, we show the direct uptake of parasites by lymph-node resident DCs followed by CD8(+) T cell-DC cluster formation, a surrogate for antigen presentation, in the DLNs. A few hours after sporozoite arrival to the DLNs, CD8(+) T cells are primed by resident CD8α(+) DCs with no apparent role for skin-derived DCs. Together, these results establish a critical role for lymph node resident CD8α(+) DCs in CD8(+) T cell priming to sporozoite antigens while emphasizing a requirement for motile sporozoites in the induction of CD8(+) T cell-mediated immunity.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Ganglios Linfáticos/inmunología , Activación de Linfocitos/inmunología , Malaria/inmunología , Esporozoítos/inmunología , Traslado Adoptivo , Animales , Presentación de Antígeno/inmunología , Antígenos de Protozoos/inmunología , Separación Celular , Células Dendríticas/inmunología , Citometría de Flujo , Inmunidad Celular/inmunología , Ganglios Linfáticos/parasitología , Ratones , Microscopía Confocal , Plasmodium berghei/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
Proc Natl Acad Sci U S A ; 111(34): 12528-32, 2014 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-25114213

RESUMEN

Malaria caused by Plasmodium falciparum kills nearly one million children each year and imposes crippling economic burdens on families and nations worldwide. No licensed vaccine exists, but infection can be prevented by antibodies against the circumsporozoite protein (CSP), the major surface protein of sporozoites, the form of the parasite injected by mosquitoes. We have used vectored immunoprophylaxis (VIP), an adeno-associated virus-based technology, to introduce preformed antibody genes encoding anti-P. falciparum CSP mAb into mice. VIP vector-transduced mice exhibited long-lived mAb expression at up to 1,200 µg/mL in serum, and up to 70% were protected from both i.v. and mosquito bite challenge with transgenic Plasmodium berghei rodent sporozoites that incorporate the P. falciparum target of the mAb in their CSP. Serum antibody levels and protection from mosquito bite challenge were dependent on the dose of the VIP vector. All individual mice expressing CSP-specific mAb 2A10 at 1 mg/mL or more were completely protected, suggesting that in this model system, exceeding that threshold results in consistent sterile protection. Our results demonstrate the potential of VIP as a path toward the elusive goal of immunization against malaria.


Asunto(s)
Técnicas de Transferencia de Gen , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/genética , Malaria Falciparum/prevención & control , Plasmodium falciparum/inmunología , Animales , Anticuerpos Monoclonales de Origen Murino/biosíntesis , Anticuerpos Monoclonales de Origen Murino/genética , Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Neutralizantes/genética , Anticuerpos Antiprotozoarios/biosíntesis , Anticuerpos Antiprotozoarios/genética , Dependovirus/genética , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos , Humanos , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Ratones , Ratones Endogámicos C57BL , Plasmodium berghei/genética , Plasmodium berghei/inmunología , Plasmodium falciparum/genética , Esporozoítos/inmunología
10.
J Infect Dis ; 212(7): 1111-9, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25762791

RESUMEN

Studies in animals and human volunteers demonstrate that antibodies against the repeat-region of the Plasmodium circumsporozoite protein (CSP) abrogate sporozoite infection. However, the realization that the N- and C- terminal regions flanking the repeats play essential roles in parasite infectivity raised the possibility that they could be targeted by protective antibodies. We characterized a monoclonal antibody (mAb5D5) specific for the N-terminus of the P. falciparum CSP, which inhibits the proteolytic cleavage of the CSP, a key requirement for parasite infection of hepatocytes. Adoptive transfer of mAb5D5 strongly inhibits the in vivo infection of sporozoites expressing the N-terminus of P. falciparum CSP, and this protection is greatly enhanced when combined with antirepeat antibodies. Our results show that antibodies interfering with molecular processes required for parasite infectivity can exert a strong in vivo protective activity and indicate that pre-erythrocytic vaccines against Plasmodium should include the CSP N-terminal region.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Antiprotozoarios/inmunología , Vacunas contra la Malaria/inmunología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Animales , Especificidad de Anticuerpos , Epítopos/inmunología , Femenino , Hepatocitos/parasitología , Humanos , Malaria Falciparum/inmunología , Malaria Falciparum/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Esporozoítos/inmunología
11.
Infect Immun ; 83(1): 268-75, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25368113

RESUMEN

Decades of success with live adenovirus vaccines suggest that replication-competent recombinant adenoviruses (rAds) could serve as effective vectors for immunization against other pathogens. To explore the potential of a live rAd vaccine against malaria, we prepared a viable adenovirus 5 (Ad5) recombinant that displays a B-cell epitope from the circumsporozoite protein (CSP) of Plasmodium falciparum on the virion surface. The recombinant induced P. falciparum sporozoite-neutralizing antibodies in mice. Human adenoviruses do not replicate in mice. Therefore, to examine immunogenicity in a system in which, as in humans, the recombinant replicates, we constructed a similar recombinant in an adenovirus mutant that replicates in monkey cells and immunized four Aotus nancymaae monkeys. The recombinant replicated in the monkeys after intratracheal instillation, the first demonstration of replication of human adenoviruses in New World monkeys. Immunization elicited antibodies both to the Plasmodium epitope and the Ad5 vector. Antibodies from all four monkeys recognized CSP on intact parasites, and plasma from one monkey neutralized sporozoites in vitro and conferred partial protection against P. falciparum sporozoite infection after passive transfer to mice. Prior enteric inoculation of two animals with antigenically wild-type adenovirus primed a response to the subsequent intratracheal inoculation, suggesting a route to optimizing performance. A vaccine is not yet available against P. falciparum, which induces the deadliest form of malaria and kills approximately one million children each year. The live capsid display recombinant described here may constitute an early step in a critically needed novel approach to malaria immunization.


Asunto(s)
Adenoviridae/genética , Anticuerpos Antiprotozoarios/sangre , Portadores de Fármacos , Vacunas contra la Malaria/inmunología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Aotidae , Técnicas de Visualización de Superficie Celular , Femenino , Vectores Genéticos , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/genética , Masculino , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
12.
J Infect Dis ; 210(9): 1508-16, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24823625

RESUMEN

It is well established that immunization with attenuated malaria sporozoites induces CD8(+) T cells that eliminate parasite-infected hepatocytes. Liver memory CD8(+) T cells induced by immunization with parasites undergo a unique differentiation program and have enhanced expression of CXCR6. Following immunization with malaria parasites, CXCR6-deficient memory CD8(+) T cells recovered from the liver display altered cell-surface expression markers as compared to their wild-type counterparts, but they exhibit normal cytokine secretion and expression of cytotoxic mediators on a per-cell basis. Most importantly, CXCR6-deficient CD8(+) T cells migrate to the liver normally after immunization with Plasmodium sporozoites or vaccinia virus, but a few weeks later their numbers severely decrease in this organ, losing their capacity to inhibit malaria parasite development in the liver. These studies are the first to show that CXCR6 is critical for the development and maintenance of protective memory CD8(+) T cells in the liver.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/fisiología , Parasitosis Hepáticas/inmunología , Receptores CXCR/fisiología , Traslado Adoptivo , Animales , Femenino , Citometría de Flujo , Malaria/inmunología , Malaria/parasitología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Plasmodium berghei/inmunología , Receptores CXCR6
13.
J Immunol ; 188(12): 6407-17, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22615208

RESUMEN

Development of subunit vaccines for malaria that elicit a strong, long-term memory response is an intensive area of research, with the focus on improving the immunogenicity of a circumsporozoite (CS) protein-based vaccine. In this study, we found that a chimeric protein, formed by fusing vaccinia virus protein 14K (A27) to the CS of Plasmodium yoelii, induces strong effector memory CD8(+) T cell responses in addition to high-affinity Abs when used as a priming agent in the absence of any adjuvant, followed by an attenuated vaccinia virus boost expressing CS in murine models. Moreover, priming with the chimeric protein improved the magnitude and polyfunctionality of cytokine-secreting CD8(+) T cells. This fusion protein formed oligomers/aggregates that led to activation of STAT-1 and IFN regulatory factor-3 in human macrophages, indicating a type I IFN response, resulting in NO, IL-12, and IL-6 induction. Furthermore, this vaccination regimen inhibited the liver stage development of the parasite, resulting in sterile protection. In summary, we propose a novel approach in designing CS based pre-erythrocytic vaccines against Plasmodium using the adjuvant-like effect of the immunogenic vaccinia virus protein 14K.


Asunto(s)
Vacunas contra la Malaria/inmunología , Proteínas Protozoarias/inmunología , Proteínas Recombinantes de Fusión/inmunología , Virus Vaccinia/inmunología , Proteínas Virales/inmunología , Adyuvantes Inmunológicos/farmacología , Animales , Antígenos de Protozoos/inmunología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Malaria/inmunología , Malaria/prevención & control , Vacunas contra la Malaria/síntesis química , Ratones , Ratones Endogámicos BALB C , Microscopía Confocal , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Infect Immun ; 81(8): 2882-7, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23716612

RESUMEN

The development of vaccine candidates against Plasmodium vivax-the most geographically widespread human malaria species-is challenged by technical difficulties, such as the lack of in vitro culture systems and availability of animal models. Chimeric rodent Plasmodium parasites are safe and useful tools for the preclinical evaluation of new vaccine formulations. We report the successful development and characterization of chimeric Plasmodium berghei parasites bearing the type I repeat region of P. vivax circumsporozoite protein (CSP). The P. berghei-P. vivax chimeric strain develops normally in mosquitoes and produces highly infectious sporozoites that produce patent infection in mice that are exposed to the bites of as few as 3 P. berghei-P. vivax-infected mosquitoes. Using this transgenic parasite, we demonstrate that monoclonal and polyclonal antibodies against P. vivax CSP strongly inhibit parasite infection and thus support the notion that these antibodies play an important role in protective immunity. The chimeric parasites we developed represent a robust model for evaluating protective immune responses against P. vivax vaccines based on CSP.


Asunto(s)
Quimera/genética , Vacunas contra la Malaria/inmunología , Plasmodium berghei/genética , Plasmodium vivax/genética , Proteínas Protozoarias/genética , Animales , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/inmunología , Quimera/inmunología , Técnica del Anticuerpo Fluorescente Indirecta , Ratones , Plasmodium berghei/inmunología , Plasmodium vivax/inmunología , Proteínas Protozoarias/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
15.
Infect Immun ; 81(3): 789-800, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23275094

RESUMEN

The Plasmodium falciparum circumsporozoite (CS) protein (CSP) is a major vaccine target for preventing malaria infection. Thus, developing strong and durable antibody and T cell responses against CSP with novel immunogens and potent adjuvants may improve upon the success of current approaches. Here, we compare four distinct full-length P. falciparum CS proteins expressed in Escherichia coli or Pichia pastoris for their ability to induce immunity and protection in mice when administered with long-chain poly(I · C) [poly(I · C)LC] as an adjuvant. CS proteins expressed in E. coli induced high-titer antibody responses against the NANP repeat region and potent CSP-specific CD4(+) T cell responses. Moreover, E. coli-derived CS proteins in combination with poly(I · C)LC induced potent multifunctional (interleukin 2-positive [IL-2(+)], tumor necrosis factor alpha-positive [TNF-α(+)], gamma interferon-positive [IFN-γ(+)]) CD4(+) effector T cell responses in blood, in spleen, and particularly in liver. Using transgenic Plasmodium berghei expressing the repeat region of P. falciparum CSP [Pb-CS(Pf)], we showed that there was a 1- to 4-log decrease in malaria rRNA in the liver following a high-dose challenge and ~50% sterilizing protection with a low-dose challenge compared to control levels. Protection was directly correlated with high-level antibody titers but not CD4(+) T cell responses. Finally, protective immunity was also induced using the Toll-like receptor 4 agonist glucopyranosyl lipid adjuvant-stable emulsion (GLA-SE) as the adjuvant, which also correlated with high antibody titers yet CD4(+) T cell immunity that was significantly less potent than that with poly(I · C)LC. Overall, these data suggest that full-length CS proteins and poly(I · C)LC or GLA-SE offer a simple vaccine formulation to be used alone or in combination with other vaccines for preventing malaria infection.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Linfocitos T CD4-Positivos/inmunología , Lípidos/farmacología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/metabolismo , Receptor Toll-Like 4/agonistas , Animales , Linfocitos T CD4-Positivos/fisiología , Relación Dosis-Respuesta Inmunológica , Emulsiones , Escherichia coli/genética , Escherichia coli/metabolismo , Regulación de la Expresión Génica , Lípidos/química , Malaria/prevención & control , Vacunas contra la Malaria/inmunología , Ratones , Organismos Modificados Genéticamente , Pichia/genética , Pichia/metabolismo , Plasmodium berghei/genética , Plasmodium berghei/metabolismo , Plasmodium falciparum/fisiología , Proteínas Protozoarias/genética , Factores de Tiempo
16.
CRISPR J ; 6(3): 243-260, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37219969

RESUMEN

Development of medicines using gene editing has been hampered by enzymological and immunological impediments. We described previously the discovery and characterization of improved, novel gene-editing systems from metagenomic data. In this study, we substantially advance this work with three such gene-editing systems, demonstrating their utility for cell therapy development. All three systems are capable of reproducible, high-frequency gene editing in primary immune cells. In human T cells, disruption of the T cell receptor (TCR) alpha-chain was induced in >95% of cells, both paralogs of the TCR beta-chain in >90% of cells, and >90% knockout of ß2-microglobulin, TIGIT, FAS, and PDCD1. Simultaneous double knockout of TRAC and TRBC was obtained at a frequency equal to that of the single edits. Gene editing with our systems had minimal effect on T cell viability. Furthermore, we integrate a chimeric antigen receptor (CAR) construct into TRAC (up to ∼60% of T cells), and demonstrate CAR expression and cytotoxicity. We next applied our novel gene-editing tools to natural killer (NK) cells, B cells, hematopoietic stem cells, and induced pluripotent stem cells, generating similarly efficient cell-engineering outcomes including the creation of active CAR-NK cells. Interrogation of our gene-editing systems' specificity reveals a profile comparable with or better than Cas9. Finally, our nucleases lack preexisting humoral and T cell-based immunity, consistent with their sourcing from nonhuman pathogens. In all, we show these new gene-editing systems have the activity, specificity, and translatability necessary for use in cell therapy development.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Humanos , Sistemas CRISPR-Cas/genética , Linfocitos T/metabolismo , Diferenciación Celular , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo
17.
PLoS Negl Trop Dis ; 16(7): e0010588, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35793354

RESUMEN

Zika virus (ZIKV) is a mosquito-borne flavivirus with maternal infection associated with preterm birth, congenital malformations, and fetal death, and adult infection associated with Guillain-Barré syndrome. Recent widespread endemic transmission of ZIKV and the potential for future outbreaks necessitate the development of an effective vaccine. We developed a ZIKV vaccine candidate based on virus-like-particles (VLPs) generated following transfection of mammalian HEK293T cells using a plasmid encoding the pre-membrane/membrane (prM/M) and envelope (E) structural protein genes. VLPs were collected from cell culture supernatant and purified by column chromatography with yields of approximately 1-2mg/L. To promote increased particle yields, a single amino acid change of phenylalanine to alanine was made in the E fusion loop at position 108 (F108A) of the lead VLP vaccine candidate. This mutation resulted in a modest 2-fold increase in F108A VLP production with no detectable prM processing by furin to a mature particle, in contrast to the lead candidate (parent). To evaluate immunogenicity and efficacy, AG129 mice were immunized with a dose titration of either the immature F108A or lead VLP (each alum adjuvanted). The resulting VLP-specific binding antibody (Ab) levels were comparable. However, geometric mean neutralizing Ab (nAb) titers using a recombinant ZIKV reporter were significantly lower with F108A immunization compared to lead. After virus challenge, all lead VLP-immunized groups showed a significant 3- to 4-Log10 reduction in mean ZIKV RNAemia levels compared with control mice immunized only with alum, but the RNAemia reduction of 0.5 Log10 for F108A groups was statistically similar to the control. Successful viral control by the lead VLP candidate following challenge supports further vaccine development for this candidate. Notably, nAb titer levels in the lead, but not F108A, VLP-immunized mice inversely correlated with RNAemia. Further evaluation of sera by an in vitro Ab-dependent enhancement assay demonstrated that the F108A VLP-induced immune sera had a significantly higher capacity to promote ZIKV infection in FcγR-expressing cells. These data indicate that a single amino acid change in the fusion loop resulted in increased VLP yields but that the immature F108A particles were significantly diminished in their capacity to induce nAbs and provide protection against ZIKV challenge.


Asunto(s)
Nacimiento Prematuro , Vacunas de Partículas Similares a Virus , Vacunas Virales , Infección por el Virus Zika , Virus Zika , Aminoácidos , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Femenino , Células HEK293 , Humanos , Recién Nacido , Mamíferos , Ratones , Mutación , Virus Zika/genética
18.
Elife ; 102021 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-34423779

RESUMEN

Arthropod-borne rickettsial pathogens cause mild and severe human disease worldwide. The tick-borne pathogen Rickettsia parkeri elicits skin lesions (eschars) and disseminated disease in humans; however, inbred mice are generally resistant to infection. We report that intradermal infection of mice lacking both interferon receptors (Ifnar1-/-;Ifngr1-/-) with as few as 10 R. parkeri elicits eschar formation and disseminated, lethal disease. Similar to human infection, eschars exhibited necrosis and inflammation, with bacteria primarily found in leukocytes. Using this model, we find that the actin-based motility factor Sca2 is required for dissemination from the skin to internal organs, and the outer membrane protein OmpB contributes to eschar formation. Immunizing Ifnar1-/-;Ifngr1-/- mice with sca2 and ompB mutant R. parkeri protects against rechallenge, revealing live-attenuated vaccine candidates. Thus, Ifnar1-/-;Ifngr1-/- mice are a tractable model to investigate rickettsiosis, virulence factors, and immunity. Our results further suggest that discrepancies between mouse and human susceptibility may be due to differences in interferon signaling.


Tick bites allow disease-causing microbes, including multiple species of Rickettsia bacteria, to pass from arthropods to humans. Being exposed to Rickettsia parkeri, for example, can cause a scab at the bite site, fever, headache and fatigue. To date, no vaccine is available against any of the severe diseases caused by Rickettsia species. Modelling human infections in animals could help to understand and combat these illnesses. R. parkeri is a good candidate for such studies, as it can give insight into more severe Rickettsia infections while being comparatively safer to handle. However, laboratory mice are resistant to this species of bacteria, limiting their use as models. To explore why this is the case, Burke et al. probed whether an immune mechanism known as interferon signalling protects laboratory rodents against R. parkeri. During infection, the immune system releases molecules called interferons that stick to 'receptors' at the surface of cells, triggering defense mechanisms that help to fight off an invader. Burke et al. injected R. parkeri into the skin of mice that had or lacked certain interferon receptors, showing that animals without two specific receptors developed scabs and saw the disease spread through their body. Further investigation showed that two R. parkeri proteins, known as OmpB or Sca2, were essential for the bacteria to cause skin lesions and damage internal organs. Burke et al. then used R. parkeri that lacked OmpB or Sca2 to test whether these modified, inoffensive microbes could act as 'vaccines'. And indeed, vulnerable laboratory mice which were first exposed to the mutant bacteria were then able to survive the 'normal' version of the microbe. Together, this work reveals that interferon signalling protects laboratory mice against R. parkeri infections. It also creates an animal model that can be used to study disease and vaccination.


Asunto(s)
Estudios de Asociación Genética , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Infecciones por Rickettsia/inmunología , Animales , Médula Ósea , Femenino , Inmunidad Innata , Inflamación , Listeria monocytogenes , Macrófagos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Interferón alfa y beta/genética , Rickettsia , Infecciones por Rickettsia/patología , Garrapatas
19.
Science ; 371(6525): 194-200, 2021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-33414220

RESUMEN

Medically important flaviviruses cause diverse disease pathologies and collectively are responsible for a major global disease burden. A contributing factor to pathogenesis is secreted flavivirus nonstructural protein 1 (NS1). Despite demonstrated protection by NS1-specific antibodies against lethal flavivirus challenge, the structural and mechanistic basis remains unknown. Here, we present three crystal structures of full-length dengue virus NS1 complexed with a flavivirus-cross-reactive, NS1-specific monoclonal antibody, 2B7, at resolutions between 2.89 and 3.96 angstroms. These structures reveal a protective mechanism by which two domains of NS1 are antagonized simultaneously. The NS1 wing domain mediates cell binding, whereas the ß-ladder triggers downstream events, both of which are required for dengue, Zika, and West Nile virus NS1-mediated endothelial dysfunction. These observations provide a mechanistic explanation for 2B7 protection against NS1-induced pathology and demonstrate the potential of one antibody to treat infections by multiple flaviviruses.


Asunto(s)
Anticuerpos Neutralizantes/química , Anticuerpos Antivirales/química , Virus del Dengue/inmunología , Proteínas no Estructurales Virales/inmunología , Virus del Nilo Occidental/inmunología , Virus Zika/inmunología , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Reacciones Cruzadas , Cristalografía por Rayos X , Dengue/prevención & control , Dengue/terapia , Endotelio/inmunología , Glicocálix/inmunología , Humanos , Ratones , Conformación Proteica en Lámina beta , Dominios Proteicos , Proteínas no Estructurales Virales/química , Fiebre del Nilo Occidental/prevención & control , Fiebre del Nilo Occidental/terapia , Infección por el Virus Zika/prevención & control , Infección por el Virus Zika/terapia
20.
Antiviral Res ; 173: 104668, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31786251

RESUMEN

Arboviral diseases caused by dengue (DENV), Zika (ZIKV) and chikungunya (CHIKV) viruses represent a major public health problem worldwide, especially in tropical areas where millions of infections occur every year. The aim of this research was to identify candidate molecules for the treatment of these diseases among the drugs currently available in the market, through in silico screening and subsequent in vitro evaluation with cell culture models of DENV and ZIKV infections. Numerous pharmaceutical compounds from antibiotics to chemotherapeutic agents presented high in silico binding affinity for the viral proteins, including ergotamine, antrafenine, natamycin, pranlukast, nilotinib, itraconazole, conivaptan and novobiocin. These five last compounds were tested in vitro, being pranlukast the one that exhibited the best antiviral activity. Further in vitro assays for this compound showed a significant inhibitory effect on DENV and ZIKV infection of human monocytic cells and human hepatocytes (Huh-7 cells) with potential abrogation of virus entry. Finally, intrinsic fluorescence analyses suggest that pranlukast may have some level of interaction with three viral proteins of DENV: envelope, capsid, and NS1. Due to its promising results, suitable accessibility in the market and reduced restrictions compared to other pharmaceuticals; the anti-asthmatic pranlukast is proposed as a drug candidate against DENV, ZIKV, and CHIKV, supporting further in vitro and in vivo assessment of the potential of this and other lead compounds that exhibited good affinity scores in silico as therapeutic agents or scaffolds for the development of new drugs against arboviral diseases.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Arbovirus/efectos de los fármacos , Simulación por Computador , Descubrimiento de Drogas/métodos , Reposicionamiento de Medicamentos , Infecciones por Arbovirus/tratamiento farmacológico , Infecciones por Arbovirus/virología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Ligandos , Modelos Moleculares , Unión Proteica , Relación Estructura-Actividad , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/química , Internalización del Virus/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA