Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 185(21): 4038-4038.e1, 2022 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-36240741

RESUMEN

In the tumor microenvironment, immune cells and tumor cells interact in a process called cancer immunoediting, giving rise to changes in gene expression, metabolism, mutational burden, and cellularity in the tumor. This SnapShot compares endogenous versus therapy-induced cancer immunoediting and outlines the molecular and cellular characteristics of interactions that result in complete tumor regression versus tumor escape and progression. To view this SnapShot, open or download the PDF.


Asunto(s)
Neoplasias/inmunología , Microambiente Tumoral , Humanos , Mutación , Neoplasias/genética , Neoplasias/terapia , Escape del Tumor
2.
Immunity ; 56(6): 1187-1203.e12, 2023 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-37160118

RESUMEN

B7 ligands (CD80 and CD86), expressed by professional antigen-presenting cells (APCs), activate the main co-stimulatory receptor CD28 on T cells in trans. However, in peripheral tissues, APCs expressing B7 ligands are relatively scarce. This raises the questions of whether and how CD28 co-stimulation occurs in peripheral tissues. Here, we report that CD8+ T cells displayed B7 ligands that interacted with CD28 in cis at membrane invaginations of the immunological synapse as a result of membrane remodeling driven by phosphoinositide-3-kinase (PI3K) and sorting-nexin-9 (SNX9). cis-B7:CD28 interactions triggered CD28 signaling through protein kinase C theta (PKCθ) and promoted CD8+ T cell survival, migration, and cytokine production. In mouse tumor models, loss of T cell-intrinsic cis-B7:CD28 interactions decreased intratumoral T cells and accelerated tumor growth. Thus, B7 ligands on CD8+ T cells can evoke cell-autonomous CD28 co-stimulation in cis in peripheral tissues, suggesting cis-signaling as a general mechanism for boosting T cell functionality.


Asunto(s)
Antígenos CD28 , Linfocitos T CD8-positivos , Ratones , Animales , Antígenos CD28/metabolismo , Antígenos CD/metabolismo , Ligandos , Membranas Sinápticas/metabolismo , Antígeno B7-2 , Glicoproteínas de Membrana/metabolismo , Antígeno B7-1/metabolismo , Moléculas de Adhesión Celular , Activación de Linfocitos
3.
Immunity ; 51(6): 1059-1073.e9, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31757674

RESUMEN

Combined immunotherapy targeting the immune checkpoint receptors cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death 1 (PD-1), or CTLA-4 and the PD-1 ligand (PD-L1) exhibits superior anti-tumor responses compared with single-agent therapy. Here, we examined the molecular basis for this synergy. Using reconstitution assays with fluorescence readouts, we found that PD-L1 and the CTLA-4 ligand CD80 heterodimerize in cis but not trans. Quantitative biochemistry and cell biology assays revealed that PD-L1:CD80 cis-heterodimerization inhibited both PD-L1:PD-1 and CD80:CTLA-4 interactions through distinct mechanisms but preserved the ability of CD80 to activate the T cell co-stimulatory receptor CD28. Furthermore, PD-L1 expression on antigen-presenting cells (APCs) prevented CTLA-4-mediated trans-endocytosis of CD80. Atezolizumab (anti-PD-L1), but not anti-PD-1, reduced cell surface expression of CD80 on APCs, and this effect was negated by co-blockade of CTLA-4 with ipilimumab (anti-CTLA-4). Thus, PD-L1 exerts an immunostimulatory effect by repressing the CTLA-4 axis; this has implications to the synergy of anti-PD-L1 and anti-CTLA-4 combination therapy.


Asunto(s)
Antígeno B7-1/metabolismo , Antígeno B7-H1/metabolismo , Antígenos CD28/metabolismo , Antígeno CTLA-4/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos/farmacología , Línea Celular Tumoral , Femenino , Células HEK293 , Humanos , Inmunoterapia/métodos , Ipilimumab/farmacología , Células Jurkat , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Neoplasias/inmunología , Neoplasias/terapia , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
4.
Bioinformatics ; 39(10)2023 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-37756695

RESUMEN

MOTIVATION: Precise identification of cancer cells in patient samples is essential for accurate diagnosis and clinical monitoring but has been a significant challenge in machine learning approaches for cancer precision medicine. In most scenarios, training data are only available with disease annotation at the subject or sample level. Traditional approaches separate the classification process into multiple steps that are optimized independently. Recent methods either focus on predicting sample-level diagnosis without identifying individual pathologic cells or are less effective for identifying heterogeneous cancer cell phenotypes. RESULTS: We developed a generalized end-to-end differentiable model, the Cell Scoring Neural Network (CSNN), which takes sample-level training data and predicts the diagnosis of the testing samples and the identity of the diagnostic cells in the sample, simultaneously. The cell-level density differences between samples are linked to the sample diagnosis, which allows the probabilities of individual cells being diagnostic to be calculated using backpropagation. We applied CSNN to two independent clinical flow cytometry datasets for leukemia diagnosis. In both qualitative and quantitative assessments, CSNN outperformed preexisting neural network modeling approaches for both cancer diagnosis and cell-level classification. Post hoc decision trees and 2D dot plots were generated for interpretation of the identified cancer cells, showing that the identified cell phenotypes match the cancer endotypes observed clinically in patient cohorts. Independent data clustering analysis confirmed the identified cancer cell populations. AVAILABILITY AND IMPLEMENTATION: The source code of CSNN and datasets used in the experiments are publicly available on GitHub (http://github.com/erobl/csnn). Raw FCS files can be downloaded from FlowRepository (ID: FR-FCM-Z6YK).


Asunto(s)
Neoplasias Hematológicas , Neoplasias , Humanos , Redes Neurales de la Computación , Neoplasias/diagnóstico , Citometría de Flujo/métodos , Programas Informáticos
5.
J Immunol ; 205(11): 3122-3129, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33077643

RESUMEN

IL-17D is a cytokine that belongs to the IL-17 family and is conserved in vertebrates and invertebrates. In contrast to IL-17A and IL-17F, which are expressed in Th17 cells, IL-17D is expressed broadly in nonimmune cells. IL-17D can promote immune responses to cancer and viruses in part by inducing chemokines and recruiting innate immune cells such as NK cells. Although bacterial infection can induce IL-17D in fish and invertebrates, the role of mammalian IL-17D in antibacterial immunity has not been established. To determine whether IL-17D has a role in mediating host defense against bacterial infections, we studied i.p. infection by group A Streptococcus (GAS) in wild-type (WT) and Il17d -/- mice. Compared with WT animals, mice deficient in IL-17D experienced decreased survival, had greater weight loss, and showed increased bacterial burden in the kidney and peritoneal cavity following GAS challenge. In WT animals, IL-17D transcript was induced by GAS infection and correlated to increased levels of chemokine CCL2 and greater neutrophil recruitment. Of note, GAS-mediated IL-17D induction in nonimmune cells required live bacteria, suggesting that processes beyond recognition of pathogen-associated molecular patterns were required for IL-17D induction. Based on our results, we propose a model in which nonimmune cells can discriminate between nonviable and viable GAS cells, responding only to the latter by inducing IL-17D.


Asunto(s)
Interleucina-27/inmunología , Infecciones Estreptocócicas/inmunología , Streptococcus/inmunología , Animales , Quimiocina CCL2/inmunología , Inmunidad Innata/inmunología , Células Asesinas Naturales/inmunología , Ratones , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Células Th17/inmunología
6.
Proc Natl Acad Sci U S A ; 115(41): 10410-10415, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30254166

RESUMEN

Natural killer (NK) cells are innate immune lymphocytes that recognize and destroy abnormal host cells, such as tumor cells or those infected by viral pathogens. To safely accomplish these functions, NK cells display activating receptors that detect stress molecules or viral ligands displayed at the cell surface, balanced by inhibitory receptors that bind to self-molecules. To date, such activating and inhibitory receptors on NK cells are not known to recognize bacterial determinants. Moreover, NK cell responses to direct interactions with extracellular bacteria are poorly explored. In this study, we observed the human neonatal pathogen group B Streptococcus (GBS) can directly engage human NK cells. The interaction was mediated through the B6N segment of streptococcal ß-protein, binding to the inhibitory receptor Siglec-7 via its amino-terminal V-set domain. Unlike classical Siglec binding, the interaction is also independent of its sialic acid recognition property. In contrast to WT GBS, mutants lacking ß-protein induced efficient pyroptosis of NK cells through the NLRP3 inflammasome, with production and secretion of the proinflammatory cytokine IL-1ß and dissemination of the cytotoxic molecule granzyme B. We postulate that GBS evolved ß-protein engagement of inhibitory human Siglec-7 to suppress the pyroptotic response of NK cells and thereby block recruitment of a broader innate immune response, i.e., by "silencing the sentinel."


Asunto(s)
Antígenos de Diferenciación Mielomonocítica/metabolismo , Proteínas de Unión al ADN/metabolismo , Inmunidad Innata/inmunología , Mediadores de Inflamación/metabolismo , Células Asesinas Naturales/patología , Lectinas/metabolismo , Piroptosis , Antígenos de Diferenciación Mielomonocítica/genética , Células Cultivadas , Proteínas de Unión al ADN/genética , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Lectinas/genética
7.
Cytometry A ; 97(3): 296-307, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31691488

RESUMEN

High-throughput single-cell cytometry technologies have significantly improved our understanding of cellular phenotypes to support translational research and the clinical diagnosis of hematological and immunological diseases. However, subjective and ad hoc manual gating analysis does not adequately handle the increasing volume and heterogeneity of cytometry data for optimal diagnosis. Prior work has shown that machine learning can be applied to classify cytometry samples effectively. However, many of the machine learning classification results are either difficult to interpret without using characteristics of cell populations to make the classification, or suboptimal due to the use of inaccurate cell population characteristics derived from gating boundaries. To date, little has been done to optimize both the gating boundaries and the diagnostic accuracy simultaneously. In this work, we describe a fully discriminative machine learning approach that can simultaneously learn feature representations (e.g., combinations of coordinates of gating boundaries) and classifier parameters for optimizing clinical diagnosis from cytometry measurements. The approach starts from an initial gating position and then refines the position of the gating boundaries by gradient descent until a set of globally-optimized gates across different samples are achieved. The learning procedure is constrained by regularization terms encoding domain knowledge that encourage the algorithm to seek interpretable results. We evaluate the proposed approach using both simulated and real data, producing classification results on par with those generated via human expertise, in terms of both the positions of the gating boundaries and the diagnostic accuracy. © 2019 The Authors. Cytometry Part A published by Wiley Periodicals, Inc. on behalf of International Society for Advancement of Cytometry.


Asunto(s)
Algoritmos , Aprendizaje Automático , Citometría de Flujo , Humanos
8.
J Immunol ; 200(12): 4012-4023, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29703862

RESUMEN

Maintenance of the regulatory T (Treg) cell pool is essential for peripheral tolerance and prevention of autoimmunity. Integrins, heterodimeric transmembrane proteins consisting of α and ß subunits that mediate cell-to-cell and cell-to-extracellular matrix interactions, play an important role in facilitating Treg cell contact-mediated suppression. In this article, we show that integrin activation plays an essential, previously unappreciated role in maintaining murine Treg cell function. Treg cell-specific loss of talin, a ß integrin-binding protein, or expression of talin(L325R), a mutant that selectively abrogates integrin activation, resulted in lethal systemic autoimmunity. This dysfunction could be attributed, in part, to a global dysregulation of the Treg cell transcriptome. Activation of integrin α4ß1 led to increased suppressive capacity of the Treg cell pool, suggesting that modulating integrin activation on Treg cells may be a useful therapeutic strategy for autoimmune and inflammatory disorders. Taken together, these results reveal a critical role for integrin-mediated signals in controlling peripheral tolerance by virtue of maintaining Treg cell function.


Asunto(s)
Integrinas/inmunología , Tolerancia Periférica/inmunología , Linfocitos T Reguladores/inmunología , Animales , Autoinmunidad/inmunología , Inflamación/inmunología , Ratones , Talina/inmunología , Transcriptoma/inmunología
9.
Cell Mol Life Sci ; 75(2): 225-240, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28744671

RESUMEN

The purpose of this review is to explore immune-mediated mechanisms of stress surveillance in cancer, with particular emphasis on the idea that all cancers have classical hallmarks (Hanahan and Weinberg in Cell 100:57-70, 67; Cell 144:646-674, 68) that could be interrelated. We postulate that hallmarks of cancer associated with cellular stress pathways (Luo et al. in Cell 136:823-837, 101) including oxidative stress, proteotoxic stress, mitotic stress, DNA damage, and metabolic stress could define and modulate the inflammatory component of cancer. As such, the overarching goal of this review is to define the types of cellular stress that cancer cells undergo, and then to explore mechanisms by which immune cells recognize, respond to, and are affected by each stress response.


Asunto(s)
Daño del ADN/inmunología , Estrés del Retículo Endoplásmico/inmunología , Vigilancia Inmunológica/inmunología , Neoplasias/inmunología , Estrés Oxidativo/inmunología , Animales , Humanos , Mitosis/inmunología , Modelos Inmunológicos , Neoplasias/genética , Neoplasias/metabolismo , Transducción de Señal/inmunología
10.
Anesth Analg ; 126(6): 1910-1913, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29570154

RESUMEN

The impact of volatile anesthetics on cancer progression has been observed for decades, but sex differences have not been described. Male and female immune systems vary considerably, and the immune system plays an important role in limiting cancer growth. Currently, mouse models describing the impact of volatile anesthetics on cancer growth are limited to same-sex models. In this brief report, we describe a sex-specific impact of isoflurane on melanoma growth observed in wild-type but not in immune-deficient mice. Future experimental designs related to anesthesia and cancer should evaluate the biological variable of sex in a systematic manner.


Asunto(s)
Anestésicos por Inhalación/efectos adversos , Inmunidad Celular/inmunología , Isoflurano/efectos adversos , Melanoma/inducido químicamente , Melanoma/inmunología , Caracteres Sexuales , Animales , Femenino , Inmunidad Celular/efectos de los fármacos , Masculino , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
11.
Biol Blood Marrow Transplant ; 23(4): 625-634, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28104514

RESUMEN

Regulatory T (Treg) cells play a central role in immune tolerance and prevention of aberrant immune responses. Several studies have suggested that the risk of graft-versus-host disease (GVHD) after allogeneic hematopoietic cell transplantation (HCT) can be ameliorated by increasing Tregs. We have developed an approach of in vivo expansion of Tregs with RGI-2001, a novel liposomal formulation of a synthetic derivative of alpha-galactosylceramide, a naturally occurring ligand that binds to CD1 and activates and expands invariant natural killer cells. In preclinical studies, a single intravenous infusion of RGI-2001 expanded Treg and could ameliorate GVHD in a mouse model of allogeneic HCT. To explore the role of RGI-2001 in clinical HCT, we initiated a phase 2A clinical trial (n = 29), testing 2 different doses of RGI-2001 administered as a single infusion on day 0 of allogeneic HCT. RGI-2001 was well tolerated and without infusion reactions or cytokine release syndrome. A subset of patients (8 of 29, 28%) responded to RGI-2001 by inducing a markedly increased number of cells with a Treg phenotype. The Treg had a high Ki-67 index and were almost exclusively Helios+ and Foxp3+, indicating that their accumulation was due to expansion of natural Treg. Notably, the incidence of grade 2 to 4 GVHD in the 8 patients who responded to RGI-2001 was 12.5%, compared with 52.4% in the 21 patients who did not respond. No grade 3 or 4 GVHD was observed in the responder group, compared with a 9.5% incidence among nonresponders. Immunosuppression with sirolimus was also associated with a low incidence of GVHD, suggesting that RGI-2001 may have synergized with sirolimus to promote Treg expansion.


Asunto(s)
Trasplante de Médula Ósea/efectos adversos , Galactosilceramidas/administración & dosificación , Enfermedad Injerto contra Huésped/prevención & control , Sirolimus/administración & dosificación , Linfocitos T Reguladores/citología , Enfermedad Aguda , Adulto , Anciano , Trasplante de Médula Ósea/métodos , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Factores de Transcripción Forkhead , Galactosilceramidas/farmacología , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Humanos , Factor de Transcripción Ikaros , Persona de Mediana Edad , Células T Asesinas Naturales/citología , Sirolimus/farmacología , Linfocitos T Reguladores/efectos de los fármacos , Trasplante Homólogo , Adulto Joven
12.
Cytokine ; 91: 10-12, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27940089

RESUMEN

Early stage immune responses can dictate the severity and outcome of inflammatory processes such as tumor growth and viral infection. Cytokines such as the interleukin 17 (IL-17) family and cellular stress defense (e.g., anti-oxidant) pathways have evolved early and regulate disease surveillance in vertebrates and invertebrates as far back as Caenorhabditis elegans. Our group has recently found a new role for nuclear factor erythroid-derived 2-like 2 (Nrf2) in regulating early anti-cancer immune responses by inducing IL-17D and recruiting natural killer (NK) cells. In this Cytokine Stimulus, we discuss recent findings that encourage boosting the Nrf2/IL-17D/NK cell axis for the treatment of cancer and viral infection.


Asunto(s)
Vigilancia Inmunológica , Interleucina-17/inmunología , Células Asesinas Naturales/inmunología , Factor 2 Relacionado con NF-E2/inmunología , Neoplasias Experimentales/inmunología , Virosis/inmunología , Animales , Células Asesinas Naturales/patología , Ratones , Neoplasias Experimentales/patología , Virosis/patología
13.
J Immunol ; 194(6): 2551-60, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25667416

RESUMEN

The link between autoimmune diseases and primary immunodeficiency syndromes has been increasingly appreciated. Immunologic evaluation of a young man with autoimmune enterocolopathy and unexplained infections revealed evidence of immunodeficiency, including IgG subclass deficiency, impaired Ag-induced lymphocyte proliferation, reduced cytokine production by CD8(+) T lymphocytes, and decreased numbers of NK cells. Genetic evaluation identified haploinsufficiency of NFAT5, a transcription factor regulating immune cell function and cellular adaptation to hyperosmotic stress, as a possible cause of this syndrome. Inhibition or deletion of NFAT5 in normal human and murine cells recapitulated several of the immune deficits identified in the patient. These results provide evidence of a primary immunodeficiency disorder associated with organ-specific autoimmunity linked to NFAT5 deficiency.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Enfermedades Gastrointestinales/inmunología , Haploinsuficiencia/inmunología , Síndromes de Inmunodeficiencia/inmunología , Factores de Transcripción/inmunología , Animales , Enfermedades Autoinmunes/diagnóstico , Enfermedades Autoinmunes/genética , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Células Cultivadas , Citocinas/inmunología , Citocinas/metabolismo , Análisis Mutacional de ADN , Enfermedades Gastrointestinales/diagnóstico , Enfermedades Gastrointestinales/genética , Expresión Génica/inmunología , Haploinsuficiencia/genética , Humanos , Immunoblotting , Síndromes de Inmunodeficiencia/diagnóstico , Síndromes de Inmunodeficiencia/genética , Células Jurkat , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Masculino , Ratones de la Cepa 129 , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Adulto Joven
14.
Proc Natl Acad Sci U S A ; 111(16): 5998-6003, 2014 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-24711415

RESUMEN

Compelling evidence for naturally occurring immunosurveillance against malignancies informs and justifies some current approaches toward cancer immunotherapy. However, some types of immune reactions have also been shown to facilitate tumor progression. For example, our previous studies showed that although experimental tumor growth is enhanced by low levels of circulating antibodies directed against the nonhuman sialic acid N-glycolyl-neuraminic acid (Neu5Gc), which accumulates in human tumors, growth could be inhibited by anti-Neu5Gc antibodies from a different source, in a different model. However, it remains generally unclear whether the immune responses that mediate cancer immunosurveillance vs. those responsible for inflammatory facilitation are qualitatively and/or quantitatively distinct. Here, we address this question using multiple murine tumor growth models in which polyclonal antibodies against tumor antigens, such as Neu5Gc, can alter tumor progression. We found that although growth was stimulated at low antibody doses, it was inhibited by high doses, over a linear and remarkably narrow range, defining an immune response curve (IRC; i.e., inverse hormesis). Moreover, modulation of immune responses against the tumor by altering antibody avidity or by enhancing innate immunity shifted the IRC in the appropriate direction. Thus, the dualistic role of immunosurveillance vs. inflammation in modulating tumor progression can be quantitatively distinguished in multiple model systems, and can occur over a remarkably narrow range. Similar findings were made in a human tumor xenograft model using a narrow range of doses of a monoclonal antibody currently in clinical use. These findings may have implications for the etiology, prevention, and treatment of cancer.


Asunto(s)
Anticuerpos Antineoplásicos/inmunología , Hormesis/inmunología , Neoplasias/inmunología , Neoplasias/patología , Inmunidad Adaptativa/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Afinidad de Anticuerpos/inmunología , Linfoma de Burkitt/inmunología , Linfoma de Burkitt/patología , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Inmunoglobulina G/inmunología , Inflamación/patología , Células Asesinas Naturales/inmunología , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Ácidos Neuramínicos/inmunología , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/metabolismo
15.
Proc Natl Acad Sci U S A ; 111(39): 14211-6, 2014 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-25225409

RESUMEN

Certain pathogenic bacteria are known to modulate the innate immune response by decorating themselves with sialic acids, which can engage the myelomonocytic lineage inhibitory receptor Siglec-9, thereby evading immunosurveillance. We hypothesized that the well-known up-regulation of sialoglycoconjugates by tumors might similarly modulate interactions with innate immune cells. Supporting this hypothesis, Siglec-9-expressing myelomonocytic cells found in human tumor samples were accompanied by a strong up-regulation of Siglec-9 ligands. Blockade of Siglec-9 enhanced neutrophil activity against tumor cells in vitro. To investigate the function of inhibitory myelomonocytic Siglecs in vivo we studied mouse Siglec-E, the murine functional equivalent of Siglec-9. Siglec-E-deficient mice showed increased in vivo killing of tumor cells, and this effect was reversed by transgenic Siglec-9 expression in myelomonocytic cells. Siglec-E-deficient mice also showed enhanced immunosurveillance of autologous tumors. However, once tumors were established, they grew faster in Siglec-E-deficient mice. In keeping with this, Siglec-E-deficient macrophages showed a propensity toward a tumor-promoting M2 polarization, indicating a secondary role of CD33-related Siglecs in limiting cancer-promoting inflammation and tumor growth. Thus, we define a previously unidentified impact of inhibitory myelomonocytic Siglecs in cancer biology, with distinct roles that reflect the dual function of myelomonocytic cells in cancer progression. In keeping with this, a human polymorphism that reduced Siglec-9 binding to carcinomas was associated with improved early survival in non-small-cell lung cancer patients, which suggests that Siglec-9 might be therapeutically targeted within the right time frame and stage of disease.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos B/metabolismo , Inmunidad Innata , Neoplasias/inmunología , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/metabolismo , Animales , Antígenos CD/genética , Antígenos de Diferenciación de Linfocitos B/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Línea Celular Tumoral , Femenino , Humanos , Ligandos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Monocitos/inmunología , Activación Neutrófila , Polimorfismo de Nucleótido Simple , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/genética , Microambiente Tumoral/inmunología
16.
Int Immunol ; 27(3): 161-6, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25568303

RESUMEN

Drak2 is a promising therapeutic target to treat organ-specific autoimmune diseases such as type 1 diabetes and multiple sclerosis without causing generalized immune suppression. Inhibition of Drak2 may also prevent graft rejection following organ transplantation. However, Drak2 may function as a critical tumor suppressor, which would challenge the prospect of targeting Drak2 for therapeutic treatment. Thus, we examined the susceptibility of Drak2 (-/-) mice in several tumor models. We show that Drak2 is not required to prevent tumor formation in a variety of settings. Therefore, Drak2 does not function as an essential tumor suppressor in in vivo tumor models. These data further validate Drak2 as a viable therapeutic target to treat autoimmune disease and graft rejection. Importantly, these data also indicate that while Drak2 may induce apoptosis when overexpressed in cell lines, it is not an essential tumor suppressor.


Asunto(s)
Diabetes Mellitus Tipo 1/tratamiento farmacológico , Rechazo de Injerto/prevención & control , Vigilancia Inmunológica , Esclerosis Múltiple/tratamiento farmacológico , Trasplante de Órganos , Proteínas Serina-Treonina Quinasas/metabolismo , Sarcoma/metabolismo , Animales , Apoptosis , Línea Celular Tumoral , Modelos Animales de Enfermedad , Rechazo de Injerto/etiología , Humanos , Terapia de Inmunosupresión , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/genética , Sarcoma/tratamiento farmacológico , Proteínas Supresoras de Tumor/metabolismo
17.
Proc Natl Acad Sci U S A ; 110(1): E15-22, 2013 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-23236148

RESUMEN

The idea of targeted therapy, whereby drug or protein molecules are delivered to specific cells, is a compelling approach to treating disease. Immunotoxins are one such targeted therapeutic, consisting of an antibody domain for binding target cells and molecules of a toxin that inhibits the proliferation of the targeted cell. One major hurdle preventing these therapies from reaching the market has been the lack of a suitable production platform that allows the cost-effective production of these highly complex molecules. The chloroplast of the green alga Chlamydomonas reinhardtii has been shown to contain the machinery necessary to fold and assemble complex eukaryotic proteins. However, the translational apparatus of chloroplasts resembles that of a prokaryote, allowing them to accumulate eukaryotic toxins that otherwise would kill a eukaryotic host. Here we show expression and accumulation of monomeric and dimeric immunotoxin proteins in algal chloroplasts. These fusion proteins contain an antibody domain targeting CD22, a B-cell surface epitope, and the enzymatic domain of exotoxin A from Pseudomonas aeruginosa. We demonstrated that algal-produced immunotoxins accumulate as soluble and enzymatically active proteins that bind target B cells and efficiently kill them in vitro. We also show that treatment with either the mono- or dimeric immunotoxins significantly prolongs the survival of mice with implanted human B-cell tumors.


Asunto(s)
Chlamydomonas reinhardtii/metabolismo , Cloroplastos/metabolismo , Inmunotoxinas/aislamiento & purificación , Inmunotoxinas/farmacología , Linfoma/tratamiento farmacológico , Ingeniería de Proteínas/métodos , Animales , Western Blotting , Cromatografía en Gel , Proteínas de Unión al ADN/genética , Electroforesis en Gel de Poliacrilamida , Femenino , Citometría de Flujo , Vectores Genéticos/genética , Humanos , Ratones , Ratones Noqueados , Organismos Modificados Genéticamente , Proteínas Recombinantes/metabolismo , Transformación Genética , Trasplante Heterólogo
18.
Proc Natl Acad Sci U S A ; 110(30): 12391-6, 2013 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-23836658

RESUMEN

The aryl hydrocarbon receptor (AhR) has become increasingly recognized for its role in the differentiation and activity of immune cell subsets; however, its role in regulating the activity of natural killer (NK) cells has not been described. Here, we show that AhR expression is induced in murine NK cells upon cytokine stimulation. We show that in the absence of AhR, NK cells have reduced cytolytic activity and reduced capacity to control RMA-S tumor formation in vivo, despite having normal development and maturation markers. Although AhR was first identified to bind the xenobiotic compound dioxin, AhR is now known to bind a variety of natural exogenous (e.g., dietary) and endogenous ligands. We show that activation of AhR with an endogenous tryptophan derivative, 6-formylindolo[3,2-b]carbazole, potentiates NK cell IFN-γ production and cytolytic activity. Further, administration of 6-formylindolo[3,2-b]carbazole in vivo enhances NK cell control of tumors in an NK cell- and AhR-dependent manner. Finally, similar effects on NK cell potency occur with AhR dietary ligands, potentially explaining the numerous associations that have been observed in the past between diet and NK cell function. Our studies introduce AhR as another regulator of NK cell activity in vivo.


Asunto(s)
Células Asesinas Naturales/inmunología , Neoplasias Experimentales/inmunología , Receptores de Hidrocarburo de Aril/fisiología , Animales , Línea Celular Tumoral , Citocinas/biosíntesis , Citocinas/inmunología , Dieta , Ratones , Ratones Endogámicos C57BL , Receptores de Hidrocarburo de Aril/genética
19.
Am J Pathol ; 184(12): 3384-93, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25310970

RESUMEN

Cancer cells condition macrophages and other inflammatory cells in the tumor microenvironment so that these cells are more permissive for cancer growth and metastasis. Conditioning of inflammatory cells reflects, at least in part, soluble mediators (such as transforming growth factor ß and IL-4) that are released by cancer cells and alter the phenotype of cells of the innate immune system. Signaling pathways in cancer cells that potentiate this activity are incompletely understood. The urokinase receptor (uPAR) is a cell-signaling receptor known to promote cancer cell survival, proliferation, metastasis, and cancer stem cell-like properties. The present findings show that uPAR expression in diverse cancer cells, including breast cancer, pancreatic cancer, and glioblastoma cells, promotes the ability of these cells to condition co-cultured bone marrow-derived macrophages so that the macrophages express significantly increased levels of arginase 1, a biomarker of the alternatively activated M2 macrophage phenotype. Expression of transforming growth factor ß was substantially increased in uPAR-expressing cancer cells via a mechanism that requires uPA-initiated cell signaling. uPAR also controlled expression of IL-4 in cancer cells via a mechanism that involves activation of ERK1/2. The ability of uPAR to induce expression of factors that condition macrophages in the tumor microenvironment may constitute an important mechanism by which uPAR promotes cancer progression.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Interleucina-4/metabolismo , Macrófagos/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Arginasa/metabolismo , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Proliferación Celular , Técnicas de Cocultivo , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Regulación Neoplásica de la Expresión Génica , Humanos , Inflamación , Ratones , Metástasis de la Neoplasia , Fenotipo , Transducción de Señal
20.
Brain Behav Immun ; 49: 49-53, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25747743

RESUMEN

OBJECTIVES: Stimuli that activate the sympathetic nervous system, such as acute psychological stress, rapidly invoke a robust mobilization of lymphocytes into the circulation. Experimental animal studies suggest that bone marrow-derived progenitor cells (PCs) also mobilize in response to sympathetic stimulation. Here we tested the effects of acute psychological stress and brief pharmacological ß-adrenergic (ßAR) stimulation on peripheral PC numbers in humans. METHODS: In two studies, we investigated PC mobilization in response to an acute speech task (n=26) and ßAR-agonist (isoproterenol) infusion (n=20). A subset of 8 participants also underwent the infusion protocol with concomitant administration of the ßAR-antagonist propranolol. Flow cytometry was used to enumerate lymphocyte subsets, total progenitor cells, total haematopoietic stem cells (HSC), early HSC (multi-lineage potential), late HSC (lineage committed), and endothelial PCs (EPCs). RESULTS: Both psychological stress and ßAR-agonist infusion caused the expected mobilization of total monocytes and lymphocytes and CD8(+) T lymphocytes. Psychological stress also induced a modest, but significant, increase in total PCs, HSCs, and EPC numbers in peripheral blood. However, infusion of a ßAR-agonist did not result in a significant change in circulating PCs. CONCLUSION: PCs are rapidly mobilized by psychological stress via mechanisms independent of ßAR-stimulation, although the findings do not exclude ßAR-stimulation as a possible cofactor. Considering the clinical and physiological relevance, further research into the mechanisms involved in stress-induced PC mobilization seems warranted.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Receptores Adrenérgicos beta/metabolismo , Estrés Psicológico/metabolismo , Agonistas Adrenérgicos beta/farmacología , Antagonistas Adrenérgicos beta/farmacología , Adulto , Ansiedad/inmunología , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/inmunología , Complejo CD3/inmunología , Linfocitos T CD8-positivos/inmunología , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Frecuencia Cardíaca/inmunología , Humanos , Isoproterenol/farmacología , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Monocitos/inmunología , Propranolol/farmacología , Habla , Estrés Psicológico/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA