Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.334
Filtrar
Más filtros

Intervalo de año de publicación
1.
Nat Immunol ; 17(9): 1075-83, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27400149

RESUMEN

Adhesion and migration of T cells are controlled by chemokines and by adhesion molecules, especially integrins, and have critical roles in the normal physiological function of T lymphocytes. Using an RNA-mediated interference screen, we identified the WNK1 kinase as a regulator of both integrin-mediated adhesion and T cell migration. We found that WNK1 is a negative regulator of integrin-mediated adhesion, whereas it acts as a positive regulator of migration via the kinases OXSR1 and STK39 and the ion co-transporter SLC12A2. WNK1-deficient T cells home less efficiently to lymphoid organs and migrate more slowly through them. Our results reveal that a pathway previously known only to regulate salt homeostasis in the kidney functions to balance T cell adhesion and migration.


Asunto(s)
Adhesión Celular/genética , Movimiento Celular/genética , Antígenos de Histocompatibilidad Menor/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores Mensajeros de Linfocitos/metabolismo , Linfocitos T/fisiología , Animales , Células Cultivadas , Homeostasis , Transporte Iónico , Riñón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Antígenos de Histocompatibilidad Menor/genética , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , Receptores Mensajeros de Linfocitos/genética , Miembro 2 de la Familia de Transportadores de Soluto 12/metabolismo , Proteína Quinasa Deficiente en Lisina WNK 1
2.
Nat Immunol ; 16(2): 207-213, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25531831

RESUMEN

Lymphocyte recruitment maintains intestinal immune homeostasis but also contributes to inflammation. The orphan chemoattractant receptor GPR15 mediates regulatory T cell homing and immunosuppression in the mouse colon. We show that GPR15 is also expressed by mouse TH17 and TH1 effector cells and is required for colitis in a model that depends on the trafficking of these cells to the colon. In humans GPR15 is expressed by effector cells, including pathogenic TH2 cells in ulcerative colitis, but is expressed poorly or not at all by colon regulatory T (Treg) cells. The TH2 transcriptional activator GATA-3 and the Treg-associated transcriptional repressor FOXP3 robustly bind human, but not mouse, GPR15 enhancer sequences, correlating with receptor expression. Our results highlight species differences in GPR15 regulation and suggest it as a potential therapeutic target for colitis.


Asunto(s)
Colitis/fisiopatología , Colon/fisiopatología , Regulación de la Expresión Génica , Receptores Acoplados a Proteínas G/metabolismo , Receptores Mensajeros de Linfocitos/metabolismo , Receptores de Péptidos/metabolismo , Animales , Células Cultivadas , Colitis/inmunología , Colon/inmunología , Modelos Animales de Enfermedad , Elementos de Facilitación Genéticos/genética , Factores de Transcripción Forkhead/metabolismo , Técnicas de Inactivación de Genes , Humanos , Ratones , Unión Proteica , Receptores Acoplados a Proteínas G/genética , Receptores de Péptidos/genética , Especificidad de la Especie
3.
Nature ; 587(7835): 619-625, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33208946

RESUMEN

Although single-cell RNA sequencing studies have begun to provide compendia of cell expression profiles1-9, it has been difficult to systematically identify and localize all molecular cell types in individual organs to create a full molecular cell atlas. Here, using droplet- and plate-based single-cell RNA sequencing of approximately 75,000 human cells across all lung tissue compartments and circulating blood, combined with a multi-pronged cell annotation approach, we create an extensive cell atlas of the human lung. We define the gene expression profiles and anatomical locations of 58 cell populations in the human lung, including 41 out of 45 previously known cell types and 14 previously unknown ones. This comprehensive molecular atlas identifies the biochemical functions of lung cells and the transcription factors and markers for making and monitoring them; defines the cell targets of circulating hormones and predicts local signalling interactions and immune cell homing; and identifies cell types that are directly affected by lung disease genes and respiratory viruses. By comparing human and mouse data, we identified 17 molecular cell types that have been gained or lost during lung evolution and others with substantially altered expression profiles, revealing extensive plasticity of cell types and cell-type-specific gene expression during organ evolution including expression switches between cell types. This atlas provides the molecular foundation for investigating how lung cell identities, functions and interactions are achieved in development and tissue engineering and altered in disease and evolution.


Asunto(s)
Células/clasificación , Células/metabolismo , Inmunidad , Pulmón/citología , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Transcriptoma/genética , Anciano , Animales , Atlas como Asunto , Biomarcadores , Comunicación Celular , Células/inmunología , Quimiocinas/metabolismo , Células Endoteliales/metabolismo , Células Epiteliales/metabolismo , Femenino , Humanos , Pulmón/inmunología , Masculino , Ratones , Persona de Mediana Edad , Receptores Mensajeros de Linfocitos/metabolismo , Transducción de Señal , Células del Estroma/metabolismo
4.
Immunity ; 45(2): 442-56, 2016 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-27521270

RESUMEN

Depending on the tissue microenvironment, T cells can differentiate into highly diverse subsets expressing unique trafficking receptors and cytokines. Studies of human lymphocytes have primarily focused on a limited number of parameters in blood, representing an incomplete view of the human immune system. Here, we have utilized mass cytometry to simultaneously analyze T cell trafficking and functional markers across eight different human tissues, including blood, lymphoid, and non-lymphoid tissues. These data have revealed that combinatorial expression of trafficking receptors and cytokines better defines tissue specificity. Notably, we identified numerous T helper cell subsets with overlapping cytokine expression, but only specific cytokine combinations are secreted regardless of tissue type. This indicates that T cell lineages defined in mouse models cannot be clearly distinguished in humans. Overall, our data uncover a plethora of tissue immune signatures and provide a systemic map of how T cell phenotypes are altered throughout the human body.


Asunto(s)
Sangre/inmunología , Movimiento Celular , Tejido Linfoide/inmunología , Espectrometría de Masas/métodos , Especificidad de Órganos , Subgrupos de Linfocitos T/inmunología , Linfocitos T Colaboradores-Inductores/fisiología , Animales , Biodiversidad , Biomarcadores/metabolismo , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Citocinas/metabolismo , Humanos , Activación de Linfocitos , Ratones , Receptores Mensajeros de Linfocitos/metabolismo , Transcriptoma
5.
Allergy ; 79(1): 15-25, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37439317

RESUMEN

Circulating skin-homing cutaneous lymphocyte-associated antigen (CLA)+ T cells constitute a small subset of human memory T cells involved in several aspects of atopic dermatitis: Staphylococcus aureus related mechanisms, the abnormal Th2 immune response, biomarkers, clinical aspects of the patients, pruritus, and the mechanism of action of targeted therapies. Superantigens, IL-13, IL-31, pruritus, CCL17 and early effects on dupilumab-treated patients have in common that they are associated with the CLA+ T cell mechanisms in atopic dermatitis patients. The function of CLA+ T cells corresponds with the role of T cells belonging to the skin-associated lymphoid tissue and could be a reason why they reflect different mechanisms of atopic dermatitis and many other T cell mediated skin diseases. The goal of this review is to gather all this translational information of atopic dermatitis pathology.


Asunto(s)
Dermatitis Atópica , Humanos , Células T de Memoria , Subgrupos de Linfocitos T , Antígenos de Diferenciación de Linfocitos T , Glicoproteínas de Membrana , Receptores Mensajeros de Linfocitos , Piel/patología , Prurito , Antígenos de Neoplasias
6.
Immunity ; 42(6): 1087-99, 2015 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-26070483

RESUMEN

Effector-T-cell-mediated immunity depends on the efficient localization of antigen-primed lymphocytes to antigen-rich non-lymphoid tissue, which is facilitated by the expression of a unique set of "homing" receptors acquired by memory T cells. We report that engagement of the hepatocyte growth factor (HGF) receptor c-Met by heart-produced HGF during priming in the lymph nodes instructs T cell cardiotropism, which was associated with a specialized homing "signature" (c-Met(+)CCR4(+)CXCR3(+)). c-Met signals facilitated T cell recruitment to the heart via the chemokine receptor CCR5 by inducing autocrine CCR5 ligand release. c-Met triggering was sufficient to support cardiotropic T cell recirculation, while CCR4 and CXCR3 sustained recruitment during heart inflammation. Transient pharmacological blockade of c-Met during T cell priming led to enhanced survival of heart, but not skin, allografts associated with impaired localization of alloreactive T cells to heart grafts. These findings suggest c-Met as a target for development of organ-selective immunosuppressive therapies.


Asunto(s)
Rechazo de Injerto/prevención & control , Trasplante de Corazón , Corazón/fisiología , Factor de Crecimiento de Hepatocito/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Linfocitos T/fisiología , Animales , Comunicación Autocrina , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Células Cultivadas , Rechazo de Injerto/etiología , Rechazo de Injerto/genética , Humanos , Memoria Inmunológica , Indoles/farmacología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/genética , Ratones , Ratones SCID , Terapia Molecular Dirigida , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/genética , ARN Interferente Pequeño/genética , Receptores CCR5/metabolismo , Receptores de Quimiocina/metabolismo , Receptores Mensajeros de Linfocitos/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Sulfonas/farmacología , Linfocitos T/efectos de los fármacos
7.
J Virol ; 96(15): e0080422, 2022 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-35852355

RESUMEN

CD4dim CD8bright T cells are a mature population of CD8+ T cells that upon activation upregulate CD4 dimly on their surface. Expression of CD4 on these cells suggests that they can be an additional source of HIV neuroinvasion and persistence in the brain. We used HIV-infected NOD/SCID/IL-2rcγ-/- (NSG) humanized mice to track CD4dim CD8bright T cell homing to the brain and define their role in HIV dissemination into the brain. We report here that CD4dim CD8bright T cells are found in the brain at a median frequency of 2.6% and in the spleen at median frequency of 7.6% of CD3+ T cells. In the brain, 10 to 20% of CD4dim CD8bright T cells contain integrated provirus, which is infectious as demonstrated by viral outgrowth assay. CD4dim CD8bright T cells in the brain exhibited significantly higher expression of the brain homing receptors CX3CR1 and CXCR3 in comparison to their single-positive CD8+ T cell counterpart. Blocking lymphocyte trafficking into the brain of humanized mice via anti-VLA4 and anti-LFA1 antibodies reduced CD4dim CD8bright T cell trafficking into the brain by 60% and diminished brain HIV proviral DNA by 72%. Collectively, our findings demonstrate that CD4dim CD8bright T cells can home to the brain and support productive HIV replication. These studies also reveal for the first time that CD4dim CD8bright T cells are capable of HIV neuroinvasion and are a reservoir for HIV. IMPORTANCE We report here a seminal finding of a novel population of T cells, termed CD4dim CD8bright T cells, that plays a role in HIV neuroinvasion and is a reservoir for HIV in the brain.


Asunto(s)
Encéfalo , Antígenos CD4 , Antígenos CD8 , Linfocitos T CD8-positivos , Movimiento Celular , Infecciones por VIH , VIH-1 , Tropismo Viral , Animales , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/virología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Receptor 1 de Quimiocinas CX3C/metabolismo , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/patogenicidad , Humanos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Ratones , Ratones Endogámicos NOD , Ratones SCID , Provirus/genética , Provirus/aislamiento & purificación , Receptores CXCR3/metabolismo , Receptores Mensajeros de Linfocitos/metabolismo
8.
Nat Immunol ; 12(2): 178-85, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21217760

RESUMEN

Type 2 helper T cells (T(H)2) are critically involved in allergies and asthma. Here we demonstrate that extracellular matrix protein-1 (ECM1) is highly and selectively expressed in T(H)2 cells. ECM1 deficiency caused impaired T(H)2 responses and reduced allergic airway inflammation in vivo. Functional analysis demonstrated that although the T(H)2 polarization of ECM1-deficient cells was unimpaired, these cells had a defect in migration and were retained in peripheral lymphoid organs. This was associated with reduced expression of KLF2 and S1P(1). We also found that ECM1 could directly bind the interleukin-2 (IL-2) receptor to inhibit IL-2 signaling and activate S1P(1) expression. Our data identify a previously unknown function of ECM1 in regulating T(H)2 cell migration through control of KLF2 and S1P(1) expression.


Asunto(s)
Proteínas de la Matriz Extracelular/metabolismo , Hipersensibilidad/inmunología , Proteínas del Tejido Nervioso/metabolismo , Receptores Mensajeros de Linfocitos/metabolismo , Células Th2/metabolismo , Traslado Adoptivo , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Movimiento Celular/genética , Movimiento Celular/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/inmunología , Regulación de la Expresión Génica/inmunología , Humanos , Ganglios Linfáticos/patología , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Receptores Mensajeros de Linfocitos/genética , Receptores Mensajeros de Linfocitos/inmunología , Transducción de Señal/inmunología , Células Th2/inmunología , Células Th2/patología , Transgenes/genética
9.
Nat Immunol ; 12(2): 167-77, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21217759

RESUMEN

Mouse CCL8 is a CC chemokine of the monocyte chemoattractant protein (MCP) family whose biological activity and receptor usage have remained elusive. Here we show that CCL8 is highly expressed in the skin, where it serves as an agonist for the chemokine receptor CCR8 but not for CCR2. This distinguishes CCL8 from all other MCP chemokines. CCL8 responsiveness defined a population of highly differentiated, CCR8-expressing inflammatory T helper type 2 (T(H)2) cells enriched for interleukin (IL)-5. Ccr8- and Ccl8-deficient mice had markedly less eosinophilic inflammation than wild-type or Ccr4-deficient mice in a model of chronic atopic dermatitis. Adoptive transfer studies established CCR8 as a key regulator of T(H)2 cell recruitment into allergen-inflamed skin. In humans, CCR8 expression also defined an IL-5-enriched T(H)2 cell subset. The CCL8-CCR8 chemokine axis is therefore a crucial regulator of T(H)2 cell homing that drives IL-5-mediated chronic allergic inflammation.


Asunto(s)
Quimiocina CCL1/metabolismo , Quimiocina CCL8/metabolismo , Dermatitis Atópica/inmunología , Piel/patología , Células Th2/metabolismo , Traslado Adoptivo , Animales , Señalización del Calcio/inmunología , Células Cultivadas , Quimiocina CCL1/genética , Quimiocina CCL1/inmunología , Quimiocina CCL8/genética , Quimiocina CCL8/inmunología , Quimiotaxis/genética , Quimiotaxis/inmunología , Clonación Molecular , Modelos Animales de Enfermedad , Humanos , Interleucina-5/inmunología , Interleucina-5/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Mensajeros de Linfocitos/inmunología , Células Th2/inmunología , Células Th2/patología
10.
J Immunol ; 207(7): 1763-1775, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34470859

RESUMEN

Regulatory T cells (Tregs) reside in nonlymphoid tissues where they carry out unique functions. The molecular mechanisms responsible for Treg accumulation and maintenance in these tissues are relatively unknown. Using an unbiased discovery approach, we identified LAYN (layilin), a C-type lectin-like receptor, to be preferentially and highly expressed on a subset of activated Tregs in healthy and diseased human skin. Expression of layilin on Tregs was induced by TCR-mediated activation in the presence of IL-2 or TGF-ß. Mice with a conditional deletion of layilin in Tregs had reduced accumulation of these cells in tumors. However, these animals somewhat paradoxically had enhanced immune regulation in the tumor microenvironment, resulting in increased tumor growth. Mechanistically, layilin expression on Tregs had a minimal effect on their activation and suppressive capacity in vitro. However, expression of this molecule resulted in a cumulative anchoring effect on Treg dynamic motility in vivo. Taken together, our results suggest a model whereby layilin facilitates Treg adhesion in skin and, in doing so, limits their suppressive capacity. These findings uncover a unique mechanism whereby reduced Treg motility acts to limit immune regulation in nonlymphoid organs and may help guide strategies to exploit this phenomenon for therapeutic benefit.


Asunto(s)
Proteínas Portadoras/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores Mensajeros de Linfocitos/metabolismo , Piel/inmunología , Linfocitos T Reguladores/inmunología , Animales , Proteínas Portadoras/genética , Movimiento Celular , Células Cultivadas , Humanos , Tolerancia Inmunológica , Activación de Linfocitos , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Inmunológicos , Especificidad de Órganos , Receptores Mensajeros de Linfocitos/genética , Factor de Crecimiento Transformador beta/metabolismo
11.
J Immunol ; 207(9): 2245-2254, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34561227

RESUMEN

Targeting interactions between α4ß7 integrin and endothelial adhesion molecule MAdCAM-1 to inhibit lymphocyte migration to the gastrointestinal tract is an effective therapy in inflammatory bowel disease (IBD). Following lymphocyte entry into the mucosa, a subset of these cells expresses αEß7 integrin, which is expressed on proinflammatory lymphocytes, to increase cell retention. The factors governing lymphocyte migration into the intestinal mucosa and αE integrin expression in healthy subjects and IBD patients remain incompletely understood. We evaluated changes in factors involved in lymphocyte migration and differentiation within tissues. Both ileal and colonic tissue from active IBD patients showed upregulation of ICAM-1, VCAM-1, and MAdCAM-1 at the gene and protein levels compared with healthy subjects and/or inactive IBD patients. ß1 and ß7 integrin expression on circulating lymphocytes was similar across groups. TGF-ß1 treatment induced expression of αE on both ß7+ and ß7- T cells, suggesting that cells entering the mucosa independently of MAdCAM-1/α4ß7 can become αEß7+ ITGAE gene polymorphisms did not alter protein induction following TGF-ß1 stimulation. Increased phospho-SMAD3, which is directly downstream of TGF-ß, and increased TGF-ß-responsive gene expression were observed in the colonic mucosa of IBD patients. Finally, in vitro stimulation experiments showed that baseline ß7 expression had little effect on cytokine, chemokine, transcription factor, and effector molecule gene expression in αE+ and αE- T cells. These findings suggest cell migration to the gut mucosa may be altered in IBD and α4ß7-, and α4ß7+ T cells may upregulate αEß7 in response to TGF-ß once within the gut mucosa.


Asunto(s)
Antígenos CD/metabolismo , Enfermedades Inflamatorias del Intestino/inmunología , Cadenas alfa de Integrinas/metabolismo , Cadenas beta de Integrinas/metabolismo , Mucosa Intestinal/inmunología , Receptores Mensajeros de Linfocitos/metabolismo , Linfocitos T/inmunología , Adulto , Anciano , Movimiento Celular , Femenino , Humanos , Cadenas beta de Integrinas/genética , Masculino , Persona de Mediana Edad , Transducción de Señal , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
12.
Int J Mol Sci ; 24(11)2023 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-37298541

RESUMEN

Integrin receptors mediate cell-cell interactions via the recognition of cell-adhesion glycoproteins, as well as via the interactions of cells with proteins of the extracellular matrix, and upon activation they transduce signals bi-directionally across the cell membrane. In the case of injury, infection, or inflammation, integrins of ß2 and α4 families participate in the recruitment of leukocytes, a multi-step process initiated by the capturing of rolling leukocytes and terminated by their extravasation. In particular, α4ß1 integrin is deeply involved in leukocyte firm adhesion preceding extravasation. Besides its well-known role in inflammatory diseases, α4ß1 integrin is also involved in cancer, being expressed in various tumors and showing an important role in cancer formation and spreading. Hence, targeting this integrin represents an opportunity for the treatment of inflammatory disorders, some autoimmune diseases, and cancer. In this context, taking inspiration from the recognition motives of α4ß1 integrin with its natural ligands FN and VCAM-1, we designed minimalist α/ß hybrid peptide ligands, with our approach being associated with a retro strategy. These modifications are expected to improve the compounds' stability and bioavailability. As it turned out, some of the ligands were found to be antagonists, being able to inhibit the adhesion of integrin-expressing cells to plates coated with the natural ligands without inducing any conformational switch and any activation of intracellular signaling pathways. An original model structure of the receptor was generated using protein-protein docking to evaluate the bioactive conformations of the antagonists via molecular docking. Since the experimental structure of α4ß1 integrin is still unknown, the simulations might also shed light on the interactions between the receptor and its native protein ligands.


Asunto(s)
Neoplasias , Peptidomiméticos , Humanos , Integrina alfa4beta1/metabolismo , Receptores Mensajeros de Linfocitos/metabolismo , Simulación del Acoplamiento Molecular , Peptidomiméticos/farmacología , Integrina beta1 , Ligandos , Integrinas/metabolismo , Adhesión Celular , Molécula 1 de Adhesión Celular Vascular/metabolismo
13.
Eur J Immunol ; 51(1): 27-38, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33301176

RESUMEN

Regulatory T cells (Tregs) have a critical role in maintaining self-tolerance and immune homeostasis. There is much interest in using Tregs as a cell therapy to re-establish tolerance in conditions such as inflammatory bowel disease and type 1 diabetes, with many ongoing clinical studies testing the safety and efficacy of this approach. Manufacturing of Tregs for therapy typically involves ex vivo expansion to obtain sufficient cell numbers for infusion and comes with the risk of altering the activity of key biological processes. However, this process also offers an opportunity to tailor Treg function to maximize in vivo activity. In this review, we focus on the roles of antigen-presenting cells (APCs) in the generation and function of Tregs in humans. In addition to stimulating the development of Tregs, APCs activate Tregs and provide signals that induce specialized functional and homing marker expression. Cross talk between Tregs and APCs is a critical, often under-appreciated, aspect of Treg biology, with APCs mediating the key properties of infectious tolerance and bystander suppression. Understanding the biology of human Treg-APC interactions will reveal new ways to optimize Treg-based therapeutic approaches.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Linfocitos T Reguladores/inmunología , Diferenciación Celular , Humanos , Tolerancia Inmunológica , Sinapsis Inmunológicas , Inmunoterapia Adoptiva , Activación de Linfocitos , Modelos Inmunológicos , Receptor Cross-Talk/inmunología , Receptores Mensajeros de Linfocitos/inmunología , Autotolerancia , Biología Sintética , Linfocitos T Reguladores/citología , Timo/citología , Timo/inmunología
15.
Allergy ; 77(9): 2737-2747, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35255168

RESUMEN

BACKGROUND: Atopic dermatitis (AD) and psoriasis represent two of the most common inflammatory skin diseases in developed countries. A hallmark of both diseases is T-cell infiltration into the skin. However, it is still not clarified to what extent these infiltrating T cells are antigen-specific skin-homing T cells or unspecific heterogeneous bystander cells. METHODS: To elucidate this, T cells from lesional skin and from blood of 9 AD and 10 psoriasis patients were compared by receptor (TCR) sequencing. Therefore, peripheral blood mononuclear cells (PBMC) were cell-sorted according to expression of the cutaneous leukocyte antigen (CLA) into skin-homing (CLA+ ) and non-skin-homing (CLA- ) subfractions. Aeroallergen-specific T-cell lines were grown from AD patients' PBMC in parallel. RESULTS: Intra-individual comparison of TCRB CDR3 regions revealed that clonally expanded T cells in skin lesions of both AD and psoriasis patients corresponded to skin-homing circulating T cells. However, in psoriasis patients, these T-cell clones were also detectable to a larger extent among CLA- circulating T cells. Up to 28% of infiltrating cells in AD skin were identified as allergen-specific by overlapping TCR sequences. CONCLUSIONS: Our data show that in line with the systemic nature of psoriasis, T-cell clones that infiltrate psoriatic skin lesions do not exclusively possess skin-homing ability and are therefore most probably specific to antigens that are not exclusively expressed or located in the skin. T cells driving AD skin inflammation appear to home nearly exclusively to the skin and are, to a certain extent, specific to aeroallergens.


Asunto(s)
Dermatitis Atópica , Psoriasis , Alérgenos , Antígenos de Diferenciación de Linfocitos T , Antígenos de Neoplasias , Humanos , Leucocitos Mononucleares/metabolismo , Glicoproteínas de Membrana , Receptores de Antígenos de Linfocitos T/genética , Receptores Mensajeros de Linfocitos
16.
Cell Mol Life Sci ; 78(21-22): 6963-6978, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34586443

RESUMEN

The endogenous chemokines CCL19 and CCL21 signal via their common receptor CCR7. CCL21 is the main lymph node homing chemokine, but a weak chemo-attractant compared to CCL19. Here we show that the 41-amino acid positively charged peptide, released through C-terminal cleavage of CCL21, C21TP, boosts the immune cell recruiting activity of CCL21 by up to 25-fold and the signaling activity via CCR7 by ~ 100-fold. Such boosting is unprecedented. Despite the presence of multiple basic glycosaminoglycan (GAG) binding motifs, C21TP boosting of CCL21 signaling does not involve interference with GAG mediated cell-surface retention. Instead, boosting is directly dependent on O-glycosylations in the CCR7 N-terminus. As dictated by the two-step binding model, the initial chemokine binding involves interaction of the chemokine fold with the receptor N-terminus, followed by insertion of the chemokine N-terminus deep into the receptor binding pocket. Our data suggest that apart from a role in initial chemokine binding, the receptor N-terminus also partakes in a gating mechanism, which could give rise to a reduced ligand activity, presumably through affecting the ligand positioning. Based on experiments that support a direct interaction of C21TP with the glycosylated CCR7 N-terminus, we propose that electrostatic interactions between the positively charged peptide and sialylated O-glycans in CCR7 N-terminus may create a more accessible version of the receptor and thus guide chemokine docking to generate a more favorable chemokine-receptor interaction, giving rise to the peptide boosting effect.


Asunto(s)
Quimiocina CCL21/metabolismo , Células Dendríticas/metabolismo , Ganglios Linfáticos/metabolismo , Receptores CCR7/metabolismo , Receptores Mensajeros de Linfocitos/metabolismo , Transducción de Señal/fisiología , Animales , Células CHO , Células Cultivadas , Cricetulus , Glicosilación , Humanos , Ligandos , Péptidos/metabolismo , Unión Proteica/fisiología , Electricidad Estática
17.
Exp Dermatol ; 30(1): 84-91, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33010063

RESUMEN

Recently, increasing attention has been paid to senescence-associated secretory phenotype (SASP), a phenomenon that senescent cells secrete molecules such as inflammatory cytokines and matrix metalloproteinases (MMPs), due to its noxious effects on the surrounding tissue. Senescent cells in the blood and liver are known to be properly depleted by macrophages. In the dermis, accumulation of senescent cells has been reported and is thought to be involved with skin ageing. In this study, to elucidate the clearance mechanism of senescent cells in the dermis, we focused on macrophage functions. Our co-culture experiments of senescent fibroblasts and macrophages revealed a two-step clearance mechanism: first, TNF-α secreted from macrophages induces apoptosis in senescent fibroblasts, and then, dead cells are phagocytosed by macrophages. Furthermore, it was suggested that SASP factors suppress both of the two steps of the senescent cell clearance by macrophages. From these findings, normally senescent cells in the dermis are thought to be removed by macrophages, but when senescent cells are excessively accumulated owing to oxidative stress, ultraviolet (UV) ray or other reasons, SASP was suggested to suppress the macrophage-dependent clearance functions and thereby cause further accumulation of senescent cells.


Asunto(s)
Fibroblastos/fisiología , Macrófagos/fisiología , Fenotipo Secretor Asociado a la Senescencia , Adulto , Anciano , Antígenos CD/genética , Antígenos de Diferenciación Mielomonocítica/genética , Apoptosis/efectos de los fármacos , Moléculas de Adhesión Celular Neuronal/genética , Línea Celular , Polaridad Celular , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Medios de Cultivo Condicionados/farmacología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Dermis/citología , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Infliximab/farmacología , Masculino , Fagocitosis , ARN/metabolismo , Receptores CCR7/genética , Receptores de Superficie Celular/genética , Receptores Mensajeros de Linfocitos/genética , Proteína de Unión al Calcio S100A4/metabolismo , Inhibidores del Factor de Necrosis Tumoral/farmacología , Adulto Joven
18.
J Immunol ; 202(12): 3447-3457, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-31053627

RESUMEN

Current therapies for gut inflammation have not reached the desired specificity and are attended by unintended immune suppression. This study aimed to provide evidence for supporting a hypothesis that direct in vivo augmentation of the induction of gut-homing regulatory T (Treg) cells is a strategy of expected specificity for the treatment of chronic intestinal inflammation (e.g., inflammatory bowel disease). We showed that dendritic cells (DCs), engineered to de novo produce high concentrations of both 1,25-dihydroxyvitamin D, the active vitamin D metabolite, and retinoic acid, an active vitamin A metabolite, augmented the induction of T cells that express both the regulatory molecule Foxp3 and the gut-homing receptor CCR9 in vitro and in vivo. In vivo, the newly generated Ag-specific Foxp3+ T cells homed to intestines. Additionally, transfer of such engineered DCs robustly suppressed ongoing experimental colitis. Moreover, CD4+ T cells from spleens of the mice transferred with the engineered DCs suppressed experimental colitis in syngeneic hosts. The data suggest that the engineered DCs enhance regulatory function in CD4+ T cell population in peripheral lymphoid tissues. Finally, we showed that colitis suppression following in vivo transfer of the engineered DCs was significantly reduced when Foxp3+ Treg cells were depleted. The data indicate that maximal colitis suppression mediated by the engineered DCs requires Treg cells. Collectively, our data support that DCs de novo overproducing both 1,25-dihydroxyvitamin D and retinoic acid are a promising novel therapy for chronic intestinal inflammation.


Asunto(s)
Colitis/terapia , Células Dendríticas/fisiología , Enfermedades Inflamatorias del Intestino/terapia , Intestinos/inmunología , Receptores CCR/metabolismo , Receptores Mensajeros de Linfocitos/metabolismo , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Células Cultivadas , Colitis/inmunología , Células Dendríticas/trasplante , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/metabolismo , Humanos , Terapia de Inmunosupresión , Enfermedades Inflamatorias del Intestino/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Linfocitos T Reguladores/trasplante , Tretinoina/metabolismo , Vitamina D/análogos & derivados , Vitamina D/metabolismo
19.
Semin Immunol ; 31: 3-10, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28802693

RESUMEN

Although natural killer (NK) cells were initially named for their spontaneous tumor-killing capacity, their concept has been greatly expanded with more than 40 years of extensive investigation. Currently, NK cells are known as a heterogeneous population of innate lymphoid cell (ILC) family, consisting of different subsets with unique phenotypic and functional features. Recent studies have shown that tissue-resident NK (trNK) cells, which are distinct from conventional NK (cNK) cells, preferentially distribute in non-lymphoid tissues, such as the liver, uterus, salivary gland, and adipose. In this review, we provide a comprehensive overview of the current knowledge about the phenotype, function and development of trNK cells across different tissues and describe the similarities and differences between diverse trNK cells and cNK cells, with a particular focus on the tissue-specific characteristics of different trNK cells.


Asunto(s)
Células Asesinas Naturales/inmunología , Subgrupos Linfocitarios/inmunología , Especificidad de Órganos , Animales , Biodiversidad , Movimiento Celular , Humanos , Inmunidad Innata , Fenotipo , Receptores Mensajeros de Linfocitos/metabolismo
20.
Br J Cancer ; 123(4): 501-509, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32595212

RESUMEN

Clever-1 (also known as Stabilin-1 and FEEL-1) is a scavenger receptor expressed on lymphatic endothelial cells, sinusoidal endothelial cells and immunosuppressive monocytes and macrophages. Its role in cancer growth and spread first became evident in Stab1-/- knockout mice, which have smaller primary tumours and metastases. Subsequent studies in mice and humans have shown that immunotherapeutic blockade of Clever-1 can activate T-cell responses, and that this response is mainly mediated by a phenotypic change in macrophages and monocytes from immunosuppressive to pro-inflammatory following Clever-1 inhibition. Analyses of human cancer cohorts have revealed marked associations between the number of Clever-1-positive macrophages and patient outcome. As hardly any reports to date have addressed the role of Clever-1 in immunotherapy resistance and T-cell dysfunction, we performed data mining using several published cancer cohorts, and observed a remarkable correlation between Clever-1 positivity and resistance to immune checkpoint therapies. This result provides impetus and potential for the ongoing clinical trial targeting Clever-1 in solid tumours, which has so far shown a shift towards immune activation when a particular epitope of Clever-1 is blocked.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/genética , Resistencia a Antineoplásicos , Neoplasias/genética , Receptores Mensajeros de Linfocitos/genética , Animales , Moléculas de Adhesión Celular Neuronal/metabolismo , Progresión de la Enfermedad , Humanos , Inmunoterapia , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Receptores Mensajeros de Linfocitos/metabolismo , Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA