Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Immunol ; 203(7): 1999-2010, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31471524

RESUMEN

To arise and progress, cancers need to evade immune elimination. Consequently, progressing tumors are often MHC class I (MHC-I) low and express immune inhibitory molecules, such as PD-L1, which allows them to avoid the main antitumor host defense, CD8+ T cells. The molecular mechanisms that led to these alterations were incompletely understood. In this study, we identify loss of the transcription factor IRF2 as a frequent underlying mechanism that leads to a tumor immune evasion phenotype in both humans and mice. We identified IRF2 in a CRISPR-based forward genetic screen for genes that controlled MHC-I Ag presentation in HeLa cells. We then found that many primary human cancers, including lung, colon, breast, prostate, and others, frequently downregulated IRF2. Although IRF2 is generally known as a transcriptional repressor, we found that it was a transcriptional activator of many key components of the MHC-I pathway, including immunoproteasomes, TAP, and ERAP1, whose transcriptional control was previously poorly understood. Upon loss of IRF2, cytosol-to-endoplasmic reticulum peptide transport and N-terminal peptide trimming become rate limiting for Ag presentation. In addition, we found that IRF2 is a repressor of PD-L1. Thus, by downregulating a single nonessential gene, tumors become harder to see (reduced Ag presentation), more inhibitory (increased checkpoint inhibitor), and less susceptible to being killed by CD8+ T cells. Importantly, we found that the loss of Ag presentation caused by IRF2 downregulation could be reversed by IFN-stimulated induction of the transcription factor IRF1. The implication of these findings for tumor progression and immunotherapy are discussed.


Asunto(s)
Presentación de Antígeno , Antígeno B7-H1/inmunología , Linfocitos T CD8-positivos/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Factor 2 Regulador del Interferón/deficiencia , Proteínas de Neoplasias/inmunología , Neoplasias , Escape del Tumor , Antígeno B7-H1/genética , Linfocitos T CD8-positivos/patología , Regulación hacia Abajo/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Células HEK293 , Células HeLa , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Factor 2 Regulador del Interferón/inmunología , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología
2.
Proc Natl Acad Sci U S A ; 116(14): 7043-7052, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30894498

RESUMEN

A human cytomegalovirus (HCMV) pentameric glycoprotein complex (PC), gH-gL-UL128-UL130-UL131A, is necessary for viral infection of clinically relevant cell types, including epithelial cells, which are important for interhost transmission and disease. We performed genome-wide CRISPR/Cas9 screens of different cell types in parallel to identify host genes specifically required for HCMV infection of epithelial cells. This effort identified a multipass membrane protein, OR14I1, as a receptor for HCMV infection. This olfactory receptor family member is required for HCMV attachment, entry, and infection of epithelial cells and is dependent on the presence of viral PC. OR14I1 is required for AKT activation and mediates endocytosis entry of HCMV. We further found that HCMV infection of epithelial cells is blocked by a synthetic OR14I1 peptide and inhibitors of adenylate cyclase and protein kinase A (PKA) signaling. Identification of OR14I1 as a PC-dependent HCMV host receptor associated with epithelial tropism and the role of the adenylate cyclase/PKA/AKT-mediated signaling pathway in HCMV infection reveal previously unappreciated targets for the development of vaccines and antiviral therapies.


Asunto(s)
Citomegalovirus/fisiología , Células Epiteliales/metabolismo , Complejos Multiproteicos/metabolismo , Transducción de Señal , Proteínas Virales/metabolismo , Tropismo Viral/fisiología , Células A549 , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células Epiteliales/patología , Células Epiteliales/virología , Células HEK293 , Células HeLa , Humanos , Complejos Multiproteicos/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Virales/genética
3.
mBio ; 10(1)2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30723129

RESUMEN

Encephalomyocarditis virus (EMCV) is a picornavirus that produces lytic infections in murine and human cells. Employing a genome-wide CRISPR-Cas9 knockout screen to find host factors required for EMCV infection, we identified a role for ADAM9 in EMCV infection. CRISPR-mediated deletion of ADAM9 in multiple human cell lines rendered the cells highly resistant to EMCV infection and cell death. Primary fibroblasts from ADAM9 KO mice were also strongly resistant to EMCV infection and cell death. In contrast, ADAM9 KO and WT cells were equally susceptible to infection with other viruses, including the picornavirus Coxsackie virus B. ADAM9 KO cells failed to produce viral progeny when incubated with EMCV. However, bypassing EMCV entry into cells through delivery of viral RNA directly to the cytosol yielded infectious EMCV virions from ADAM9 KO cells, suggesting that ADAM9 is not required for EMCV replication post-entry. These findings establish that ADAM9 is required for the early stage of EMCV infection, likely for virus entry or viral genome delivery to the cytosol.IMPORTANCE Viral myocarditis is a leading cause of death in the United States, contributing to numerous unexplained deaths in people ≤35 years old. Enteroviruses contribute to many cases of human myocarditis. Encephalomyocarditis virus (EMCV) infection causes viral myocarditis in rodent models, but its receptor requirements have not been fully identified. CRISPR-Cas9 screens can identify host dependency factors essential for EMCV infection and enhance our understanding of key events that follow viral infection, potentially leading to new strategies for preventing viral myocarditis. Using a CRISPR-Cas9 screen, we identified adisintegrin and metalloproteinase 9 domain (ADAM9) as a major factor required for the early stages of EMCV infection in both human and murine infection.


Asunto(s)
Proteínas ADAM/metabolismo , Infecciones por Cardiovirus/genética , Resistencia a la Enfermedad , Virus de la Encefalomiocarditis/crecimiento & desarrollo , Proteínas de la Membrana/metabolismo , Animales , Línea Celular , Técnicas de Inactivación de Genes , Pruebas Genéticas , Humanos , Ratones , Ratones Noqueados , Modelos Biológicos
4.
PLoS Pathog ; 15(1): e1007532, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30640957

RESUMEN

Late endosome-resident interferon-induced transmembrane protein 3 (IFITM3) inhibits fusion of diverse viruses, including Influenza A virus (IAV), by a poorly understood mechanism. Despite the broad antiviral activity of IFITM3, viruses like Lassa virus (LASV), are fully resistant to its inhibitory effects. It is currently unclear whether resistance arises from a highly efficient fusion machinery that is capable of overcoming IFITM3 restriction or the ability to enter from cellular sites devoid of this factor. Here, we constructed and validated a functional IFITM3 tagged with EGFP or other fluorescent proteins. This breakthrough allowed live cell imaging of virus co-trafficking and fusion with endosomal compartments in cells expressing fluorescent IFITM3. Three-color single virus and endosome tracking revealed that sensitive (IAV), but not resistant (LASV), viruses become trapped within IFITM3-positive endosomes where they underwent hemifusion but failed to release their content into the cytoplasm. IAV fusion with IFITM3-containing compartments could be rescued by amphotericin B treatment, which has been previously shown to antagonize the antiviral activity of this protein. By comparison, virtually all LASV particles trafficked and fused with endosomes lacking detectable levels of fluorescent IFITM3, implying that this virus escapes restriction by utilizing endocytic pathways that are distinct from the IAV entry pathways. The importance of virus uptake and transport pathways is further reinforced by the observation that LASV glycoprotein-mediated cell-cell fusion is inhibited by IFITM3 and other members of the IFITM family expressed in target cells. Together, our results strongly support a model according to which IFITM3 accumulation at the sites of virus fusion is a prerequisite for its antiviral activity and that this protein traps viral fusion at a hemifusion stage by preventing the formation of fusion pores. We conclude that the ability to utilize alternative endocytic pathways for entry confers IFITM3-resistance to otherwise sensitive viruses.


Asunto(s)
Endosomas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/fisiología , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/fisiología , Células A549/metabolismo , Animales , Antivirales/metabolismo , Células COS/metabolismo , Chlorocebus aethiops , Endosomas/virología , Células HEK293/metabolismo , Interacciones Huésped-Patógeno , Humanos , Virus de la Influenza A/patogenicidad , Interferones/metabolismo , Virus Lassa/patogenicidad , Imagen Óptica/métodos , Transporte de Proteínas , Internalización del Virus
5.
Nature ; 538(7625): 350-355, 2016 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-27680706

RESUMEN

Clostridium difficile toxin B (TcdB) is a critical virulence factor that causes diseases associated with C. difficile infection. Here we carried out CRISPR-Cas9-mediated genome-wide screens and identified the members of the Wnt receptor frizzled family (FZDs) as TcdB receptors. TcdB binds to the conserved Wnt-binding site known as the cysteine-rich domain (CRD), with the highest affinity towards FZD1, 2 and 7. TcdB competes with Wnt for binding to FZDs, and its binding blocks Wnt signalling. FZD1/2/7 triple-knockout cells are highly resistant to TcdB, and recombinant FZD2-CRD prevented TcdB binding to the colonic epithelium. Colonic organoids cultured from FZD7-knockout mice, combined with knockdown of FZD1 and 2, showed increased resistance to TcdB. The colonic epithelium in FZD7-knockout mice was less susceptible to TcdB-induced tissue damage in vivo. These findings establish FZDs as physiologically relevant receptors for TcdB in the colonic epithelium.


Asunto(s)
Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Colon/metabolismo , Epitelio/metabolismo , Receptores Frizzled/metabolismo , Animales , Antígenos/metabolismo , Proteínas Bacterianas/química , Toxinas Bacterianas/química , Sitios de Unión , Células CHO , Sistemas CRISPR-Cas , Línea Celular , Clostridioides difficile/patogenicidad , Cricetulus , Femenino , Receptores Frizzled/química , Receptores Frizzled/deficiencia , Receptores Frizzled/genética , Técnicas de Inactivación de Genes , Humanos , Masculino , Ratones , Ratones Noqueados , Oligopéptidos/química , Oligopéptidos/metabolismo , Organoides/metabolismo , Dominios Proteicos , Proteoglicanos/metabolismo , Factores de Virulencia/metabolismo , Proteínas Wnt/metabolismo
6.
Cell Rep ; 16(1): 232-246, 2016 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-27342126

RESUMEN

The flaviviruses dengue virus (DENV) and Zika virus (ZIKV) are severe health threats with rapidly expanding ranges. To identify the host cell dependencies of DENV and ZIKV, we completed orthologous functional genomic screens using RNAi and CRISPR/Cas9 approaches. The screens recovered the ZIKV entry factor AXL as well as multiple host factors involved in endocytosis (RAB5C and RABGEF), heparin sulfation (NDST1 and EXT1), and transmembrane protein processing and maturation, including the endoplasmic reticulum membrane complex (EMC). We find that both flaviviruses require the EMC for their early stages of infection. Together, these studies generate a high-confidence, systems-wide view of human-flavivirus interactions and provide insights into the role of the EMC in flavivirus replication.


Asunto(s)
Virus del Dengue/genética , Genómica/métodos , Virus Zika/genética , Sistemas CRISPR-Cas , Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Pruebas Genéticas , Células HeLa , Interacciones Huésped-Patógeno/genética , Humanos , Membranas Intracelulares/metabolismo , Unión Proteica , Mapas de Interacción de Proteínas , Interferencia de ARN , Replicación Viral
7.
Cell Rep ; 15(11): 2323-30, 2016 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-27268505

RESUMEN

Zika virus has emerged as a severe health threat with a rapidly expanding range. The IFITM family of restriction factors inhibits the replication of a broad range of viruses, including the closely related flaviruses West Nile virus and dengue virus. Here, we show that IFITM1 and IFITM3 inhibit Zika virus infection early in the viral life cycle. Moreover, IFITM3 can prevent Zika-virus-induced cell death. These results suggest that strategies to boost the actions and/or levels of the IFITMs might be useful for inhibiting a broad range of emerging viruses.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Unión al ARN/metabolismo , Replicación Viral/fisiología , Virus Zika/fisiología , Células A549 , Animales , Efecto Citopatogénico Viral , Eliminación de Gen , Sitios Genéticos , Células HeLa , Humanos , Ratones , Transporte de Proteínas , Infección por el Virus Zika
8.
Adv Virus Res ; 94: 1-51, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26997589

RESUMEN

Over the last several years a wealth of transformative human-virus interaction discoveries have been produced using loss-of-function functional genomics. These insights have greatly expanded our understanding of how human pathogenic viruses exploit our cells to replicate. Two technologies have been at the forefront of this genetic revolution, RNA interference (RNAi) and random retroviral insertional mutagenesis using haploid cell lines (haploid cell screening), with the former technology largely predominating. Now the cutting edge gene editing of the CRISPR/Cas9 system has also been harnessed for large-scale functional genomics and is poised to possibly displace these earlier methods. Here we compare and contrast these three screening approaches for elucidating host-virus interactions, outline their key strengths and weaknesses including a comparison of an arrayed multiple orthologous RNAi reagent screen to a pooled CRISPR/Cas9 human rhinovirus 14-human cell interaction screen, and recount some notable insights made possible by each. We conclude with a brief perspective on what might lie ahead for the fast evolving field of human-virus functional genomics.


Asunto(s)
Sistemas CRISPR-Cas , Genómica/métodos , Haploidia , Interacciones Huésped-Patógeno/genética , Interferencia de ARN , Virus/patogenicidad , Proteínas Bacterianas , Proteína 9 Asociada a CRISPR , Endonucleasas , Técnicas de Inactivación de Genes , Pruebas Genéticas/métodos , Humanos/virología , Mutagénesis Insercional , ARN Interferente Pequeño/genética
9.
Cell Rep ; 12(5): 850-63, 2015 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-26212330

RESUMEN

Human rhinovirus (HRV) causes upper respiratory infections and asthma exacerbations. We screened multiple orthologous RNAi reagents and identified host proteins that modulate HRV replication. Here, we show that RNASEK, a transmembrane protein, was needed for the replication of HRV, influenza A virus, and dengue virus. RNASEK localizes to the cell surface and endosomal pathway and closely associates with the vacuolar ATPase (V-ATPase) proton pump. RNASEK is required for endocytosis, and its depletion produces enlarged clathrin-coated pits (CCPs) at the cell surface. These enlarged CCPs contain endocytic cargo and are bound by the scissioning GTPase, DNM2. Loss of RNASEK alters the localization of multiple V-ATPase subunits and lowers the levels of the ATP6AP1 subunit. Together, our results show that RNASEK closely associates with the V-ATPase and is required for its function; its loss prevents the early events of endocytosis and the replication of multiple pathogenic viruses.


Asunto(s)
Virus del Dengue/fisiología , Endorribonucleasas/metabolismo , Virus de la Influenza A/fisiología , Rhinovirus/fisiología , ATPasas de Translocación de Protón Vacuolares/metabolismo , Replicación Viral/fisiología , Endocitosis/fisiología , Endorribonucleasas/genética , Células HeLa , Humanos , ATPasas de Translocación de Protón Vacuolares/genética
10.
Nat Struct Mol Biol ; 20(8): 973-81, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23851458

RESUMEN

Physiological Ca(2+) signaling in T lymphocytes and other cells depends on the STIM-ORAI pathway of store-operated Ca(2+) entry. STIM1 and STIM2 are Ca(2+) sensors in the endoplasmic reticulum (ER) membrane, with ER-luminal domains that monitor cellular Ca(2+) stores and cytoplasmic domains that gate ORAI channels in the plasma membrane. The STIM ER-luminal domain dimerizes or oligomerizes upon dissociation of Ca(2+), but the mechanism transmitting activation to the STIM cytoplasmic domain was previously undefined. Using Tb(3+)-acceptor energy transfer, we show that dimerization of STIM1 ER-luminal domains causes an extensive conformational change in mouse STIM1 cytoplasmic domains. The conformational change, triggered by apposition of the predicted coiled-coil 1 (CC1) regions, releases the ORAI-activating domains from their interaction with the CC1 regions and allows physical extension of the STIM1 cytoplasmic domain across the gap between ER and plasma membrane and communication with ORAI channels.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio/fisiología , Retículo Endoplásmico/metabolismo , Glicoproteínas de Membrana/química , Modelos Moleculares , Conformación Proteica , Animales , Dimerización , Electroforesis en Gel de Poliacrilamida , Glicoproteínas de Membrana/metabolismo , Ratones , Proteína ORAI1 , Ingeniería de Proteínas , Colorantes de Rosanilina , Espectrometría de Fluorescencia , Molécula de Interacción Estromal 1
11.
EMBO J ; 30(19): 3895-912, 2011 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-21847095

RESUMEN

Cell polarization enables restriction of signalling into microdomains. Polarization of lymphocytes following formation of a mature immunological synapse (IS) is essential for calcium-dependent T-cell activation. Here, we analyse calcium microdomains at the IS with total internal reflection fluorescence microscopy. We find that the subplasmalemmal calcium signal following IS formation is sufficiently low to prevent calcium-dependent inactivation of ORAI channels. This is achieved by localizing mitochondria close to ORAI channels. Furthermore, we find that plasma membrane calcium ATPases (PMCAs) are re-distributed into areas beneath mitochondria, which prevented PMCA up-modulation and decreased calcium export locally. This nano-scale distribution-only induced following IS formation-maximizes the efficiency of calcium influx through ORAI channels while it decreases calcium clearance by PMCA, resulting in a more sustained NFAT activity and subsequent activation of T cells.


Asunto(s)
Señalización del Calcio , Calcio/química , Linfocitos T/citología , Canales de Calcio/metabolismo , Membrana Celular/enzimología , Citoesqueleto/metabolismo , Electrofisiología , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Sinapsis Inmunológicas , Células Jurkat , Activación de Linfocitos , Microscopía Fluorescente/métodos , Mitocondrias/metabolismo , Proteína ORAI1 , Estructura Terciaria de Proteína
12.
Nat Struct Mol Biol ; 17(1): 112-6, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20037597

RESUMEN

Store-operated Ca(2+) entry through the plasma membrane Ca(2+) release-activated Ca(2+) (CRAC) channel in mammalian T cells and mast cells depends on the sensor protein stromal interaction molecule 1 (STIM1) and the channel subunit ORAI1. To study STIM1-ORAI1 signaling in vitro, we have expressed human ORAI1 in a sec6-4 strain of the yeast Saccharomyces cerevisiae and isolated sealed membrane vesicles carrying ORAI1 from the Golgi compartment to the plasma membrane. We show by in vitro Ca(2+) flux assays that bacterially expressed recombinant STIM1 opens wild-type ORAI1 channels but not channels assembled from the ORAI1 pore mutant E106Q or the ORAI1 severe combined immunodeficiency (SCID) mutant R91W. These experiments show that the STIM1-ORAI1 interaction is sufficient to gate recombinant human ORAI1 channels in the absence of other proteins of the human ORAI1 channel complex, and they set the stage for further biochemical and biophysical dissection of ORAI1 channel gating.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Activación del Canal Iónico/fisiología , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Recombinantes/metabolismo , Canales de Calcio/genética , Cromatografía en Gel , Humanos , Activación del Canal Iónico/genética , Mutación/genética , Proteína ORAI1 , Saccharomyces cerevisiae , Vesículas Secretoras/metabolismo , Molécula de Interacción Estromal 1
13.
Sci Signal ; 2(99): ra79, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19952372

RESUMEN

Rap1-guanosine triphosphate (GTP)-interacting adaptor molecule (RIAM) plays a critical role in actin reorganization and inside-out activation of integrins in lymphocytes and platelets. We investigated the role of RIAM in T cell receptor (TCR)-mediated signaling. Although phosphorylation of the kinase ZAP-70 and formation of a signalosome recruited to the adaptor protein LAT were unaffected, elimination of endogenous RIAM by short hairpin RNA impaired generation of inositol 1,4,5-trisphosphate, mobilization of intracellular calcium ions (Ca(2+)), and translocation of the transcription factor NFAT to the nucleus. The activation of Ras guanine nucleotide-releasing protein 1 was also impaired, which led to the diminished expression of the gene encoding interleukin-2. These events were associated with the impaired translocation of phosphorylated phospholipase C-gamma1 (PLC-gamma1) to the actin cytoskeleton, which was required to bring PLC-gamma1 close to its substrate phosphatidylinositol 4,5-bisphosphate, and were reversed by reconstitution of cells with RIAM. Thus, by regulating the localization of PLC-gamma1, RIAM plays a central role in TCR signaling and the transcription of target genes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Citoesqueleto/enzimología , Proteínas de la Membrana/fisiología , Fosfolipasa C gamma/metabolismo , Células Cultivadas , Humanos , Células Jurkat , Fosfatidilinositol 4,5-Difosfato , Fosfatos de Fosfatidilinositol/metabolismo , Transporte de Proteínas , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Transcripción Genética
14.
J Immunol ; 178(4): 2262-71, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17277131

RESUMEN

It is unclear whether peptide-MHC class II (pMHC) complexes on distinct types of APCs differ in their capacity to trigger TCRs. In this study, we show that individual cognate pMHC complexes displayed by dendritic cells (DCs), as compared with nonprofessional APCs, are far better in productively triggering Ag-specific TCRs independently of conventional costimulation. As we further show, this is accomplished by the unique ability of DCs to robustly activate the Src family kinases (SFKs) Lck and Fyn in T cells even in the absence of cognate peptide. Instead, this form of SFK activation depends on interactions of DC-displayed MHC with TCRs of appropriate restriction, suggesting a central role of self-pMHC recognition. DC-mediated SFK activation leads to "TCR licensing," a process that dramatically increases sensitivity and magnitude of the TCR response to cognate pMHC. Thus, TCR licensing, besides costimulation, is a main mechanism of DCs to present Ag effectively.


Asunto(s)
Células Dendríticas/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/inmunología , Proteínas Proto-Oncogénicas c-fyn/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Presentación de Antígeno/inmunología , Complejo Antígeno-Anticuerpo/inmunología , Complejo Antígeno-Anticuerpo/metabolismo , Autoinmunidad , Línea Celular Transformada , Células Dendríticas/enzimología , Activación Enzimática/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Péptidos/inmunología , Péptidos/metabolismo , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/metabolismo
15.
Blood ; 109(11): 4777-85, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-17289814

RESUMEN

The discovery of marker proteins of human blood (BECs) and lymphatic endothelial cells (LECs) has allowed researchers to isolate these cells. So far, efforts to unravel their transcriptional and functional programs made use of cultured cells only. Hence, it is unknown to which extent previously identified LEC- and BEC-specific programs are representative of the in vivo situation. Here, we define the human BEC- and LEC-specific in vivo transcriptomes by comparative genomewide expression profiling of freshly isolated cutaneous EC subsets and of non-EC skin cells (fibroblasts, mast cells, dendritic cells, epithelial cells). Interestingly, the expression of most of the newly identified EC subset-discriminating genes depends strictly on the in vivo tissue environment as revealed by comparative analyses of freshly isolated and cultured EC subsets. The identified environment-dependent, EC subset-restricted gene expression regulates lineage fidelity, fluid exchange, and MHC class II-dependent antigen presentation. As an example for a BEC-restricted in vivo function, we show that non-activated BECs in situ, but not in vitro, assemble and display MHC class II protein complexes loaded with self-peptides. Thus, our data demonstrate the key importance of using precisely defined native ECs for the global identification of in vivo relevant cell functions.


Asunto(s)
Células Endoteliales/citología , Perfilación de la Expresión Génica , Melanoma/inmunología , Diferenciación Celular , Separación Celular , Células Cultivadas/metabolismo , Genoma , Antígenos de Histocompatibilidad Clase II/química , Humanos , Microcirculación , Hibridación de Ácido Nucleico , Análisis de Secuencia por Matrices de Oligonucleótidos , Péptidos/química , Transcripción Genética
16.
J Immunol ; 173(11): 6592-602, 2004 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-15557149

RESUMEN

Plasmacytoid dendritic cells (pDCs) recognize microbes, viruses in particular, and provide unique means of innate defense against them. The mechanism of pDC tissue recruitment remained enigmatic because the ligands of CXCR3, the cardinal chemokine receptor on pDCs, have failed to induce in vitro chemotaxis of pDCs in the absence of additional chemokines. In this study, we demonstrate that CXCR3 is sufficient to induce pDC migration, however, by a migratory mechanism that amalgamates the features of haptotaxis and chemorepulsion. To mediate "haptorepulsion" of pDCs, CXCR3 requires the encounter of its cognate ligands immobilized, optimally by heparan sulfate, in a form of a negative gradient. This is the first report of the absolute requirement of chemokine immobilization and presentation for its in vitro promigratory activity. The paradigmatic example of pDC haptorepulsion described here may represent a new pathophysiologically relevant migratory mechanism potentially used by other cells in response to other chemokines.


Asunto(s)
Movimiento Celular/inmunología , Células Dendríticas/citología , Células Dendríticas/inmunología , Receptores de Quimiocina/fisiología , Presentación de Antígeno , Adhesión Celular/inmunología , Membrana Celular/inmunología , Membrana Celular/metabolismo , Inhibición de Migración Celular , Quimiocina CXCL10 , Quimiocina CXCL12 , Quimiocinas/farmacología , Quimiocinas CXC/inmunología , Quimiocinas CXC/metabolismo , Quimiocinas CXC/fisiología , Células Dendríticas/metabolismo , Células Dendríticas/patología , Endotelio Vascular/citología , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Heparitina Sulfato/inmunología , Heparitina Sulfato/metabolismo , Herpes Zóster/inmunología , Herpes Zóster/patología , Herpesvirus Humano 3/inmunología , Humanos , Interferón-alfa/farmacología , Ligandos , Toxina del Pertussis/farmacología , Fosforilación , Unión Proteica/inmunología , Receptores CCR7 , Receptores CXCR3 , Receptores de Quimiocina/biosíntesis , Simplexvirus/inmunología , Solubilidad , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología , Tirosina/metabolismo
17.
J Immunol ; 171(5): 2714-24, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12928425

RESUMEN

Therapeutic vaccination against cutaneous T cell lymphoma (CTCL) requires the characterization of cancer cell-specific CTL epitopes. Despite reported evidence for tumor-reactive cytotoxicity in CTCL patients, the nature of the recognized determinants remains elusive. The clonotypic TCR of CTCL cells is a promising candidate tumor-specific Ag. In this study, we report that the clonotypic and framework regions of the TCRs expressed in the malignant T cell clones of six CTCL patients contain multiple peptides with anchor residues fitting the patients' MHC class I molecules. We demonstrate that TCR peptide-specific T cells from the blood of healthy donors and patients can be induced to become cytotoxic effectors after repeated stimulation with 6 of 11 selected peptides with experimentally proven affinity for HLA-A*0201. Importantly, 4 of these 6 CTL lines reproducibly recognize and lyse autologous primary CTCL cells in MHC class I/CD8-dependent fashion. These tumoricidal CTL lines are directed against epitopes from V, hypervariable, and C regions of TCRalpha. We therefore conclude that recombined as well as V framework regions of the tumor cell TCRs contain predictable epitopes for CTL-mediated attack of CTCL cells. Our data further suggest that such peptides represent valuable tools for future anti-CTCL vaccination approaches.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Citotoxicidad Inmunológica/inmunología , Epítopos de Linfocito T/uso terapéutico , Linfoma de Células T/inmunología , Receptores de Antígenos de Linfocitos T/uso terapéutico , Neoplasias Cutáneas/inmunología , Linfocitos T Citotóxicos/inmunología , Secuencia de Aminoácidos , Presentación de Antígeno/inmunología , Vacunas contra el Cáncer/metabolismo , Vacunas contra el Cáncer/uso terapéutico , Línea Celular Tumoral , Células Clonales , Endopeptidasas/metabolismo , Epítopos de Linfocito T/sangre , Epítopos de Linfocito T/inmunología , Epítopos de Linfocito T/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Hidrólisis , Activación de Linfocitos , Linfoma de Células T/sangre , Linfoma de Células T/enzimología , Linfoma de Células T/prevención & control , Datos de Secuencia Molecular , Fragmentos de Péptidos/sangre , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo , Unión Proteica/inmunología , Receptores de Antígenos de Linfocitos T/sangre , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Neoplasias Cutáneas/sangre , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/prevención & control , Linfocitos T Citotóxicos/enzimología , Linfocitos T Citotóxicos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...