Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Front Immunol ; 15: 1292059, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38370404

RESUMEN

Background: Previous studies have demonstrated enhanced efficacy of vaccine formulations that incorporate the chemokine macrophage inflammatory protein 3α (MIP-3α) to direct vaccine antigens to immature dendritic cells. To address the reduction in vaccine efficacy associated with a mutation in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mutants, we have examined the ability of receptor-binding domain vaccines incorporating MIP-3α to sustain higher concentrations of antibody when administered intramuscularly (IM) and to more effectively elicit lung T-cell responses when administered intranasally (IN). Methods: BALB/c mice aged 6-8 weeks were immunized intramuscularly or intranasally with DNA vaccine constructs consisting of the SARS-CoV-2 receptor-binding domain alone or fused to the chemokine MIP-3α. In a small-scale (n = 3/group) experiment, mice immunized IM with electroporation were followed up for serum antibody concentrations over a period of 1 year and for bronchoalveolar antibody levels at the termination of the study. Following IN immunization with unencapsulated plasmid DNA (n = 6/group), mice were evaluated at 11 weeks for serum antibody concentrations, quantities of T cells in the lungs, and IFN-γ- and TNF-α-expressing antigen-specific T cells in the lungs and spleen. Results: At 12 months postprimary vaccination, recipients of the IM vaccine incorporating MIP-3α had significantly, approximately threefold, higher serum antibody concentrations than recipients of the vaccine not incorporating MIP-3α. The area-under-the-curve analyses of the 12-month observation interval demonstrated significantly greater antibody concentrations over time in recipients of the MIP-3α vaccine formulation. At 12 months postprimary immunization, only recipients of the fusion vaccine had concentrations of serum-neutralizing activity deemed to be effective. After intranasal immunization, only recipients of the MIP-3α vaccine formulations developed T-cell responses in the lungs significantly above those of PBS controls. Low levels of serum antibody responses were obtained following IN immunization. Conclusion: Although requiring separate IM and IN immunizations for optimal immunization, incorporating MIP-3α in a SARS-CoV-2 vaccine construct demonstrated the potential of a stable and easily produced vaccine formulation to provide the extended antibody and T-cell responses that may be required for protection in the setting of emerging SARS-CoV-2 variants. Without electroporation, simple, uncoated plasmid DNA incorporating MIP-3α administered intranasally elicited lung T-cell responses.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Animales , Ratones , Formación de Anticuerpos , Quimiocinas , COVID-19/prevención & control , Vacunas contra la COVID-19/inmunología , ADN , Pulmón , SARS-CoV-2 , Linfocitos T
2.
Res Sq ; 2023 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-37645859

RESUMEN

Previous studies in the B16F10 mouse melanoma model have demonstrated that combining a DNA vaccine comprised of regions of gp100 and tyrosinase-related protein 2 fused to Macrophage-inflammatory protein 3-alpha (MIP3α) with recombinant Interferon alpha (IFN) and 5-Aza-2'-Deoxycytidine (5Aza) treatments resulted in significantly greater anti-tumor activity and immunogenicity in the tumor microenvironment (TME). This brief report details that the combination of vaccine with treatments IFN and 5Aza results in both the upregulation of genes expressing CD11c-interacting proteins and an increase in the TME of a distinct CD11c+ CD8+ T cell population. This cell population correlates with tumor size, is primarily comprised of effector or effector memory T cells, and has a more robust response to ex vivo stimulation as compared to CD11c- CD8+ T cells as measured by surface activation markers 4-1BB (CD137) and KLRG1 (Killer cell lectin-like receptor G1) and intracellular IFNγ production. In conclusion, this combination therapy results in greater presence of highly active effector CD8+ T-cells expressing CD11c in the TME that correlate with and are likely primary contributors to treatment efficacy.

3.
Front Immunol ; 13: 972266, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36189260

RESUMEN

Lengthy tuberculosis (TB) treatment is required to overcome the ability of a subpopulation of persistent Mycobacterium tuberculosis (Mtb) to remain in a non-replicating, antibiotic-tolerant state characterized by metabolic remodeling, including induction of the RelMtb-mediated stringent response. We developed a novel therapeutic DNA vaccine containing a fusion of the relMtb gene with the gene encoding the immature dendritic cell-targeting chemokine, MIP-3α/CCL20. To augment mucosal immune responses, intranasal delivery was also evaluated. We found that intramuscular delivery of the MIP-3α/relMtb (fusion) vaccine or intranasal delivery of the relMtb (non-fusion) vaccine potentiate isoniazid activity more than intramuscular delivery of the DNA vaccine expressing relMtb alone in a chronic TB mouse model (absolute reduction of Mtb burden: 0.63 log10 and 0.5 log10 colony-forming units, respectively; P=0.0002 and P=0.0052), inducing pronounced Mtb-protective immune signatures. The combined approach involving intranasal delivery of the DNA MIP-3α/relMtb fusion vaccine demonstrated the greatest mycobactericidal activity together with isoniazid when compared to each approach alone (absolute reduction of Mtb burden: 1.13 log10, when compared to the intramuscular vaccine targeting relMtb alone; P<0.0001), as well as robust systemic and local Th1 and Th17 responses. This DNA vaccination strategy may be a promising adjunctive approach combined with standard therapy to shorten curative TB treatment, and also serves as proof of concept for treating other chronic bacterial infections.


Asunto(s)
Tuberculosis , Vacunas de ADN , Animales , Antibacterianos , Células Dendríticas , Isoniazida , Ratones
4.
Front Immunol ; 13: 1074644, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36741387

RESUMEN

Introduction: DNA vaccines containing a fusion of the gene encoding chemokine MIP-3α (CCL20), the ligand for CCR6 on immature dendritic cells (DCs), to melanoma-associated antigen genes have enhanced anti-tumor immunity and efficacy compared to those lacking the chemokine gene. Previous work has shown that type-I interferon (IFNα or IFN) and 5-Aza-2'-deoxycytidine (5Aza) significantly enhance the therapeutic benefit of DNA vaccines as measured by reduced tumor burden and improved mouse survival. Methods: Here, we explored mouse intratumoral immune correlates underlying the therapeutic benefit of this combination regimen (vaccine, IFN, and 5Aza) as compared to vaccine alone and IFN and 5Aza without vaccine, focusing on chemokine mRNA expression by qRT-PCR and inflammatory cellular infiltration into the tumor microenvironment (TME) by flow cytometry and immunohistochemistry (IHC). Results: The combination group significantly upregulated intratumoral mRNA expression of key immune infiltration chemokines XCL1 and CXCL10. Flow cytometric analyses of tumor suspensions exhibited greater tumor infiltration of CD8+ DCs, CCR7+ DCs, and NK cells in the combination group, as well as reduced levels of myeloid-derived suppressor cells (MDSCs) in vaccinated groups. The mice receiving combination therapy also had greater proportions of effector/memory T-cells (Tem), in addition to showing an enhanced infiltration of Tem and central memory CD8+ T-cells, (Tcm). Tem and Tcm populations both correlated with smaller tumor size. Immunohistochemical analysis of tumors confirmed that CD8+ cells were more abundant overall and especially in the tumor parenchyma with combination therapy. Discussion: Efficient targeting of antigen to immature DCs with a chemokine-fusion vaccine offers a potential alternative approach to classic and dendritic cell-based vaccines. Combining this approach with IFNα and 5Aza treatments significantly improved vaccine efficacy. This treatment creates an environment of increased inflammatory chemokines that facilitates the trafficking of CD8+ DCs, NK cells, and CD8+ T-cells, especially memory cells, while reducing the number of MDSCs. Importantly, in the combination group, CD8+ cells were more able to penetrate the tumor mass in addition to being more numerous. Further analysis of the pathways engaged by our combination therapy is expected to provide additional insights into melanoma pathogenesis and facilitate the development of novel treatment strategies.


Asunto(s)
Vacunas contra el Cáncer , Melanoma , Vacunas de ADN , Animales , Ratones , Decitabina/farmacología , Interferón-alfa , ARN Mensajero , Microambiente Tumoral
5.
Sci Rep ; 11(1): 1220, 2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33441615

RESUMEN

Infants and young children are the groups at greatest risk for severe disease resulting from Plasmodium falciparum infection. We previously demonstrated in mice that a protein vaccine composed of the chemokine macrophage inflammatory protein 3α genetically fused to the minimally truncated circumsporozoite protein of P. falciparum (MCSP) elicits high concentrations of specific antibody and significant reduction of liver sporozoite load in a mouse model system. In the current study, a squalene based adjuvant (AddaVax, InvivoGen, San Diego, Ca) equivalent to the clinically approved MF59 (Seqiris, Maidenhead, UK) elicited greater antibody responses in mice than the previously employed adjuvant polyinosinic:polycytidylic acid, ((poly(I:C), InvivoGen, San Diego, Ca) and the clinically approved Aluminum hydroxide gel (Alum, Invivogen, San Diego, Ca) adjuvant. Use of the AddaVax adjuvant also expanded the range of IgG subtypes elicited by mouse vaccination. Sera passively transferred into mice from MCSP/AddaVax immunized 1 and 6 month old macaques significantly reduced liver sporozoite load upon sporozoite challenge. Protective antibody concentrations attained by passive transfer in the mice were equivalent to those observed in infant macaques 18 weeks after the final immunization. The efficacy of this vaccine in a relevant non-human primate model indicates its potential usefulness for the analogous high risk human population.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Formación de Anticuerpos/inmunología , Quimiocinas/inmunología , Células Dendríticas/inmunología , Macaca/inmunología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/inmunología , Esporozoítos/inmunología , Adyuvantes Inmunológicos/farmacología , Animales , Células Dendríticas/parasitología , Modelos Animales de Enfermedad , Femenino , Inmunización/métodos , Macaca/parasitología , Malaria Falciparum/parasitología , Ratones , Ratones Endogámicos C57BL , Plasmodium falciparum/inmunología , Poli I-C/inmunología , Proteínas Protozoarias/inmunología , Vacunación/métodos
6.
Front Immunol ; 11: 680, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32411131

RESUMEN

The lengthy and complicated current regimen required to treat drug-susceptible tuberculosis (TB) reflects the ability of Mycobacterium tuberculosis (Mtb) to persist in host tissues. The stringent response pathway, governed by the dual (p)ppGpp synthetase/hydrolase, Rel Mtb , is a major mechanism underlying Mtb persistence and antibiotic tolerance. In the current study, we addressed the hypothesis that Rel Mtb is a "persistence antigen" presented during TB chemotherapy and that enhanced immunity to Rel Mtb can enhance the tuberculocidal activity of the first-line anti-TB drug, isoniazid, which has reduced efficacy against Mtb persisters. C57BL/6 mice and Hartley guinea pigs were aerosol-infected with M. tuberculosis (Mtb) and, 4 weeks later, received either human-equivalent daily doses of isoniazid alone, or isoniazid in combination with a DNA vaccine targeting relMtb . After isoniazid treatment, there was a significant reduction in dominant antigen ESAT6-reactive CD4+ or TB10.4-reactive CD8+ T cells in the lungs and spleens of mice. However, the total number of Rel Mtb -reactive CD4+ T cells remained stable in mouse lungs and spleens, as did the number of Rel Mtb -reactive CD8+T cells. Therapeutic vaccination with relMtb DNA vaccine enhanced the activity of isoniazid in Mtb-infected C57BL/6 mice and guinea pigs. When treatment with isoniazid was discontinued, mice immunized with the relMtb DNA vaccine showed a lower mean lung bacterial burden at relapse compared to the control group. Our work shows that antitubercular treatment shapes the antigenic environment, and that therapeutic vaccination targeting the Mtb stringent response may represent a novel approach to enhance immunity against Mtb persisters, with the ultimate goal of shortening curative TB treatment.


Asunto(s)
Antituberculosos/uso terapéutico , Isoniazida/uso terapéutico , Mycobacterium tuberculosis/inmunología , Vacunas contra la Tuberculosis/uso terapéutico , Tuberculosis/tratamiento farmacológico , Tuberculosis/prevención & control , Vacunación/métodos , Vacunas de ADN/uso terapéutico , Animales , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular , Enfermedad Crónica/tratamiento farmacológico , Enfermedad Crónica/prevención & control , Farmacorresistencia Bacteriana/inmunología , Femenino , Guanosina Pentafosfato/metabolismo , Cobayas , Hidrolasas/inmunología , Hidrolasas/metabolismo , Ligasas/inmunología , Ligasas/metabolismo , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Mycobacterium tuberculosis/enzimología , Resultado del Tratamiento , Tuberculosis/inmunología , Tuberculosis/microbiología , Vacunas de ADN/inmunología
7.
Cancer Immunol Immunother ; 69(4): 569-580, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31980915

RESUMEN

BACKGROUND: The chemokine MIP-3α (CCL20) binds to CCR6 on immature dendritic cells. DNA vaccines fusing MIP-3α to melanoma-associated antigens have shown improved efficacy and immunogenicity in the B16F10 mouse melanoma model. Here, we report that the combination of type-I interferon therapy (IFNα) with 5-Aza-2'-deoxycitidine (5Aza) profoundly enhanced the therapeutic efficacy of a MIP-3α-Gp100-Trp2 DNA vaccine. METHODS: Beginning on day 5 post-transplantation of B16F10 melanoma, vaccine was administered intramuscularly (i.m.) by electroporation. CpG adjuvant was given 2 days later. 5Aza was given intraperitoneally at 1 mg/kg and IFNα therapy either intratumorally or i.m. as noted. Tumor sizes, tumor growth, and mouse survival were assessed. Tumor lysate gene expression levels and tumor-infiltrating lymphocytes (TILs) were assessed by qRT-PCR and flow cytometry, respectively. RESULTS: Adding IFNα and 5Aza treatments to mice vaccinated with MIP-3α-Gp100-Trp2 leads to reduced tumor burden and increased median survival (39% over vaccine and 95% over controls). Tumor lysate expression of CCL19 and CCR7 were upregulated ten and fivefold over vaccine, respectively. Vaccine-specific and overall CD8+ TILs were increased over vaccine (sevenfold and fourfold, respectively), as well as the proportion of TILs that were CD8+ (twofold). CONCLUSIONS: Efficient targeting of antigen to immature dendritic cells with a chemokine-fusion vaccine offers an alternative to classic and dendritic cell vaccines. Combining this approach with IFNα and 5Aza treatment significantly improved vaccine efficacy. This improved efficacy correlated with changes in chemokine gene expression and CD8+ TIL infiltration and was dependent on the presence of all therapeutic components.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Decitabina/inmunología , Células Dendríticas/inmunología , Interferón-alfa/inmunología , Melanoma Experimental/inmunología , Animales , Vacunas contra el Cáncer/administración & dosificación , Línea Celular Tumoral , Quimiocina CCL19/genética , Quimiocina CCL19/inmunología , Metilación de ADN/efectos de los fármacos , Decitabina/administración & dosificación , Células Dendríticas/citología , Femenino , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Inmunoterapia/métodos , Interferón-alfa/administración & dosificación , Estimación de Kaplan-Meier , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma Experimental/genética , Melanoma Experimental/terapia , Ratones Endogámicos C57BL , Receptores CCR7/genética , Receptores CCR7/inmunología
8.
Nat Microbiol ; 4(12): 2552-2564, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31659299

RESUMEN

Innate immunity is the first line of host defence against pathogens. Suppression of innate immune responses is essential for the survival of all viruses. However, the interplay between innate immunity and HIV/SIV is only poorly characterized. We have discovered Vpx as a novel inhibitor of innate immune activation that associates with STING signalosomes and interferes with the nuclear translocation of NF-κB and the induction of innate immune genes. This new function of Vpx could be separated from its role in mediating degradation of the antiviral factor SAMHD1, and is conserved among diverse HIV-2/SIV Vpx. Vpx selectively suppressed cGAS-STING-mediated nuclear factor-κB signalling. Furthermore, Vpx and Vpr had complementary activities against cGAS-STING activity. Since SIVMAC lacking both Vpx and Vpr was less pathogenic than SIV deficient for Vpr or Vpx alone, suppression of innate immunity by HIV/SIV is probably a key pathogenic determinant, making it a promising target for intervention.


Asunto(s)
VIH-2/metabolismo , Proteínas de la Membrana/metabolismo , FN-kappa B/metabolismo , Nucleotidiltransferasas/metabolismo , Transducción de Señal , Virus de la Inmunodeficiencia de los Simios/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Adulto , Secuencia de Aminoácidos , Animales , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-2/genética , Células HeLa , Humanos , Inmunidad Innata/genética , Masculino , Ratones Endogámicos BALB C , Proteína 1 que Contiene Dominios SAM y HD/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/genética , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas Reguladoras y Accesorias Virales/inmunología , Adulto Joven , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/inmunología , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/metabolismo
9.
Eye (Lond) ; 33(5): 845-849, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30651593

RESUMEN

BACKGROUND/OBJECTIVES: Retinopathy of prematurity (ROP) is a potentially blinding disorder affecting premature infants. Our Eye Unit supports two neonatal intensive care units (NICUs), one provides neonatal surgical and medical facilities and the other is exclusively medical. Our objectives were to (1) to identify the annual rate of ROP treatments during the period 2009-2015 and (2) to estimate the incidence of ROP treatment in babies born very prematurely (<27 weeks). SUBJECTS/METHODS: Records for all infants treated for ROP by our unit during the period 2009-2015 were reviewed. We calculated numbers treated in each year. Records of babies born under 27 weeks of gestation and cared for in the non-surgical NICU were also reviewed. Their requirement for laser treatments for ROP was calculated by the week of gestation at birth. RESULTS: In the two NICUs combined, 95 infants were treated for ROP between 2009 and 2015. The numbers treated increased from 9/158 (5.7%) of babies screened in 2009 to 22/159 (13.8%) in 2015 (ptrend = 0.004). The rate of laser treatment for ROP increased as gestation at birth decreased: from 12/100 (12%) of babies born at 26 weeks to 17/29 (59%) of babies born at 23 weeks (ptrend = 0.001). CONCLUSION: The number of laser treatments for ROP carried out by this unit has increased steadily between 2009 and 2015 and this may in part be due to the increased need for ROP treatment in extremely preterm babies, whose survival has increased in the same period. These data may aid planning for ROP services.


Asunto(s)
Coagulación con Láser/estadística & datos numéricos , Retinopatía de la Prematuridad/epidemiología , Retinopatía de la Prematuridad/cirugía , Femenino , Edad Gestacional , Humanos , Incidencia , Lactante , Recién Nacido de Bajo Peso , Recién Nacido , Recien Nacido Prematuro , Unidades de Cuidado Intensivo Neonatal , Masculino , Estudios Retrospectivos , Factores de Riesgo , Índice de Severidad de la Enfermedad , Centros de Atención Terciaria
11.
J Immunother ; 41(4): 181-189, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29334492

RESUMEN

The chemokine MIP3α (CCL20) binds to CCR6 on immature dendritic cells. Vaccines fusing MIP3α to gp100 have been shown to be effective in therapeutically reducing melanoma tumor burden and prolonging survival in a mouse model. Other studies have provided evidence that interleukin-10 (IL-10) neutralizing antibodies (αIL-10) enhance immunologic melanoma therapies by modulating the tolerogenic tumor microenvironment. In the current study, we have utilized the B16F10 syngeneic mouse melanoma model to demonstrate for the first time that a therapy neutralizing IL-10 enhances the antitumor efficacy of a MIP3α-gp100 DNA vaccine, leading to significantly smaller tumors, slower growing tumors, and overall increases in mouse survival. The additive effects of αIL-10 were not shown to be correlated to vaccine-specific tumor-infiltrating lymphocytes (TILs), total TILs, or regulatory T cells. However, we discovered an upregulation of IFNα-4 transcripts in tumors and a correlation of increased plasmacytoid dendritic cell numbers with reduced tumor burden in αIL-10-treated mice. Interferon α receptor knockout (IFNαR1) mice received no benefit from αIL-10 treatment, demonstrating that the additional therapeutic value of αIL-10 is primarily mediated by type I IFNs. Efficient targeting of antigen to immature dendritic cells with a chemokine-fusion vaccine provides an effective anticancer therapeutic. Combining this approach with an IL-10 neutralizing antibody therapy enhances the antitumor efficacy of the therapy in a manner dependent upon the activity of type I IFNs. This combination of a vaccine and immunomodulatory agent provides direction for future optimization of a novel cancer vaccine therapy.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Quimiocina CCL20/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Interferón Tipo I/metabolismo , Interleucina-10/antagonistas & inhibidores , Antígeno gp100 del Melanoma/inmunología , Animales , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Inmunoterapia , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma Experimental , Ratones , Ratones Noqueados , Resultado del Tratamiento , Vacunación
12.
Virology ; 511: 23-29, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28803141

RESUMEN

The lentiviral accessory protein Vpx is critical for viral infection of myeloid cells and acts by hijacking CRL4(DCAF1) E3 ubiquitin ligase to induce the degradation of the host restriction factor SAMHD1. It has been observed that the sequences from HIV-2 and SIVsmm/SIVmac Vpx contain a poly-proline tail which is distinct from other SIV Vpx proteins. However, the role of this region in Vpx function is controversial. Herein, we found proteasome-dependent degradation of a Vpx mutant lacking the poly-proline tail in the nucleus in a CRL4(DCAF1) E3 ligase-independent fashion. Unlike wild-type Vpx, the poly-proline tail mutant Vpx is partly defective in enhancing viral infection in macrophages. Our findings suggest that during Vpx evolution, Vpx of the HIV-2/SIVsm/SIVmac lineage is targeted by a CRL4(DCAF1) E3 ligase-independent ubiquitination pathway, and have gained this interesting region, allowing them to maintain nuclear accumulation as part of their adaptation to host cell regulation.


Asunto(s)
Inhibidores Enzimáticos/metabolismo , Interacciones Huésped-Patógeno , Péptidos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Virus de la Inmunodeficiencia de los Simios/efectos de los fármacos , Proteínas Reguladoras y Accesorias Virales/metabolismo
13.
J Virol ; 91(13)2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28424289

RESUMEN

Coxsackievirus A16 (CV-A16), CV-A6, and enterovirus D68 (EV-D68) belong to the Picornaviridae family and are major causes of hand, foot, and mouth disease (HFMD) and pediatric respiratory disease worldwide. The biological characteristics of these viruses, especially their interplay with the host innate immune system, have not been well investigated. In this study, we discovered that the 3Cpro proteins from CV-A16, CV-A6, and EV-D68 bind melanoma differentiation-associated gene 5 (MDA5) and inhibit its interaction with MAVS. Consequently, MDA5-triggered type I interferon (IFN) signaling in the retinoic acid-inducible gene I-like receptor (RLR) pathway was blocked by the CV-A16, CV-A6, and EV-D68 3Cpro proteins. Furthermore, the CV-A16, CV-A6, and EV-D68 3Cpro proteins all cleave transforming growth factor ß-activated kinase 1 (TAK1), resulting in the inhibition of NF-κB activation, a host response also critical for Toll-like receptor (TLR)-mediated signaling. Thus, our data demonstrate that circulating HFMD-associated CV-A16 and CV-A6, as well as severe respiratory disease-associated EV-D68, have developed novel mechanisms to subvert host innate immune responses by targeting key factors in the RLR and TLR pathways. Blocking the ability of 3Cpro proteins from diverse enteroviruses and coxsackieviruses to interfere with type I IFN induction should restore IFN antiviral function, offering a potential novel antiviral strategy.IMPORTANCE CV-A16, CV-A6, and EV-D68 are emerging pathogens associated with hand, foot, and mouth disease and pediatric respiratory disease worldwide. The pathogenic mechanisms of these viruses are largely unknown. Here we demonstrate that the CV-A16, CV-A6, and EV-D68 3Cpro proteins block MDA5-triggered type I IFN induction. The 3Cpro proteins of these viruses bind MDA5 and inhibit its interaction with MAVS. In addition, the CV-A16, CV-A6, and EV-D68 3Cpro proteins cleave TAK1 to inhibit the NF-κB response. Thus, our data demonstrate that circulating HFMD-associated CV-A16 and CV-A6, as well as severe respiratory disease-associated EV-D68, have developed a mechanism to subvert host innate immune responses by simultaneously targeting key factors in the RLR and TLR pathways. These findings indicate the potential merit of targeting the CV-A16, CV-A6, and EV-D68 3Cpro proteins as an antiviral strategy.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Enterovirus/inmunología , Enterovirus/patogenicidad , Interacciones Huésped-Patógeno , Evasión Inmune , Inmunidad Innata , Helicasa Inducida por Interferón IFIH1/antagonistas & inhibidores , Proteínas Virales/metabolismo , Proteasas Virales 3C , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Proteolisis , Transducción de Señal
14.
Vaccine ; 35(18): 2358-2364, 2017 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-28342669

RESUMEN

Mouse studies evaluating candidate malaria vaccines have typically examined protective efficacy over the relatively short time frames of several weeks after the final of multiple immunizations. The current study examines the protective ability in a mouse model system of a novel protein vaccine construct in which the adjuvant polyinosinic polycytidilic acid (poly(I:C)) is used in combination with a vaccine in which the immature dendritic cell targeting chemokine, macrophage inflammatory protein 3 alpha (MIP3α), is fused to the circumsporozoite protein (CSP) of Plasmodium falciparum (P. falciparum). Two vaccinations, three weeks apart, elicited extraordinarily high, MIP3α-dependent antibody responses. MIP3α was able to target the vaccine to the CCR6 receptor found predominantly on immature dendritic cells and significantly enhanced the cellular influx at the vaccination site. At three and 23 weeks after the final of two immunizations, mice were challenged by intravenous injection of 5×103 transgenic Plasmodium berghei sporozoites expressing P. falciparum CSP, a challenge dose approximately one order of magnitude greater than that which is encountered after mosquito bite in the clinical setting. A ninety-seven percent reduction in liver sporozoite load was observed at both time points, 23 weeks being the last time point tested.


Asunto(s)
Células Dendríticas/inmunología , Vacunas contra la Malaria/inmunología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/genética , Adyuvantes Inmunológicos/metabolismo , Animales , Quimiocina CCL20/genética , Quimiocina CCL20/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Hígado/parasitología , Malaria/prevención & control , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/genética , Ratones Endogámicos C57BL , Carga de Parásitos , Plasmodium berghei/inmunología , Plasmodium berghei/aislamiento & purificación , Poli I-C/administración & dosificación , Proteínas Protozoarias/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
15.
J Virol ; 91(9)2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28202763

RESUMEN

The lentiviral accessory proteins Vpx and Vpr are known to utilize CRL4 (DCAF1) E3 ligase to induce the degradation of the host restriction factor SAMHD1 or host helicase transcription factor (HLTF), respectively. Selective disruption of viral CRL4 (DCAF1) E3 ligase could be a promising antiviral strategy. Recently, we have determined that posttranslational modification (neddylation) of Cullin-4 is required for the activation of Vpx-CRL4 (DCAF1) E3 ligase. However, the mechanism of Vpx/Vpr-CRL4 (DCAF1) E3 ligase assembly is still poorly understood. Here, we report that zinc coordination is an important regulator of Vpx-CRL4 E3 ligase assembly. Residues in a conserved zinc-binding motif of Vpx were essential for the recruitment of the CRL4 (DCAF1) E3 complex and Vpx-induced SAMHD1 degradation. Importantly, altering the intracellular zinc concentration by treatment with the zinc chelator N,N,N'-tetrakis-(2'-pyridylmethyl)ethylenediamine (TPEN) potently blocked Vpx-mediated SAMHD1 degradation and inhibited wild-type SIVmac (simian immunodeficiency virus of macaques) infection of myeloid cells, even in the presence of Vpx. TPEN selectively inhibited Vpx and DCAF1 binding but not the Vpx-SAMHD1 interaction or Vpx virion packaging. Moreover, we have shown that zinc coordination is also important for the assembly of the HIV-1 Vpr-CRL4 E3 ligase. In particular, Vpr zinc-binding motif mutation or TPEN treatment efficiently inhibited Vpr-CRL4 (DCAF1) E3 ligase assembly and Vpr-mediated HLTF degradation or Vpr-induced G2 cell cycle arrest. Collectively, our study sheds light on a conserved strategy by the viral proteins Vpx and Vpr to recruit host CRL4 (DCAF1) E3 ligase, which represents a target for novel anti-human immunodeficiency virus (HIV) drug development.IMPORTANCE The Vpr and its paralog Vpx are accessory proteins encoded by different human immunodeficiency virus (HIV)/simian immunodeficiency virus (SIV) lentiviruses. To facilitate viral replication, Vpx has evolved to induce SAMHD1 degradation and Vpr to mediate HLTF degradation. Both Vpx and Vpr perform their functions by recruiting CRL4 (DCAF1) E3 ligase. In this study, we demonstrate that the assembly of the Vpx- or Vpr-CRL4 E3 ligase requires a highly conserved zinc-binding motif. This motif is specifically required for the DCAF1 interaction but not for the interaction of Vpx or Vpr with its substrate. Selective disruption of Vpx- or Vpr-CRL4 E3 ligase function was achieved by zinc sequestration using N,N,N'-tetrakis-(2'-pyridylmethyl)ethylenediamine (TPEN). At the same time, zinc sequestration had no effect on zinc-dependent cellular protein functions. Therefore, information obtained from this study may be important for novel anti-HIV drug development.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/metabolismo , Línea Celular , Proteínas de Unión al ADN/metabolismo , Etilenodiaminas/farmacología , Puntos de Control de la Fase G2 del Ciclo Celular , Células HEK293 , Infecciones por VIH/virología , VIH-1/metabolismo , Interacciones Huésped-Patógeno , Humanos , Células Mieloides/virología , Proteínas Serina-Treonina Quinasas , Estructura Terciaria de Proteína , Proteína 1 que Contiene Dominios SAM y HD , Virus de la Inmunodeficiencia de los Simios/metabolismo , Factores de Transcripción/metabolismo , Proteínas Reguladoras y Accesorias Virales/genética , Replicación Viral , Zinc/metabolismo , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/genética
16.
J Immunother Cancer ; 4: 96, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28018602

RESUMEN

BACKGROUND: Although therapeutic cancer vaccines have been mostly disappointing in the clinic, the advent of novel immunotherapies and the future promise of neoantigen-based therapies have created the need for new vaccine modalities that can easily adapt to current and future developments in cancer immunotherapy. One such novel platform is a DNA vaccine fusing the chemokine Macrophage Inflammatory Protein-3α (MIP-3α) to an antigen, here melanoma antigen gp100. Previous published work has indicated that MIP-3α targets nascent peptides to immature dendritic cells, leading to processing by class I and II MHC pathways. This platform has shown enhanced efficacy in prophylactic melanoma and therapeutic lymphoma model systems. METHODS: The B16F10 melanoma syngeneic mouse model system was utilized, with a standard therapeutic protocol: challenge with lethal dose of B16F10 cells (5 × 104) on day 0 and then vaccinate by intramuscular electroporation with 50 µg plasmid on days three, 10, and 17. Efficacy was assessed by analysis of tumor burden, tumor growth, and mouse survival, using the statistical tests ANOVA, mixed effects regression, and log-rank, respectively. Immunogenicity was assessed by ELISA and flow cytometric methods, including intracellular cytokine staining to assess vaccine-specific T-cell responses, all tested by ANOVA. RESULTS: We demonstrate that the addition of MIP3α to gp100 significantly enhances systemic anti-gp100 immunological parameters. Further, chemokine-fusion vaccine therapy significantly reduces tumor burden, slows tumor growth, and enhances mouse overall survival compared to antigen-only, irrelevant-antigen, and mock vaccines, with efficacy mediated by both CD4+ and CD8+ effector T cells. Antigen-only, irrelevant-antigen, and chemokine-fusion vaccines elicit significantly higher and similar CD4+ and CD8+ tumor-infiltrating lymphocyte (TIL) levels compared to mock vaccine. However, vaccine-specific CD8+ TILs are significantly higher in the chemokine-fusion vaccine group, indicating that the critical step induced by the fusion vaccine construct is the enhancement of vaccine-specific T-cell effectors. CONCLUSIONS: The current study shows that fusion of MIP3α to melanoma antigen gp100 enhances the immunogenicity and efficacy of a DNA vaccine in a therapeutic B16F10 mouse melanoma model. This study analyzes an adaptable and easily produced MIP3α-antigen modular vaccine platform that could lend itself to a variety of functionalities, including combination treatments and neoantigen vaccination in the pursuit of personalized cancer therapy.

17.
EBioMedicine ; 8: 230-236, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27428433

RESUMEN

Recent studies have identified human myxovirus resistance protein 2 (MxB or Mx2) as an interferon induced inhibitor of HIV-1 replication. However, whether HIV-1 can overcome MxB restriction without compromise of viral fitness has been undefined. Here, we have discovered that naturally occurring capsid (CA) variants can render HIV-1 resistant to the activity of MxB without losing viral infectivity or the ability to escape from interferon induction. Moreover, these MxB resistant HIV-1 variants do not lose MxB recognition. Surprisingly, MxB resistant CA variants are most commonly found in the Clade C HIV-1 that is the most rapidly expanding Clade throughout the world. Accumulation of MxB resistant mutations is also observed during HIV-1 spreading in human populations. These findings support a potential role for MxB as a selective force during HIV-1 transmission and evolution.


Asunto(s)
Fármacos Anti-VIH/farmacología , Proteínas de la Cápside/genética , Farmacorresistencia Viral , Infecciones por VIH/epidemiología , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , VIH-1/genética , Proteínas de Resistencia a Mixovirus/farmacología , Proteínas de la Cápside/química , China/epidemiología , Aptitud Genética , Variación Genética , Geografía , Humanos , Modelos Moleculares , Vigilancia de la Población , Conformación Proteica , Replicación Viral
18.
PLoS One ; 10(12): e0144208, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26630383

RESUMEN

Enterovirus 68 (EVD68) causes respiratory illness, mostly in children. Despite a reported low-level of transmission, the occurrence of several recent outbreaks worldwide including the 2014 outbreak in North America has raised concerns regarding the pathogenesis and evolution of EVD68. To elucidate the phylogenetic features of EVD68 and possible causes for the 2014 outbreak, 216 EVD68 strain sequences were retrieved from Genbank, including 22 from the 2014 outbreak. Several geographic and genotypic origins were established for these 22 strains, 19 of which were classified as Clade B. Of these 19 strains, 17 exhibited subsequent clustering and variation in protein residues involved in host-receptor interaction and/or viral antigenicity. Approximately 18 inter-clade variations were detected in VP1, which led to the identification of a new Clade D in EVD68 strains. The classification of this new clade was also verified by the re-construction of a Neighbor-Joining tree during the phylogenetic analysis. In addition, our results indicate that members of Clade B containing highly specific alterations in VP1 protein residues were the foremost contributors to the 2014 outbreak in the US. Altered host-receptor interaction and/or host immune recognition may explain the evolution of EVD68 as well as the global emergence and ongoing adaptation of this virus.


Asunto(s)
Enterovirus Humano D/genética , Secuencia de Aminoácidos , Evolución Biológica , Proteínas de la Cápside/genética , Análisis por Conglomerados , Brotes de Enfermedades , Infecciones por Enterovirus/genética , Infecciones por Enterovirus/virología , Genotipo , Humanos , América del Norte/epidemiología , Filogenia , ARN Viral/genética , Análisis de Secuencia de ADN/métodos
19.
J Acquir Immune Defic Syndr ; 69(3): 278-85, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25828964

RESUMEN

BACKGROUND: Poor adherence to prevention regimens for gel-based anti-HIV-1 microbicides has been a major obstacle to more effective pre-exposure prophylaxis. Concern persists that the antiretroviral drug containing microbicides might promote development of antiretroviral resistance. METHODS: Using in vitro transwell systems and a humanized mouse model of HIV-1 sexual transmission, we examined, as candidate microbicides, antibodies targeting the heterodimeric leukocyte function-associated antigen 1 (LFA-1), a non-virally encoded protein acquired by the virus that also plays a critical role cell movement across endothelial and epithelial barriers. LFA-1-specific single domain variable regions from alpaca heavy-chain only antibodies (VHH) were identified and evaluated for their ability to inhibit HIV-1 transmission in the in vitro transwell system. RESULTS: Monoclonal antibodies targeting the CD11a and CD18 components of LFA-1 significantly reduced cell-free and cell-associated HIV-1 transmission in the in vitro transwell culture system and prevented virus transmission in the humanized mouse model of vaginal transmission. The broadly neutralizing monoclonal antibody b12 was unable to block transmission of cell-free virus. CD11a-specific VHH were isolated and expressed and the purified variable region protein domains reduced in vitro transepithelial transmission with an efficacy comparable with that of the CD11a monoclonal antibody. CONCLUSIONS: Targeting integrins acquired by HIV-1 during budding and which are critical to interactions between epithelial cells and lymphocytes can reduce viral movement across epithelial barriers and prevent transmission in a humanized mouse model of sexual transmission. VHH capable of being produced by transformed bacteria can significantly reduce transepithelial virus transmission in in vitro model systems.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Infecciones por VIH/transmisión , VIH-1/fisiología , Cadenas beta de Integrinas/inmunología , Animales , Especificidad de Anticuerpos , Antígeno CD11a/inmunología , Antígenos CD18/inmunología , Camélidos del Nuevo Mundo , Línea Celular , Células Epiteliales/virología , Infecciones por VIH/prevención & control , VIH-1/inmunología , Humanos , Células Jurkat , Leucocitos Mononucleares , Ratones , Ratones SCID , Biblioteca de Péptidos
20.
Biochem Biophys Res Commun ; 459(2): 214-219, 2015 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-25704090

RESUMEN

Although the Vpr protein of human immunodeficiency virus type 1 (HIV-1) has been shown to act as a transcriptional activator of the HIV-1 LTR and certain host genes, the current study demonstrates that it can also function as a potent inhibitor of the cytomegalovirus (CMV) promoter. Previous studies have shown that the cell cycle arrest and apoptotic functions of Vpr required recruitment of the CRL4(DCAF1) E3 ligase, but this complex is shown not to be required for inhibition of the CMV promoter. We identified conserved sites (A30/V31) from diverse Vpr from HIV/SIV that were critical for blocking the CMV promoter activity. Interestingly, the Vpr mutant A30S/V31S protein also impaired the ability of Vpr to down-regulate transcription of the host UNG2 gene. Our findings shed light on the dual functions of Vpr on the transcription of HIV-1, other viruses and host genes which may contribute to viral replication and disease progression in vivo.


Asunto(s)
Citomegalovirus/genética , VIH-1/genética , VIH-1/metabolismo , Regiones Promotoras Genéticas , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/metabolismo , Proteínas Portadoras/metabolismo , ADN Glicosilasas/genética , Células HEK293 , Duplicado del Terminal Largo de VIH , VIH-1/patogenicidad , Células HeLa , Interacciones Huésped-Patógeno/genética , Humanos , Modelos Moleculares , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación , Proteínas Nucleares/metabolismo , Conformación Proteica , Proteínas Serina-Treonina Quinasas , Ubiquitina-Proteína Ligasas/metabolismo , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA