Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Radiol Prot ; 42(1)2022 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-35037901

RESUMEN

The field of radiation countermeasures is growing, however, currently there are no effective and non-toxic compounds which could be administered orally to the individuals post exposure to high doses of ionising radiation. The pigment melanin is ubiquitous through all kingdoms of life and provides selective advantage under radiation stress through its role as a chemical and physical shield, and its capacity to respond and react to exposures. Soluble allomelanin was administered to mice following whole-body exposure to lethal or sublethal doses of gamma radiation to determine its capacity to mitigate the effects of acute radiation syndrome, and its utility as a radiation countermeasure. Allomelanin has shown a trend to improve survival post an 8 Gy sublethal radiation exposure when administered up to 48 h post-irradiation. Furthermore, it improved median and overall survival to a 10 Gy lethal radiation exposure, specifically when administered at 24 h post-irradiation. Histological analysis on the jejunum region of the small intestine of this treatment group indicated that alterations of the mucosal and submucosal architecture, and disruption of the lymphatic system associated with lethal radiation exposure were mitigated when allomelanin was administered at 24 h post-irradiation. Based on this work soluble allomelanin derived from a fungal source could serve as an easily sourced, cost-effective, and viable countermeasure to accidental radiation exposure and merits further investigation.


Asunto(s)
Síndrome de Radiación Aguda , Melaninas , Animales , Rayos gamma , Ratones , Dosis de Radiación , Irradiación Corporal Total/efectos adversos
2.
J Proteome Res ; 18(3): 1145-1161, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30706717

RESUMEN

Respiratory syncytial virus (RSV) is a significant cause of mortality and morbidity in infants, the elderly, immunocompromised individuals, and patients with congenital heart diseases. Despite extensive efforts, a vaccine against RSV is still not available. We have previously reported the development of a subunit vaccine (ΔF/TriAdj) composed of a truncated version of the fusion protein (ΔF) and a polymer-based combination adjuvant (TriAdj). We compared inflammatory responses of ΔF/TriAdj-vaccinated and unvaccinated mice following intranasal challenge with RSV. Rapid and early inflammatory responses were observed in lung samples from both groups but modulated in the vaccinated group 7 days after the viral challenge. The underlying mechanism of action of ΔF/TriAdj was further studied through LC-MS-based metabolomic profiling by using 12C- or 13C-dansyl labeling for the amine/phenol submetabolome. RSV infection predominantly affected the amino acid biosynthesis pathways and urea cycle, whereas ΔF/TriAdj modulated the concentrations of almost all of the altered metabolites. Tryptophan metabolites were significantly affected, including indole, l-kynurenine, xanthurenic acid, serotonin, 5-hydroxyindoleacetic acid, and 6-hydroxymelatonin. The results from the present study provide further mechanistic insights into the mode of action of this RSV vaccine candidate and have important implications in the design of metabolic therapeutic interventions.


Asunto(s)
Inmunización/métodos , Metabolómica/métodos , Infecciones por Virus Sincitial Respiratorio/metabolismo , Vacunas de Subunidad/metabolismo , Adyuvantes Inmunológicos/metabolismo , Administración Intranasal , Animales , Anticuerpos Antivirales/inmunología , Humanos , Pulmón/efectos de los fármacos , Pulmón/virología , Ratones , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/metabolismo , Virus Sincitial Respiratorio Humano/patogenicidad , Vacunas de Subunidad/genética
3.
J Virol ; 92(15)2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29769345

RESUMEN

VP8, the UL47 gene product in bovine herpesvirus-1 (BoHV-1), is a major tegument protein that is essential for virus replication in vivo The major DNA damage response protein, ataxia telangiectasia mutated (ATM), phosphorylates Nijmegen breakage syndrome (NBS1) and structural maintenance of chromosome-1 (SMC1) proteins during the DNA damage response. VP8 was found to interact with ATM and NBS1 during transfection and BoHV-1 infection. However, VP8 did not interfere with phosphorylation of ATM in transfected or BoHV-1-infected cells. In contrast, VP8 inhibited phosphorylation of both NBS1 and SMC1 in transfected cells, as well as in BoHV-1-infected cells, but not in cells infected with a VP8 deletion mutant (BoHV-1ΔUL47). Inhibition of NBS1 and SMC1 phosphorylation was observed at 4 h postinfection by nuclear VP8. Furthermore, UV light-induced cyclobutane pyrimidine dimer (CPD) repair was reduced in the presence of VP8, and VP8 in fact enhanced etoposide or UV-induced apoptosis. This suggests that VP8 blocks the ATM/NBS1/SMC1 pathway and inhibits DNA repair. VP8 induced apoptosis in VP8-transfected cells through caspase-3 activation. The fact that BoHV-1 is known to induce apoptosis through caspase-3 activation is in agreement with this observation. The role of VP8 was confirmed by the observation that BoHV-1 induced significantly more apoptosis than BoHV-1ΔUL47. These data reveal a potential role of VP8 in the modulation of the DNA damage response pathway and induction of apoptosis during BoHV-1 infection.IMPORTANCE To our knowledge, the effect of BoHV-1 infection on the DNA damage response has not been characterized. Since BoHV-1ΔUL47 was previously shown to be avirulent in vivo, VP8 is critical for the progression of viral infection. We demonstrated that VP8 interacts with DNA damage response proteins and disrupts the ATM-NBS1-SMC1 pathway by inhibiting phosphorylation of DNA repair proteins NBS1 and SMC1. Furthermore, interference of VP8 with DNA repair was correlated with decreased cell viability and increased DNA damage-induced apoptosis. These data show that BoHV-1 VP8 developed a novel strategy to interrupt the ATM signaling pathway and to promote apoptosis. These results further enhance our understanding of the functions of VP8 during BoHV-1 infection and provide an additional explanation for the reduced virulence of BoHV-1ΔUL47.


Asunto(s)
Apoptosis , Proteínas de la Cápside/metabolismo , Daño del ADN , Herpesvirus Bovino 1/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de la Cápside/genética , Caspasa 3/genética , Caspasa 3/metabolismo , Bovinos , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Células HEK293 , Células HeLa , Herpesvirus Bovino 1/genética , Humanos
4.
Parasite Immunol ; 37(7): 368-75, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25824598

RESUMEN

Isolation of the soluble exogenous antigens (SEAgs), its immune response study and proteome profiling is an essential prerequisite for understanding the molecular pathogenesis of Leishmania donovani. The immunostimulatory potential of L. donovani SEAgs, purified from culture of L. donovani clinical isolate, was evaluated for their ability to induce cellular responses in treated/cured hamsters. SEAgs induced significant proliferative responses in lymphocytes (SI 5.6 ± 2.3; P < 0.01) isolated from cured hamster. In addition, significant NO production in response to SEAgs was also noticed in macrophages of hamsters, mouse and human cell lines (J774A-1 and THP1). Western blot analyses with antibodies against proteophosphoglycan (PPG; surface-expressed and secreted molecule) of L. donovani revealed that PPG molecules are also present in L. donovani SEAgs. Mass spectrometry (MS)-based proteome analysis of 12 protein bands of SEAgs through MALDI-TOF/TOF endorsed the identification of some Th1-stimulatory immunogenic proteins. These immunogenic proteins may offer increased hope for the discovery of new promising vaccine candidates against visceral leishmaniasis (VL). The overall results suggest that immunostimulatory molecules are present in the SEAgs, which may be further exploited, for developing a subunit vaccine against VL a fatal human disease.


Asunto(s)
Antígenos de Protozoos/inmunología , Leishmania donovani/inmunología , Leishmaniasis Visceral/inmunología , Macrófagos/inmunología , Animales , Anticuerpos/inmunología , Antígenos de Protozoos/aislamiento & purificación , Línea Celular , Proliferación Celular , Cricetinae , Humanos , Inmunización , Activación de Linfocitos/inmunología , Linfocitos/inmunología , Activación de Macrófagos/inmunología , Masculino , Ratones , Proteoma/inmunología
5.
Vaccine ; 2024 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-38769033

RESUMEN

The emergence and ongoing evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has highlighted the need for rapid vaccine development platforms that can be updated to counteract emerging variants of currently circulating and future emerging coronaviruses. Here we report the development of a "train model" subunit vaccine platform that contains a SARS-CoV-2 Wuhan S1 protein (the "engine") linked to a series of flexible receptor binding domains (RBDs; the "cars") derived from SARS-CoV-2 variants of concern (VOCs). We demonstrate that these linked subunit vaccines when combined with Sepivac SWE™, a squalene in water emulsion (SWE) adjuvant, are immunogenic in Syrian hamsters and subsequently provide protection from infection with SARS-CoV-2 VOCs Omicron (BA.1), Delta, and Beta. Importantly, the bivalent and trivalent vaccine candidates offered protection against some heterologous SARS-CoV-2 VOCs that were not included in the vaccine design, demonstrating the potential for broad protection against a range of different VOCs. Furthermore, these formulated vaccine candidates were stable at 2-8 °C for up to 13 months post-formulation, highlighting their utility in low-resource settings. Indeed, our vaccine platform will enable the development of safe and broadly protective vaccines against emerging betacoronaviruses that pose a significant health risk for humans and agricultural animals.

6.
Cancer Med ; 10(3): 1128-1140, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33347715

RESUMEN

PURPOSE: Despite the availability of new drugs, many patients with acute myeloid leukemia (AML) do not achieve remission and outcomes remain poor. Venetoclax is a promising new therapy approved for use in combination with a hypomethylating agent or with low-dose cytarabine for the treatment of newly diagnosed older AML patients or those ineligible for intensive chemotherapy. 225 Actinium-lintuzumab (225 Ac-lintuzumab) is a clinical stage radioimmunotherapy targeting CD33 that has shown evidence of single-agent activity in relapsed/refractory AML. Increased expression of MCL-1 is a mediator of resistance to venetoclax in cancer. EXPERIMENTAL DESIGN: Here we investigated the potential for 225 Ac-lintuzumab-directed DNA damage to suppress MCL-1 levels as a possible mechanism of reversing resistance to venetoclax in two preclinical in vivo models of AML. RESULTS: We demonstrated that 225 Ac-lintuzumab in combination with venetoclax induced a synergistic increase in tumor cell killing compared to treatment with either drug alone in venetoclax-resistant AML cell lines through both an induction of double-stranded DNA breaks (DSBs) and depletion of MCL-1 protein levels. Further, this combination led to significant tumor growth control and prolonged survival benefit in venetoclax-resistant in vivo AML models. CONCLUSIONS: There results suggest that the combination of 225 Ac-lintuzumab with venetoclax is a promising therapeutic strategy for the treatment of patients with venetoclax-resistant AML. Clinical trial of this combination therapy (NCT03867682) is currently ongoing.


Asunto(s)
Actinio/química , Anticuerpos Monoclonales Humanizados/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Resistencia a Antineoplásicos , Leucemia Mieloide Aguda/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Lectina 3 Similar a Ig de Unión al Ácido Siálico/inmunología , Sulfonamidas/farmacología , Animales , Anticuerpos Monoclonales Humanizados/química , Antineoplásicos/farmacología , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/farmacología , Apoptosis , Proliferación Celular , Femenino , Humanos , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Ratones SCID , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Oncotarget ; 11(39): 3571-3581, 2020 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-33062193

RESUMEN

Chimeric antigen receptor (CAR) T cell therapies, and adoptive cell therapy (ACT) in general, represent one of the most promising anti-cancer strategies. Conditioning has been shown to improve the immune homeostatic environment to enable successful ACT or CAR-T engraftment and expansion in vivo following infusion, and represents potential point of intervention to decrease serious toxicities following CAR-T treatment. In contrast to relatively non-specific chemotherapy-derived lymphodepletion, targeted lymphodepletion with radioimmunotherapy (RIT) directed to CD45 may be a safer and more effective alternative to target and deplete immune cells. Here we describe the results of preclinical studies with an anti-mouse CD45 antibody 30F11, labeled with two different beta-emitters 131Iodine (131I) and 177Lutetium (177Lu), to investigate the effect of anti-CD45 RIT lymphodepletion on immune cell types and on tumor control in a model of adoptive cell therapy. Treatment of mice with 3.7 MBq 131I-30F11 or 1.48 MBq 177Lu-30F11 safely depleted immune cells such as spleen CD4+ and CD8+ T Cells, B and NK cells as well as Tregs in OT I tumor model while sparing RBC and platelets and enabled E. G7 tumor control. Our results support the application of CD45-targeted RIT lymphodepletion with a non-myeloablative dose of 131I-30F11 or 177Lu-30F11 antibody prior to adoptive cell therapy.

8.
Nucl Med Biol ; 82-83: 80-88, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32113033

RESUMEN

BACKGROUND: cART has significantly improved the life expectancy of people living with HIV (PLWH). However, it fails to eliminate the long-lived reservoir of latent HIV-infected cells. Radioimmunotherapy (RIT) relies on antigen-specific monoclonal antibodies (mAbs) for targeted delivery of lethal doses of ionizing radiation to cells. Previously, we have demonstrated that human mAb 2556 against HIV gp41 conjugated with 213Bismuth radioisotope (t1/2 = 46 min, alpha-emitter) selectively killed HIV-infected cells. 225Actinium (t1/2 = 9.92 d, alpha-emitter) and 177Lutetium (t1/2 = 6.7 d, beta-emitter) are two long-lived clinically proven radioisotopes for cancer treatment which might be more effective in killing infected cells systemically and in CNS. METHODS: In this study we have conjugated 2556 mAb with 213Bi, 225Ac and 177Lu, and compared their ability to kill HIV-infected human peripheral blood mononuclear cells (PBMCs) and monocytes. PBMCs and monocytes from healthy donors were infected with HIVp49.5 and treated in vitro with increasing concentrations of 213Bi (4-20 µCi)-, 225Ac (20-100 nCi)- and 177Lu (4-50 µCi)-2556 mAb. RESULTS: After three days post-treatment of infected PBMCs and monocytes, 213Bi- and 177Lu-conjugated 2556 mAb reduced virus production measured by p24 level in a dose-dependent manner, whereas, 225Ac-2556 showed minimal effect. However, seven days post-treatment all three radioisotopes showed significantly more pronounced reduction of virus replication as compared to control labeled mAb with 225Ac-2556 showing the least non-specific killing. CONCLUSION: These results indicate that RIT holds promise as a novel treatment option for the eradication of HIV-infected cells that merits further study in combination with cART and reactivation drugs.


Asunto(s)
Anticuerpos Monoclonales/inmunología , VIH-1/fisiología , Glicoproteínas de Membrana/inmunología , Anticuerpos Monoclonales/química , Línea Celular , Roturas del ADN de Doble Cadena/efectos de la radiación , VIH-1/efectos de la radiación , Humanos , Marcaje Isotópico , Leucocitos Mononucleares/virología , Monocitos/virología
9.
Expert Rev Vaccines ; 18(5): 505-521, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31009255

RESUMEN

INTRODUCTION: Adjuvants form an integral component in most of the inactivated and subunit vaccine formulations. Careful and proper selection of adjuvants helps in promoting appropriate immune responses against target pathogens at both innate and adaptive levels such that protective immunity can be elicited. Areas covered: Herein, we describe the recent progress in our understanding of the mode of action of adjuvants that are licensed for use in human vaccines or in clinical or pre-clinical stages at both innate and adaptive levels. Different pathogens have distinct characteristics, which require the host to mount an appropriate immune response against them. Adjuvants can be selected to elicit a tailor-made immune response to specific pathogens based on their unique properties. Identification of biomarkers of adjuvanticity for several candidate vaccines using omics-based technologies can unravel the mechanism of action of modern and experimental adjuvants. Expert opinion: Adjuvant technology has been revolutionized over the last two decades. In-depth understanding of the role of adjuvants in activating the innate immune system, combined with systems vaccinology approaches, have led to the development of next-generation, novel adjuvants that can be used in vaccines against challenging pathogens and in specific target populations.


Asunto(s)
Inmunidad Adaptativa , Adyuvantes Inmunológicos/farmacología , Inmunidad Innata , Vacunas de Productos Inactivados/inmunología , Vacunas de Subunidad/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Investigación Biomédica/tendencias , Humanos , Biología de Sistemas/tendencias , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Subunidad/administración & dosificación
10.
Antiviral Res ; 171: 104594, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31470041

RESUMEN

Respiratory syncytial virus (RSV) is responsible for a large proportion of acute lower respiratory tract infections, specifically in children. Pneumonia virus of mice (PVM) causes similar lung pathology and clinical disease in rodents, and is therefore an appropriate model of RSV infection. Previously, we demonstrated that a single intranasal dose of P-I-P, a novel immunomodulator composed of the toll-like receptor 3 agonist poly(I:C), an innate defense regulator peptide and a polyphosphazene, confers protection in Balb/c mice for up to 3 days from lethal PVM-15 infection. In the present study a dual intranasal treatment with P-I-P was shown to extend the duration of the protection conferred by P-I-P from PVM-15 challenge. Balb/c mice treated twice with P-I-P showed higher survival rates and milder clinical signs when compared to animals that received a single P-I-P dose. While the mice treated with two consecutive doses of P-I-P experienced some weight loss, they all recovered. The dual P-I-P treatment mediated infiltration of several innate immune cells into the BALF and lung, including alveolar macrophages, neutrophils, and γδ T cells. Partial depletion of alveolar macrophages decreased survival rates and exacerbated clinical signs of mice subjected to the P-I-P dual treatment regime followed by PVM-15 challenge. This suggests that the alveolar macrophage is at least partially responsible for the protection elicited by this novel prophylactic treatment strategy.


Asunto(s)
Inmunidad Innata , Factores Inmunológicos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Virus de la Neumonía Murina/efectos de los fármacos , Virus de la Neumonía Murina/inmunología , Infecciones por Pneumovirus/inmunología , Infecciones por Pneumovirus/virología , Animales , Línea Celular , Citocinas/biosíntesis , Citocinas/sangre , Femenino , Interacciones Huésped-Patógeno , Factores Inmunológicos/administración & dosificación , Macrófagos/metabolismo , Macrófagos/virología , Ratones , Infecciones por Pneumovirus/tratamiento farmacológico , Infecciones por Pneumovirus/mortalidad
11.
Vaccine ; 36(17): 2326-2336, 2018 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-29559168

RESUMEN

Respiratory syncytial virus (RSV) causes acute respiratory tract infections in infants, the elderly and immunocompromised individuals. No licensed vaccine is available against RSV. We previously reported that intranasal immunization of rodents and lambs with a RSV vaccine candidate (ΔF/TriAdj) induces protective immunity with a good safety profile. ΔF/TriAdj promoted innate immune responses in respiratory mucosal tissues in vivo, by local chemokine and cytokine production, as well as infiltration and activation of immune cells including macrophages. The macrophage is an important cell type in context of both innate and adaptive immune responses against RSV. Therefore, we characterized the effects of ΔF/TriAdj on a murine macrophage cell line, RAW264.7, and bone marrow-derived macrophages (BMMs). A gene expression study of pattern recognition receptors (PRRs) revealed induction of endosomal and cytosolic receptors in RAW264.7 cells and BMMs by ΔF/TriAdj, but no up-regulation by ΔF in PBS. As a secondary response to the PRR gene expression, induction of several chemokines and pro-inflammatory cytokines, as well as up-regulation of MHC-II and co-stimulatory immune markers, was observed. To further investigate the mechanisms involved in ΔF/TriAdj-mediated secondary responses, we used relevant signal transduction pathway inhibitors. Based on inhibition studies at both transcript and protein levels, JNK, ERK1/2, CaMKII, PI3K and JAK pathways were clearly responsible for ΔF/TriAdj-mediated chemokine and pro-inflammatory cytokine responses, while the p38 and NF-κB pathways appeared to be not or minimally involved. ΔF/TriAdj induced IFN-ß, which may participate in the JAK-STAT pathway to further amplify CXCL-10 production, which was strongly up-regulated. Blocking this pathway by a JAK inhibitor almost completely abrogated CXCL-10 production and caused a significant reduction in the cell surface expression of MHC-II and co-stimulatory immune markers. These data demonstrate that ΔF/TriAdj induces multiple signaling pathways in macrophages.


Asunto(s)
Macrófagos/inmunología , Polímeros/química , Infecciones por Virus Sincitial Respiratorio/inmunología , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Transducción de Señal/inmunología , Proteínas Virales de Fusión/inmunología , Adyuvantes Inmunológicos/química , Animales , Biomarcadores/metabolismo , Línea Celular , Quimiocinas/inmunología , Inmunidad Innata/inmunología , Inmunización/métodos , Inflamación/inmunología , Inflamación/metabolismo , Macrófagos/virología , Ratones , Células RAW 264.7 , Infecciones por Virus Sincitial Respiratorio/metabolismo , Vacunas contra Virus Sincitial Respiratorio/química , Vacunación/métodos , Proteínas Virales de Fusión/química
12.
Trends Parasitol ; 23(7): 317-24, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17531536

RESUMEN

Recent studies have shown that both Leishmania and HIV type-1 (HIV-1) hijack dendritic cell (DC) functions to escape immune surveillance using an array of elaborate strategies. Leishmania has developed a variety of adaptations to disrupt cellular defense mechanisms, whereas HIV-1 targets DCs to achieve a more efficient dissemination. The capacity of Leishmania and HIV-1 to target DCs through a common cell-surface molecule, namely DC-SIGN (dendritic cell specific ICAM-3-grabbing non-integrin), points to a possible dangerous liaison between these two pathogens. This review explores our knowledge of how Leishmania and HIV-1 interact dynamically with DCs, and how they exploit this cell type for their reciprocal benefit.


Asunto(s)
Células Dendríticas/parasitología , Células Dendríticas/virología , Infecciones por VIH/inmunología , VIH-1/inmunología , Leishmania infantum/inmunología , Leishmaniasis Visceral/inmunología , Animales , Moléculas de Adhesión Celular/inmunología , Citocinas/inmunología , Células Dendríticas/inmunología , Infecciones por VIH/parasitología , Humanos , Leishmaniasis Visceral/virología , Receptores de Superficie Celular/inmunología , Linfocitos T/inmunología , Linfocitos T/parasitología , Linfocitos T/virología
13.
Vaccine ; 35(27): 3466-3473, 2017 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-28527686

RESUMEN

Bovine viral diarrhea virus (BVDV) is one of the most serious pathogens in cattle. Recently, we developed a novel adjuvant platform (TriAdj) that includes a toll-like receptor 3 agonist, poly (I:C); an innate defense regulatory peptide; and water-soluble polymer, poly[di(sodiumcarboxylatoethylphenoxy)]-phosphazene (PCEP). To develop a needle-free intradermal (ID) subunit vaccine, the BVDV type-2 E2 protein was formulated with TriAdj, and immune protection was evaluated in calves against a BVDV-2 strain. Intradermal delivery of E2/TriAdj elicited robust virus neutralizing antibodies and cell-mediated immune responses including CD4+ and CD8+ T-cell responses. The development of CD8+ T-cell responses in vaccinated calves indicates that TriAdj promotes cross-presentation. Upon challenge with virulent BVDV-2, the vaccinated calves showed no weight loss, leukopenia or virus shedding, and almost no temperature increase, in contrast to the control animals, which had severe clinical disease and shed virus for three to six days in nasal fluids and white blood cells. Intradermal vaccination was shown to attract various immune cell populations including dendritic cells, the most important antigen presenting cells. These data demonstrate that ID delivery is suitable as an administration route in cattle and that ID delivered, TriAdj-formulated E2 can protect cattle from BVDV-2.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Diarrea Mucosa Bovina Viral/prevención & control , Virus de la Diarrea Viral Bovina Tipo 1/inmunología , Inmunidad Innata , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Peso Corporal , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Bovinos , Fiebre/prevención & control , Inyecciones Intradérmicas , Leucopenia/prevención & control , Vacunas Virales/administración & dosificación , Esparcimiento de Virus
14.
Vaccine ; 35(35 Pt A): 4486-4489, 2017 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-28599794

RESUMEN

Adjuvants are crucial components of many vaccines. They are used to improve the immunogenicity of vaccines with the aim of conferring long-term protection, to enhance the efficacy of vaccines in newborns, elderly or immunocompromised persons, and to reduce the amount of antigen or the number of doses required to elicit effective immunity. Novel combination adjuvants have been tested in both candidate animals and humans vaccines and have generated encouraging results. Recently, we developed a combination adjuvant platform (TriAdj) comprising of three components, namely a TLR agonist, either polyI:C or CpG oligodeoxynucleotides (ODN), host defense peptide and polyphosphazene. This adjuvant platform is stable and highly effective in a wide range of animal and human vaccines tested in mice, cotton rats, pigs, sheep, and koalas. TriAdj with various vaccines antigens induced effective long-term humoral and cellular immunity. Moreover, the adjuvant platform is suitable for maternal immunization and highly effective in neonates even in the presence of maternal antibodies. This novel vaccine platform, offers excellent opportunity for use in present and future generations of vaccines against multiple infectious agents and targets challenging populations.


Asunto(s)
Adyuvantes Inmunológicos/química , Oligodesoxirribonucleótidos/química , Compuestos Organofosforados/química , Polímeros/química , Vacunas Combinadas/química , Vacunas Combinadas/inmunología , Animales , Diseño de Fármacos , Femenino , Humanos , Inmunidad Celular/inmunología , Inmunidad Humoral/inmunología , Inmunidad Materno-Adquirida/inmunología , Lactante , Embarazo , Vacunas Combinadas/síntesis química
15.
Indian J Med Res ; 123(3): 439-54, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16778322

RESUMEN

Visceral leishmaniases (VL) or kala-azar is the most dreaded and devastating amongst the various forms of leishmaniases. The disease, though localized in certain areas only, has gained immense importance because of high mortality rate, mainly in children. The parasite is responsible for a spectrum of clinical syndromes, which can, in most extreme cases, go from an asymptomatic infection to a fatal form of VL. Chemotherapeutic measures, alone are not sufficient to control and contain the disease. As an alternate strategy, vaccination is also under experimental and clinical trails. The situation unquestionably demands the use of proper screening system, rationale chemical synthesis, vaccine development and targeted vaccine delivery. Thus, development of an acceptable vaccine is not an easy task. While the factors, which determine clinical outcomes, are in part, a feature of the parasite, it is the nature of the host and its genetic make up and immune status that play crucial role. The prerequisite of reliable animal model is that it should have a considerably good correlation with the clinical situation and is expected to mimic the pathological features and immunological responses observed in humans when exposed to a variety of Leishmania spp. with different pathogenic characteristics. Many experimental animal models like rodents, dogs and monkeys have been developed, each with specific features, but none accurately reproduces what happens in humans. In addition to the nature of the host, the major difference between natural and experimental infections is the parasite inoculum; in natural conditions, the infected sand fly vector deposits a few hundred metacyclic promastigotes into the dermis of the host, whereas experimental infections are induced by the injection (subcutaneous or intravenous) of millions of promastigotes grown in axenic cultures in vitro or amastigotes recovered from infected spleens. In public health terms, VL is the disease of humans and dogs (which may be considered secondary or 'accidental' hosts in the leishmanial life cycle) who often exhibit severe clinical signs and symptoms when infected, whereas reservoir hosts generally show a few, minor or no signs. This situation makes the definition of a suitable laboratory model a difficult one since the various experimental hosts may behave either like a reservoir or an accidental host. This review discusses the concept of animal models for VL and provides a critical evaluation of the most common experimental models and their respective advantages and disadvantages. Particular emphasis is given to the value of using mouse, hamster, dog and primate models, especially in the context of testing potential antileishmanial vaccines.


Asunto(s)
Leishmaniasis Visceral/prevención & control , Vacunas Antiprotozoos/química , Animales , Cricetinae , Modelos Animales de Enfermedad , Perros , Diseño de Fármacos , Industria Farmacéutica , Haplorrinos , Humanos , Leishmania/metabolismo , Leishmaniasis Visceral/parasitología , Leishmaniasis Visceral/terapia , Ratones , Resultado del Tratamiento
16.
Vaccine ; 34(42): 5114-5124, 2016 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-27591951

RESUMEN

Respiratory syncytial virus (RSV) causes serious upper and lower respiratory tract infections in newborns and infants. Presently, there is no licensed vaccine against RSV. We previously reported the safety and efficacy of a novel vaccine candidate (ΔF/TriAdj) in rodent and lamb models following intranasal immunization. However, the effects of the vaccine on the innate immune system in the upper and lower respiratory tracts, when delivered intranasally, have not been characterized. In the present study, we found that ΔF/TriAdj triggered transient production of chemokines, cytokines and interferons in the nasal tissues and lungs of BALB/c mice. The types of chemokines produced were consistent with the populations of immune cells recruited, i.e. dendritic cells, macrophages and neutrophils, in the nose-associated lymphoid tissue (NALT), lung and their draining lymph nodes of the ΔF/TriAdj-immunized group. In addition, ΔF/TriAdj stimulated cellular activation with generation of mucosal and systemic antibody responses, and conferred complete protection from viral infection in the lungs upon RSV challenge. The effect of ΔF/TriAdj was short-lived in the nasal tissues and more prolonged in the lungs. In addition, both innate and adaptive immune responses were lower when mice were immunized with ΔF alone. These results suggest that ΔF/TriAdj modulates the innate mucosal environment in both upper and lower respiratory tracts, which contributes to robust adaptive immune responses and long-term protective efficacy of this novel vaccine formulation.


Asunto(s)
Inmunidad Adaptativa , Adyuvantes Inmunológicos , Inmunidad Innata , Infecciones por Virus Sincitial Respiratorio/inmunología , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitial Respiratorio Humano/química , Proteínas Virales de Fusión/inmunología , Administración Intranasal , Animales , Anticuerpos Antivirales/sangre , Quimiocinas/biosíntesis , Citocinas/biosíntesis , Interferones/biosíntesis , Ratones , Ratones Endogámicos BALB C , Polímeros/química , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Virus Sincitial Respiratorio Humano/genética , Virus Sincitial Respiratorio Humano/inmunología , Virus Sincitiales Respiratorios/química , Virus Sincitiales Respiratorios/genética , Virus Sincitiales Respiratorios/inmunología , Sistema Respiratorio/inmunología , Sistema Respiratorio/virología , Proteínas Virales de Fusión/administración & dosificación , Proteínas Virales de Fusión/química
17.
Virology ; 499: 288-297, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27721128

RESUMEN

Respiratory syncytial virus (RSV) causes serious respiratory illness in infants and elderly. RSV infection induces short-lived immunity, which leaves people prone to re-infection. In contrast, the RSV fusion (F) protein formulated with a novel adjuvant (∆F/TriAdj) elicits long term protective immunity. A comparison of RSV-immunized mice to mice vaccinated with a single dose of ∆F/TriAdj showed no difference in IgG1 and IgG2a production; however, local IgA secreting memory B cell development and B cell IgA production were significantly lower in RSV vaccinated mice than in ∆F/TriAdj-immunized mice. This indicates a potential reason as to why long-term immunity is not induced by RSV infection. The comparison also revealed that germinal center lymphocyte populations were higher in ∆F/TriAdj-vaccinated mice. Furthermore, ∆F/TriAdj induced higher gene expression of activation-induced cytidine deaminase (AID), as well as IL-6, IL-21, TGF-ß cytokines, which are key players in IgA class switch recombination, ultimately leading to a sustained long-term memory response.


Asunto(s)
Anticuerpos Antivirales/inmunología , Inmunoglobulina A/inmunología , Mucosa Respiratoria/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Virus Sincitial Respiratorio Humano/inmunología , Vacunas de Subunidad/administración & dosificación , Vacunas Virales/administración & dosificación , Administración Intranasal , Animales , Linfocitos B/inmunología , Citocinas/inmunología , Composición de Medicamentos , Femenino , Humanos , Inmunización , Ratones Endogámicos BALB C , Mucosa Respiratoria/virología , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/virología , Vacunas contra Virus Sincitial Respiratorio/genética , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitial Respiratorio Humano/genética , Vacunas de Subunidad/genética , Vacunas de Subunidad/inmunología , Vacunas Virales/genética , Vacunas Virales/inmunología
18.
Antiviral Res ; 135: 108-119, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27771388

RESUMEN

Respiratory syncytial virus (RSV) is the leading cause of acute lower respiratory tract infections in infants and young children. There are no licensed RSV vaccines available, and the few treatment options for high-risk individuals are either extremely costly or cause severe side effects and toxicity. Immunomodulation mediated by a novel formulation consisting of the toll-like receptor 3 agonist poly(I:C), an innate defense regulator peptide and a polyphosphazene (P-I-P) was evaluated in the context of lethal infection with pneumonia virus of mice (PVM). Intranasal delivery of a single dose of P-I-P protected adult mice against PVM when given 24 h prior to challenge. These animals experienced minimal weight loss, no clinical disease, 100% survival, and reduced lung pathology. Similar clinical outcomes were observed in mice treated up to 3 days prior to infection. P-I-P pre-treatment induced early mRNA and protein expression of key chemokine and cytokine genes, reduced the recruitment of neutrophils and eosinophils, decreased virus titers in the lungs, and modulated the delayed exacerbated nature of PVM disease without any short-term side effects. On day 14 post-infection, P-I-P-treated mice were confirmed to be PVM-free. These results demonstrate the capacity of this formulation to prevent PVM and possibly other viral respiratory infections.


