Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 54(9): 2143-2158.e15, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34453881

RESUMEN

Neutralizing antibodies (NAbs) are effective in treating COVID-19, but the mechanism of immune protection is not fully understood. Here, we applied live bioluminescence imaging (BLI) to monitor the real-time effects of NAb treatment during prophylaxis and therapy of K18-hACE2 mice intranasally infected with SARS-CoV-2-nanoluciferase. Real-time imaging revealed that the virus spread sequentially from the nasal cavity to the lungs in mice and thereafter systemically to various organs including the brain, culminating in death. Highly potent NAbs from a COVID-19 convalescent subject prevented, and also effectively resolved, established infection when administered within three days. In addition to direct neutralization, depletion studies indicated that Fc effector interactions of NAbs with monocytes, neutrophils, and natural killer cells were required to effectively dampen inflammatory responses and limit immunopathology. Our study highlights that both Fab and Fc effector functions of NAbs are essential for optimal in vivo efficacy against SARS-CoV-2.


Asunto(s)
Anticuerpos Neutralizantes/metabolismo , Anticuerpos Antivirales/metabolismo , Encéfalo/patología , COVID-19/inmunología , Pulmón/patología , SARS-CoV-2/fisiología , Testículo/patología , Enzima Convertidora de Angiotensina 2/genética , Animales , Anticuerpos Neutralizantes/genética , Anticuerpos Antivirales/genética , Encéfalo/virología , COVID-19/terapia , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Luciferasas/genética , Mediciones Luminiscentes , Pulmón/virología , Masculino , Ratones , Ratones Transgénicos , Testículo/virología
2.
J Cell Sci ; 137(10)2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38682259

RESUMEN

SARS-CoV-2 interferes with antigen presentation by downregulating major histocompatibility complex (MHC) II on antigen-presenting cells, but the mechanism mediating this process is unelucidated. Herein, analysis of protein and gene expression in human antigen-presenting cells reveals that MHC II is downregulated by the SARS-CoV-2 main protease, NSP5. This suppression of MHC II expression occurs via decreased expression of the MHC II regulatory protein CIITA. CIITA downregulation is independent of the proteolytic activity of NSP5, and rather, NSP5 delivers HDAC2 to the transcription factor IRF3 at an IRF-binding site within the CIITA promoter. Here, HDAC2 deacetylates and inactivates the CIITA promoter. This loss of CIITA expression prevents further expression of MHC II, with this suppression alleviated by ectopic expression of CIITA or knockdown of HDAC2. These results identify a mechanism by which SARS-CoV-2 limits MHC II expression, thereby delaying or weakening the subsequent adaptive immune response.


Asunto(s)
Antígenos de Histocompatibilidad Clase II , Histona Desacetilasa 2 , Proteínas Nucleares , Regiones Promotoras Genéticas , SARS-CoV-2 , Transactivadores , Humanos , Presentación de Antígeno/genética , Células Presentadoras de Antígenos/metabolismo , Células Presentadoras de Antígenos/inmunología , COVID-19/virología , COVID-19/inmunología , COVID-19/genética , COVID-19/metabolismo , Cisteína Endopeptidasas/metabolismo , Cisteína Endopeptidasas/genética , Regulación hacia Abajo/genética , Células HEK293 , Antígenos de Histocompatibilidad Clase II/metabolismo , Antígenos de Histocompatibilidad Clase II/genética , Histona Desacetilasa 2/metabolismo , Histona Desacetilasa 2/genética , Factor 3 Regulador del Interferón/metabolismo , Factor 3 Regulador del Interferón/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Regiones Promotoras Genéticas/genética , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , SARS-CoV-2/inmunología , Transactivadores/metabolismo , Transactivadores/genética , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética
4.
PLoS Pathog ; 17(12): e1010092, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34914812

RESUMEN

The development of safe and effective vaccines to prevent SARS-CoV-2 infections remains an urgent priority worldwide. We have used a recombinant vesicular stomatitis virus (rVSV)-based prime-boost immunization strategy to develop an effective COVID-19 vaccine candidate. We have constructed VSV genomes carrying exogenous genes resulting in the production of avirulent rVSV carrying the full-length spike protein (SF), the S1 subunit, or the receptor-binding domain (RBD) plus envelope (E) protein of SARS-CoV-2. Adding the honeybee melittin signal peptide (msp) to the N-terminus enhanced the protein expression, and adding the VSV G protein transmembrane domain and the cytoplasmic tail (Gtc) enhanced protein incorporation into pseudotype VSV. All rVSVs expressed three different forms of SARS-CoV-2 spike proteins, but chimeras with VSV-Gtc demonstrated the highest rVSV-associated expression. In immunized mice, rVSV with chimeric S protein-Gtc derivatives induced the highest level of potent neutralizing antibodies and T cell responses, and rVSV harboring the full-length msp-SF-Gtc proved to be the superior immunogen. More importantly, rVSV-msp-SF-Gtc vaccinated animals were completely protected from a subsequent SARS-CoV-2 challenge. Overall, we have developed an efficient strategy to induce a protective response in SARS-CoV-2 challenged immunized mice. Vaccination with our rVSV-based vector may be an effective solution in the global fight against COVID-19.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Vacunas contra la COVID-19/administración & dosificación , COVID-19/prevención & control , Glicoproteína de la Espiga del Coronavirus/inmunología , Virus de la Estomatitis Vesicular Indiana/genética , Enzima Convertidora de Angiotensina 2/genética , Animales , COVID-19/inmunología , COVID-19/virología , Vacunas contra la COVID-19/genética , Chlorocebus aethiops , Humanos , Inmunización , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Células Vero , Proteínas Virales/genética , Proteínas Virales/inmunología
5.
Virologie (Montrouge) ; 26(1): 55-71, 2022 01 01.
Artículo en Francés | MEDLINE | ID: mdl-35766094

RESUMEN

Résumé La thérapie anti-rétrovirale peut contrôler la réplication du virus de l'immunodéficience humaine de type 1 (VIH-1) chez les individus vivant avec le VIH. Par contre, ces traitements ne constituent pas une guérison et aucune approche pour une guérison du VIH-1 n'a encore montré de succès lors des études cliniques. Les approches de guérison sont souvent contrées in vivo par des barrières développées par le VIH-1. L'inhibition pharmacologique de la protéine accessoire Nef du VIH-1 représente une approche ambitieuse et prometteuse pour développer une nouvelle stratégie de guérison. Des petites molécules inhibitrices de Nef peuvent inverser les défauts reliés à l'infection par le VIH dans la signalisation des récepteurs des cellules T et les kinases, l'apoptose, l'autophagie et surtout, la présentation d'antigène. Ensemble, ces activités démontrent la grande capacité des inhibiteurs de Nef à être appliqués comme agents thérapeutiques dans un traitement contre le VIH-1. Dans cette revue, nous présentons les motifs pour lesquels Nef constitue une cible thérapeutique et nous soulignons les progrès effectués dans l'identification et le développement d'inhibiteurs de Nef.


Asunto(s)
Seropositividad para VIH , VIH-1 , Humanos , Ácido Láctico , Receptor PAR-1 , Replicación Viral , Productos del Gen nef del Virus de la Inmunodeficiencia Humana
6.
Virologie (Montrouge) ; 26(1): 17-33, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-35766095

RESUMEN

Antiretroviral therapy can control human immunodeficiency virus type 1 (HIV-1) replication in people living with HIV; however, these treatments are not curative and no practical approach for an HIV-1 cure has yet shown success in clinical trials. Counteracting the multiple barriers HIV-1 presents against a practical cure is a direct means to functionalize these curative approaches in vivo. Pharmacological inhibition of the HIV-1 accessory protein, Nef, represents a particularly promising and ambitious approach, with Nef inhibitors holding the potential to reverse HIV-1-related defects in T cell receptor and kinase signaling, apoptosis, autophagy and most importantly, antigen presentation. Together, the capacity for Nef inhibitors to restore these activities underscores their potential as supportive agents in a practical HIV-1 cure. In this review, we outline a rationale for pharmacologically targeting Nef and review the progress made in the identification and development of Nef inhibitors.


Asunto(s)
Infecciones por VIH , VIH-1 , Presentación de Antígeno , Infecciones por VIH/tratamiento farmacológico , VIH-1/fisiología , Humanos , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo
7.
Hum Brain Mapp ; 39(4): 1489-1499, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29271016

RESUMEN

The purpose of this study was to use non-invasive proton magnetic resonance spectroscopy (MRS) and diffusion tensor imaging (DTI) to monitor changes in prefrontal white matter metabolite levels and tissue microstructure in female rugby players with and without concussion (ages 18-23, n = 64). Evaluations including clinical tests and 3 T MRI were performed at the beginning of a season (in-season) and followed up at the end of the season (off-season). Concussed athletes were additionally evaluated 24-72 hr (n = 14), three months (n = 11), and six months (n = 8) post-concussion. Reduced glutamine at 24-72 hr and three months post-concussion, and reduced glutamine/creatine at three months post-concussion were observed. In non-concussed athletes (n = 46) both glutamine and glutamine/creatine were lower in the off-season compared to in-season. Within the MRS voxel, an increase in fractional anisotropy (FA) and decrease in radial diffusivity (RD) were also observed in the non-concussed athletes, and correlated with changes in glutamine and glutamine/creatine. Decreases in glutamine and glutamine/creatine suggest reduced oxidative metabolism. Changes in FA and RD may indicate neuroinflammation or re-myelination. The observed changes did not correlate with clinical test scores suggesting these imaging metrics may be more sensitive to brain injury and could aid in assessing recovery of brain injury from concussion.


Asunto(s)
Atletas , Conmoción Encefálica/metabolismo , Encéfalo/metabolismo , Fútbol Americano/lesiones , Fútbol Americano/fisiología , Glutamina/metabolismo , Adolescente , Encéfalo/diagnóstico por imagen , Conmoción Encefálica/diagnóstico por imagen , Conmoción Encefálica/etiología , Creatina/metabolismo , Imagen de Difusión Tensora , Femenino , Humanos , Imagen por Resonancia Magnética , Espectroscopía de Resonancia Magnética , Estudios Prospectivos , Adulto Joven
8.
Cell Immunol ; 332: 7-23, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30017085

RESUMEN

In response to micro-environmental cues such as microbial infections or T-helper 1 and 2 (TH1 and TH2) cytokines, macrophages (Mϕs) develop into M1- or M2-like phenotypes. Phenotypic polarization/activation of Mϕs are also essentially regulated by autocrine signals. Type-A γ-aminobutyric acid receptor (GABAAR)-mediated autocrine signaling is critical for phenotypic differentiation and transformation of various cell types. The present study explored whether GABAAR signaling regulates lung Mϕ (LMϕ) phenotypic activation under M1/TH1 and M2/TH2 environments. Results showed that GABAAR subunits were expressed by primary LMϕ of mice and the mouse Mϕ cell line RAW264.7. The expression levels of GABAAR subunits in mouse LMϕs and RAW264.7 cells decreased or increased concurrently with classical (M1) or alternative (M2) activation, respectively. Moreover, activation or blockade of GABAARs distinctively influenced the phenotypic characteristics of Mϕ. These results suggested that microenvironments leading to LMϕ phenotypic polarization concurrently modulates autocrine GABA signaling and its role in Mϕ activation.


Asunto(s)
Comunicación Autocrina/fisiología , Activación de Macrófagos/fisiología , Macrófagos Alveolares/metabolismo , Transducción de Señal/fisiología , Ácido gamma-Aminobutírico/metabolismo , Animales , Línea Celular , Citocinas/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Células RAW 264.7 , Receptores de GABA/metabolismo , Células TH1/metabolismo , Células Th2/metabolismo
9.
Virol J ; 15(1): 82, 2018 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-29743079

RESUMEN

BACKGROUND: Adverse drug reactions (ADRs) are a significant problem for HIV patients, with the risk of developing ADRs increasing as the infection progresses to AIDS. However, the pathophysiology underlying ADRs remains unknown. Sulphamethoxazole (SMX) via its active metabolite SMX-hydroxlyamine, when used prophylactically for pneumocystis pneumonia in HIV-positive individuals, is responsible for a high incidence of ADRs. We previously demonstrated that the HIV infection and, more specifically, that the HIV-1 Tat protein can exacerbate SMX-HA-mediated ADRs. In the current study, Jurkat T cell lines expressing Tat and its deletion mutants were used to determine the effect of Tat on the thiol proteome in the presence and absence of SMX-HA revealing drug-dependent changes in the disulfide proteome in HIV infected cells. Protein lysates from HIV infected Jurkat T cells and Jurkat T cells stably transfected with HIV Tat and Tat deletion mutants were subjected to quantitative slot blot analysis, western blot analysis and redox 2 dimensional (2D) gel electrophoresis to analyze the effects of SMX-HA on the thiol proteome. RESULTS: Redox 2D gel electrophoresis demonstrated that untreated, Tat-expressing cells contain a number of proteins with oxidized thiols. The most prominent of these protein thiols was identified as peroxiredoxin. The untreated, Tat-expressing cell lines had lower levels of peroxiredoxin compared to the parental Jurkat E6.1 T cell line. Conversely, incubation with SMX-HA led to a 2- to 3-fold increase in thiol protein oxidation as well as a significant reduction in the level of peroxiredoxin in all the cell lines, particularly in the Tat-expressing cell lines. CONCLUSION: SMX-HA is an oxidant capable of inducing the oxidation of reactive protein cysteine thiols, the majority of which formed intermolecular protein bonds. The HIV Tat-expressing cell lines showed greater levels of oxidative stress than the Jurkat E6.1 cell line when treated with SMX-HA. Therefore, the combination of HIV Tat and SMX-HA appears to alter the activity of cellular proteins required for redox homeostasis and thereby accentuate the cytopathic effects associated with HIV infection of T cells that sets the stage for the initiation of an ADR.


Asunto(s)
Oxidantes/farmacología , Peroxirredoxinas/genética , Sulfametoxazol/análogos & derivados , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Apoptosis/efectos de los fármacos , Disulfuros , Expresión Génica/efectos de los fármacos , VIH-1 , Humanos , Células Jurkat , Mutación , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Peroxirredoxinas/antagonistas & inhibidores , Peroxirredoxinas/metabolismo , Plásmidos/química , Plásmidos/metabolismo , Proteoma/genética , Proteoma/metabolismo , Sulfametoxazol/farmacología , Compuestos de Sulfhidrilo/antagonistas & inhibidores , Compuestos de Sulfhidrilo/química , Compuestos de Sulfhidrilo/metabolismo , Transgenes , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo
10.
AIDS Res Ther ; 14(1): 53, 2017 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-28893294

RESUMEN

The success of many current vaccines relies on a formulation that incorporates an immune activating adjuvant. This will hold true for the design of a successful therapeutic HIV vaccine targeted at controlling reactivated virus following cessation of combined antiretroviral therapy (cART). The HIV accessory protein Nef functions by interfering with HIV antigen presentation through the major histocompatibility complex I (MHC-I) pathway thereby suppressing CD8+ cytotoxic T cell (CTL)-mediated killing of HIV infected cells. Thus, this important impediment to HIV vaccine success must be circumvented. This review covers our current knowledge of Nef inhibitors that may serve as immune adjuvants that will specifically restore and enhance CTL-mediated killing of reactivated HIV infected cells as part of an overall vaccine strategy to affect a cure for HIV infection.


Asunto(s)
Adyuvantes Inmunológicos , Infecciones por VIH/inmunología , VIH-1/inmunología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/antagonistas & inhibidores , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/inmunología , Animales , Presentación de Antígeno , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Infecciones por VIH/tratamiento farmacológico , VIH-1/genética , VIH-1/fisiología , Humanos , Ratones , Linfocitos T Citotóxicos/inmunología , Latencia del Virus , Replicación Viral , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética
11.
Proc Natl Acad Sci U S A ; 111(1): E159-67, 2014 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-24374633

RESUMEN

Estimates of the apparent transverse relaxation rate (R2*) can be used to quantify important properties of biological tissue. Surprisingly, the mechanism of R2* dependence on tissue orientation is not well understood. The primary goal of this paper was to characterize orientation dependence of R2* in gray and white matter and relate it to independent measurements of two other susceptibility based parameters: the local Larmor frequency shift (fL) and quantitative volume magnetic susceptibility (Δχ). Through this comparative analysis we calculated scaling relations quantifying R2' (reversible contribution to the transverse relaxation rate from local field inhomogeneities) in a voxel given measurements of the local Larmor frequency shift. R2' is a measure of both perturber geometry and density and is related to tissue microstructure. Additionally, two methods (the Generalized Lorentzian model and iterative dipole inversion) for calculating Δχ were compared in gray and white matter. The value of Δχ derived from fitting the Generalized Lorentzian model was then connected to the observed R2* orientation dependence using image-registered optical density measurements from histochemical staining. Our results demonstrate that the R2* and fL of white and cortical gray matter are well described by a sinusoidal dependence on the orientation of the tissue and a linear dependence on the volume fraction of myelin in the tissue. In deep brain gray matter structures, where there is no obvious symmetry axis, R2* and fL have no orientation dependence but retain a linear dependence on tissue iron concentration and hence Δχ.


Asunto(s)
Ganglios Basales/fisiología , Encéfalo/fisiología , Vaina de Mielina/química , Fibras Nerviosas Mielínicas/fisiología , Animales , Axones/química , Medios de Contraste , Procesamiento de Imagen Asistido por Computador , Hierro/metabolismo , Imagen por Resonancia Magnética , Masculino , Modelos Neurológicos , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
12.
Int Immunol ; 24(1): 29-41, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22190576

RESUMEN

The clinical application of dendritic cells (DC) as adjuvants in immunotherapies such as the cell-based cancer vaccine continues to gain interest. The overall efficacy of this emerging immunotherapy, however, remains low. Studies suggest the stage of maturation and activation of ex vivo-prepared DC immediately prior to patient administration is critical to subsequent DC migration in vivo, which ultimately affects overall vaccine efficacy. While it is possible to generate mature and activated DC ex vivo using various stimulatory cocktails, in the case of cancer patients, the qualitative and quantitative assessment of which DC stimulatory cocktail works most effectively to enhance subsequent DC migration in vivo is difficult. Thus, a non-invasive imaging modality capable of monitoring the real-time migration of DC in long-term studies is required. In this paper, we address whether cellular magnetic resonance imaging (MRI) is sufficiently sensitive to quantitatively detect differences in the migratory abilities of two different DC preparations: untreated (resting) versus ex vivo matured in a mouse model. In order to distinguish our ex vivo-generated DC of interest from surrounding tissues in magnetic resonance (MR) images, DC were labeled in vitro with the superparamagnetic iron oxide (SPIO) nanoparticle FeREX®. Characterization of DC phenotype and function following addition of a cytokine maturation cocktail and the toll-like receptor ligand CpG, both in the presence and in the absence of SPIO, were also carried out. Conventional histological techniques were used to verify the quantitative data obtained from MR images. This study provides important information relevant to tracking the in vivo migration of ex vivo-prepared and stimulated DC.


Asunto(s)
Células de la Médula Ósea/inmunología , Movimiento Celular/inmunología , Células Dendríticas/inmunología , Imagen por Resonancia Magnética/métodos , Adyuvantes Inmunológicos/metabolismo , Adyuvantes Inmunológicos/farmacología , Animales , Células de la Médula Ósea/metabolismo , Vacunas contra el Cáncer/inmunología , Células Cultivadas , Citocinas/inmunología , Citocinas/metabolismo , Citocinas/farmacología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Compuestos Férricos/química , Citometría de Flujo , Humanos , Inmunoterapia/métodos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Nanopartículas/química , Oligodesoxirribonucleótidos/inmunología , Oligodesoxirribonucleótidos/metabolismo , Oligodesoxirribonucleótidos/farmacología , Ovalbúmina/inmunología , Ovalbúmina/metabolismo , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo , Reproducibilidad de los Resultados , Linfocitos T/inmunología , Linfocitos T/metabolismo , Receptor Toll-Like 9/inmunología , Receptor Toll-Like 9/metabolismo
13.
Virol J ; 10: 240, 2013 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-23870437

RESUMEN

BACKGROUND: Dendritic cells (DCs) are promising mediators of anti-tumor immune responses due to their potent antigen-presentation capacity. Unfortunately, cancer cells can often disarm differentiated DCs by rendering them incapable of maturation or by promoting their apoptosis. DC vaccine regimens attempt to generate functional DCs and preload them with Tumor-Associated Antigens (TAAs) to target various malignancies. Despite these efforts, the efficacy of DC vaccines in clinical trials is still rather disappointing to date. In addition to undergoing cancer-induced apoptosis, it is well established that DCs are intrinsically short-lived cell types. It is likely that a significant portion of infused DCs undergo apoptosis prior to locating and activating naïve TAA-reactive T cells. METHODS: In our current study, we constructed and investigated novel bicistronic lentivectors (LVs) encoding the cDNA for the xeno-TAA, rat HER-2/neu (rHER-2), along with five candidate mouse DC survival factors (c-FLIPS, c-FLIPL, Bcl-XL, M11L, and AKT-1) that operate in both the extrinsic and intrinsic cycles of apoptosis. The murine DC cell line, DC2.4 was transduced separately with each novel LV construct. Infected cells were enriched via flow cytometric methods based on rHER-2 expression. Transduced DC2.4 cell lines were then exposed to Fetal Calf Serum (FCS) withdrawal and to specific pharmacological apoptosis-inducing agents. DC2.4 cell death was assayed based on Annexin V and PI double-positive staining via flow cytometry. The phenotype and function of transduced DC2.4 cells and primary bone marrow-derived DCs were then assessed via expression and secretion of DC markers and cytokines, respectively. RESULTS: DC2.4 cells transduced with LVs encoding cDNAs for c-FLIPS, c-FLIPL, Bcl-XL, and M11L were protected from apoptosis when exposed to low FCS-containing culture media. When treated with an anti-CD95 antibody, only DC2.4 cells transduced with LVs encoding c-FLIPS and c-FLIPL were protected from apoptosis. In contrast, only DC2.4 cells transduced with LVs encoding Bcl-XL and M11L were protected from effects of staurosporine (STS) treatment. Also, LV-modified DCs maintained their original phenotype and function. CONCLUSIONS: We present evidence that by employing novel recombinant bicistronic LVs we can simultaneously load DCs with a relevant TAA and block apoptosis; thereby confirming the usage of such LVs in the modulation of DC lifespan and function.


Asunto(s)
Antígenos de Neoplasias/inmunología , Apoptosis , Células Dendríticas/inmunología , Células Dendríticas/fisiología , Ingeniería Genética/métodos , Vectores Genéticos , Lentivirus/genética , Animales , Antígenos de Neoplasias/genética , Supervivencia Celular , Células Dendríticas/virología , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Transducción Genética
14.
Eur Radiol Exp ; 7(1): 42, 2023 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-37580614

RESUMEN

BACKGROUND: Despite widespread study of dendritic cell (DC)-based cancer immunotherapies, the in vivo postinjection fate of DC remains largely unknown. Due in part to a lack of quantifiable imaging modalities, this is troubling as the amount of DC migration to secondary lymphoid organs correlates with therapeutic efficacy. Magnetic particle imaging (MPI) has emerged as a suitable modality to quantify in vivo migration of superparamagnetic iron oxide (SPIO)-labeled DC. Herein, we describe a popliteal lymph node (pLN)-focused MPI scan to quantify DC in vivo migration accurately and consistently. METHODS: Adenovirus (Ad)-transduced SPIO+ (Ad SPIO+) and SPIO+ C57BL/6 bone marrow-derived DC were generated and assessed for viability and phenotype, then fluorescently labeled and injected into mouse hind footpads (n = 6). Two days later, in vivo DC migration was quantified using whole animal, pLN-focused, and ex vivo pLN MPI scans. RESULTS: No significant differences in viability, phenotype and in vivo pLN migration were noted for Ad SPIO+ and SPIO+ DC. Day 2 pLN-focused MPI quantified DC migration in all instances while whole animal MPI only quantified pLN migration in 75% of cases. Ex vivo MPI and fluorescence microscopy confirmed that pLN MPI signal was due to originally injected Ad SPIO+ and SPIO+ DC. CONCLUSION: We overcame a reported limitation of MPI by using a pLN-focused MPI scan to quantify pLN-migrated Ad SPIO+ and SPIO+ DC in 100% of cases and detected as few as 1000 DC (4.4 ng Fe) in vivo. MPI is a suitable preclinical imaging modality to assess DC-based cancer immunotherapeutic efficacy. RELEVANCE STATEMENT: Tracking the in vivo fate of DC using noninvasive quantifiable magnetic particle imaging can potentially serve as a surrogate marker of therapeutic effectiveness. KEY POINTS: • Adenoviral-transduced and iron oxide-labeled dendritic cells are in vivo migration competent. • Magnetic particle imaging is a suitable modality to quantify in vivo dendritic cell migration. • Magnetic particle imaging focused field of view overcomes dynamic range limitation.


Asunto(s)
Médula Ósea , Imagen por Resonancia Magnética , Animales , Ratones , Movimiento Celular , Imagen por Resonancia Magnética/métodos , Ratones Endogámicos C57BL , Adenoviridae , Células Dendríticas , Fenómenos Magnéticos
15.
Mol Imaging Biol ; 24(6): 886-897, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35648316

RESUMEN

PURPOSE: The purpose of this study was to evaluate magnetic particle imaging (MPI) as a method for the in vivo tracking of dendritic cells (DC). DC are used in cancer immunotherapy and must migrate from the site of implantation to lymph nodes to be effective. The magnitude of the ensuing T cell response is proportional to the number of lymph node-migrated DC. With current protocols, less than 10% of DC are expected to reach target nodes. Therefore, imaging techniques for studying DC migration must be sensitive and quantitative. Here, we describe the first study using MPI to detect and track DC injected into the footpads of C57BL/6 mice migrating to the popliteal lymph nodes (pLNs). PROCEDURES: DC were labelled with Synomag-D™ and injected into each hind footpad of C57BL/6 mice (n = 6). In vivo MPI was conducted immediately and repeated 48 h later. The MPI signal was measured from images and related to the signal from a known number of cells to calculate iron content. DC numbers were estimated by dividing iron content in the image by the iron per cell measured from a separate cell sample. The presence of SPIO-labeled DC in nodes was validated by ex vivo MPI, histology, and fluorescence microscopy. RESULTS: Day 2 imaging showed a decrease in MPI signal in the footpads and an increase in signal at the pLNs, indicating DC migration. MPI signal was detected in the left pLN in four of the six mice and two of the six mice showed MPI signal in the right pLN. Ex vivo imaging detected signal in 11/12 nodes. We report a sensitivity of approximately 4000 cells (0.015 µg Fe) in vivo and 2000 cells (0.007 µg Fe) ex vivo. CONCLUSIONS: Here, we describe the first study to use MPI to detect and track DC in a migration model with immunotherapeutic applications. We also bring attention to the issue of resolving unequal signals within close proximity, a challenge for any pre-clinical study using a highly concentrated tracer bolus that shadows nearby lower signals.


Asunto(s)
Células Dendríticas , Nanopartículas de Magnetita , Ratones , Animales , Ratones Endogámicos C57BL , Movimiento Celular , Imagen por Resonancia Magnética/métodos , Hierro , Fenómenos Magnéticos , Nanopartículas de Magnetita/química
16.
medRxiv ; 2022 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-32995803

RESUMEN

SARS-CoV-2 precipitates respiratory distress by infection of airway epithelial cells and is often accompanied by acute kidney injury. We report that Kidney Injury Molecule-1/T cell immunoglobulin mucin domain 1 (KIM-1/TIM-1) is expressed in lung and kidney epithelial cells in COVID-19 patients and is a receptor for SARS-CoV-2. Human and mouse lung and kidney epithelial cells express KIM-1 and endocytose nanoparticles displaying the SARS-CoV-2 spike protein (virosomes). Uptake was inhibited by anti-KIM-1 antibodies and TW-37, a newly discovered inhibitor of KIM-1-mediated endocytosis. Enhanced KIM-1 expression by human kidney tubuloids increased uptake of virosomes. KIM-1 binds to the SARS-CoV-2 Spike protein in vitro . KIM-1 expressing cells, not expressing angiotensin-converting enzyme 2 (ACE2), are permissive to SARS-CoV-2 infection. Thus, KIM-1 is an alternative receptor to ACE2 for SARS-CoV-2. KIM-1 targeted therapeutics may prevent and/or treat COVID-19.

17.
iScience ; 25(11): 105316, 2022 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-36254158

RESUMEN

The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) spike glycoprotein (S) binds to angiotensin-converting enzyme 2 (ACE2) to mediate membrane fusion via two distinct pathways: 1) a surface, serine protease-dependent or 2) an endosomal, cysteine protease-dependent pathway. In this study, we found that SARS-CoV-2 S has a wider protease usage and can also be activated by TMPRSS13 and matrix metalloproteinases (MMPs). We found that MMP-2 and MMP-9 played roles in SARS-CoV-2 S cell-cell fusion and TMPRSS2- and cathepsin-independent viral entry in cells expressing high MMP levels. MMP-dependent viral entry required cleavage at the S1/S2 junction in viral producer cells, and differential processing of variants of concern S dictated its usage; the efficiently processed Delta S preferred metalloproteinase-dependent entry when available, and less processed Omicron S was unable to us metalloproteinases for entry. As MMP-2/9 are released during inflammation, they may play roles in S-mediated cytopathic effects, tropism, and disease outcome.

18.
Cytotherapy ; 13(10): 1234-48, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21923625

RESUMEN

BACKGROUND AIMS. The use of dendritic cells (DC) as an adjuvant in cell-based immunotherapeutic cancer vaccines is a growing field of interest. A reliable and non-invasive method to track the fate of autologous DC following their administration to patients is required in order to confirm that clinically sufficient numbers are reaching the lymph node (LN). We demonstrate that an immunocompromised mouse model can be used to conduct translational studies employing cellular magnetic resonance imaging (MRI). Such studies can provide clinically relevant information regarding the migration potential of clinical-grade DC used in cancer immunotherapies. METHODS. Human monocyte-derived dendritic cells (mo-DC) were generated from negatively selected monocytes obtained from either healthy donors or cancer patients. DC were labeled with superparamagnetic iron oxide (SPIO) nanoparticles in order to track them in vivo in a CB17scid mouse model using cellular MRI. SPIO did not have any adverse effects on DC phenotype or function, independent of donor type. Cellular MRI readily detected migration of SPIO-loaded DC in CB17scid mice. No differences in migration were observed between DC obtained from healthy donors and those obtained from donors undergoing autologous stem cell transplant for cancer therapy. CONCLUSIONS. Cellular MRI provided semi-quantitative image data that corresponded with data obtained by digital morphometry, validating cellular MRI's potential to assess DC migration in DC-based cancer immunotherapy clinical trials.


Asunto(s)
Vacunas contra el Cáncer , Movimiento Celular , Células Dendríticas/metabolismo , Inmunoterapia Adoptiva , Neoplasias/terapia , Animales , Antígenos de Diferenciación/metabolismo , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/trasplante , Modelos Animales de Enfermedad , Estudios de Factibilidad , Humanos , Imagen por Resonancia Magnética/métodos , Ratones , Ratones SCID , Monocitos/citología , Neoplasias/inmunología , Neoplasias/patología
19.
Front Immunol ; 12: 775447, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34858434

RESUMEN

CD11d/CD18 is the most recently discovered and least understood ß2 integrin. Known CD11d adhesive mechanisms contribute to both extravasation and mesenchymal migration - two key aspects for localizing peripheral leukocytes to sites of inflammation. Differential expression of CD11d induces differences in monocyte/macrophage mesenchymal migration including impacts on macrophage sub-set migration. The participation of CD11d/CD18 in leukocyte localization during atherosclerosis and following neurotrauma has sparked interest in the development of CD11d-targeted therapeutic agents. Whereas the adhesive properties of CD11d have undergone investigation, the signalling pathways induced by ligand binding remain largely undefined. Underlining each adhesive and signalling function, CD11d is under unique transcriptional control and expressed on a sub-set of predominately tissue-differentiated innate leukocytes. The following review is the first to capture the nearly three decades of CD11d research and discusses the emerging role of CD11d in leukocyte migration and retention during the progression of a staged immune response.


Asunto(s)
Antígenos CD11/genética , Antígenos CD18/genética , Quimiotaxis de Leucocito/genética , Quimiotaxis de Leucocito/inmunología , Regulación de la Expresión Génica , Cadenas alfa de Integrinas/genética , Leucocitos/fisiología , Animales , Antígenos CD11/química , Antígenos CD11/metabolismo , Antígenos CD18/química , Antígenos CD18/metabolismo , Susceptibilidad a Enfermedades , Desarrollo de Medicamentos , Humanos , Cadenas alfa de Integrinas/química , Cadenas alfa de Integrinas/metabolismo , Linfopoyesis/genética , Terapia Molecular Dirigida , Especificidad de Órganos/genética , Fagocitosis/genética , Fagocitosis/inmunología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Relación Estructura-Actividad , Factores de Transcripción
20.
Front Bioeng Biotechnol ; 9: 642465, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33816453

RESUMEN

Cell-based therapies involving the delivery of adipose-derived stromal cells (ASCs) on decellularized adipose tissue (DAT) scaffolds are a promising approach for soft tissue augmentation and reconstruction. Our lab has recently shown that culturing human ASCs on DAT scaffolds within a perfusion bioreactor prior to implantation can enhance their capacity to stimulate in vivo adipose tissue regeneration. Building from this previous work, the current study investigated the effects of bioreactor preconditioning on the ASC phenotype and secretory profile in vitro, as well as host cell recruitment following implantation in an athymic nude mouse model. Immunohistochemical analyses indicated that culturing within the bioreactor increased the percentage of ASCs co-expressing inducible nitric oxide synthase (iNOS) and arginase-1 (Arg-1), as well as tumor necrosis factor-alpha (TNF-α) and interleukin-10 (IL-10), within the peripheral regions of the DAT relative to statically cultured controls. In addition, bioreactor culture altered the expression levels of a range of immunomodulatory factors in the ASC-seeded DAT. In vivo testing revealed that culturing the ASCs on the DAT within the perfusion bioreactor prior to implantation enhanced the infiltration of host CD31+ endothelial cells and CD26+ cells into the DAT implants, but did not alter CD45+F4/80+CD68+ macrophage recruitment. However, a higher fraction of the CD45+ cell population expressed the pro-regenerative macrophage marker CD163 in the bioreactor group, which may have contributed to enhanced remodeling of the scaffolds into host-derived adipose tissue. Overall, the findings support that bioreactor preconditioning can augment the capacity of human ASCs to stimulate regeneration through paracrine-mediated mechanisms.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA