RESUMEN
The continuous migration of immune cells between lymphoid and nonlymphoid organs is a key feature of the immune system, facilitating the distribution of effector cells within nearly all compartments of the body. Furthermore, reaching their correct position within primary, secondary, or tertiary lymphoid organs is a prerequisite to ensure immune cells' unimpaired differentiation, maturation, and selection, as well as their activation or functional silencing. The superfamilies of chemokines and chemokine receptors are of major importance in guiding immune cells to and within lymphoid and nonlymphoid tissues. In this review we focus on the role of the chemokine system in the migration dynamics of immune cells within lymphoid organs at the steady state and on how these dynamics are affected by infectious and inflammatory processes.
Asunto(s)
Quimiocinas/inmunología , Sistema Inmunológico , Infecciones/inmunología , Inflamación/inmunología , Linfocitos/inmunología , Tejido Linfoide/inmunología , Receptores de Quimiocina/inmunología , Animales , Comunicación Celular , Movimiento Celular , Humanos , Activación de LinfocitosRESUMEN
Carcinoma-associated fibroblasts (CAFs) are abundant and heterogeneous stromal cells in tumor microenvironment that are critically involved in cancer progression. Here, we demonstrate that two cell-surface molecules, CD10 and GPR77, specifically define a CAF subset correlated with chemoresistance and poor survival in multiple cohorts of breast and lung cancer patients. CD10+GPR77+ CAFs promote tumor formation and chemoresistance by providing a survival niche for cancer stem cells (CSCs). Mechanistically, CD10+GPR77+ CAFs are driven by persistent NF-κB activation via p65 phosphorylation and acetylation, which is maintained by complement signaling via GPR77, a C5a receptor. Furthermore, CD10+GPR77+ CAFs promote successful engraftment of patient-derived xenografts (PDXs), and targeting these CAFs with a neutralizing anti-GPR77 antibody abolishes tumor formation and restores tumor chemosensitivity. Our study reveals a functional CAF subset that can be defined and isolated by specific cell-surface markers and suggests that targeting the CD10+GPR77+ CAF subset could be an effective therapeutic strategy against CSC-driven solid tumors.
Asunto(s)
Transformación Celular Neoplásica/inmunología , Resistencia a Antineoplásicos/inmunología , Fibroblastos/inmunología , Neoplasias/inmunología , Células Madre Neoplásicas/inmunología , Neprilisina/inmunología , Receptores de Quimiocina/inmunología , Microambiente Tumoral/inmunología , Células A549 , Transformación Celular Neoplásica/patología , Fibroblastos/patología , Humanos , Células MCF-7 , Proteínas de Neoplasias/inmunología , Neoplasias/patología , Células Madre Neoplásicas/patología , Receptor de Anafilatoxina C5aRESUMEN
Neutrophils are expanded and abundant in cancer-bearing hosts. Under the influence of CXCR1 and CXCR2 chemokine receptor agonists and other chemotactic factors produced by tumors, neutrophils, and granulocytic myeloid-derived suppressor cells (MDSCs) from cancer patients extrude their neutrophil extracellular traps (NETs). In our hands, CXCR1 and CXCR2 agonists proved to be the major mediators of cancer-promoted NETosis. NETs wrap and coat tumor cells and shield them from cytotoxicity, as mediated by CD8+ T cells and natural killer (NK) cells, by obstructing contact between immune cells and the surrounding target cells. Tumor cells protected from cytotoxicity by NETs underlie successful cancer metastases in mice and the immunotherapeutic synergy of protein arginine deiminase 4 (PAD4) inhibitors, which curtail NETosis with immune checkpoint inhibitors. Intravital microscopy provides evidence of neutrophil NETs interfering cytolytic cytotoxic T lymphocytes (CTLs) and NK cell contacts with tumor cells.
Asunto(s)
Trampas Extracelulares/metabolismo , Neoplasias Experimentales/terapia , Receptores de Quimiocina/agonistas , Receptores de Interleucina-8A/agonistas , Receptores de Interleucina-8B/agonistas , Animales , Línea Celular Tumoral , Citotoxicidad Inmunológica/inmunología , Células HT29 , Humanos , Microscopía Intravital/métodos , Células Asesinas Naturales/inmunología , Ligandos , Ratones , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/metabolismo , Receptores de Quimiocina/inmunología , Receptores de Quimiocina/metabolismo , Receptores de Interleucina-8A/inmunología , Receptores de Interleucina-8A/metabolismo , Receptores de Interleucina-8B/inmunología , Receptores de Interleucina-8B/metabolismo , Linfocitos T Citotóxicos/inmunologíaRESUMEN
Dendritic cells (DCs) that orchestrate mucosal immunity have been studied in mice. Here we characterized human gut DC populations and defined their relationship to previously studied human and mouse DCs. CD103(+)Sirpα(-) DCs were related to human blood CD141(+) DCs and to mouse intestinal CD103(+)CD11b(-) DCs and expressed markers of cross-presenting DCs. CD103(+)Sirpα(+) DCs aligned with human blood CD1c(+) DCs and mouse intestinal CD103(+)CD11b(+) DCs and supported the induction of regulatory T cells. Both CD103(+) DC subsets induced the TH17 subset of helper T cells, while CD103(-)Sirpα(+) DCs induced the TH1 subset of helper T cells. Comparative analysis of transcriptomes revealed conserved transcriptional programs among CD103(+) DC subsets and identified a selective role for the transcriptional repressors Bcl-6 and Blimp-1 in the specification of CD103(+)CD11b(-) DCs and intestinal CD103(+)CD11b(+) DCs, respectively. Our results highlight evolutionarily conserved and divergent programming of intestinal DCs.
Asunto(s)
Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Mucosa Intestinal/inmunología , Transcriptoma/inmunología , Animales , Antígenos CD/inmunología , Antígenos CD/metabolismo , Antígenos CD1/inmunología , Antígenos CD1/metabolismo , Antígeno CD11b/inmunología , Antígeno CD11b/metabolismo , Diferenciación Celular/genética , Células Cultivadas , Análisis por Conglomerados , Reactividad Cruzada/genética , Reactividad Cruzada/inmunología , Células Dendríticas/metabolismo , Citometría de Flujo , Glicoproteínas/inmunología , Glicoproteínas/metabolismo , Humanos , Cadenas alfa de Integrinas/inmunología , Cadenas alfa de Integrinas/metabolismo , Integrinas/genética , Integrinas/inmunología , Ratones , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores de Quimiocina/genética , Receptores de Quimiocina/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Transcriptoma/genéticaRESUMEN
The paradigm that macrophages that reside in steady-state tissues are derived from embryonic precursors has never been investigated in the intestine, which contains the largest pool of macrophages. Using fate-mapping models and monocytopenic mice, together with bone marrow chimera and parabiotic models, we found that embryonic precursor cells seeded the intestinal mucosa and demonstrated extensive in situ proliferation during the neonatal period. However, these cells did not persist in the intestine of adult mice. Instead, they were replaced around the time of weaning by the chemokine receptor CCR2-dependent influx of Ly6C(hi) monocytes that differentiated locally into mature, anti-inflammatory macrophages. This process was driven largely by the microbiota and had to be continued throughout adult life to maintain a normal intestinal macrophage pool.
Asunto(s)
Mucosa Intestinal/inmunología , Intestinos/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Animales , Animales Recién Nacidos , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/inmunología , Antígenos de Diferenciación/metabolismo , Antígenos Ly/inmunología , Antígenos Ly/metabolismo , Trasplante de Médula Ósea , Antígeno CD11b/genética , Antígeno CD11b/inmunología , Antígeno CD11b/metabolismo , Receptor 1 de Quimiocinas CX3C , Diferenciación Celular/inmunología , Proliferación Celular , Citometría de Flujo , Expresión Génica/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Intestinos/citología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Inmunológicos , Monocitos/metabolismo , Parabiosis , Receptores CCR2/genética , Receptores CCR2/inmunología , Receptores CCR2/metabolismo , Receptores de Quimiocina/genética , Receptores de Quimiocina/inmunología , Receptores de Quimiocina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de TiempoRESUMEN
Respiratory syncytial virus (RSV) is the major cause of lower respiratory tract infections in infants and is characterized by pulmonary infiltration of B cells in fatal cases. We analyzed the B cell compartment in human newborns and identified a population of neonatal regulatory B lymphocytes (nBreg cells) that produced interleukin 10 (IL-10) in response to RSV infection. The polyreactive B cell receptor of nBreg cells interacted with RSV protein F and induced upregulation of chemokine receptor CX3CR1. CX3CR1 interacted with RSV glycoprotein G, leading to nBreg cell infection and IL-10 production that dampened T helper 1 (Th1) cytokine production. In the respiratory tract of neonates with severe RSV-induced acute bronchiolitis, RSV-infected nBreg cell frequencies correlated with increased viral load and decreased blood memory Th1 cell frequencies. Thus, the frequency of nBreg cells is predictive of the severity of acute bronchiolitis disease and nBreg cell activity may constitute an early-life host response that favors microbial pathogenesis.
Asunto(s)
Linfocitos B Reguladores/inmunología , Bronquiolitis Viral/inmunología , Receptores de Quimiocina/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Linfocitos B Reguladores/virología , Bronquiolitis Viral/patología , Linfocitos T CD4-Positivos/inmunología , Receptor 1 de Quimiocinas CX3C , Ensayo de Inmunoadsorción Enzimática , Ensayo de Immunospot Ligado a Enzimas , Perfilación de la Expresión Génica , Humanos , Recién Nacido , Activación de Linfocitos/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , Infecciones por Virus Sincitial Respiratorio/patología , Virus Sincitiales Respiratorios , TranscriptomaRESUMEN
Signaling through the G protein-coupled receptors for the complement fragments C3a and C5a (C3aR and C5aR, respectively) by dendritic cells and CD4(+) cells provides costimulatory and survival signals to effector T cells. Here we found that when signals from C3aR and C5aR were not transduced into CD4(+) cells, signaling via the kinases PI(3)Kγ, Akt and mTOR ceased, activation of the kinase PKA increased, autoinductive signaling by transforming growth factor-ß1 (TGF-ß1) initiated and CD4(+) T cells became Foxp3(+) induced regulatory T cells (iT(reg) cells). Endogenous TGF-ß1 suppressed signaling through C3aR and C5aR by preventing the production of C3a and C5a and upregulating C5L2, an alternative receptor for C5a. The absence of signaling via C3aR and C5aR resulted in lower expression of costimulatory molecules and interleukin 6 (IL-6) and more production of IL-10. The resulting iT(reg) cells exerted robust suppression, had enhanced stability and suppressed ongoing autoimmune disease. Antagonism of C3aR and C5aR can also induce functional human iT(reg) cells.
Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Receptor de Anafilatoxina C5a/metabolismo , Receptores de Complemento/metabolismo , Transducción de Señal/inmunología , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Comunicación Celular/inmunología , Diferenciación Celular , Fosfatidilinositol 3-Quinasa Clase Ib/inmunología , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Complemento C3a/inmunología , Complemento C3a/metabolismo , Complemento C5a/inmunología , Complemento C5a/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/inmunología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Factores de Transcripción Forkhead/inmunología , Regulación de la Expresión Génica , Interleucina-10/inmunología , Interleucina-10/metabolismo , Interleucina-6/inmunología , Interleucina-6/metabolismo , Ratones , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-akt/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor Cross-Talk/inmunología , Receptor de Anafilatoxina C5a/inmunología , Receptores de Quimiocina/inmunología , Receptores de Quimiocina/metabolismo , Receptores de Complemento/inmunología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Serina-Treonina Quinasas TOR/inmunología , Serina-Treonina Quinasas TOR/metabolismo , Factor de Crecimiento Transformador beta1/inmunologíaRESUMEN
Infections induce pathogen-specific T cell differentiation into diverse effectors (Teff) that give rise to memory (Tmem) subsets. The cell-fate decisions and lineage relationships that underlie these transitions are poorly understood. Here, we found that the chemokine receptor CX3CR1 identifies three distinct CD8+ Teff and Tmem subsets. Classical central (Tcm) and effector memory (Tem) cells and their corresponding Teff precursors were CX3CR1- and CX3CR1high, respectively. Viral infection also induced a numerically stable CX3CR1int subset that represented â¼15% of blood-borne Tmem cells. CX3CR1int Tmem cells underwent more frequent homeostatic divisions than other Tmem subsets and not only self-renewed, but also contributed to the expanding CX3CR1- Tcm pool. Both Tcm and CX3CR1int cells homed to lymph nodes, but CX3CR1int cells, and not Tem cells, predominantly surveyed peripheral tissues. As CX3CR1int Tmem cells present unique phenotypic, homeostatic, and migratory properties, we designate this subset peripheral memory (tpm) cells and propose that tpm cells are chiefly responsible for the global surveillance of non-lymphoid tissues.
Asunto(s)
Linfocitos T CD8-positivos/inmunología , Homeostasis/inmunología , Vigilancia Inmunológica/inmunología , Receptores de Quimiocina/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Receptor 1 de Quimiocinas CX3C , Separación Celular , Citometría de Flujo , Memoria Inmunológica/inmunología , Ratones , Ratones Endogámicos C57BLRESUMEN
The dynamics of when and where CD4(+) T cells provide help for CD8(+) T cell priming and which dendritic cells (DCs) activate CD4(+) T cells in vivo after localized infection are poorly understood. By using a cutaneous herpes simplex virus infection model combined with intravital 2-photon imaging of the draining lymph node (LN) to concurrently visualize pathogen-specific CD4(+) and CD8(+) T cells, we found that early priming of CD4(+) T cells involved clustering with migratory skin DCs. CD8(+) T cells did not interact with migratory DCs and their activation was delayed, requiring later clustering interactions with LN-resident XCR1(+) DCs. CD4(+) T cells interacted with these late CD8(+) T cell clusters on resident XCR1(+) DCs. Together, these data reveal asynchronous T cell activation by distinct DC subsets and highlight the key role of XCR1(+) DCs as the central platform for cytotoxic T lymphocyte activation and the delivery of CD4(+) T cell help.
Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Comunicación Celular/inmunología , Células Dendríticas/inmunología , Ganglios Linfáticos/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Movimiento Celular/inmunología , Células Dendríticas/metabolismo , Citometría de Flujo , Colorantes Fluorescentes/química , Herpes Simple/inmunología , Herpes Simple/metabolismo , Herpes Simple/virología , Interacciones Huésped-Patógeno/inmunología , Ganglios Linfáticos/citología , Ganglios Linfáticos/virología , Activación de Linfocitos/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Microscopía de Fluorescencia por Excitación Multifotónica , Receptores de Quimiocina/inmunología , Receptores de Quimiocina/metabolismo , Rodaminas/química , Simplexvirus/inmunología , Simplexvirus/fisiologíaRESUMEN
Mutations in MECP2, encoding the epigenetic regulator methyl-CpG-binding protein 2, are the predominant cause of Rett syndrome, a disease characterized by both neurological symptoms and systemic abnormalities. Microglial dysfunction is thought to contribute to disease pathogenesis, and here we found microglia become activated and subsequently lost with disease progression in Mecp2-null mice. Mecp2 was found to be expressed in peripheral macrophage and monocyte populations, several of which also became depleted in Mecp2-null mice. RNA-seq revealed increased expression of glucocorticoid- and hypoxia-induced transcripts in Mecp2-deficient microglia and peritoneal macrophages. Furthermore, Mecp2 was found to regulate inflammatory gene transcription in response to TNF stimulation. Postnatal re-expression of Mecp2 using Cx3cr1(creER) increased the lifespan of otherwise Mecp2-null mice. These data suggest that Mecp2 regulates microglia and macrophage responsiveness to environmental stimuli to promote homeostasis. Dysfunction of tissue-resident macrophages might contribute to the systemic pathologies observed in Rett syndrome.
Asunto(s)
Islas de CpG/inmunología , Epigénesis Genética , Macrófagos Peritoneales/inmunología , Proteína 2 de Unión a Metil-CpG/inmunología , Microglía/inmunología , Síndrome de Rett/inmunología , Animales , Receptor 1 de Quimiocinas CX3C , Metilación de ADN , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Homeostasis/inmunología , Humanos , Integrasas/genética , Integrasas/inmunología , Longevidad/inmunología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/patología , Masculino , Proteína 2 de Unión a Metil-CpG/deficiencia , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/efectos de los fármacos , Microglía/patología , Receptores de Quimiocina/genética , Receptores de Quimiocina/inmunología , Síndrome de Rett/genética , Síndrome de Rett/patología , Transducción de Señal , Factor de Necrosis Tumoral alfa/farmacologíaRESUMEN
About 500 million years ago, a new type of adaptive immune defense emerged in basal jawed vertebrates, accompanied by morphological innovations, including the thymus. Did these evolutionary novelties arise de novo or from elaboration of ancient genetic networks? We reconstructed the genetic changes underlying thymopoiesis by comparative genome and expression analyses in chordates and basal vertebrates. The derived models of genetic networks were experimentally verified in bony fishes. Ancestral networks defining circumscribed regions of the pharyngeal epithelium of jawless vertebrates expanded in cartilaginous fishes to incorporate novel genes, notably those encoding chemokines. Correspondingly, novel networks evolved in lymphocytes of jawed vertebrates to control the expression of additional chemokine receptors. These complementary changes enabled unprecedented Delta/Notch signaling between pharyngeal epithelium and lymphoid cells that was exploited for specification to the T cell lineage. Our results provide a framework elucidating the evolution of key features of the adaptive immune system in jawed vertebrates.
Asunto(s)
Evolución Biológica , Redes Reguladoras de Genes , Timo/inmunología , Vertebrados/genética , Vertebrados/inmunología , Animales , Quimiocinas/genética , Quimiocinas/inmunología , Cordados no Vertebrados/genética , Cordados no Vertebrados/inmunología , Peces/genética , Peces/inmunología , Humanos , Lampreas/genética , Lampreas/inmunología , Linfocitos/inmunología , Datos de Secuencia Molecular , Receptores de Quimiocina/genética , Receptores de Quimiocina/inmunologíaRESUMEN
The relationship between dendritic cells (DCs) and macrophages is often debated. Here we ask whether steady-state, lymphoid-tissue-resident conventional DCs (cDCs), plasmacytoid DCs (pDCs), and macrophages share a common macrophage-DC-restricted precursor (MDP). Using new clonal culture assays combined with adoptive transfer, we found that MDP fractions isolated by previous strategies are dominated by precursors of macrophages and monocytes, include some multipotent precursors of other hematopoietic lineages, but contain few precursors of resident cDCs and pDCs and no detectable common precursors restricted to these DC types and macrophages. Overall we find no evidence for a common restricted MDP leading to both macrophages and FL-dependent, resident cDCs and pDCs.
Asunto(s)
Linaje de la Célula/inmunología , Células Dendríticas/citología , Tejido Linfoide/citología , Macrófagos/citología , Células Precursoras de Monocitos y Macrófagos/citología , Traslado Adoptivo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Receptor 1 de Quimiocinas CX3C , Diferenciación Celular/inmunología , Células Cultivadas , Citocinas/biosíntesis , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Granulocitos/citología , Granulocitos/inmunología , Factor Estimulante de Colonias de Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Células Precursoras de Monocitos y Macrófagos/inmunología , Monocitos/citología , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Receptores de Quimiocina/inmunologíaRESUMEN
Interleukin-22 (IL-22) plays a critical role in mucosal defense, although the molecular mechanisms that ensure IL-22 tissue distribution remain poorly understood. We show that the CXCL16-CXCR6 chemokine-chemokine receptor axis regulated group 3 innate lymphoid cell (ILC3) diversity and function. CXCL16 was constitutively expressed by CX3CR1(+) intestinal dendritic cells (DCs) and coexpressed with IL-23 after Citrobacter rodentium infection. Intestinal ILC3s expressed CXCR6 and its ablation generated a selective loss of the NKp46(+) ILC3 subset, a depletion of intestinal IL-22, and the inability to control C. rodentium infection. CD4(+) ILC3s were unaffected by CXCR6 deficiency and remained clustered within lymphoid follicles. In contrast, the lamina propria of Cxcr6(-/-) mice was devoid of ILC3s. The loss of ILC3-dependent IL-22 epithelial stimulation reduced antimicrobial peptide expression that explained the sensitivity of Cxcr6(-/-) mice to C. rodentium. Our results delineate a critical CXCL16-CXCR6 crosstalk that coordinates the intestinal topography of IL-22 secretion required for mucosal defense.
Asunto(s)
Quimiocina CXCL6/inmunología , Infecciones por Enterobacteriaceae/inmunología , Interleucinas/inmunología , Membrana Mucosa/inmunología , Receptores CXCR/inmunología , Animales , Antígenos Ly/biosíntesis , Linfocitos T CD4-Positivos/inmunología , Receptor 1 de Quimiocinas CX3C , Quimiocina CXCL16 , Quimiocina CXCL6/biosíntesis , Citrobacter rodentium/inmunología , Células Dendríticas/inmunología , Interleucina-23/biosíntesis , Interleucinas/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptor 1 Gatillante de la Citotoxidad Natural/biosíntesis , Receptores CXCR/biosíntesis , Receptores CXCR/genética , Receptores CXCR6 , Receptores de Quimiocina/biosíntesis , Receptores de Quimiocina/inmunología , Interleucina-22RESUMEN
Esophagogastric adenocarcinomas (EAC) are obesity-associated malignancies underpinned by severe immune dysregulation and inflammation. Our previous work indicates that NK cells migrate to EAC omentum, where they undergo phenotypic and functional alterations and apoptosis. In this study, we investigate whether such erroneous chemotaxis to omentum is paralleled by compromised NK cell infiltration of EAC patient tumor and examine the role of the inflammatory chemokine fractalkine in shaping the NK cell-mediated response. Our data show diminished NK cell frequencies in EAC tumor compared with those in the circulation and reveal that intratumoral NK cell frequencies decline as visceral obesity increases in EAC patients. Our in vitro findings demonstrate that antagonism of fractalkine receptor CX3CR1 significantly reduces NK cell migration to EAC patient-derived, omental adipose tissue-conditioned media, but not toward tumor-conditioned media. These data suggest fractalkine is a key driver of NK cell chemotaxis to omentum but has a lesser role in NK cell homing to tumor in EAC. We propose that this may offer a novel therapeutic strategy to limit NK cell depletion in the omentum of obese EAC patients, and our data suggest the optimal timing for CX3CR1 antagonism is after neoadjuvant chemoradiotherapy. Our functional studies demonstrate that fractalkine induces the conversion from CX3CR1+CD27- to CX3CR1-CD27+ NK cells and increases their IFN-γ and TNF-α production, indicative of its role in shaping the dominant NK cell phenotype in EAC omentum. This study uncovers crucial and potentially druggable pathways underpinning NK cell dysfunction in obesity-associated cancer and provides compelling insights into fractalkine's diverse biological functions.
Asunto(s)
Quimiocina CX3CL1/inmunología , Quimiotaxis/inmunología , Células Asesinas Naturales/inmunología , Obesidad/inmunología , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Adenocarcinoma/inmunología , Tejido Adiposo/inmunología , Movimiento Celular/inmunología , Neoplasias Esofágicas/inmunología , Femenino , Humanos , Inflamación/inmunología , Masculino , Persona de Mediana Edad , Fenotipo , Receptores de Quimiocina/inmunología , Neoplasias Gástricas/inmunologíaRESUMEN
Circulatory antigens transit through the small intestine via the fenestrated capillaries in the lamina propria prior to entering into the draining lymphatics. But whether or how this process controls mucosal immune responses remains unknown. Here we demonstrate that dendritic cells (DCs) of the lamina propria can sample and process both circulatory and luminal antigens. Surprisingly, antigen cross-presentation by resident CX3CR1(+) DCs induced differentiation of precursor cells into CD8(+) T cells that expressed interleukin-10 (IL-10), IL-13, and IL-9 and could migrate into adjacent compartments. We conclude that lamina propria CX3CR1(+) DCs facilitate the surveillance of circulatory antigens and act as a conduit for the processing of self- and intestinally absorbed antigens, leading to the induction of CD8(+) T cells, that partake in the control of T cell activation during mucosal immune responses.
Asunto(s)
Presentación de Antígeno/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Mucosa Intestinal/inmunología , Activación de Linfocitos/inmunología , Animales , Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/citología , Receptor 1 de Quimiocinas CX3C , Diferenciación Celular/inmunología , Reactividad Cruzada/inmunología , Células Dendríticas/metabolismo , Enteritis/inmunología , Enteritis/prevención & control , Epítopos de Linfocito T/inmunología , Mucosa Intestinal/citología , Intestino Delgado/inmunología , Ratones , Receptores de Quimiocina/inmunología , Receptores de Quimiocina/metabolismoRESUMEN
CD103+ dendritic cells (DCs) carry bacteria from the small intestine and can present antigens to T cells. Yet they have not been recorded sampling luminal bacteria or presenting bacterial antigens in mesentery lymph nodes. We used 2-photon microscopy in live Cx3cr1(+/gfp) ×Cd11c-YFP mice to study these processes. At steady state, sparse CD103+ DCs occupied the epithelium. They patrolled among enterocytes while extending dendrites toward the lumen, likely using tight-junction proteins to penetrate the epithelium. Challenge with Salmonella triggered chemokine- and toll-like receptor (TLR)-dependent recruitment of additional DCs from the lamina propria (LP). The DCs efficiently phagocytosed the bacteria using intraepithelial dendrites. Noninvasive bacteria were similarly sampled. In contrast, CD103+ DCs sampled soluble luminal antigen inefficiently. In mice harboring CD103+ DCs, antigen-specific CD8 T cells were subsequently activated in MLNs. Intestinal CD103+ DCs are therefore equipped with unique mechanisms to independently complete the processes of uptake, transportation, and presentation of bacterial antigens.
Asunto(s)
Presentación de Antígeno/inmunología , Antígenos Bacterianos/inmunología , Antígenos CD/inmunología , Células Dendríticas/inmunología , Cadenas alfa de Integrinas/inmunología , Mucosa Intestinal/inmunología , Animales , Antígenos CD/metabolismo , Antígeno CD11c/genética , Antígeno CD11c/inmunología , Antígeno CD11c/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Receptor 1 de Quimiocinas CX3C , Línea Celular Tumoral , Movimiento Celular/inmunología , Células Cultivadas , Células Dendríticas/metabolismo , Citometría de Flujo , Interacciones Huésped-Patógeno/inmunología , Cadenas alfa de Integrinas/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía de Fluorescencia por Excitación Multifotónica , Membrana Mucosa/inmunología , Membrana Mucosa/metabolismo , Membrana Mucosa/microbiología , Receptores de Quimiocina/genética , Receptores de Quimiocina/inmunología , Receptores de Quimiocina/metabolismo , Salmonella typhi/inmunología , Salmonella typhi/fisiología , Salmonella typhimurium/inmunología , Salmonella typhimurium/fisiología , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismoRESUMEN
Monocyte-derived macrophages are essential for recovery after spinal cord injury, but their homing mechanism is poorly understood. Here, we show that although of common origin, the homing of proinflammatory (M1) and the "alternatively activated" anti-inflammatory (M2) macrophages to traumatized spinal cord (SC) was distinctly regulated, neither being through breached blood-brain barrier. The M1 macrophages (Ly6c(hi)CX3CR1(lo)) derived from monocytes homed in a CCL2 chemokine-dependent manner through the adjacent SC leptomeninges. The resolving M2 macrophages (Ly6c(lo)CX3CR1(hi)) derived from monocytes trafficked through a remote blood-cerebrospinal-fluid (CSF) barrier, the brain-ventricular choroid plexus (CP), via VCAM-1-VLA-4 adhesion molecules and epithelial CD73 enzyme for extravasation and epithelial transmigration. Blockage of these determinants, or mechanical CSF flow obstruction, inhibited M2 macrophage recruitment and impaired motor-function recovery. The CP, along with the CSF and the central canal, provided an anti-inflammatory supporting milieu, potentially priming the trafficking monocytes. Overall, our finding demonstrates that the route of monocyte entry to central nervous system provides an instructional environment to shape their function.
Asunto(s)
Plexo Coroideo/inmunología , Macrófagos/inmunología , Traumatismos de la Médula Espinal/inmunología , Médula Espinal/inmunología , 5'-Nucleotidasa/antagonistas & inhibidores , 5'-Nucleotidasa/genética , 5'-Nucleotidasa/inmunología , Adenosina Difosfato/análogos & derivados , Adenosina Difosfato/farmacología , Animales , Antígenos Ly/inmunología , Antígenos Ly/metabolismo , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/metabolismo , Receptor 1 de Quimiocinas CX3C , Movimiento Celular/genética , Movimiento Celular/inmunología , Plexo Coroideo/metabolismo , Inhibidores Enzimáticos/farmacología , Citometría de Flujo , Expresión Génica/inmunología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Integrina alfa4beta1/genética , Integrina alfa4beta1/inmunología , Antígenos Comunes de Leucocito/inmunología , Antígenos Comunes de Leucocito/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Meninges/inmunología , Meninges/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/metabolismo , Receptores de Quimiocina/genética , Receptores de Quimiocina/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/líquido cefalorraquídeo , Traumatismos de la Médula Espinal/genética , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/inmunologíaRESUMEN
Targeting Ag to surface receptors on conventional type 1 dendritic cells can enhance induction of Ab and T cell responses. However, it is unclear to what extent the targeted receptor influences the resulting responses. In this study, we target Ag to Xcr1, Clec9A, or DEC-205, surface receptors that are expressed on conventional type 1 dendritic cells, and compare immune responses in BALB/c and C57BL/6 mice in vitro and in vivo after intradermal DNA vaccination. Targeting hemagglutinin from influenza A to Clec9A induced Ab responses with higher avidity that more efficiently neutralized influenza virus compared with Xcr1 and DEC-205 targeting. In contrast, targeting Xcr1 resulted in higher IFN-γ+CD8+ T cell responses in spleen and lung and stronger cytotoxicity. Both Clec9A and Xcr1 targeting induced Th1-polarized Ab responses, although the Th1 polarization of CD4+ T cells was more pronounced after Xcr1 targeting. Targeting DEC-205 resulted in poor Ab responses in BALB/c mice and a more mixed Th response. In an influenza challenge model, targeting either Xcr1 or Clec9A induced full and long-term protection against influenza infection, whereas only partial short-term protection was obtained when targeting DEC-205. In summary, the choice of targeting receptor, even on the same dendritic cell subpopulation, may strongly influence the resulting immune response, suggesting that different targeting strategies should be considered depending on the pathogen.
Asunto(s)
Antígenos CD/inmunología , Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Lectinas Tipo C/inmunología , Antígenos de Histocompatibilidad Menor/inmunología , Receptores de Superficie Celular/inmunología , Receptores de Quimiocina/inmunología , Receptores Inmunológicos/inmunología , Células TH1/inmunología , Animales , Femenino , Células HEK293 , Humanos , Interferón gamma/inmunología , Ratones , Ratones Endogámicos BALB CRESUMEN
As main drivers of leukocyte recruitment during inflammatory reactions, chemokines act as mediatrs of alarmins in priming host defense responses after tissue exposure to toxic or infectious agents, immunomediated damage, and in inflammation-driven tumors. Chemokines can therefore be considered alarm signals generated by tissues in a broad number of conditions, and mechanisms controlling chemokines biological activities are therefore key to regulate tissue reactions induced by alarmins. By transporting, presenting or scavenging different sets of chemokines, atypical chemokine receptors represent an emerign subfamily of chemokine receptors which operates in tissues as chemokine gatekeepers in order to establish and shape their gradients and coordinate leukocyte recruitment.
Asunto(s)
Alarminas/inmunología , Quimiocinas/inmunología , Inflamación/inmunología , Neoplasias/inmunología , Alarminas/metabolismo , Animales , Quimiocinas/metabolismo , Humanos , Inflamación/metabolismo , Leucocitos/inmunología , Leucocitos/metabolismo , Modelos Inmunológicos , Neoplasias/metabolismo , Receptores de Quimiocina/inmunología , Receptores de Quimiocina/metabolismo , Transducción de Señal/inmunologíaRESUMEN
Analysis of chemokine receptor, and atypical chemokine receptor, expression is frequently hampered by the lack of availability of high-quality antibodies and the species specificity of those that are available. We have previously described methodology utilizing Alexa-Fluor-labeled chemokine ligands as versatile reagents to detect receptor expression. Previously this has been limited to hematopoietic cells and methodology for assessing expression of receptors on stromal cells has been lacking. Among chemokine receptors, the ones most frequently expressed on stromal cells belong to the atypical chemokine receptor subfamily. These receptors do not signal in the classic sense in response to ligand but scavenge their ligands and degrade them and thus sculpt in vivo chemokine gradients. Here, we demonstrate the ability to use either intratracheal or intravenous, Alexa-Fluor-labeled chemokine administration to detect stromal cell populations expressing the atypical chemokine receptor ACKR2. Using this methodology, we demonstrate, for the first time, expression of ACKR2 on blood endothelial cells. This observation sets the lung aside from other tissues in which ACKR2 is exclusively expressed on lymphatic endothelial cells and suggest unique roles for ACKR2 in the pulmonary environment.