Asunto(s)
Inmunidad Innata , Factores Inmunológicos/administración & dosificación , Virus de la Neumonía Murina/inmunología , Compuestos Organofosforados/administración & dosificación , Infecciones por Pneumovirus/prevención & control , Poli I-C/administración & dosificación , Polímeros/administración & dosificación , Adyuvantes Inmunológicos , Administración Intranasal , Animales , Citocinas/inmunología , Factores Inmunológicos/química , Factores Inmunológicos/inmunología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Compuestos Organofosforados/inmunología , Infecciones por Pneumovirus/inmunología , Poli I-C/inmunología , Receptor Toll-Like 3/agonistas
19.
Peptides ; 25(11): 1873-81, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15501518

RESUMEN

The characteristic feature of visceral leishmaniasis (VL) is the profound impairment of immune system of the infected host, which contributes significantly to the partial success of antileishmanial chemotherapy. Since in VL, cure is the combinatorial effect of drug and immune status of the host, the rationale approach towards antileishmanial chemotherapy would be to potentiate the immune functioning of the host to extract desired results. Towards this direction several rationally designed analogues of human beta-casein fragment (54-59) were evaluated for their ability to stimulate the non-specific resistance in hamsters against Leishmania donovani infection. By virtue of being derived from the food protein casein derivatives may be devoid of unwanted side effects associated with the substances of microbial origin, e.g. muramyl dipeptide (MDP). Out of this one peptide Val-Glu-Gly-Ile-Pro-Tyr (compound 89/215) had been reported to have such activity. In this communication, the prophylactic and therapeutic efficacy of the peptide along with its natural sequence has been evaluated in detail against experimental VL in hamsters. Their use as an adjunct to chemotherapy was also explored. Human beta-casein fragment, compound 89/215 and MDP were tested in vivo at various dose levels wherein compound 89/215 showed superiority over MDP at 3 mg/kg x 2 given intraperitoneally (i.p.). Compound 89/215 sensitized peritoneal macrophages acquired considerable resistance and only 24% of the cells were found infected in comparison to control peritoneal macrophages where 76.4% of the cells were found infected. Similarly, the efficacy of sodium antimony gluconate (SAG) in hamsters pretreated with compound 89/215 enhanced significantly (P < 0.001). This peptide also exhibited considerably good therapeutic efficacy when evaluated either alone or in combination with SAG in established infection of L. donovani.


Asunto(s)
Antiprotozoarios/farmacología , Caseínas/farmacología , Leishmania donovani/efectos de los fármacos , Mesocricetus/parasitología , Fragmentos de Péptidos/farmacología , Acetilmuramil-Alanil-Isoglutamina/farmacología , Secuencia de Aminoácidos , Animales , Gluconato de Sodio Antimonio/uso terapéutico , Caseínas/síntesis química , Caseínas/química , Células Cultivadas , Cricetinae , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Ensayo de Inmunoadsorción Enzimática , Interacciones Huésped-Parásitos , Humanos , Inyecciones Intraperitoneales , Leishmania donovani/aislamiento & purificación , Leishmaniasis Visceral/terapia , Macrófagos Peritoneales/efectos de los fármacos , Masculino , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/química , Factores de Tiempo
20.
Vaccine ; 32(50): 6758-64, 2014 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-25454860

RESUMEN

Bovine viral diarrhea virus (BVDV) is still one of the most serious pathogens in cattle, meriting the development of improved vaccines. Recently, we developed a new adjuvant consisting of poly[di(sodium carboxylatoethylphenoxy)]-phosphazene (PCEP), either CpG ODN or poly(I:C), and an immune defense regulator (IDR) peptide. As this adjuvant has been shown to mediate the induction of robust, balanced immune responses, it was evaluated in an E2 subunit vaccine against BVDV in lambs and calves. The BVDV type 2 E2 protein was produced at high levels in a mammalian expression system and purified. When formulated with either CpG ODN or poly(I:C), together with IDR and PCEP, the E2 protein elicited high antibody titers and production of IFN-γ secreting cells in lambs. As the immune responses were stronger when poly(I:C) was used, the E2 protein with poly(I:C), IDR and PCEP was subsequently tested in cattle. Robust virus neutralizing antibodies as well as cell-mediated immune responses, including CD8(+) cytotoxic T cell (CTL) responses, were induced. The fact that CTL responses were demonstrated in calves vaccinated with an E2 protein subunit vaccine indicates that this adjuvant formulation promotes cross-presentation. Furthermore, upon challenge with a high dose of virulent BVDV-2, the vaccinated calves showed almost no temperature response, weight loss, leukopenia or virus replication, in contrast to the control animals, which had severe clinical disease. These data suggest that this E2 subunit formulation induces significant protection from BVDV-2 challenge, and thus is a promising BVDV vaccine candidate; in addition, the adjuvant platform has applications in bovine vaccines in general.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Enfermedades de los Bovinos/prevención & control , Virus de la Diarrea Viral Bovina Tipo 2/inmunología , Infecciones por Pestivirus/veterinaria , Enfermedades de las Ovejas/prevención & control , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Bovinos , Enfermedades de los Bovinos/patología , Interferón gamma/metabolismo , Leucocitos Mononucleares/inmunología , Infecciones por Pestivirus/patología , Infecciones por Pestivirus/prevención & control , Ovinos , Enfermedades de las Ovejas/patología , Linfocitos T Citotóxicos/inmunología , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología , Vacunas Virales/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA