Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cells ; 12(2)2023 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-36672182

RESUMEN

The efficacy of CD19-specific CAR T cells in the treatment of leukemia/lymphoma relies, at least in part, on the unique properties of the particular CAR and the presence of healthy B cells that enhance the target cell lysis and cytokine secretion through repetitive stimulation. Here, we report to apply the same CAR to target solid tumors, such as ErbB2+ carcinoma. CD19 CAR T cells are redirected towards the ErbB2+ cells by a fusion protein that is composed of the herceptin-derived anti-ErbB2 scFv 4D5 linked to the CD19 exodomain. The CD19-4D5scFv engager enabled CD19 CAR T cells to recognize the ErbB2+ cancer cells and to suppress the ErbB2+ tumor growth. The primary killing capacity by the ErbB2-redirected CD19 CAR T cells was as efficient as by the ErbB2 CAR T cells, however, adding CD19+ B cells furthermore reinforced the activation of the CD19 CAR T cells, thereby improving the anti-tumor activities. The ErbB2-redirected CD19 CAR T cells, moreover, showed a 100-fold superior selectivity in targeting cancer cells versus healthy fibroblasts, which was not the case for the ErbB2 CAR T cells. The data demonstrate that the CD19 CAR T cells can be high-jacked by a CD19-scFv engager protein to attack specifically solid cancer, thereby expanding their application beyond the B cell malignancies.


Asunto(s)
Neoplasias , Humanos , Neoplasias/terapia , Trastuzumab , Linfocitos B , Proteínas Adaptadoras Transductoras de Señales , Linfocitos T , Receptor ErbB-2
2.
Oncoimmunology ; 11(1): 2111904, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35990518

RESUMEN

B cell lymphoma therapy has been transformed by CD19-targeting cellular therapeutics that induce high clinical response rates and impressive remissions in relapsed and refractory patients. However, approximately half of all patients who respond to CD19-directed cell therapy relapse, the majority within 6 months. One characteristic of relapse is loss or reduction of CD19 expression on malignant B cells. We designed a unique therapeutic to prevent and reverse relapses due to lost or reduced CD19 expression. This novel biologic, a CAR T Engager, binds CD20 and displays the CD19 extracellular domain. This approach increases the apparent CD19 antigen density on CD19-positive/CD20-positive lymphoma cells, and prevents antigen-loss induced relapse, as CD19 bound to CD20 remains present on the cell surface. We demonstrate that this novel therapeutic prevents and reverses lymphoma relapse in vitro and prevents CD19-negative lymphoma growth and relapse in vivo.


Asunto(s)
Linfoma , Receptores Quiméricos de Antígenos , Antígenos CD19 , Antígenos CD20 , Humanos , Linfoma/terapia , Recurrencia Local de Neoplasia , Receptores de Antígenos de Linfocitos T , Linfocitos T
3.
Mol Cancer Ther ; 20(10): 2071-2081, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34253594

RESUMEN

Refractory acute myeloid leukemia (AML) remains an incurable malignancy despite the clinical use of novel targeted therapies, new antibody-based therapies, and cellular therapeutics. Here, we describe the preclinical development of a novel cell therapy that targets the antigen CLEC12A with a biparatopic bridging protein. Bridging proteins are designed as "CAR-T cell engagers," with a CAR-targeted protein fused to antigen binding domains derived from antibodies. Here, we created a CD19-anti-CLEC12A bridging protein that binds to CAR19 T cells and to the antigen CLEC12A. Biparatopic targeting increases the potency of bridging protein-mediated cytotoxicity by CAR19 T cells. Using CAR19 T cells that secrete the bridging protein we demonstrate potent activity against aggressive leukemic cell lines in vivo This CAR-engager platform is facile and modular, as illustrated by activity of a dual-antigen bridging protein targeting CLEC12A and CD33, designed to counter tumor heterogeneity and antigen escape, and created without the need for extensive CAR T-cell genetic engineering. CAR19 T cells provide an optimal cell therapy platform with well-understood inherent persistence and fitness characteristics.


Asunto(s)
Antígenos CD19/inmunología , Inmunoterapia Adoptiva/métodos , Inmunoterapia/métodos , Lectinas Tipo C/inmunología , Leucemia Mieloide Aguda/tratamiento farmacológico , Receptores Mitogénicos/inmunología , Lectina 3 Similar a Ig de Unión al Ácido Siálico/inmunología , Linfocitos T/inmunología , Animales , Deriva y Cambio Antigénico , Apoptosis , Proliferación Celular , Citotoxicidad Inmunológica/inmunología , Humanos , Técnicas In Vitro , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
4.
PLoS One ; 16(3): e0247701, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33735268

RESUMEN

Successful CAR T cell therapy for the treatment of solid tumors requires exemplary CAR T cell expansion, persistence and fitness, and the ability to target tumor antigens safely. Here we address this constellation of critical attributes for successful cellular therapy by using integrated technologies that simplify development and derisk clinical translation. We have developed a CAR-CD19 T cell that secretes a CD19-anti-Her2 bridging protein. This cell therapy strategy exploits the ability of CD19-targeting CAR T cells to interact with CD19 on normal B cells to drive expansion, persistence and fitness. The secreted bridging protein potently binds to Her2-positive tumor cells, mediating CAR-CD19 T cell cytotoxicity in vitro and in vivo. Because of its short half-life, the secreted bridging protein will selectively accumulate at the site of highest antigen expression, ie. at the tumor. Bridging proteins that bind to multiple different tumor antigens have been created. Therefore, antigen-bridging CAR-CD19 T cells incorporate critical attributes for successful solid tumor cell therapy. This platform can be exploited to attack tumor antigens on any cancer.


Asunto(s)
Antígenos CD19/genética , Inmunoterapia Adoptiva/métodos , Linfoma de Células B/terapia , Receptor ErbB-2/genética , Receptores Quiméricos de Antígenos/genética , Linfocitos T/inmunología , Animales , Antígenos CD19/inmunología , Linfocitos B/inmunología , Linfocitos B/patología , Línea Celular Tumoral , Proliferación Celular , Técnicas de Cocultivo , Citotoxicidad Inmunológica , Receptores ErbB/genética , Receptores ErbB/inmunología , Expresión Génica , Vectores Genéticos/inmunología , Vectores Genéticos/metabolismo , Humanos , Lentivirus/genética , Lentivirus/inmunología , Activación de Linfocitos , Linfoma de Células B/genética , Linfoma de Células B/inmunología , Linfoma de Células B/patología , Ratones , Ratones SCID , Unión Proteica , Receptor ErbB-2/inmunología , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/citología , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Glycobiology ; 31(7): 859-872, 2021 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-33403396

RESUMEN

N-glycosylated proteins produced in human embryonic kidney 293 (HEK 293) cells often carry terminal N-acetylgalactosamine (GalNAc) and only low levels of sialylation. On therapeutic proteins, such N-glycans often trigger rapid clearance from the patient's bloodstream via efficient binding to asialoglycoprotein receptor (ASGP-R) and mannose receptor (MR). This currently limits the use of HEK 293 cells for therapeutic protein production. To eliminate terminal GalNAc, we knocked-out GalNAc transferases B4GALNT3 and B4GALNT4 by CRISPR/Cas9 in FreeStyle 293-F cells. The resulting cell line produced a coagulation factor VII-albumin fusion protein without GalNAc but with increased sialylation. This glyco-engineered protein bound less efficiently to both the ASGP-R and MR in vitro and it showed improved recovery, terminal half-life and area under the curve in pharmacokinetic rat experiments. By overexpressing sialyltransferases ST6GAL1 and ST3GAL6 in B4GALNT3 and B4GALNT4 knock-out cells, we further increased factor VII-albumin sialylation; for ST6GAL1 even to the level of human plasma-derived factor VII. Simultaneous knock-out of B4GALNT3 and B4GALNT4 and overexpression of ST6GAL1 further lowered factor VII-albumin binding to ASGP-R and MR. This novel glyco-engineered cell line is well-suited for the production of factor VII-albumin and presumably other therapeutic proteins with fully human N-glycosylation and superior pharmacokinetic properties.


Asunto(s)
Glicoproteínas , Sialiltransferasas , Animales , Técnicas de Inactivación de Genes , Glicoproteínas/biosíntesis , Glicoproteínas/genética , Glicoproteínas/farmacocinética , Glicosilación , Células HEK293 , Humanos , Ratas , Sialiltransferasas/genética , Sialiltransferasas/metabolismo
6.
Biochemistry ; 58(48): 4869-4881, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31702909

RESUMEN

The B-cell surface protein CD19 is present throughout the cell life cycle and is uniformly expressed in leukemias, making it a target for chimeric antigen receptor engineered immune cell therapy. Identifying the sequence dependence of the binding of CD19 to antibodies empowers fundamental study and more tailored development of CD19-targeted therapeutics. To identify the antibody-binding epitopes on CD19, we screened a comprehensive single-site saturation mutation library of the human CD19 extracellular domain to identify mutations detrimental to binding FMC63-the dominant CD19 antibody used in chimeric antigen receptor development-as well as 4G7-2E3 and 3B10, which have been used in various types of CD19 research and development. All three antibodies had partially overlapping, yet distinct, epitopes near the published epitope of antibody B43. The FMC63 conformational epitope spans spatially adjacent, but genetically distant, loops in exons 3 and 4. The 3B10 epitope is a linear peptide sequence that binds CD19 with 440 pM affinity. Along with their primary goal of epitope mapping, the mutational tolerance data also empowered additional CD19 variant design and analysis. A designed CD19 variant with all N-linked glycosylation sites removed successfully bound antibody in the yeast display context, which provides a lead for aglycosylated applications. Screening for thermally stable variants identified mutations to guide further CD19 stabilization for fusion protein applications and revealed evolutionary affinity-stability trade-offs. These fundamental insights into CD19 sequence-function relationships enhance our understanding of antibody-mediated CD19-targeted therapeutics.


Asunto(s)
Antígenos CD19/química , Antígenos CD19/inmunología , Anticuerpos Monoclonales/análisis , Anticuerpos Monoclonales/inmunología , Antígenos CD19/genética , Mapeo Epitopo , Exones , Humanos , Mutación , Dominios Proteicos
7.
Mol Pharm ; 16(8): 3544-3558, 2019 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-31242389

RESUMEN

CD19-targeted chimeric antigen receptor (CAR) T-cells (CAR19s) show remarkable efficacy in the treatment of relapsed/refractory acute lymphocytic leukemia and Non-Hodgkin's lymphoma. However, the use of CAR T-cell therapy against CD19-negative hematological cancers and solid tumors has been challenging. We propose CD19-fusion proteins (CD19-FPs) to leverage the benefits of CAR19s while retargeting this validated cellular therapy to alternative tumor antigens. We demonstrate the ability of a fusion of CD19 extracellular domain (ECD) and a human epidermal growth factor receptor 2 (HER2) single-chain antibody fragment to retarget CAR19s to kill HER2+ CD19- tumor cells. To enhance the modularity of this technology, we engineered a more robust CD19 ECD via deep mutational scanning with yeast display and flow cytometric selections for improved protease resistance and anti-CD19 antibody binding. These enhanced CD19 ECDs significantly increase, and in some cases recover, fusion protein expression while maintaining target antigen affinity. Importantly, CD19-FPs retarget CAR19s to kill tumor cells expressing multiple distinct antigens, including HER2, CD20, EGFR, BCMA, and Clec12A as N- or C-terminal fusions and linked to both antibody fragments and fibronectin ligands. This study provides fundamental insights into CD19 sequence-function relationships and defines a flexible and modular platform to retarget CAR19s to any tumor antigen.


Asunto(s)
Antígenos CD19/metabolismo , Inmunoterapia Adoptiva/métodos , Neoplasias/terapia , Proteínas Recombinantes de Fusión/metabolismo , Anticuerpos de Cadena Única/metabolismo , Linfocitos T/inmunología , Antígenos CD19/genética , Antígenos CD19/inmunología , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Células HEK293 , Humanos , Mutagénesis , Neoplasias/inmunología , Neoplasias/patología , Dominios Proteicos/genética , Ingeniería de Proteínas , Receptor ErbB-2/antagonistas & inhibidores , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología , Linfocitos T/metabolismo , Linfocitos T/trasplante
8.
J Virol ; 91(18)2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28659487

RESUMEN

The mumps virus (MuV) small hydrophobic protein (SH) is a type I membrane protein expressed in infected cells. SH has been reported to interfere with innate immunity by inhibiting tumor necrosis factor alpha (TNF-α)-mediated apoptosis and NF-κB activation. To elucidate the underlying mechanism, we generated recombinant MuVs (rMuVs) expressing the SH protein with an N-terminal FLAG epitope or lacking SH expression due to the insertion of three stop codons into the SH gene. Using these viruses, we were able to show that SH reduces the phosphorylation of IKKß, IκBα, and p65 as well as the translocation of p65 into the nucleus of infected A549 cells. Reporter gene assays revealed that SH interferes not only with TNF-α-mediated NF-κB activation but also with IL-1ß- and poly(I·C)-mediated NF-κB activation, and that this inhibition occurs upstream of the NF-κB pathway components TRAF2, TRAF6, and TAK1. Since SH coimmunoprecipitated with tumor necrosis factor receptor 1 (TNFR1), RIP1, and IRAK1, we hypothesize that SH exerts its inhibitory function by interacting with TNFR1, interleukin-1 receptor type 1 (IL-1R1), and TLR3 complexes in the plasma membrane of infected cells.IMPORTANCE The MuV SH has been shown to impede TNF-α-mediated NF-κB activation and is therefore thought to contribute to viral immune evasion. However, the mechanisms by which SH mediates NF-κB inhibition remained largely unknown. In this study, we show that SH interacts with TNFR1, IL-1R1, and TLR3 complexes in infected cells. We thereby not only shed light on the mechanisms of SH-mediated NF-κB inhibition but also reveal that SH interferes with NF-κB activation induced by interleukin-1ß (IL-1ß) and double-stranded RNA.


Asunto(s)
Interacciones Huésped-Patógeno , Virus de la Parotiditis/inmunología , FN-kappa B/antagonistas & inhibidores , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptor Toll-Like 3/metabolismo , Proteínas Virales/metabolismo , Animales , Línea Celular , Humanos , Receptores Tipo I de Interleucina-1
9.
Virol J ; 13: 21, 2016 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-26837541

RESUMEN

BACKGROUND: Rubella virus (RV) infection is usually a mild illness in children and adults. However, maternal infection during the first trimester of pregnancy can lead to congenital rubella syndrome (CRS) in the infant. Fetuses with CRS show damage to the endothelium of the heart and blood vessels; thus, it has been speculated that the clinical manifestations associated with CRS may be a result of endothelial cells persistently infected with RV. Here, we compared the effects of RV infection on gene expression in primary endothelial cells of fetal (HUVEC) and of adult (HSaVEC) origin by transcriptional profiling. RESULTS: More than 75 % of the genes differentially regulated following RV infection were identical in both cell types. Gene Ontology (GO) analysis of these commonly regulated genes showed an enrichment of terms involved in cytokine production and cytokine regulation. Increased accumulation of inflammatory cytokines following RV infection was verified by protein microarray. Interestingly, the chemokine CCL14, which is implicated in supporting embryo implantation at the fetal-maternal interface, was down-regulated following RV infection only in HUVEC. Most noticeably, when analyzing the uniquely regulated transcripts for each cell type, GO term-based cluster analysis of the down-regulated genes of HUVEC revealed an enrichment of the GO terms "sensory organ development", "ear development" and "eye development". CONCLUSION: Since impairment in vision and hearing are the most prominent clinical manifestations observed in CRS patients, the here detected down-regulated genes involved in the development of sensory organs sheds light on the molecular mechanisms that may contribute to the teratogenic effect of RV.


Asunto(s)
Células Endoteliales/metabolismo , Células Endoteliales/virología , Perfilación de la Expresión Génica , Virus de la Rubéola/fisiología , Transcriptoma , Línea Celular , Quimiocinas/genética , Biología Computacional , Regulación de la Expresión Génica , Ontología de Genes , Humanos , Rubéola (Sarampión Alemán)/genética , Rubéola (Sarampión Alemán)/virología , Síndrome de Rubéola Congénita/genética , Síndrome de Rubéola Congénita/virología , Replicación Viral
10.
Nat Rev Drug Discov ; 14(8): 561-84, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26228759

RESUMEN

Targeting immune checkpoints such as programmed cell death protein 1 (PD1), programmed cell death 1 ligand 1 (PDL1) and cytotoxic T lymphocyte antigen 4 (CTLA4) has achieved noteworthy benefit in multiple cancers by blocking immunoinhibitory signals and enabling patients to produce an effective antitumour response. Inhibitors of CTLA4, PD1 or PDL1 administered as single agents have resulted in durable tumour regression in some patients, and combinations of PD1 and CTLA4 inhibitors may enhance antitumour benefit. Numerous additional immunomodulatory pathways as well as inhibitory factors expressed or secreted by myeloid and stromal cells in the tumour microenvironment are potential targets for synergizing with immune checkpoint blockade. Given the breadth of potential targets in the immune system, critical questions to address include which combinations should move forward in development and which patients will benefit from these treatments. This Review discusses the leading drug targets that are expressed on tumour cells and in the tumour microenvironment that allow enhancement of the antitumour immune response.


Asunto(s)
Antineoplásicos/farmacología , Inmunoterapia/métodos , Neoplasias/terapia , Animales , Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Antígeno CTLA-4/antagonistas & inhibidores , Antígeno CTLA-4/inmunología , Humanos , Terapia Molecular Dirigida , Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Microambiente Tumoral/inmunología
11.
Proc Natl Acad Sci U S A ; 111(35): E3699-707, 2014 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-25136083

RESUMEN

Accumulating evidence indicates that T-cell immunoglobulin (Ig) and mucin domain (TIM) proteins play critical roles in viral infections. Herein, we report that the TIM-family proteins strongly inhibit HIV-1 release, resulting in diminished viral production and replication. Expression of TIM-1 causes HIV-1 Gag and mature viral particles to accumulate on the plasma membrane. Mutation of the phosphatidylserine (PS) binding sites of TIM-1 abolishes its ability to block HIV-1 release. TIM-1, but to a much lesser extent PS-binding deficient mutants, induces PS flipping onto the cell surface; TIM-1 is also found to be incorporated into HIV-1 virions. Importantly, TIM-1 inhibits HIV-1 replication in CD4-positive Jurkat cells, despite its capability of up-regulating CD4 and promoting HIV-1 entry. In addition to TIM-1, TIM-3 and TIM-4 also block the release of HIV-1, as well as that of murine leukemia virus (MLV) and Ebola virus (EBOV); knockdown of TIM-3 in differentiated monocyte-derived macrophages (MDMs) enhances HIV-1 production. The inhibitory effects of TIM-family proteins on virus release are extended to other PS receptors, such as Axl and RAGE. Overall, our study uncovers a novel ability of TIM-family proteins to block the release of HIV-1 and other viruses by interaction with virion- and cell-associated PS. Our work provides new insights into a virus-cell interaction that is mediated by TIMs and PS receptors.


Asunto(s)
Linfocitos T CD4-Positivos/fisiología , Infecciones por VIH/metabolismo , VIH-1/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Receptores Virales/metabolismo , Linfocitos T CD4-Positivos/virología , Membrana Celular/metabolismo , Membrana Celular/virología , Técnicas de Silenciamiento del Gen , Células HEK293 , Infecciones por VIH/virología , VIH-1/crecimiento & desarrollo , Células HeLa , Receptor Celular 1 del Virus de la Hepatitis A , Receptor 2 Celular del Virus de la Hepatitis A , Humanos , Células Jurkat , Glicoproteínas de Membrana/genética , Proteínas de la Membrana/genética , Fosfatidilserinas/metabolismo , ARN Interferente Pequeño/genética , Receptores Virales/genética , Virión/crecimiento & desarrollo , Virión/metabolismo , Replicación Viral/fisiología
12.
Immunity ; 40(4): 542-53, 2014 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-24703780

RESUMEN

Selectins play a central role in leukocyte trafficking by mediating tethering and rolling on vascular surfaces. Here we have reported that T cell immunoglobulin and mucin domain 1 (TIM-1) is a P-selectin ligand. We have shown that human and murine TIM-1 binds to P-selectin, and that TIM-1 mediates tethering and rolling of T helper 1 (Th1) and Th17, but not Th2 and regulatory T cells on P-selectin. Th1 and Th17 cells lacking the TIM-1 mucin domain showed reduced rolling in thrombin-activated mesenteric venules and inflamed brain microcirculation. Inhibition of TIM-1 had no effect on naive T cell homing, but it reduced T cell recruitment in a skin hypersensitivity model and blocked experimental autoimmune encephalomyelitis. Uniquely, the TIM-1 immunoglobulin variable domain was also required for P-selectin binding. Our data demonstrate that TIM-1 is a major P-selectin ligand with a specialized role in T cell trafficking during inflammatory responses and the induction of autoimmune disease.


Asunto(s)
Encéfalo/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Hipersensibilidad/inmunología , Proteínas de la Membrana/metabolismo , Selectina-P/metabolismo , Subgrupos de Linfocitos T/inmunología , Células TH1/inmunología , Traslado Adoptivo , Animales , Movimiento Celular/genética , Proliferación Celular , Células Cultivadas , Receptor Celular 1 del Virus de la Hepatitis A , Ligandos , Glicoproteínas de Membrana/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Glicoproteína Mielina-Oligodendrócito/inmunología , Fragmentos de Péptidos/inmunología
13.
J Virol ; 88(12): 6702-13, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24696470

RESUMEN

UNLABELLED: T-cell immunoglobulin and mucin domain 1 (TIM-1) and other TIM family members were recently identified as phosphatidylserine (PtdSer)-mediated virus entry-enhancing receptors (PVEERs). These proteins enhance entry of Ebola virus (EBOV) and other viruses by binding PtdSer on the viral envelope, concentrating virus on the cell surface, and promoting subsequent internalization. The PtdSer-binding activity of the immunoglobulin-like variable (IgV) domain is essential for both virus binding and internalization by TIM-1. However, TIM-3, whose IgV domain also binds PtdSer, does not effectively enhance virus entry, indicating that other domains of TIM proteins are functionally important. Here, we investigate the domains supporting enhancement of enveloped virus entry, thereby defining the features necessary for a functional PVEER. Using a variety of chimeras and deletion mutants, we found that in addition to a functional PtdSer-binding domain PVEERs require a stalk domain of sufficient length, containing sequences that promote an extended structure. Neither the cytoplasmic nor the transmembrane domain of TIM-1 is essential for enhancing virus entry, provided the protein is still plasma membrane bound. Based on these defined characteristics, we generated a mimic lacking TIM sequences and composed of annexin V, the mucin-like domain of α-dystroglycan, and a glycophosphatidylinositol anchor that functioned as a PVEER to enhance transduction of virions displaying Ebola, Chikungunya, Ross River, or Sindbis virus glycoproteins. This identification of the key features necessary for PtdSer-mediated enhancement of virus entry provides a basis for more effective recognition of unknown PVEERs. IMPORTANCE: T-cell immunoglobulin and mucin domain 1 (TIM-1) and other TIM family members are recently identified phosphatidylserine (PtdSer)-mediated virus entry-enhancing receptors (PVEERs). These proteins enhance virus entry by binding the phospholipid, PtdSer, present on the viral membrane. While it is known that the PtdSer binding is essential for the PVEER function of TIM-1, TIM-3 shares this binding activity but does not enhance virus entry. No comprehensive studies have been done to characterize the other domains of TIM-1. In this study, using a variety of chimeric proteins and deletion mutants, we define the features necessary for a functional PVEER. With these features in mind, we generated a TIM-1 mimic using functionally similar domains from other proteins. This mimic, like TIM-1, effectively enhanced transduction. These studies provide insight into the key features necessary for PVEERs and will allow for more effective identification of unknown PVEERs.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Receptores Virales/metabolismo , Virosis/metabolismo , Internalización del Virus , Fenómenos Fisiológicos de los Virus , Línea Celular , Ebolavirus/genética , Ebolavirus/fisiología , Receptor Celular 1 del Virus de la Hepatitis A , Humanos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Fosfatidilserinas/metabolismo , Estructura Terciaria de Proteína , Receptores Virales/química , Receptores Virales/genética , Acoplamiento Viral , Virosis/genética , Virosis/virología , Virus/genética
14.
J Virol ; 87(15): 8327-41, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23698310

RESUMEN

The cell surface receptor T cell immunoglobulin mucin domain 1 (TIM-1) dramatically enhances filovirus infection of epithelial cells. Here, we showed that key phosphatidylserine (PtdSer) binding residues of the TIM-1 IgV domain are critical for Ebola virus (EBOV) entry through direct interaction with PtdSer on the viral envelope. PtdSer liposomes but not phosphatidylcholine liposomes competed with TIM-1 for EBOV pseudovirion binding and transduction. Further, annexin V (AnxV) substituted for the TIM-1 IgV domain, supporting a PtdSer-dependent mechanism. Our findings suggest that TIM-1-dependent uptake of EBOV occurs by apoptotic mimicry. Additionally, TIM-1 enhanced infection of a wide range of enveloped viruses, including alphaviruses and a baculovirus. As further evidence of the critical role of enveloped-virion-associated PtdSer in TIM-1-mediated uptake, TIM-1 enhanced internalization of pseudovirions and virus-like proteins (VLPs) lacking a glycoprotein, providing evidence that TIM-1 and PtdSer-binding receptors can mediate virus uptake independent of a glycoprotein. These results provide evidence for a broad role of TIM-1 as a PtdSer-binding receptor that mediates enveloped-virus uptake. Utilization of PtdSer-binding receptors may explain the wide tropism of many of these viruses and provide new avenues for controlling their virulence.


Asunto(s)
Ebolavirus/fisiología , Glicoproteínas de Membrana/metabolismo , Fosfatidilserinas/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores Virales/metabolismo , Internalización del Virus , Alphavirus/química , Alphavirus/fisiología , Animales , Anexina A5/metabolismo , Baculoviridae/química , Baculoviridae/fisiología , Línea Celular , Ebolavirus/química , Receptor Celular 1 del Virus de la Hepatitis A , Interacciones Huésped-Patógeno , Humanos , Transducción Genética
15.
Immunol Lett ; 141(1): 28-35, 2011 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-21911007

RESUMEN

T cell, immunoglobulin domain and mucin domain-1 (TIM-1) is the nominant member of a small family of related proteins that regulate immune cell activities. TIM-1 was initially characterized in a mouse congenic analysis of Th2 T cell responses and related pathology. Data accumulated to date suggest that TIM-1 regulates effector T cell function, and may play distinct roles in the activities of B cells, invariant NKT cells and epithelial cells. In addition, a variety of ligands for TIM-1 have been proposed. In this review I discuss recent data that have accumulated on the function of TIM-1, propose a model to explain how TIM-1 regulates effector T cell activity through recognition of distinct ligands, and review others functions of this increasingly fascinating protein. Of considerable interest are the novel findings that TIM-1 mediates virus entry and virulence.


Asunto(s)
Asma/inmunología , Glicoproteínas de Membrana , Proteínas de la Membrana , Receptores Inmunológicos/metabolismo , Receptores Virales , Linfocitos T , Animales , Células Dendríticas/inmunología , Filoviridae/metabolismo , Receptor Celular 1 del Virus de la Hepatitis A , Virus de la Hepatitis A/metabolismo , Humanos , Hipótesis de la Higiene , Infecciones/inmunología , Activación de Linfocitos/inmunología , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica/inmunología , Isoformas de Proteínas , Receptores Virales/inmunología , Receptores Virales/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo
16.
Proc Natl Acad Sci U S A ; 108(20): 8426-31, 2011 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-21536871

RESUMEN

The glycoproteins (GP) of enveloped viruses facilitate entry into the host cell by interacting with specific cellular receptors. Despite extensive study, a cellular receptor for the deadly filoviruses Ebolavirus and Marburgvirus has yet to be identified and characterized. Here, we show that T-cell Ig and mucin domain 1 (TIM-1) binds to the receptor binding domain of the Zaire Ebola virus (EBOV) glycoprotein, and ectopic TIM-1 expression in poorly permissive cells enhances EBOV infection by 10- to 30-fold. Conversely, reduction of cell-surface expression of TIM-1 by RNAi decreased infection of highly permissive Vero cells. TIM-1 expression within the human body is broader than previously appreciated, with expression on mucosal epithelia from the trachea, cornea, and conjunctiva--tissues believed to be important during in vivo transmission of filoviruses. Recognition that TIM-1 serves as a receptor for filoviruses on these mucosal epithelial surfaces provides a mechanistic understanding of routes of entry into the human body via inhalation of aerosol particles or hand-to-eye contact. ARD5, a monoclonal antibody against the IgV domain of TIM-1, blocked EBOV binding and infection, suggesting that antibodies or small molecules directed against this cellular receptor may provide effective filovirus antivirals.


Asunto(s)
Ebolavirus , Marburgvirus , Glicoproteínas de Membrana/análisis , Receptores Virales/análisis , Sitios de Unión , Fiebre Hemorrágica Ebola , Receptor Celular 1 del Virus de la Hepatitis A , Humanos , Membrana Mucosa/química , Unión Proteica
17.
Mol Immunol ; 48(12-13): 1424-31, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21513984

RESUMEN

The gene encoding T cell immunoglobulin and mucin domain-1 (Tim-1) is linked to atopy and asthma susceptibility in mice and humans. Tim-1 is a transmembrane protein expressed on activated lymphocytes and appears to have a role as a co-stimulatory receptor in T cells. The protein has not been shown to have enzymatic activity but contains a site within its cytoplasmic tail predicted to be a target for tyrosine kinases. Here, we show that Tim-1 can associate with the kinase Fyn, a member of the Src family of tyrosine kinases. This association does not require Fyn's kinase activity and is independent of the phosphorylation of a conserved tyrosine present within the cytoplasmic tail of Tim-1. Fyn is necessary for phosphorylation of this tyrosine in Tim-1 and the phosphorylation of Tim-1 varies with the levels of Fyn present in cells. These data suggest a role for Fyn in the signaling downstream of Tim-1.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Receptores Virales/metabolismo , Linfocitos T/metabolismo , Animales , Linfocitos B/metabolismo , Línea Celular , Células Epiteliales , Receptor Celular 1 del Virus de la Hepatitis A , Humanos , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos BALB C , Fosforilación , Proteínas Proto-Oncogénicas c-fyn/genética , ARN Interferente Pequeño , Receptores Virales/genética , Transducción de Señal
18.
J Clin Invest ; 120(8): 2767-81, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20628202

RESUMEN

Studies in mice and humans have revealed that the T cell, immunoglobulin, mucin (TIM) genes are associated with several atopic diseases. TIM-1 is a type I membrane protein that is expressed on T cells upon stimulation and has been shown to modulate their activation. In addition to a recently described interaction with dendritic cells, TIM-1 has also been identified as a phosphatidylserine recognition molecule, and several protein ligands have been proposed. Our understanding of its activity is complicated by the possibility that TIM-1 possesses multiple and diverse binding partners. In order to delineate the function of TIM-1, we generated monoclonal antibodies directed to a cleft formed within the IgV domain of TIM-1. We have shown here that antibodies that bind to this defined cleft antagonize TIM-1 binding to specific ligands and cells. Notably, these antibodies exhibited therapeutic activity in a humanized SCID model of experimental asthma, ameliorating inflammation, and airway hyperresponsiveness. Further experiments demonstrated that the effects of the TIM-1-specific antibodies were mediated via suppression of Th2 cell proliferation and cytokine production. These results demonstrate that modulation of the TIM-1 pathway can critically influence activated T cells in a humanized disease model, suggesting that TIM-1 antagonists may provide potent therapeutic benefit in asthma and other immune-mediated disorders.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Asma/prevención & control , Glicoproteínas de Membrana/antagonistas & inhibidores , Proteínas de la Membrana/antagonistas & inhibidores , Receptores Virales/antagonistas & inhibidores , Animales , Asma/inmunología , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Receptor Celular 1 del Virus de la Hepatitis A , Humanos , Activación de Linfocitos , Glicoproteínas de Membrana/fisiología , Proteínas de la Membrana/fisiología , Ratones , Ratones SCID , Fosfatidilserinas/metabolismo , Receptores Virales/fisiología
19.
Am J Physiol Gastrointest Liver Physiol ; 295(1): G16-G26, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18436621

RESUMEN

Inoculation of BALB/c mice with rhesus rotavirus (RRV) in the newborn period results in biliary epithelial cell (cholangiocyte) infection and the murine model of biliary atresia. Rotavirus infection of a cell requires attachment, which is governed in part by cell-surface expression of integrins such as alpha2beta1. We hypothesized that cholangiocytes were susceptible to RRV infection because they express alpha2beta1. RRV attachment and replication was measured in cell lines derived from cholangiocytes and hepatocytes. Flow cytometry was performed on these cell lines to determine whether alpha2beta1 was present. Cholangiocytes were blocked with natural ligands, a monoclonal antibody, or small interfering RNA against the alpha2-subunit and were infected with RRV. The extrahepatic biliary tract of newborn mice was screened for the expression of the alpha2beta1-integrin. Newborn mice were pretreated with a monoclonal antibody against the alpha2-subunit and were inoculated with RRV. RRV attached and replicated significantly better in cholangiocytes than in hepatocytes. Cholangiocytes, but not hepatocytes, expressed alpha2beta1 in vitro and in vivo. Blocking assays led to a significant reduction in attachment and yield of virus in RRV-infected cholangiocytes. Pretreatment of newborn pups with an anti-alpha2 monoclonal antibody reduced the ability of RRV to cause biliary atresia in mice. Cell-surface expression of the alpha2beta1-integrin plays a role in the mechanism that confers cholangiocyte susceptibility to RRV infection.


Asunto(s)
Conductos Biliares/citología , Conductos Biliares/metabolismo , Atresia Biliar/virología , Integrina alfa2beta1/metabolismo , Rotavirus/fisiología , Animales , Animales Recién Nacidos , Anticuerpos Monoclonales , Anticuerpos Antivirales , Línea Celular , Modelos Animales de Enfermedad , Silenciador del Gen , Hepatocitos/metabolismo , Integrina alfa2beta1/genética , Hígado/metabolismo , Ratones , Ratones Endogámicos BALB C , Replicación Viral
20.
J Immunol ; 180(7): 4361-5, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18354155

RESUMEN

Topical transcutaneous immunization (TCI) presents many clinical advantages, but its underlying mechanism remains unknown. TCI induced Ag-specific IgA Ab-secreting cells expressing CCR9 and CCR10 in the small intestine in a retinoic acid-dependent manner. These intestinal IgA Abs were maintained in Peyer's patch-null mice but abolished in the Peyer's patch- and lymph node-null mice. The mesenteric lymph node (MLN) was shown to be the site of IgA isotype class switching after TCI. Unexpectedly, langerin(+)CD8alpha(-) dendritic cells emerged in the MLN after TCI; they did not migrate from the skin but rather differentiated rapidly from bone marrow precursors. Depletion of langerin(+) cells impaired intestinal IgA Ab responses after TCI. Taken together, these findings suggest that MLN is indispensable for the induction of intestinal IgA Abs following skin immunization and that cross-talk between the skin and gut immune systems might be mediated by langerin(+) dendritic cells in the MLN.


Asunto(s)
Antígenos de Superficie/inmunología , Células Dendríticas/inmunología , Intestinos/inmunología , Lectinas Tipo C/inmunología , Ganglios Linfáticos/inmunología , Lectinas de Unión a Manosa/inmunología , Mesenterio/inmunología , Piel/inmunología , Animales , Células Presentadoras de Antígenos/efectos de los fármacos , Células Cultivadas , Inmunización , Inmunoglobulina A/inmunología , Ratones , Ratones Endogámicos C57BL , Tretinoina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...