Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Exp Eye Res ; 168: 12-18, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29288646

RESUMO

Autophagy protects cells from intracellular pathogens, but can be exploited by some infectious agents to their benefit. Currently it is not known if bacteria induce autohpagy in cells of the cornea. The goal of this study was to develop an ocular surface autophagy reporter cell line and determine whether ocular bacterial pathogens influence host responses through autophagy induction. The cell line was made using lentivirus transduction of an LC3-GFP fusion protein in human corneal limbal epithelial (HCLE) cells. LC3-GFP puncta in HCLEs were induced by rapamycin and ammonium chloride treatments, and prevented by the autophagy inhibitors 3-methyladenine (3'MA) and bafilomycin. Importantly, secretomes from Escherichia coli, Serratia marcescens, Staphylococcus aureus, methicillin sensitive (MSSA) and resistant (MRSA), were found to induce autophagy, whereas other bacteria, including Acinetobacter baumannii, Achromobacter xylosoxidans, Enterococcus faecalis, Klebsiella pneumoniae, Moraxella sp., and Stenotrophomonas maltophilia, did not. Our data indicates differences between tested ocular isolates of MRSA and MSSA in the activation of autophagy. HCLEs treated with 3'MA were slightly more susceptible to cytotoxic factors produced by S. marcescens and MRSA keratitis isolates, by contrast, bafilomycin A1 treatment caused no difference. This work demonstrates the successful development and validation of an autophagy reporter corneal cell line and indicates differences between ocular bacterial isolates in the activation of autophagy.


Assuntos
Autofagia/fisiologia , Bactérias/patogenicidade , Fenômenos Fisiológicos Bacterianos , Células Epiteliais/fisiologia , Ceratite/microbiologia , Antibacterianos/farmacologia , Linhagem Celular , Células Epiteliais/efeitos dos fármacos , Humanos , Limbo da Córnea/citologia
2.
J Immunol ; 194(12): 5937-47, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25972487

RESUMO

The development of effective cancer vaccines remains an urgent, but as yet unmet, clinical need. This deficiency is in part due to an incomplete understanding of how to best invoke dendritic cells (DC) that are crucial for the induction of tumor-specific CD8(+) T cells capable of mediating durable protective immunity. In this regard, elevated expression of the transcription factor X box-binding protein 1 (XBP1) in DC appears to play a decisive role in promoting the ability of DC to cross-present Ags to CD8(+) T cells in the therapeutic setting. Delivery of DNA vaccines encoding XBP1 and tumor Ag to skin DC resulted in increased IFN-α production by plasmacytoid DC (pDC) from skin/tumor draining lymph nodes and the cross-priming of Ag-specific CD8(+) T cell responses associated with therapeutic benefit. Antitumor protection was dependent on cross-presenting Batf3(+) DC, pDC, and CD8(+) T cells. CD103(+) DC from the skin/tumor draining lymph nodes of the immunized mice appeared responsible for activation of Ag-specific naive CD8(+) T cells, but were dependent on pDC for optimal effectiveness. Similarly, human XBP1 improved the capacity of human blood- and skin-derived DC to activate human T cells. These data support an important intrinsic role for XBP1 in DC for effective cross-priming and orchestration of Batf3(+) DC-pDC interactions, thereby enabling effective vaccine induction of protective antitumor immunity.


Assuntos
Vacinas Anticâncer/imunologia , Apresentação Cruzada/imunologia , Células Dendríticas/imunologia , Neoplasias/imunologia , Vacinas de DNA/imunologia , Animais , Apresentação de Antígeno , Antígenos CD/metabolismo , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Medula Óssea/imunologia , Medula Óssea/patologia , Vacinas Anticâncer/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Feminino , Proteínas de Choque Térmico HSP72/genética , Proteínas de Choque Térmico HSP72/imunologia , Humanos , Cadeias alfa de Integrinas/metabolismo , Interferon-alfa/biossíntese , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/imunologia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/secundário , Ativação Linfocitária , Melanoma Experimental , Camundongos , Camundongos Knockout , Neoplasias/mortalidade , Neoplasias/patologia , Neoplasias/terapia , Fenótipo , Ratos , Fatores de Transcrição de Fator Regulador X , Proteínas Repressoras/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Receptor 3 Toll-Like/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Proteína 1 de Ligação a X-Box
3.
J Neurosci ; 34(24): 8268-76, 2014 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-24920630

RESUMO

Persistent neurogenesis in the olfactory epithelium provides a unique model to study neural stem cell self-renewal and fate determination. In the olfactory neuroepithelium, globose basal cells (GBCs) are considered to be the direct progenitors of olfactory neurons. However, the study of neurogenesis from GBCs has been impeded by the paucity of GBC-specific markers. Here we report that Lgr5, a recently discovered adult stem cell marker, is exclusively expressed in GBCs in neonatal and adult mice. Lgr5(+) cells display characteristics of cycling stem cells, including Ki67 expression and EdU incorporation. Lineage tracing analysis demonstrates that Lgr5(+) GBCs regenerate multiple cell types under normal turnover condition or after olfactory lesion. Furthermore, upregulation or downregulation of Wnt signaling in vivo indicates a key role of Wnt signaling not only in maintaining Lgr5(+) cell proliferation and promoting neuroregeneration, but also in delaying sensory neuron maturation. Together, our observations provided new insights into the dynamics of neurogenesis in the olfactory epithelium.


Assuntos
Células-Tronco Multipotentes/fisiologia , Neurônios Receptores Olfatórios/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Animais , Animais Recém-Nascidos , Cápsulas Bacterianas/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/fisiologia , Citometria de Fluxo , Proteína GAP-43/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Antígeno Ki-67/metabolismo , Camundongos , Camundongos Transgênicos , Proteína de Marcador Olfatório/metabolismo , Mucosa Olfatória/citologia , Receptores Acoplados a Proteínas G/genética , beta-Galactosidase/metabolismo
4.
Mol Ther ; 20(2): 432-42, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21934655

RESUMO

In vivo dendritic cells (DC) targeting is an attractive approach with potential advantages in vaccine efficacy, cost, and availability. Identification of molecular adjuvants to in vivo "modulate " DC to coordinately render improved Th1 and CD8 T cell immunity, and attenuated deleterious Treg effects, is a critical challenge. Here, we report that in vivo genetic targeting of the active transcription factor XBP1s to DC (XBP1s/DC) potentiated vaccine-induced prophylactic and therapeutic antitumor immunity in multiple tumor models. This immunization strategy is based on a genetic vaccine encoding both cytomegalovirus (CMV)-driven vaccine Aghsp70 and DC-specific CD11c-driven XBP1s. The novel targeted vaccine induced durable Th1 and CD8 T cell responses to poorly immunogenic self/tumor antigen (Ag) and attenuated tumor-associated Treg suppressive function. Bone marrow (BM)-derived DC genetically modified to simultaneously overexpress XBP1s and express Aghsp70 upregulated CD40, CD70, CD86, interleukin (IL)-15, IL-15Rα, and CCR7 expression, and increased IL-6, IL-12, and tumor necrosis factor (TNF)-α production in vitro. XBP1s/DC elevated functional DEC205(+)CD8α(+)DC in the draining lymph nodes (DLN). The data suggest a novel role for XBP1s in modulating DC to potentiate tumor vaccine efficacy via overcoming two major obstacles to tumor vaccines (i.e., T cell hyporesponsiveness against poorly immunologic self/tumor Ag and tumor-associated Treg-mediated suppression) and improving DEC205(+)CD8α(+)DC.


Assuntos
Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Proteínas de Ligação a DNA/genética , Células Dendríticas/imunologia , Marcação de Genes , Neoplasias Experimentais/imunologia , Fatores de Transcrição/genética , Animais , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/uso terapêutico , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Proteínas de Ligação a DNA/metabolismo , Células Dendríticas/metabolismo , Feminino , Expressão Gênica , Ordem dos Genes , Interferon gama/biossíntese , Linfonodos/imunologia , Linfonodos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/mortalidade , Fatores de Transcrição de Fator Regulador X , Análise de Sobrevida , Linfócitos T Reguladores , Canais de Cátion TRPC/imunologia , Células Th1/imunologia , Fatores de Transcrição/metabolismo , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Proteína 1 de Ligação a X-Box
5.
Elife ; 102021 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-33506763

RESUMO

Glaucoma is a leading cause of irreversible blindness. In this study, we investigated if transplanted stem cells are able to rescue a glaucoma mouse model with transgenic myocilin Y437H mutation and explored the possible mechanisms. Human trabecular meshwork stem cells (TMSCs) were intracamerally transplanted which reduced mouse intraocular pressure, increased outflow facility, protected the retinal ganglion cells and preserved their function. TMSC transplantation also significantly increased the TM cellularity, promoted myocilin secretion from TM cells into the aqueous humor to reduce endoplasmic reticulum stress, repaired the TM tissue with extracellular matrix modulation and ultrastructural restoration. Co-culturing TMSCs with myocilin mutant TM cells in vitro promoted TMSCs differentiating into phagocytic functional TM cells. RNA sequencing revealed that TMSCs had upregulated genes related to TM regeneration and neuroprotection. Our results uncovered therapeutic potential of TMSCs for curing glaucoma and elucidated possible mechanisms by which TMSCs achieve the treatment effect.


Assuntos
Glaucoma de Ângulo Aberto/terapia , Transplante de Células-Tronco , Malha Trabecular/transplante , Animais , Proteínas do Citoesqueleto/metabolismo , Modelos Animais de Doenças , Proteínas do Olho/metabolismo , Feminino , Glicoproteínas/metabolismo , Humanos , Masculino , Camundongos
6.
Autophagy ; 17(10): 3140-3159, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33404293

RESUMO

Retinal ganglion cell axons are heavily myelinated (98%) and myelin damage in the optic nerve (ON) severely affects vision. Understanding the molecular mechanism of oligodendrocyte progenitor cell (OPC) differentiation into mature oligodendrocytes will be essential for developing new therapeutic approaches for ON demyelinating diseases. To this end, we developed a new method for isolation and culture of ON-derived oligodendrocyte lineage cells and used it to study OPC differentiation. A critical aspect of cellular differentiation is macroautophagy/autophagy, a catabolic process that allows for cell remodeling by degradation of excess or damaged cellular molecules and organelles. Knockdown of ATG9A and BECN1 (pro-autophagic proteins involved in the early stages of autophagosome formation) led to a significant reduction in proliferation and survival of OPCs. We also found that autophagy flux (a measure of autophagic degradation activity) is significantly increased during progression of oligodendrocyte differentiation. Additionally, we demonstrate a significant change in mitochondrial dynamics during oligodendrocyte differentiation, which is associated with a significant increase in programmed mitophagy (selective autophagic clearance of mitochondria). This process is mediated by the mitophagy receptor BNIP3L (BCL2/adenovirus E1B interacting protein 3-like). BNIP3L-mediated mitophagy plays a crucial role in the regulation of mitochondrial network formation, mitochondrial function and the viability of newly differentiated oligodendrocytes. Our studies provide novel evidence that proper mitochondrial dynamics is required for establishment of functional mitochondria in mature oligodendrocytes. These findings are significant because targeting BNIP3L-mediated programmed mitophagy may provide a novel therapeutic approach for stimulating myelin repair in ON demyelinating diseases.Abbreviations: A2B5: a surface antigen of oligodendrocytes precursor cells, A2B5 clone 105; ACTB: actin, beta; APC: an antibody to label mature oligodendrocytes, anti-adenomatous polyposis coli clone CC1; ATG5: autophagy related 5; ATG7: autophagy related 7; ATG9A: autophagy related 9A; AU: arbitrary units; BafA1: bafilomycin A1; BCL2: B cell leukemia/lymphoma 2; BECN1: beclin 1, autophagy related; BNIP3: BCL2/adenovirus E1B interacting protein 3; BNIP3L/NIX: BCL2/adenovirus E1B interacting protein 3-like; CASP3: caspase 3; CNP: 2',3'-cyclic nucleotide 3'-phosphodiesterase; Ctl: control; COX8: cytochrome c oxidase subunit; CSPG4/NG2: chondroitin sulfate proteoglycan 4; DAPI: 4'6-diamino-2-phenylindole; DNM1L: dynamin 1-like; EGFP: enhanced green fluorescent protein; FACS: fluorescence-activated cell sorting; FIS1: fission, mitochondrial 1; FUNDC1: FUN14 domain containing 1; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFAP: glial fibrillary growth factor; GFP: green fluorescent protein; HsESC: human embryonic stem cell; IEM: immunoelectron microscopy; LAMP1: lysosomal-associated membrane protein 1; LC3B: microtubule-associated protein 1 light chain 3; MBP: myelin basic protein; MFN2: mitofusin 2; Mito-Keima: mitochondria-targeted monomeric keima-red; Mito-GFP: mitochondria-green fluorescent protein; Mito-RFP: mitochondria-red fluorescent protein; MitoSOX: red mitochondrial superoxide probe; MKI67: antigen identified by monoclonal antibody Ki 67; MMP: mitochondrial membrane potential; O4: oligodendrocyte marker O4; OLIG2: oligodendrocyte transcription factor 2; ON: optic nerve; OPA1: OPA1, mitochondrial dynamin like GTPase; OPC: oligodendrocyte progenitor cell; PDL: poly-D-lysine; PINK1: PTEN induced putative kinase 1; PRKN/Parkin: parkin RBR E3 ubiquitin protein ligase; RFP: red fluorescent protein; RGC: retinal ganglion cell; ROS: reactive oxygen species; RT-PCR: real time polymerase chain reaction; SEM: standard error of the mean; SOD2: superoxide dismutase 2, mitochondrial; SQSTM1/p62: sequestosome 1; TEM: transmission electron microscopy; TMRM: tetramethylrhodamine methyl ester; TOMM20: translocase of outer mitochondrial membrane 20; TUBB: tubulin, beta; TUBB3: tubulin, beta 3 class III.


Assuntos
Autofagia , Mitofagia , Autofagia/fisiologia , Diferenciação Celular/fisiologia , Mitocôndrias/metabolismo , Mitofagia/fisiologia , Oligodendroglia/metabolismo , Nervo Óptico/metabolismo
7.
Mol Ther ; 17(7): 1274-81, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19417739

RESUMO

Tumor-primed CD4(+) T cells from splenocytes of tumor-rejection mice in combination with in vivo glucocorticoid-induced tumor necrosis factor receptor (GITR) ligation (the combination therapy) elicited effective host CD8(+) T cell-dependent therapeutic immunity against a murine breast tumor. GITR ligation in vitro enhanced tumor-primed CD4(+) T-cell activity and partially abrogated regulatory T cells (Treg) suppressor function. Dendritic cells (DCs) from tumor-draining lymph nodes (TDLNs) of tumor-bearing mice treated by the combination therapy stimulated Ag-specific T cells and produced interleukin (IL)-12 ex vivo. Whereas tumor-primed CD4(+) T cells or in vivo GITR ligation alone induced a tumor-specific interferon (IFN)-gamma-producing cellular response, the combination therapy enhanced and sustained it. Furthermore, the combination therapy in vivo attenuated Treg's ability to suppress IL-12 production by DCs and IFN-gamma production by effectors ex vivo. Importantly, tumor-primed CD4(+) CD25(-) T cells from splenocytes of untreated tumor-bearing mice in combination with in vivo GITR ligation also elicited an effective therapeutic effect in this model. These data suggest that the combination therapy may improve DC function, accentuate tumor-specific T-cell responses, and attenuate Treg suppressor function, thereby eliciting effective therapeutic immunity.


Assuntos
Neoplasias da Mama/imunologia , Neoplasias da Mama/terapia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/transplante , Proteína Relacionada a TNFR Induzida por Glucocorticoide/metabolismo , Imunoterapia Adotiva/métodos , Animais , Anticorpos Monoclonais/uso terapêutico , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Feminino , Imunidade Celular/efeitos dos fármacos , Imunidade Celular/imunologia , Interferon gama/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Receptores do Fator de Necrose Tumoral/imunologia , Linfócitos T Reguladores/imunologia
8.
Invest Ophthalmol Vis Sci ; 60(2): 731-740, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30786277

RESUMO

Purpose: Krüppel-like factor 4 (KLF4) promotes corneal epithelial (CE) cell fate while suppressing mesenchymal properties. TGF-ß plays a crucial role in cell differentiation and development, and if dysregulated, it induces epithelial-mesenchymal transition (EMT). As KLF4 and TGF-ß regulate each other in a context-dependent manner, we evaluated the role of the crosstalk between KLF4 and TGF-ß-signaling in CE homeostasis. Methods: We used spatiotemporally regulated ablation of Klf4 within the adult mouse CE in ternary transgenic Klf4Δ/ΔCE (Klf4LoxP/LoxP/ Krt12rtTA/rtTA/ Tet-O-Cre) mice and short hairpin RNA (shRNA)-mediated knockdown or lentiviral vector-mediated overexpression of KLF4 in human corneal limbal epithelial (HCLE) cells to evaluate the crosstalk between KLF4 and TGF-ß-signaling components. Expression of TGF-ß signaling components and cyclin-dependent kinase (CDK) inhibitors was quantified by quantitative PCR, immunoblots, and/or immunofluorescent staining. Results: CE-specific ablation of Klf4 resulted in (1) upregulation of TGF-ß1, -ß2, -ßR1, and -ßR2; (2) downregulation of inhibitory Smad7; (3) hyperphosphorylation of Smad2/3; (4) elevated nuclear localization of phospho-Smad2/3 and Smad4; and (5) downregulation of CDK inhibitors p16 and p27. Consistently, shRNA-mediated knockdown of KLF4 in HCLE cells resulted in upregulation of TGF-ß1 and -ß2, hyperphosphorylation and nuclear localization of SMAD2/3, downregulation of SMAD7, and elevated SMAD4 nuclear localization. Furthermore, overexpression of KLF4 in HCLE cells resulted in downregulation of TGF-ß1, -ßR1, and -ßR2 and upregulation of SMAD7, p16, and p27. Conclusions: Collectively, these results demonstrate that KLF4 regulates CE cell cycle progression by suppressing canonical TGF-ß signaling and overcomes the undesirable concomitant decrease in TGF-ß-dependent CDK inhibitors p16 and p27 expression by directly upregulating them.


Assuntos
Ciclo Celular/fisiologia , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor de Quinase Dependente de Ciclina p27/genética , Epitélio Corneano/metabolismo , Fatores de Transcrição Kruppel-Like/fisiologia , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Células Epiteliais , Técnica Indireta de Fluorescência para Anticorpo , Regulação da Expressão Gênica/fisiologia , Inativação Gênica/fisiologia , Vetores Genéticos , Humanos , Immunoblotting , Fator 4 Semelhante a Kruppel , Limbo da Córnea/citologia , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase em Tempo Real , Transfecção , Regulação para Cima
9.
Aging Cell ; 14(4): 635-43, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25990970

RESUMO

Current knowledge indicates that the adult mammalian retina lacks regenerative capacity. Here, we show that the adult stem cell marker, leucine-rich repeat-containing G-protein-coupled receptor 5 (Lgr5), is expressed in the retina of adult mice. Lgr5(+) cells are generated at late stages of retinal development and exhibit properties of differentiated amacrine interneurons (amacrine cells). Nevertheless, Lgr5(+) amacrine cells contribute to regeneration of new retinal cells in the adult stage. The generation of new retinal cells, including retinal neurons and Müller glia from Lgr5(+) amacrine cells, begins in early adulthood and continues as the animal ages. Together, these findings suggest that the mammalian retina is not devoid of regeneration as previously thought. It is rather dynamic, and Lgr5(+) amacrine cells function as an endogenous regenerative source. The identification of such cells in the mammalian retina may provide new insights into neuronal regeneration and point to therapeutic opportunities for age-related retinal degenerative diseases.


Assuntos
Células-Tronco Adultas/metabolismo , Envelhecimento/metabolismo , Células Amácrinas/metabolismo , Neurogênese/genética , Receptores Acoplados a Proteínas G/genética , Regeneração/genética , Células-Tronco Adultas/citologia , Envelhecimento/genética , Células Amácrinas/citologia , Animais , Diferenciação Celular , Proliferação de Células , Células Ependimogliais/citologia , Células Ependimogliais/metabolismo , Expressão Gênica , Técnicas de Introdução de Genes , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Transgênicos , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
10.
Oncoimmunology ; 3(10): e959321, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25941586

RESUMO

IL-15 supports improved antitumor immunity. How to best incorporate IL-15 into vaccine formulations for superior cancer immunotherapy remains a challenge. DC-derived IL-15 (DCIL-15) notably has the capacity to activate DC, to substitute for CD4+ Th and to potentiate vaccine efficacy making IL-15-based therapies attractive treatment options. We observed in transplantable melanoma, glioma and metastatic breast carcinoma models that DCIL-15-based DNA vaccines in which DC specifically express IL-15 and simultaneously produce tumor Aghsp70 were able to mediate potent therapeutic efficacy that required both host Batf3+ DC and CD8+ T cells. In an inducible BrafV600E/Pten-driven murine melanoma model, DCIL-15 (not rIL-15)-based DNA vaccines elicited durable therapeutic CD8+ T cell-dependent antitumor immunity. DCIL-15 was found to be superior to rIL-15 in "licensing" both mouse and human DC, and for activating CD8+ T cells. Such activation occurred even in the presence of Treg, without a need for CD4+ Th, but was IL-15/IL-15Rα-dependent. A single low-dose of DCIL-15 (not rIL-15)-based DC vaccines induced therapeutic antitumor immunity. CD14+ DC emigrating from human skin explants genetically-immunized by IL-15 and Aghsp70 were more effective than similar DC emigrating from the explants genetically-immunized by Aghsp70 in the presence of rIL-15 in expressing membrane-bound IL-15/IL-15Rα and activating CD8+ T cells. These results support future clinical use of DCIL-15 as a therapeutic agent in battling cancer.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA