Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
bioRxiv ; 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38659921

RESUMEN

Synthetic receptors that mediate antigen-dependent cell responses are transforming therapeutics, drug discovery, and basic research. However, established technologies such as chimeric antigen receptors (CARs) can only detect immobilized antigens, have limited output scope, and lack built-in drug control. Here, we engineer synthetic G protein-coupled receptors (GPCRs) capable of driving a wide range of native or nonnative cellular processes in response to user-defined antigen. We achieve modular antigen gating by engineering and fusing a conditional auto-inhibitory domain onto GPCR scaffolds. Antigen binding to a fused nanobody relieves auto-inhibition and enables receptor activation by drug, thus generating Programmable Antigen-gated G protein-coupled Engineered Receptors (PAGERs). We create PAGERs responsive to more than a dozen biologically and therapeutically important soluble and cell surface antigens, in a single step, from corresponding nanobody binders. Different PAGER scaffolds permit antigen binding to drive transgene expression, real-time fluorescence, or endogenous G protein activation, enabling control of cytosolic Ca 2+ , lipid signaling, cAMP, and neuronal activity. Due to its modular design and generalizability, we expect PAGER to have broad utility in discovery and translational science.

2.
Elife ; 112022 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-35311648

RESUMEN

Technologies for detecting cell-cell contacts are powerful tools for studying a wide range of biological processes, from neuronal signaling to cancer-immune interactions within the tumor microenvironment. Here, we report TRACC (Transcriptional Readout Activated by Cell-cell Contacts), a GPCR-based transcriptional recorder of cellular contacts, which converts contact events into stable transgene expression. TRACC is derived from our previous protein-protein interaction recorders, SPARK (Kim et al., 2017) and SPARK2 (Kim et al., 2019), reported in this journal. TRACC incorporates light gating via the light-oxygen-voltage-sensing (LOV) domain, which provides user-defined temporal control of tool activation and reduces background. We show that TRACC detects cell-cell contacts with high specificity and sensitivity in mammalian cell culture and that it can be used to interrogate interactions between neurons and glioma, a form of brain cancer.


Asunto(s)
Luz , Transducción de Señal , Animales , Mamíferos
3.
Proc Natl Acad Sci U S A ; 117(46): 28763-28774, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33139573

RESUMEN

The molecular mechanisms by which receptor tyrosine kinases (RTKs) and heterotrimeric G proteins, two major signaling hubs in eukaryotes, independently relay signals across the plasma membrane have been extensively characterized. How these hubs cross-talk has been a long-standing question, but answers remain elusive. Using linear ion-trap mass spectrometry in combination with biochemical, cellular, and computational approaches, we unravel a mechanism of activation of heterotrimeric G proteins by RTKs and chart the key steps that mediate such activation. Upon growth factor stimulation, the guanine-nucleotide exchange modulator dissociates Gαi•ßγ trimers, scaffolds monomeric Gαi with RTKs, and facilitates the phosphorylation on two tyrosines located within the interdomain cleft of Gαi. Phosphorylation triggers the activation of Gαi and inhibits second messengers (cAMP). Tumor-associated mutants reveal how constitutive activation of this pathway impacts cell's decision to "go" vs. "grow." These insights define a tyrosine-based G protein signaling paradigm and reveal its importance in eukaryotes.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Animales , Células COS , Chlorocebus aethiops , Receptores ErbB/metabolismo , Células HEK293 , Células HeLa , Proteínas de Unión al GTP Heterotriméricas/fisiología , Humanos , Fosforilación , Proteínas Tirosina Quinasas Receptoras/fisiología , Transducción de Señal , Tirosina/metabolismo
4.
iScience ; 23(6): 101209, 2020 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-32535026

RESUMEN

Cells perceive and respond to the extracellular matrix via integrin receptors; their dysregulation has been implicated in inflammation and cancer metastasis. Here we show that a guanine nucleotide-exchange modulator of trimeric-GTPase Gαi, GIV (a.k.a Girdin), directly binds the integrin adaptor Kindlin-2. A non-canonical short linear motif within the C terminus of GIV binds Kindlin-2-FERM3 domain at a site that is distinct from the binding site for the canonical NPxY motif on the -integrin tail. Binding of GIV to Kindlin-2 allosterically enhances Kindlin-2's affinity for ß1-integrin. Consequently, integrin activation and clustering are maximized, which augments cell adhesion, spreading, and invasion. Findings elucidate how the GIV•Kindlin-2 complex has a 2-fold impact: it allosterically synergizes integrin activation and enables ß1-integrins to indirectly access and modulate trimeric GTPases via the complex. Furthermore, Cox proportional-hazard models on tumor transcriptomics provide trans-scale evidence of synergistic interactions between GIV•Kindlin-2•ß1-integrin on time to progression to metastasis.

5.
Proc Natl Acad Sci U S A ; 116(33): 16394-16403, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31363053

RESUMEN

Heterotrimeric G proteins are key molecular switches that control cell behavior. The canonical activation of G proteins by agonist-occupied G protein-coupled receptors (GPCRs) has recently been elucidated from the structural perspective. In contrast, the structural basis for GPCR-independent G protein activation by a novel family of guanine-nucleotide exchange modulators (GEMs) remains unknown. Here, we present a 2.0-Å crystal structure of Gαi in complex with the GEM motif of GIV/Girdin. Nucleotide exchange assays, molecular dynamics simulations, and hydrogen-deuterium exchange experiments demonstrate that GEM binding to the conformational switch II causes structural changes that allosterically propagate to the hydrophobic core of the Gαi GTPase domain. Rearrangement of the hydrophobic core appears to be a common mechanism by which GPCRs and GEMs activate G proteins, although with different efficiency. Atomic-level insights presented here will aid structure-based efforts to selectively target the noncanonical G protein activation.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/química , Proteínas de Unión al GTP Heterotriméricas/química , Proteínas de Microfilamentos/química , Receptores Acoplados a Proteínas G/química , Proteínas de Transporte Vesicular/química , Regulación Alostérica/genética , Cristalografía por Rayos X , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Factores de Intercambio de Guanina Nucleótido/química , Factores de Intercambio de Guanina Nucleótido/genética , Células HeLa , Proteínas de Unión al GTP Heterotriméricas/genética , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Proteínas de Microfilamentos/genética , Simulación de Dinámica Molecular , Unión Proteica/genética , Conformación Proteica , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/genética , Proteínas de Transporte Vesicular/genética
6.
Sci Signal ; 11(519)2018 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-29487190

RESUMEN

Cellular proliferation, differentiation, and morphogenesis are shaped by multiple signaling cascades, and their dysregulation plays an integral role in cancer progression. Three cascades that contribute to oncogenic potential are those mediated by Wnt proteins and the receptor Frizzled (FZD), growth factor receptor tyrosine kinases (RTKs), and heterotrimeric G proteins and associated GPCRs. Daple is a guanine nucleotide exchange factor (GEF) for the G protein Gαi Daple also binds to FZD and the Wnt/FZD mediator Dishevelled (Dvl), and it enhances ß-catenin-independent Wnt signaling in response to Wnt5a-FZD7 signaling. We identified Daple as a substrate of multiple RTKs and non-RTKs and, hence, as a point of convergence for the three cascades. We found that phosphorylation near the Dvl-binding motif in Daple by both RTKs and non-RTKs caused Daple/Dvl complex dissociation and augmented the ability of Daple to bind to and activate Gαi, which potentiated ß-catenin-independent Wnt signals and stimulated epithelial-mesenchymal transition (EMT) similarly to Wnt5a/FZD7 signaling. Although Daple acts as a tumor suppressor in the healthy colon, the concurrent increased abundance of Daple and epidermal growth factor receptor (EGFR) in colorectal tumors was associated with poor patient prognosis. Thus, the Daple-dependent activation of Gαi and the Daple-dependent enhancement of ß-catenin-independent Wnt signals are not only stimulated by Wnt5a/FZD7 to suppress tumorigenesis but also hijacked by growth factor-activated RTKs to enhance tumor progression. These findings identify a cross-talk paradigm among growth factor RTKs, heterotrimeric G proteins, and the Wnt/FZD pathway in cancer.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/genética , Proteínas de Unión al GTP Heterotriméricas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Wnt/genética , Vía de Señalización Wnt/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Proteínas Dishevelled/genética , Proteínas Dishevelled/metabolismo , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células HeLa , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Humanos , Estimación de Kaplan-Meier , Fosforilación , Unión Proteica , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Wnt/metabolismo
7.
Curr Protoc Chem Biol ; 8(4): 265-298, 2016 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-27925669

RESUMEN

Canonical signal transduction via heterotrimeric G proteins is spatiotemporally restricted, i.e., triggered exclusively at the plasma membrane, only by agonist activation of G protein-coupled receptors via a finite process that is terminated within a few hundred milliseconds. Recently, a rapidly emerging paradigm has revealed a noncanonical pathway for activation of heterotrimeric G proteins via the nonreceptor guanidine-nucleotide exchange factor, GIV/Girdin. Biochemical, biophysical, and functional studies evaluating this pathway have unraveled its unique properties and distinctive spatiotemporal features. As in the case of any new pathway/paradigm, these studies first required an in-depth optimization of tools/techniques and protocols, governed by rationale and fundamentals unique to the pathway, and more specifically to the large multimodular GIV protein. Here we provide the most up-to-date overview of protocols that have generated most of what we know today about noncanonical G protein activation by GIV and its relevance in health and disease. © 2016 by John Wiley & Sons, Inc.


Asunto(s)
Técnica del Anticuerpo Fluorescente/métodos , Factores de Intercambio de Guanina Nucleótido/análisis , Immunoblotting/métodos , Inmunoprecipitación/métodos , Animales , Biofisica/métodos , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Transducción de Señal
8.
Bioessays ; 38(4): 379-93, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26879989

RESUMEN

Canonical signal transduction via heterotrimeric G proteins is spatially and temporally restricted, that is, triggered exclusively at the plasma membrane (PM), only by agonist activation of G protein-coupled receptors (GPCRs) via a process that completes within a few hundred milliseconds. Recently, a rapidly emerging paradigm has revealed a non-canonical pathway for activation of heterotrimeric G proteins by the non-receptor guanidine-nucleotide exchange factor (GEF), GIV/Girdin. This pathway has distinctive temporal and spatial features and an unusual profile of receptor engagement: diverse classes of receptors, not just GPCRs can engage with GIV to trigger such activation. Such activation is spatially and temporally unrestricted, that is, can occur both at the PM and on internal membranes discontinuous with the PM, and can continue for prolonged periods of time. Here, we provide the most complete up-to-date review of the molecular mechanisms that govern the unique spatiotemporal aspects of non-canonical G protein activation by GIV and the relevance of this new paradigm in health and disease.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Diabetes Mellitus/metabolismo , Proteínas de Microfilamentos/metabolismo , Neoplasias/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/genética , Proteínas de Transporte Vesicular/metabolismo , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/patología , Enfermedades Cardiovasculares/terapia , Membrana Celular/metabolismo , Diabetes Mellitus/genética , Diabetes Mellitus/patología , Diabetes Mellitus/terapia , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Membranas Intracelulares , Proteínas de Microfilamentos/genética , Modelos Moleculares , Terapia Molecular Dirigida , Neoplasias/genética , Neoplasias/patología , Neoplasias/terapia , Mapeo de Interacción de Proteínas , Receptores Acoplados a Proteínas G/genética , Factores de Tiempo , Proteínas de Transporte Vesicular/genética
9.
Biochem Biophys Res Commun ; 468(1-2): 287-93, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26514725

RESUMEN

Insulin resistance (IR) is a metabolic disorder characterized by impaired glucose uptake in response to insulin. The current paradigm for insulin signaling centers upon the insulin receptor (InsR) and its substrate IRS1; the latter is believed to be the chief conduit for post-receptor signaling. We recently demonstrated that GIV, a Guanidine Exchange Factor (GEF) for the trimeric G protein, Gαi, is a major hierarchical conduit for the metabolic insulin response. By virtue of its ability to directly bind the InsR, IRS1 and PI3K, GIV enhances the InsR-IRS1-Akt-AS160 (RabGAP) signaling cascade and cellular glucose uptake via its GEF function. Phosphoinhibition of GIV-GEF by the fatty-acid/PKCθ pathway inhibits the cascade and impairs glucose uptake. Here we show that GIV directly and constitutively binds the exocyst complex subunit Exo-70 and also associates with GLUT4-storage vesicles (GSVs) exclusively upon insulin stimulation. Without GIV or its GEF function, membrane association of Exo-70 as well as exocytosis of GSVs in response to insulin are impaired. Thus, GIV is an essential component within the insulin signaling cascade that couples upstream signal transducers within the InsR and G-Protein signaling cascade to downstream vesicular trafficking events within the exocytic pathway. These findings suggest a role of GIV in coordinating key signaling and trafficking events of metabolic insulin response.


Asunto(s)
Transportador de Glucosa de Tipo 4/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animales , Línea Celular , Exocitosis , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células HeLa , Humanos , Insulina/metabolismo , Unión Proteica , Transporte de Proteínas , Ratas , Transducción de Señal
10.
Mol Biol Cell ; 26(24): 4313-24, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26446841

RESUMEN

GIV/Girdin is a multimodular signal transducer and a bona fide metastasis-related protein. As a guanidine exchange factor (GEF), GIV modulates signals initiated by growth factors (chemical signals) by activating the G protein Gαi. Here we report that mechanical signals triggered by the extracellular matrix (ECM) also converge on GIV-GEF via ß1 integrins and that focal adhesions (FAs) serve as the major hubs for mechanochemical signaling via GIV. GIV interacts with focal adhesion kinase (FAK) and ligand-activated ß1 integrins. Phosphorylation of GIV by FAK enhances PI3K-Akt signaling, the integrity of FAs, increases cell-ECM adhesion, and triggers ECM-induced cell motility. Activation of Gαi by GIV-GEF further potentiates FAK-GIV-PI3K-Akt signaling at the FAs. Spatially restricted signaling via tyrosine phosphorylated GIV at the FAs is enhanced during cancer metastasis. Thus GIV-GEF serves as a unifying platform for integration and amplification of adhesion (mechanical) and growth factor (chemical) signals during cancer progression.


Asunto(s)
Adhesiones Focales/metabolismo , Proteínas de Unión al GTP/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Secuencia de Aminoácidos , Animales , Células COS , Línea Celular Tumoral , Movimiento Celular/fisiología , Quinasa 1 de Adhesión Focal/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células HeLa , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Unión Proteica , Transducción de Señal , Tirosina/metabolismo
11.
Mol Biol Cell ; 26(23): 4209-23, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26378251

RESUMEN

Insulin resistance (IR) is a metabolic disorder characterized by impaired insulin signaling and cellular glucose uptake. The current paradigm for insulin signaling centers upon the insulin receptor (InsR) and its substrate IRS1; the latter is believed to be the sole conduit for postreceptor signaling. Here we challenge that paradigm and show that GIV/Girdin, a guanidine exchange factor (GEF) for the trimeric G protein Gαi, is another major hierarchical conduit for the metabolic insulin response. By virtue of its ability to directly bind InsR, IRS1, and phosphoinositide 3-kinase, GIV serves as a key hub in the immediate postreceptor level, which coordinately enhances the metabolic insulin response and glucose uptake in myotubes via its GEF function. Site-directed mutagenesis or phosphoinhibition of GIV-GEF by the fatty acid/protein kinase C-theta pathway triggers IR. Insulin sensitizers reverse phosphoinhibition of GIV and reinstate insulin sensitivity. We also provide evidence for such reversible regulation of GIV-GEF in skeletal muscles from patients with IR. Thus GIV is an essential upstream component that couples InsR to G-protein signaling to enhance the metabolic insulin response, and impairment of such coupling triggers IR. We also provide evidence that GIV-GEF serves as therapeutic target for exogenous manipulation of physiological insulin response and reversal of IR in skeletal muscles.


Asunto(s)
Reguladores de Proteínas de Unión al GTP/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Resistencia a la Insulina/fisiología , Proteínas de Microfilamentos/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Células Cultivadas , Ácidos Grasos/metabolismo , Femenino , Humanos , Insulina/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Músculo Esquelético/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Receptor de Insulina/metabolismo , Transducción de Señal
12.
Proc Natl Acad Sci U S A ; 112(20): E2602-10, 2015 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-25926659

RESUMEN

In eukaryotes, receptor tyrosine kinases (RTKs) and trimeric G proteins are two major signaling hubs. Signal transduction via trimeric G proteins has long been believed to be triggered exclusively by G protein-coupled receptors (GPCRs). This paradigm has recently been challenged by several studies on a multimodular signal transducer, Gα-Interacting Vesicle associated protein (GIV/Girdin). We recently demonstrated that GIV's C terminus (CT) serves as a platform for dynamic association of ligand-activated RTKs with Gαi, and for noncanonical transactivation of G proteins. However, exogenous manipulation of this platform has remained beyond reach. Here we developed cell-permeable GIV-CT peptides by fusing a TAT-peptide transduction domain (TAT-PTD) to the minimal modular elements of GIV that are necessary and sufficient for activation of Gi downstream of RTKs, and used them to engineer signaling networks and alter cell behavior. In the presence of an intact GEF motif, TAT-GIV-CT peptides enhanced diverse processes in which GIV's GEF function has previously been implicated, e.g., 2D cell migration after scratch-wounding, invasion of cancer cells, and finally, myofibroblast activation and collagen production. Furthermore, topical application of TAT-GIV-CT peptides enhanced the complex, multireceptor-driven process of wound repair in mice in a GEF-dependent manner. Thus, TAT-GIV peptides provide a novel and versatile tool to manipulate Gαi activation downstream of growth factors in a diverse array of pathophysiologic conditions.


Asunto(s)
Péptidos de Penetración Celular/metabolismo , Proteínas de Unión al GTP/metabolismo , Productos del Gen tat/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de Microfilamentos/metabolismo , Modelos Moleculares , Transducción de Señal/fisiología , Proteínas de Transporte Vesicular/metabolismo , Animales , Péptidos de Penetración Celular/uso terapéutico , Transferencia Resonante de Energía de Fluorescencia , Productos del Gen tat/química , Productos del Gen tat/genética , Ingeniería Genética/métodos , Células HeLa , Humanos , Ratones , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/genética , Reacción en Cadena de la Polimerasa , Transducción Genética/métodos , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/genética
13.
Cancer Immunol Immunother ; 62(8): 1303-13, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23661160

RESUMEN

Hu14.18-IL2 is an immunocytokine (IC) consisting of human IL-2 linked to hu14.18 mAb, which recognizes GD2 disialoganglioside. Phase II clinical trials of intravenous-hu14.18-IL2 (IV-IC) in neuroblastoma and melanoma are underway, and have already demonstrated activity in neuroblastoma. In our Phase II trial, lower neuroblastoma burden at the time of treatment was associated with a greater likelihood of clinical response to IV-IC. We have previously shown that intratumoral-hu14.18-IL2 (IT-IC) compared to IV-IC results in enhanced local and systemic antitumor activity in tumor-bearing mice. We utilized a mouse model to investigate the impact of tumor burden on hu14.18-IL2 treatment efficacy in IV- versus IT-treated animals. Studies presented here describe the analyses of tumor burden at the initiation of treatment and its effects on treatment efficacy, survival, and tumor-infiltrating leukocytes in A/J mice bearing subcutaneous NXS2 neuroblastoma. We show that smaller tumor burden at treatment initiation is associated with increased infiltration of NK and CD8+ T cells and increased overall survival. NXS2 tumor shrinkage shortly after completion of the 3 days of hu14.18-IL2 treatment is necessary for long-term survival. This model demonstrates that tumor size is a strong predictor of hu14.18-IL2-induced lymphocyte infiltration and treatment outcome.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Linfocitos T CD8-positivos/inmunología , Interleucina-2/inmunología , Células Asesinas Naturales/inmunología , Neuroblastoma/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/genética , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Inmunoterapia/métodos , Interleucina-2/administración & dosificación , Interleucina-2/genética , Células Asesinas Naturales/metabolismo , Ratones , Ratones Endogámicos , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Neuroblastoma/patología , Neuroblastoma/terapia , Pronóstico , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/inmunología , Análisis de Supervivencia , Factores de Tiempo , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología
14.
J Immunol ; 189(5): 2656-64, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22844125

RESUMEN

hu14.18-IL-2 (IC) is an immunocytokine consisting of human IL-2 linked to hu14.18 mAb, which recognizes the GD2 disialoganglioside. Phase 2 clinical trials of i.v. hu14.18-IL-2 (i.v.-IC) in neuroblastoma and melanoma are underway and have already demonstrated activity in neuroblastoma. We showed previously that intratumoral hu14.18-IL-2 (IT-IC) results in enhanced antitumor activity in mouse models compared with i.v.-IC. The studies presented in this article were designed to determine the mechanisms involved in this enhanced activity and to support the future clinical testing of intratumoral administration of immunocytokines. Improved survival and inhibition of growth of both local and distant tumors were observed in A/J mice bearing s.c. NXS2 neuroblastomas treated with IT-IC compared with those treated with i.v.-IC or control mice. The local and systemic antitumor effects of IT-IC were inhibited by depletion of NK cells or T cells. IT-IC resulted in increased NKG2D receptors on intratumoral NKG2A/C/E⁺ NKp46⁺ NK cells and NKG2A/C/E⁺ CD8⁺ T cells compared with control mice or mice treated with i.v.-IC. NKG2D levels were augmented more in tumor-infiltrating lymphocytes compared with splenocytes, supporting the localized nature of the intratumoral changes induced by IT-IC treatment. Prolonged retention of IC at the tumor site was seen with IT-IC compared with i.v.-IC. Overall, IT-IC resulted in increased numbers of activated T and NK cells within tumors, better IC retention in the tumor, enhanced inhibition of tumor growth, and improved survival compared with i.v.-IC.


Asunto(s)
Antineoplásicos/uso terapéutico , Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Neuroblastoma/inmunología , Neuroblastoma/terapia , Proteínas Recombinantes de Fusión/uso terapéutico , Subgrupos de Linfocitos T/inmunología , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Inyecciones Intralesiones , Interleucina-2/administración & dosificación , Interleucina-2/metabolismo , Interleucina-2/uso terapéutico , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/patología , Ratones , Ratones Endogámicos A , Ratones Endogámicos C57BL , Neuroblastoma/patología , Distribución Aleatoria , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/metabolismo , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología
15.
Cancer Immunol Immunother ; 61(10): 1683-97, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22392192

RESUMEN

Tumor growth is often accompanied by the accumulation of myeloid cells in the tumors and lymphoid organs. These cells can suppress T cell immunity, thereby posing an obstacle to T cell-targeted cancer immunotherapy. In this study, we tested the possibility of activating tumor-associated myeloid cells to mediate antitumor effects. Using the peritoneal model of B16 melanoma, we show that peritoneal cells (PEC) in tumor-bearing mice (TBM) had reduced ability to secrete nitric oxide (NO) following in vitro stimulation with interferon gamma and lipopolysaccharide, as compared to PEC from control mice. This reduced function of PEC was accompanied by the influx of CD11b(+) Gr-1(+) myeloid cells to the peritoneal cavity. Nonadherent PEC were responsible for most of the NO production in TBM, whereas in naïve mice NO was mainly secreted by adherent CD11b(+) F4/80(+) macrophages. Sorted CD11b(+) Gr-1(-) monocytic and CD11b(+) Gr-1(+) granulocytic PEC from TBM had a reduced ability to secrete NO following in vitro stimulation (compared to naïve PEC), but effectively suppressed proliferation of tumor cells in vitro. In vivo, treatment of mice bearing established peritoneal B16 tumors with anti-CD40 and CpG resulted in activation of tumor-associated PEC, reduction in local tumor burden and prolongation of mouse survival. Inhibition of NO did not abrogate the antitumor effects of stimulated myeloid cells. Taken together, the results indicate that in tumor-bearing hosts, tumor-associated myeloid cells can be activated to mediate antitumor effects.


Asunto(s)
Melanoma Experimental/inmunología , Células Mieloides/inmunología , Neoplasias Peritoneales/inmunología , Traslado Adoptivo/métodos , Animales , Antígenos CD40/inmunología , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Islas de CpG/inmunología , Femenino , Inmunoterapia/métodos , Interferón gamma/farmacología , Lipopolisacáridos/farmacología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Óxido Nítrico/metabolismo
16.
Artículo en Inglés | MEDLINE | ID: mdl-19964260

RESUMEN

The majority of the focus related to the modernization of medical records is placed on developed countries. However, developing countries are also progressing from paper-based records to electronic records. The requirements of their systems can be dramatically different from those of the developed world. This paper describes briefly the benefits of EMRs in developing countries. It focuses on the basic EMR information, including types of EMRs, components of EMRs, and already existing case studies, in order to establish which EMR systems would be feasible and effective for specific situations.


Asunto(s)
Países en Desarrollo , Registros Electrónicos de Salud , Estudios de Factibilidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA