Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Semin Immunol ; 69: 101799, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37413923

RESUMEN

Siglecs (sialic acid-binding immunoglobulin-like lectins) are a family of vertebrate glycan-binding cell-surface proteins. The majority mediate cellular inhibitory activity once engaged by specific ligands or ligand-mimicking molecules. As a result, Siglec engagement is now of interest as a strategy to therapeutically dampen unwanted cellular responses. When considering allergic inflammation, human eosinophils and mast cells express overlapping but distinct patterns of Siglecs. For example, Siglec-6 is selectively and prominently expressed on mast cells while Siglec-8 is highly specific for both eosinophils and mast cells. This review will focus on a subset of Siglecs and their various endogenous or synthetic sialoside ligands that regulate eosinophil and mast cell function and survival. It will also summarize how certain Siglecs have become the focus of novel therapies for allergic and other eosinophil- and mast cell-related diseases.


Asunto(s)
Eosinófilos , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico , Humanos , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/metabolismo , Mastocitos , Antígenos CD/química , Ligandos
2.
Int J Mol Sci ; 24(4)2023 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36835342

RESUMEN

After exposure to an antigen, CD8 T cells reach a decision point about their fate: to become either short-lived effector cells (SLECs) or memory progenitor effector cells (MPECs). SLECs are specialized in providing an immediate effector function but have a shorter lifespan and lower proliferative capacity compared to MPECs. Upon encountering the cognate antigen during an infection, CD8 T cells rapidly expand and then contract to a level that is maintained for the memory phase after the peak of the response. Studies have shown that the contraction phase is mediated by TGFß and selectively targets SLECs, while sparing MPECs. The aim of this study is to investigate how the CD8 T cell precursor stage determines TGFß sensitivity. Our results demonstrate that MPECs and SLECs have differential responses to TGFß, with SLECs being more sensitive to TGFß than MPECs. This difference in sensitivity is associated with the levels of TGFßRI and RGS3, and the SLEC-related transcriptional activator T-bet binding to the TGFßRI promoter may provide a molecular basis for increased TGFß sensitivity in SLECs.


Asunto(s)
Linfocitos T CD8-positivos , Memoria Inmunológica , Subgrupos de Linfocitos T , Factor de Crecimiento Transformador beta , Animales , Ratones , Antígenos/metabolismo , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular , Ratones Endogámicos C57BL , Subgrupos de Linfocitos T/inmunología , Factor de Crecimiento Transformador beta/inmunología
3.
J Am Chem Soc ; 141(36): 14032-14037, 2019 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-31460762

RESUMEN

The Siglec family of cell surface receptors have emerged as attractive targets for cell-directed therapies due to their restricted expression on immune cells, endocytic properties, and ability to modulate receptor signaling. Human Siglec-8, for instance, has been identified as a therapeutic target for the treatment of eosinophil and mast cell disorders. A promising strategy to target Siglecs involves the use of liposomal nanoparticles with a multivalent display of Siglec ligands. A key challenge for this approach is the identification of a high affinity ligand for the target Siglec. Here, we report the development of a ligand of Siglec-8 and its closest murine functional orthologue Siglec-F that is capable of targeting liposomes to cells expressing Siglec-8 or -F. A glycan microarray library of synthetic 9-N-sulfonyl sialoside analogues was screened to identify potential lead compounds. The best ligand, 9-N-(2-naphthyl-sulfonyl)-Neu5Acα2-3-[6-O-sulfo]-Galß1-4GlcNAc (6'-O-sulfo NSANeu5Ac) combined the lead 2-naphthyl sulfonyl C-9 substituent with the preferred sulfated scaffold. The ligand 6'-O-sulfo NSANeu5Ac was conjugated to lipids for display on liposomes to evaluate targeted delivery to cells. Targeted liposomes showed strong in vitro binding/uptake and selectivity to cells expressing Siglec-8 or -F and, when administered to mice, exhibit in vivo targeting to Siglec-F+ eosinophils.


Asunto(s)
Antígenos de Diferenciación Mielomonocítica/metabolismo , Linfocitos B/efectos de los fármacos , Lectinas/antagonistas & inhibidores , Ácidos Siálicos/farmacología , Sulfonamidas/farmacología , Linfocitos T/efectos de los fármacos , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos B/metabolismo , Linfocitos B/metabolismo , Células CHO , Cricetulus , Humanos , Lectinas/metabolismo , Liposomas/química , Liposomas/metabolismo , Ratones , Conformación Molecular , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico , Ácidos Siálicos/química , Sulfonamidas/química , Linfocitos T/metabolismo
4.
J Allergy Clin Immunol ; 141(2): 505-517, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29045815

RESUMEN

The goal of this series is to offer a survey of the latest literature for clinicians and scientists alike, providing a list of important recent advances relevant to the broad field of allergy and immunology. This particular assignment was to cover the topic of eosinophils. In an attempt to highlight major ideas, themes, trends, and advances relevant to basic and clinical aspects of eosinophil biology, a search of articles published since 2015 in the Journal of Allergy and Clinical Immunology and other high-impact journals was performed. Articles were then reviewed and organized, and then key findings were summarized. Given space limitations, many outstanding articles could not be included, but the hope is that what follows provides a succinct overview of recently published work that has significantly added to our knowledge of eosinophils and eosinophil-associated diseases.


Asunto(s)
Eosinófilos/inmunología , Eosinófilos/patología , Enfermedades del Sistema Inmune/inmunología , Humanos , Enfermedades del Sistema Inmune/patología
5.
J Allergy Clin Immunol ; 141(5): 1774-1785.e7, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-28734845

RESUMEN

BACKGROUND: Sialic acid-binding immunoglobulin-like lectin (Siglec)-8 is a cell-surface protein expressed selectively on human eosinophils, mast cells, and basophils, making it an ideal target for the treatment of diseases involving these cell types. However, the effective delivery of therapeutic agents to these cells requires an understanding of the dynamics of Siglec-8 surface expression. OBJECTIVES: We sought to determine whether Siglec-8 is endocytosed in human eosinophils and malignant mast cells, identify mechanisms underlying its endocytosis, and demonstrate whether a toxin can be targeted to Siglec-8-bearing cells to kill these cells. METHODS: Siglec-8 surface dynamics were examined by flow cytometry using peripheral blood eosinophils, mast cell lines, and Siglec-8-transduced cells in the presence of inhibitors targeting components of endocytic pathways. Siglec-8 intracellular trafficking was followed by confocal microscopy. The ribosome-inhibiting protein saporin was conjugated to a Siglec-8-specific antibody to examine the targeting of an agent to these cells through Siglec-8 endocytosis. RESULTS: Siglec-8 endocytosis required actin rearrangement, tyrosine kinase and protein kinase C activities, and both clathrin and lipid rafts. Internalized Siglec-8 localized to the lysosomal compartment. Maximal endocytosis in Siglec-8-transduced HEK293T cells required an intact immunoreceptor tyrosine-based inhibitory motif. Siglec-8 was also shuttled to the surface via a distinct pathway. Sialidase treatment of eosinophils revealed that Siglec-8 is partially masked by sialylated cis ligands. Targeting saporin to Siglec-8 consistently caused extensive cell death in eosinophils and the human mast cell leukemia cell line HMC-1.2. CONCLUSIONS: Therapeutic payloads can be targeted selectively to eosinophils and malignant mast cells by exploiting this Siglec-8 endocytic pathway.


Asunto(s)
Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos B/inmunología , Eosinófilos/inmunología , Lectinas/inmunología , Mastocitos/inmunología , Actinas/inmunología , Basófilos/inmunología , Muerte Celular/inmunología , Línea Celular , Línea Celular Tumoral , Clatrina/inmunología , Endocitosis/inmunología , Citometría de Flujo/métodos , Células HEK293 , Humanos , Ligandos , Microdominios de Membrana/inmunología , Proteína Quinasa C/inmunología , Proteínas Tirosina Quinasas/inmunología
6.
J Allergy Clin Immunol ; 141(6): 2196-2207, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-28888781

RESUMEN

BACKGROUND: Siglec-8 is a CD33 subfamily cell-surface receptor selectively expressed on human eosinophils. After cytokine priming, Siglec-8 mAb or glycan ligand binding causes eosinophil apoptosis associated with reactive oxygen species (ROS) production. Most CD33-related Siglecs function as inhibitory receptors, but the ability of Siglec-8 to stimulate eosinophil ROS production and apoptosis suggests that Siglec-8 might instead function as an activating receptor. OBJECTIVE: We sought to determine the role of IL-5 priming and identify the signaling molecules involved in Siglec-8 function for human eosinophils. METHODS: We used an mAb and/or a multimeric synthetic sulfated sialoglycan ligand recognizing Siglec-8 in combination with integrin blocking antibodies, pharmacologic inhibitors, phosphoproteomics, and Western blot analysis to define the necessity of various proteins involved in Siglec-8 function for human eosinophils. RESULTS: Cytokine priming was required to elicit the unanticipated finding that Siglec-8 engagement promotes rapid ß2-integrin-dependent eosinophil adhesion. Also novel was the finding that this adhesion was necessary for subsequent ROS production and apoptosis. Siglec-8-mediated ROS was generated through reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activation because pretreatment of eosinophils with catalase (an extracellular superoxide scavenger) or NSC 23766 (a Rac GTPase inhibitor) completely inhibited Siglec-8-mediated eosinophil apoptosis. Finally, engagement of Siglec-8 on IL-5-primed eosinophils resulted in increased phosphorylation of Akt, p38, and c-Jun N-terminal kinase 1 that was also ß2-integrin dependent; pharmacologic inhibition of these kinases completely prevented Siglec-8-mediated eosinophil apoptosis. CONCLUSIONS: These data demonstrate that Siglec-8 functions uniquely as an activating receptor on IL-5-primed eosinophils through a novel pathway involving regulation of ß2-integrin-dependent adhesion, NADPH oxidase, and a subset of protein kinases.


Asunto(s)
Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos B/inmunología , Antígenos CD18/inmunología , Eosinófilos/inmunología , Lectinas/inmunología , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos B/metabolismo , Apoptosis/inmunología , Antígenos CD18/metabolismo , Adhesión Celular/inmunología , Degranulación de la Célula/inmunología , Eosinófilos/metabolismo , Humanos , Lectinas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
7.
J Immunol ; 188(8): 3639-47, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22430740

RESUMEN

CD8(+) T cell responses have been shown to be regulated by dendritic cells (DCs) and CD4(+) T cells, leading to the tenet that CD8(+) T cells play a passive role in their own differentiation. In contrast, by using a DNA vaccination model, to separate the events of vaccination from those of CD8(+) T cell priming, we demonstrate that CD8(+) T cells, themselves, actively limit their own memory potential through CD8(+) T cell-derived IFN-γ-dependent modification of the IL-12/IL-15Rα axis on DCs. Such CD8(+) T cell-driven cytokine alterations result in increased T-bet and decreased Bcl-2 expression, and thus decreased memory progenitor formation. These results identify an unrecognized role for CD8(+) T cells in the regulation of their own effector differentiation fate and a previously uncharacterized relationship between the balance of inflammation and memory formation.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Memoria Inmunológica , Receptores de Interleucina-12/inmunología , Receptores de Interleucina-15/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/inmunología , Células Dendríticas/metabolismo , Regulación de la Expresión Génica/inmunología , Humanos , Interferón gamma , Interleucina-12/genética , Interleucina-12/inmunología , Interleucina-15/genética , Interleucina-15/inmunología , Activación de Linfocitos , Melanoma Experimental/inmunología , Melanoma Experimental/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Interleucina-12/genética , Receptores de Interleucina-15/genética , Transducción de Señal/inmunología , Vacunación , Vacunas de ADN/inmunología
8.
J Immunol ; 186(1): 275-83, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21106849

RESUMEN

Treatment of tumor-bearing mice with a stimulatory Ab to glucocorticoid-induced TNFR family-related receptor (GITR) has previously been shown to elicit protective T cell responses against poorly immunogenic tumors. However, the role of GITR stimulation on CD8 T cells and the nature of tumor rejection Ags have yet to be determined. In this study, we show that a stimulatory mAb to GITR (clone DTA-1) acts directly on CD8 T cells, but not on CD4(+)CD25(+) regulatory T (T(reg)) cells, in B16 tumor-bearing mice to induce concomitant immunity against secondary B16 tumors, as well as protective memory following surgical excision of the primary tumor. Melanoma growth itself induced GITR expression on tumor-specific CD8 T cells, providing a mechanism whereby these cells may respond to stimulatory anti-GITR. Unexpectedly, in contrast to T(reg) cell depletion therapy with anti-CD4, GITR stimulation induced very weak CD8 T cell responses to melanocyte differentiation Ags expressed by the tumor, and did not induce autoimmune vitiligo. Accordingly, GITR-stimulated hosts that were primed with B16 melanoma rejected B16, but not the unrelated JBRH melanoma, indicating that tumor rejection Ags are tumor-specific rather than shared. In support of this, we show that GITR stimulation induces CD8 T cell responses to a tumor-specific Ag, and that these responses are of higher functional avidity compared with those induced by T(reg) cell depletion. We conclude that stimulation of GITR on effector CD8 T cells results in high-avidity T cell responses to tumor-specific Ags, thereby inducing potent antitumor immunity in the absence of autoimmunity.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Epítopos de Linfocito T/inmunología , Activación de Linfocitos/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Animales , Linfocitos T CD8-positivos/patología , Adhesión Celular/inmunología , Línea Celular Tumoral , Femenino , Proteína Relacionada con TNFR Inducida por Glucocorticoide , Depleción Linfocítica , Masculino , Melanoma Experimental/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores de Factor de Crecimiento Nervioso/deficiencia , Receptores de Factor de Crecimiento Nervioso/inmunología , Receptores del Factor de Necrosis Tumoral/deficiencia , Receptores del Factor de Necrosis Tumoral/inmunología , Linfocitos T Reguladores/inmunología
9.
J Immunol ; 186(6): 3309-16, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21289306

RESUMEN

A main goal of cancer immunology research is the formation of Ag-specific memory T cell immunity capable of activation upon tumor re-encounter. The requirements necessary to overcome the inhibitory signals present in the tumor microenvironment and form such memory T cell responses are unknown. In contrast to previous studies targeting tumors expressing highly immunogenic model Ags, we demonstrate that alleviating tumor-induced suppression along with vaccination against authentic Ags during the perioperative period provides long-lasting protection against a highly suppressive and poorly immunogenic melanoma. In this study, we employed DNA vaccination with an immunologically optimized mouse melanoma-shared Ag, Trp1ee/ng, combined with systemic TGF-ß blockade during the perioperative period of primary tumor resection, to confer protection against B16 melanoma, and against JBRH, an independently derived melanoma unrelated to B16. Importantly, we demonstrate that correlative to memory responses, perioperative immunotherapy increases the formation of tumor-infiltrating and tumor-reactive CD8(+) T cells expressing low levels of the transcription factor T-bet, defined as memory precursor effector cells. We show that conditions for an immunologically fertile environment are met when TGF-ß blockade and vaccination are applied during the perioperative period of primary tumor resection. These findings address limitations of current CD8(+) T cell immunotherapies against cancer by generating effective CD8(+) T cell memory recall responses.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Memoria Inmunológica , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma Experimental/cirugía , Melanoma Experimental/terapia , Glicoproteínas de Membrana/uso terapéutico , Oxidorreductasas/uso terapéutico , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/uso terapéutico , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/patología , Carcinoma Pulmonar de Lewis/cirugía , Quimioterapia Combinada , Inmunización Secundaria/métodos , Memoria Inmunológica/genética , Inmunoterapia Adoptiva/métodos , Linfocitos Infiltrantes de Tumor/patología , Masculino , Melanoma Experimental/patología , Glicoproteínas de Membrana/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oxidorreductasas/administración & dosificación , Periodo Perioperatorio/métodos , Células Madre/inmunología , Células Madre/patología , Factor de Crecimiento Transformador beta/fisiología , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética , Vacunas de ADN/uso terapéutico
10.
Front Immunol ; 14: 1283370, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37928558

RESUMEN

Sialic acid-binding immunoglobulin-like lectin (Siglec)-8 is a sialoside-binding receptor expressed by eosinophils and mast cells that exhibits priming status- and cell type-dependent inhibitory activity. On eosinophils that have been primed with IL-5, GM-CSF, or IL-33, antibody ligation of Siglec-8 induces cell death through a pathway involving the ß2 integrin-dependent generation of reactive oxygen species (ROS) via NADPH oxidase. In contrast, Siglec-8 engagement on mast cells inhibits cellular activation and mediator release but reportedly does not impact cell viability. The differences in responses between cytokine-primed and unprimed eosinophils, and between eosinophils and mast cells, to Siglec-8 ligation are not understood. We previously found that Siglec-8 binds to sialylated ligands present on the surface of the same cell (so-called cis ligands), preventing Siglec-8 ligand binding in trans. However, the functional relevance of these cis ligands has not been elucidated. We therefore explored the potential influence of cis ligands of Siglec-8 on both eosinophils and mast cells. De-sialylation using exogenous sialidase profoundly altered the consequences of Siglec-8 antibody engagement on both cell types, eliminating the need for cytokine priming of eosinophils to facilitate cell death and enabling Siglec-8-dependent mast cell death without impacting anti-Siglec-8 antibody binding. The cell death process licensed by de-sialylation resembled that characterized in IL-5-primed eosinophils, including CD11b upregulation, ROS production, and the activities of Syk, PI3K, and PLC. These results implicate cis ligands in restraining Siglec-8 function on eosinophils and mast cells and reveal a promising approach to the selective depletion of mast cells in patients with mast cell-mediated diseases.


Asunto(s)
Eosinófilos , Mastocitos , Humanos , Ligandos , Especies Reactivas de Oxígeno/metabolismo , Interleucina-5/metabolismo , Antígenos CD/metabolismo , Muerte Celular , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/metabolismo , Citocinas/metabolismo
11.
Mol Immunol ; 155: 1-6, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36634520

RESUMEN

CD8 T cells play a critical role in immunity against intracellular pathogens and cancer. A primary objective of T cell-based vaccine strategies is the induction of durable and effective immune responses. Achieving this goal involves more than simply boosting the numbers of responding T cells. Of particular interest is the induction of CD8 T cells with polycytokine capability, specifically with the ability of CD8 T cells to co-produce IFNγ, TNFα and IL-2. The presence of these polycytokine-producing CD8 T cells correlates strongly with protection against foreign pathogens and cancer. Therefore, approaches capable of inducing such polyfunctional responses are needed. NKG2D engagement on CD8 T cells has been shown to result in increased effector response. However, the manner in which NKG2D engagement results in improved CD8 T cell effector response is unclear. Here we demonstrate in vitro and in vivo that NKG2D engagement by its natural ligand, Rae-1ε, shifts the balance from single cytokine to polycytokine (IL-2, IFNγ, and TFNα) production. These data define a previously unrecognized process in which NKG2D costimulation on CD8 T cells results in improved effector responses.


Asunto(s)
Citocinas , Neoplasias , Humanos , Subfamilia K de Receptores Similares a Lectina de Células NK , Interleucina-2 , Linfocitos T CD8-positivos
12.
Mol Aspects Med ; 90: 101104, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-35835621

RESUMEN

The term "allergic diseases" encompasses several common, IgE-mediated conditions that range from being annoying to those that are life-threatening. Available treatments include active avoidance of the instigating allergen and the use of a variety of oral, inhaled, intranasal, intraocular and injected agents. While most individuals with allergies do well with existing therapies, there are still unmet therapeutic needs. Siglecs (sialic acid-binding, immunoglobulin-like lectins) are a family of single-pass transmembrane I-type lectins found on various subsets of cells, especially those of the immune system. All Siglecs have extracellular domains recognizing sialoside ligands, and most contain cytoplasmic domains with inhibitory signaling activity. This review focuses on Siglecs that likely play a role in regulating allergic and asthmatic responses, and how specific Siglecs, expressed on cells such as eosinophils and mast cells, are being targeted for therapeutic benefit.


Asunto(s)
Asma , Hipersensibilidad , Humanos , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/metabolismo , Antígenos CD , Transducción de Señal , Asma/terapia
13.
Expert Opin Drug Deliv ; 20(2): 205-222, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36629456

RESUMEN

INTRODUCTION: Current and developing mast cell therapeutics are reliant on small molecule drugs and biologics, but few are truly selective for mast cells. Most have cellular and disease-specific limitations that require innovation to overcome longstanding challenges to selectively targeting and modulating mast cell behavior. This review is designed to serve as a frame of reference for new approaches that utilize nanotechnology or combine different drugs to increase mast cell selectivity and therapeutic efficacy. AREAS COVERED: Mast cell diseases include allergy and related conditions as well as malignancies. Here, we discuss the targets of existing and developing therapies used to treat these disease pathologies, classifying them into cell surface, intracellular, and extracellular categories. For each target discussed, we discuss drugs that are either the current standard of care, under development, or have indications for potential use. Finally, we discuss how novel technologies and tools can be used to take existing therapeutics to a new level of selectivity and potency against mast cells. EXPERT OPINION: There are many broadly and very few selectively targeted therapeutics for mast cells in allergy and malignant disease. Combining existing targeting strategies with technology like nanoparticles will provide novel platforms to treat mast cell disease more selectively.


Asunto(s)
Productos Biológicos , Hipersensibilidad , Trastornos de la Activación de los Mastocitos , Neoplasias , Humanos , Sistemas de Liberación de Medicamentos , Mastocitos/metabolismo , Mastocitos/patología , Neoplasias/tratamiento farmacológico , Hipersensibilidad/tratamiento farmacológico , Hipersensibilidad/metabolismo , Hipersensibilidad/patología
14.
Cells ; 11(7)2022 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-35406705

RESUMEN

Mast cells are tissue-resident cells that contribute to allergic diseases, among others, due to excessive or inappropriate cellular activation and degranulation. Therapeutic approaches to modulate mast cell activation are urgently needed. Siglec-6 is an immunoreceptor tyrosine-based inhibitory motif (ITIM)-bearing receptor selectively expressed by mast cells, making it a promising target for therapeutic intervention. However, the effects of its engagement on mast cells are poorly defined. Siglec-6 expression and endocytosis on primary human mast cells and mast cell lines were assessed by flow cytometry. SIGLEC6 mRNA expression was examined by single-cell RNAseq in esophageal tissue biopsy samples. The ability of Siglec-6 engagement or co-engagement to prevent primary mast cell activation was determined based on assessments of mediator and cytokine secretion and degranulation markers. Siglec-6 was highly expressed by all mast cells examined, and the SIGLEC6 transcript was restricted to mast cells in esophageal biopsy samples. Siglec-6 endocytosis occurred with delayed kinetics relative to the related receptor Siglec-8. Co-crosslinking of Siglec-6 with FcεRIα enhanced the inhibition of mast cell activation and diminished downstream ERK1/2 and p38 phosphorylation. The selective, stable expression and potent inhibitory capacity of Siglec-6 on human mast cells are favorable for its use as a therapeutic target in mast cell-driven diseases.


Asunto(s)
Antígenos CD , Antígenos de Diferenciación Mielomonocítica , Lectinas , Mastocitos , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/genética , Línea Celular , Humanos , Lectinas/genética , Mastocitos/metabolismo , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/genética
15.
Cancer Immunol Immunother ; 60(11): 1543-51, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21681376

RESUMEN

While the effects of TCR affinity and TGFß on CD8(+) T-cell function have been studied individually, the manner in which TCR affinity dictates susceptibility to TGFß-mediated suppression remains unknown. To address this issue, we utilized OVA altered peptide ligands (APLs) of different affinities in the OT-I model. We demonstrate that while decreased TCR ligand affinity initially results in weakened responses, such interactions prime the resultant effector cells to respond more strongly to cognate antigen upon secondary exposure. Despite this, responses by CD8(+) T cells primed with lower-affinity TCR ligands are more effectively regulated by TGFß. Susceptibility to TGFß-mediated suppression is associated with downregulation of RGS3, a recently recognized negative regulator of TGFß signaling, but not expression of TGFß receptors I/II. These results suggest a novel tolerance mechanism whereby CD8(+) T cells are discriminately regulated by TGFß according to the affinity of the ligand on which they were initially primed. In addition, because of the major role played by TGFß in tumor-induced immune suppression, these results identify the affinity of the priming ligand as a primary concern in CD8(+) T-cell-mediated cancer immunotherapeutic strategies.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Oligopéptidos/inmunología , Factor de Crecimiento Transformador beta/inmunología , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Regulación hacia Abajo , Proteínas de Unión al GTP/biosíntesis , Proteínas de Unión al GTP/inmunología , Proteínas Activadoras de GTPasa/biosíntesis , Proteínas Activadoras de GTPasa/inmunología , Humanos , Ligandos , Ratones , Ratones Endogámicos C57BL , Oligopéptidos/farmacología , Proteínas RGS , Transducción de Señal , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología
16.
Cancer Immunol Immunother ; 60(2): 291-7, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21193909

RESUMEN

CD8+ T cell function depends on a finely orchestrated balance of activation/suppression signals. While the stimulatory role of the CD8 co-receptor and pleiotropic capabilities of TGF-ß have been studied individually, the influence of CD8 co-receptor on TGF-ß function in CD8+ T cells is unknown. Here, we show that while CD8 enhances T cell activation, it also enhances susceptibility to TGF-ß-mediated immune suppression. Using Jurkat cells expressing a full-length, truncated or no αßCD8 molecule, we demonstrate that cells expressing full-length αßCD8 were highly susceptible, αßCD8-truncated cells were partially susceptible, and CD8-deficient cells were completely resistant to suppression by TGF-ß. Additionally, we determined that inhibition of Lck rendered mouse CD8+ T cells highly resistant to TGF-ß suppression. Resistance was not associated with TGF-ß receptor expression but did correlate with decreased Smad3 and increased Smad7 levels. These findings highlight a previously unrecognized third role for CD8 co-receptor which appears to prepare activated CD8+ T cells for response to TGF-ß. Based on the important role which TGF-ß-mediated suppression plays in tumor immunology, these findings unveil necessary considerations in formulation of CD8+ T cell-related cancer immunotherapy strategies.


Asunto(s)
Antígenos CD8/inmunología , Linfocitos T CD8-positivos/inmunología , Tolerancia Inmunológica/inmunología , Factor de Crecimiento Transformador beta/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Antígenos CD8/biosíntesis , Humanos , Células Jurkat , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/antagonistas & inhibidores , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/biosíntesis , Ratones , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/inmunología , Proteínas Smad/metabolismo
17.
Front Immunol ; 12: 737988, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34721399

RESUMEN

Sialic acid-binding immunoglobulin-like lectin (Siglec)-8 is a glycan-binding receptor bearing immunoreceptor tyrosine-based inhibitory and switch motifs (ITIM and ITSM, respectively) that is selectively expressed on eosinophils, mast cells, and, to a lesser extent, basophils. Previous work has shown that engagement of Siglec-8 on IL-5-primed eosinophils causes cell death via CD11b/CD18 integrin-mediated adhesion and NADPH oxidase activity and identified signaling molecules linking adhesion, reactive oxygen species (ROS) production, and cell death. However, the proximal signaling cascade activated directly by Siglec-8 engagement has remained elusive. Most members of the Siglec family possess similar cytoplasmic signaling motifs and recruit the protein tyrosine phosphatases SHP-1/2, consistent with ITIM-mediated signaling, to dampen cellular activation. However, the dependence of Siglec-8 function in eosinophils on these phosphatases has not been studied. Using Siglec-8 antibody engagement and pharmacological inhibition in conjunction with assays to measure cell-surface upregulation and conformational activation of CD11b integrin, ROS production, and cell death, we sought to identify molecules involved in Siglec-8 signaling and determine the stage of the process in which each molecule plays a role. We demonstrate here that the enzymatic activities of Src family kinases (SFKs), Syk, SHIP1, PAK1, MEK1, ERK1/2, PLC, PKC, acid sphingomyelinase/ceramidase, and Btk are all necessary for Siglec-8-induced eosinophil cell death, with no apparent role for SHP-1/2, SHIP2, or c-Raf. While most of these signaling molecules are necessary for Siglec-8-induced upregulation of CD11b integrin at the eosinophil cell surface, Btk is phosphorylated and activated later in the signaling cascade and is instead necessary for CD11b activation. In contrast, SFKs and ERK1/2 are phosphorylated far earlier in the process, consistent with their role in augmenting cell-surface levels of CD11b. In addition, pretreatment of eosinophils with latrunculin B or jasplakinolide revealed that actin filament disassembly is necessary and sufficient for surface CD11b integrin upregulation and that actin polymerization is necessary for downstream ROS production. These results show that Siglec-8 signals through an unanticipated set of signaling molecules in IL-5-primed eosinophils to induce cell death and challenges the expectation that ITIM-bearing Siglecs signal through inhibitory pathways involving protein tyrosine phosphatases to achieve their downstream functions.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos B/metabolismo , Eosinófilos/metabolismo , Lectinas/metabolismo , Agammaglobulinemia Tirosina Quinasa/metabolismo , Antígeno CD11b/metabolismo , Muerte Celular , Células Cultivadas , Eosinófilos/efectos de los fármacos , Eosinófilos/inmunología , Eosinófilos/patología , Humanos , Interleucina-5/farmacología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/metabolismo , Proteína Quinasa C/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Quinasa Syk/metabolismo , Fosfolipasas de Tipo C/metabolismo , Quinasas p21 Activadas/metabolismo , Familia-src Quinasas/metabolismo
18.
BMC Res Notes ; 13(1): 211, 2020 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-32276656

RESUMEN

OBJECTIVE: Little has been reported regarding the reliability of methods for the purification of human blood eosinophils. We retrospectively reviewed our experience with 350 consecutive eosinophil isolations. RESULTS: Between January 2014 and December 2018, we conducted 350 eosinophil purifications from 83 donors. Absolute eosinophil count (AEC), calculated from hospital complete blood counts when available (n = 289), ranged from 32 to 1352 eosinophils/µL ([Formula: see text]: 179 ± 136/µL). Eosinophil yields ranged from 0.4 to 24.4 million cells per 20 mL of blood drawn ([Formula: see text]: 3.1 ± 1.9 million eosinophils) with > 98% purity. Comparing AEC to actual yield, recovery was 87% ± 29% ([Formula: see text]) and AEC strongly correlated with yield. To explore the reproducibility of yield, a subsequent analysis was limited to those donors drawn ≥ 3 times (N = 35), and there was no difference in the average coefficient of variation for yield between allergic and non-allergic donors. Viability of isolated eosinophils was consistently > 95% and after 24 h of culture did not differ between allergic and non-allergic donors. We conclude that this immunomagnetic separation method for human eosinophil isolation from whole blood is a reliable, reproducible technique for obtaining an average of 87% yield with high purity and viability.


Asunto(s)
Centrifugación por Gradiente de Densidad , Eosinófilos , Separación Inmunomagnética , Adulto , Centrifugación por Gradiente de Densidad/normas , Femenino , Humanos , Separación Inmunomagnética/normas , Masculino , Persona de Mediana Edad , Reproducibilidad de los Resultados , Estudios Retrospectivos , Adulto Joven
19.
J Leukoc Biol ; 108(1): 73-81, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31965606

RESUMEN

Mast cells and eosinophils are innate immune cells involved in both acute and chronic inflammatory responses. Siglecs are a family of cell surface receptors that share sialic acid binding activity. Over the past 20 years, our knowledge of the expression and function of Siglecs on cells of the immune system and others has greatly expanded, as has our understanding of their signaling, ligands, and possible roles in disease pathophysiology. Because of this, Siglecs have garnered interest as potential drug targets using strategies ranging from biologics to ligand-directed nanoparticles. This mini-review will highlight the state of our knowledge regarding human eosinophil and mast cell Siglecs, their biology, what they recognize, tools developed for in vitro and preclinical experimentation, and the status of ongoing efforts to develop drugs that engage eosinophil and mast cell Siglecs for potential therapeutic benefit.


Asunto(s)
Sistemas de Liberación de Medicamentos , Eosinófilos/metabolismo , Mastocitos/metabolismo , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/metabolismo , Animales , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Humanos
20.
J Leukoc Biol ; 108(1): 43-58, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32134149

RESUMEN

Eosinophils are important multifunctional granulocytes. When studying eosinophil function and its contribution to diseases, mouse models are often used. Mouse eosinophils selectively express sialic acid-binding immunoglobulin-like lectin (Siglec)-F. Its closest functional paralog on human eosinophils is Siglec-8. These Siglecs are being used to target eosinophils when exploring their mechanistic roles in disease and for potential therapeutic benefit. In order to facilitate preclinical studies of human Siglec-8, we developed transgenic mouse strains expressing human Siglec-8 only on the surface of eosinophils with or without endogenous Siglec-F and have begun characterizing various cellular functions in vitro and in vivo. Eosinophils from Siglec-8+ mice, with or without Siglec-F, responded to Siglec-8 antibody engagement in vitro by up-regulating surface CD11b, whereas Siglec-F antibody had no such effect. Engagement of Siglec-F or Siglec-8 with respective antibodies in vitro resulted in only modest increases in cell death. Administration of rat Siglec-F antibodies to mice led to a significant decrease in Siglec-F surface expression on eosinophils due to internalization, and thus appeared to decrease eosinophil numbers based on Siglec-F+ cells, but with proper gaiting strategies did not in fact result in significant eosinophil depletion. In marked contrast, administration of mouse Siglec-8 antibodies rapidly and effectively depleted eosinophils from blood and spleens of mice, but an F(ab')2 version did not, indicating an Fc-mediated mechanism for eosinophil depletion in vivo. Siglec-8 expressing mice with or without endogenous Siglec-F will be useful to study Siglec-8-based therapeutics, and may be a preferred approach when acute or chronic eosinophil depletion is needed.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Eosinófilos/metabolismo , Animales , Anticuerpos/metabolismo , Anticuerpos/farmacología , Biomarcadores/metabolismo , Células de la Médula Ósea/citología , Antígeno CD11b/metabolismo , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Inyecciones , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Ácido N-Acetilneuramínico/metabolismo , Fenotipo , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico , Bazo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA