Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nat Immunol ; 22(8): 1042-1051, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34267375

RESUMEN

Pathogens and vaccines that produce persisting antigens can generate expanded pools of effector memory CD8+ T cells, described as memory inflation. While properties of inflating memory CD8+ T cells have been characterized, the specific cell types and tissue factors responsible for their maintenance remain elusive. Here, we show that clinically applied adenovirus vectors preferentially target fibroblastic stromal cells in cultured human tissues. Moreover, we used cell-type-specific antigen targeting to define critical cells and molecules that sustain long-term antigen presentation and T cell activity after adenovirus vector immunization in mice. While antigen targeting to myeloid cells was insufficient to activate antigen-specific CD8+ T cells, genetic activation of antigen expression in Ccl19-cre-expressing fibroblastic stromal cells induced inflating CD8+ T cells. Local ablation of vector-targeted cells revealed that lung fibroblasts support the protective function and metabolic fitness of inflating memory CD8+ T cells in an interleukin (IL)-33-dependent manner. Collectively, these data define a critical fibroblastic niche that underpins robust protective immunity operating in a clinically important vaccine platform.


Asunto(s)
Adenoviridae/inmunología , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/inmunología , Interleucina-33/inmunología , Activación de Linfocitos/inmunología , Células del Estroma/inmunología , Adenoviridae/genética , Animales , Línea Celular Tumoral , Quimiocina CCL19/metabolismo , Quimera/genética , Epítopos de Linfocito T/inmunología , Fibroblastos/citología , Fibroblastos/metabolismo , Vectores Genéticos/inmunología , Humanos , Pulmón/citología , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vacunación
2.
Nat Immunol ; 17(12): 1388-1396, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27798617

RESUMEN

Fibroblastic reticular cells (FRCs) of secondary lymphoid organs form distinct niches for interaction with hematopoietic cells. We found here that production of the cytokine IL-15 by FRCs was essential for the maintenance of group 1 innate lymphoid cells (ILCs) in Peyer's patches and mesenteric lymph nodes. Moreover, FRC-specific ablation of the innate immunological sensing adaptor MyD88 unleashed IL-15 production by FRCs during infection with an enteropathogenic virus, which led to hyperactivation of group 1 ILCs and substantially altered the differentiation of helper T cells. Accelerated clearance of virus by group 1 ILCs precipitated severe intestinal inflammatory disease with commensal dysbiosis, loss of intestinal barrier function and diminished resistance to colonization. In sum, FRCs act as an 'on-demand' immunological 'rheostat' by restraining activation of group 1 ILCs and thereby preventing immunopathological damage in the intestine.


Asunto(s)
Citrobacter rodentium/inmunología , Infecciones por Coronavirus/inmunología , Infecciones por Enterobacteriaceae/inmunología , Fibroblastos/inmunología , Interleucina-15/metabolismo , Linfocitos/inmunología , Virus de la Hepatitis Murina/inmunología , Animales , Células Cultivadas , Inmunidad Innata , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Ganglios Linfáticos Agregados/patología , Células TH1/inmunología , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo
3.
Nat Immunol ; 15(10): 973-81, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25151489

RESUMEN

Fibroblastic reticular cells (FRCs) are known to inhabit T cell-rich areas of lymphoid organs, where they function to facilitate interactions between T cells and dendritic cells. However, in vivo manipulation of FRCs has been limited by a dearth of genetic tools that target this lineage. Here, using a mouse model to conditionally ablate FRCs, we demonstrated their indispensable role in antiviral T cell responses. Unexpectedly, loss of FRCs also attenuated humoral immunity due to impaired B cell viability and follicular organization. Follicle-resident FRCs established a favorable niche for B lymphocytes via production of the cytokine BAFF. Thus, our study indicates that adaptive immunity requires an intact FRC network and identifies a subset of FRCs that control B cell homeostasis and follicle identity.


Asunto(s)
Linfocitos B/inmunología , Fibroblastos/inmunología , Homeostasis/inmunología , Linfocitos T/inmunología , Animales , Factor Activador de Células B/inmunología , Factor Activador de Células B/metabolismo , Linfocitos B/metabolismo , Movimiento Celular/inmunología , Supervivencia Celular/inmunología , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Fibroblastos/metabolismo , Citometría de Flujo , Inmunidad Humoral/inmunología , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Linfocitos T/metabolismo
4.
Immunity ; 44(3): 622-633, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26921107

RESUMEN

Stromal cells generate a complex cellular scaffold that provides specialized microenvironments for lymphocyte activation in secondary lymphoid organs. Here, we assessed whether local activation of stromal cells in the central nervous system (CNS) is mandatory to transfer immune recognition from secondary lymphoid organs into the infected tissue. We report that neurotropic virus infection in mice triggered the establishment of such stromal cell niches in the CNS. CNS stromal cell activation was dominated by a rapid and vigorous production of CC-motif chemokine receptor (CCR) 7 ligands CCL19 and CCL21 by vascular endothelial cells and adjacent fibroblastic reticular cell (FRC)-like cells in the perivascular space. Moreover, CCR7 ligands produced by CNS stromal cells were crucial to support recruitment and local re-activation of antiviral CD8(+) T cells and to protect the host from lethal neuroinflammatory disease, indicating that CNS stromal cells generate confined microenvironments that control protective T cell immunity.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Sistema Nervioso Central/inmunología , Endotelio Vascular/inmunología , Virus de la Hepatitis A/inmunología , Hepatitis A/inmunología , Inflamación Neurogénica/parasitología , Receptores CCR7/metabolismo , Células del Estroma/inmunología , Animales , Movimiento Celular , Microambiente Celular , Sistema Nervioso Central/virología , Quimiocina CCL19/metabolismo , Quimiocina CCL21/metabolismo , Endotelio Vascular/virología , Hepatitis A/complicaciones , Inmunidad Celular , Inmunomodulación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Inflamación Neurogénica/etiología , Receptores CCR7/genética , Células del Estroma/virología , Tropismo Viral
5.
Mol Ther ; 32(2): 426-439, 2024 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-38058126

RESUMEN

Harnessing the immune system to eradicate tumors requires identification and targeting of tumor antigens, including tumor-specific neoantigens and tumor-associated self-antigens. Tumor-associated antigens are subject to existing immune tolerance, which must be overcome by immunotherapies. Despite many novel immunotherapies reaching clinical trials, inducing self-antigen-specific immune responses remains challenging. Here, we systematically investigate viral-vector-based cancer vaccines encoding a tumor-associated self-antigen (TRP2) for the treatment of established melanomas in preclinical mouse models, alone or in combination with adoptive T cell therapy. We reveal that, unlike foreign antigens, tumor-associated antigens require replication of lymphocytic choriomeningitis virus (LCMV)-based vectors to break tolerance and induce effective antigen-specific CD8+ T cell responses. Immunization with a replicating LCMV vector leads to complete tumor rejection when combined with adoptive TRP2-specific T cell transfer. Importantly, immunization with replicating vectors leads to extended antigen persistence in secondary lymphoid organs, resulting in efficient T cell priming, which renders previously "cold" tumors open to immune infiltration and reprograms the tumor microenvironment to "hot." Our findings have important implications for the design of next-generation immunotherapies targeting solid cancers utilizing viral vectors and adoptive cell transfer.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Ratones , Animales , Virus de la Coriomeningitis Linfocítica/genética , Linfocitos T CD8-positivos , Neoplasias/tratamiento farmacológico , Antígenos de Neoplasias/genética , Autoantígenos , Microambiente Tumoral
6.
Immunity ; 43(5): 974-86, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26588782

RESUMEN

Tissue damage caused by viral hepatitis is a major cause of morbidity and mortality worldwide. Using a mouse model of viral hepatitis, we identified virus-induced early transcriptional changes in the redox pathways in the liver, including downregulation of superoxide dismutase 1 (Sod1). Sod1(-/-) mice exhibited increased inflammation and aggravated liver damage upon viral infection, which was independent of T and NK cells and could be ameliorated by antioxidant treatment. Type I interferon (IFN-I) led to a downregulation of Sod1 and caused oxidative liver damage in Sod1(-/-) and wild-type mice. Genetic and pharmacological ablation of the IFN-I signaling pathway protected against virus-induced liver damage. These results delineate IFN-I mediated oxidative stress as a key mediator of virus-induced liver damage and describe a mechanism of innate-immunity-driven pathology, linking IFN-I signaling with antioxidant host defense and infection-associated tissue damage. VIDEO ABSTRACT.


Asunto(s)
Hepatocitos/inmunología , Interferón Tipo I/inmunología , Estrés Oxidativo/inmunología , Superóxido Dismutasa/inmunología , Animales , Antioxidantes/metabolismo , Hepatitis Viral Animal/inmunología , Inmunidad Innata/inmunología , Inflamación/inmunología , Células Asesinas Naturales/inmunología , Hígado/inmunología , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción , Transducción de Señal/inmunología , Superóxido Dismutasa-1 , Linfocitos T/inmunología , Transcripción Genética/inmunología
7.
Immunity ; 38(5): 1013-24, 2013 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-23623380

RESUMEN

The stromal scaffold of the lymph node (LN) paracortex is built by fibroblastic reticular cells (FRCs). Conditional ablation of lymphotoxin-ß receptor (LTßR) expression in LN FRCs and their mesenchymal progenitors in developing LNs revealed that LTßR-signaling in these cells was not essential for the formation of LNs. Although T cell zone reticular cells had lost podoplanin expression, they still formed a functional conduit system and showed enhanced expression of myofibroblastic markers. However, essential immune functions of FRCs, including homeostatic chemokine and interleukin-7 expression, were impaired. These changes in T cell zone reticular cell function were associated with increased susceptibility to viral infection. Thus, myofibroblasic FRC precursors are able to generate the basic T cell zone infrastructure, whereas LTßR-dependent maturation of FRCs guarantees full immunocompetence and hence optimal LN function during infection.


Asunto(s)
Infecciones por Coronavirus/inmunología , Ganglios Linfáticos/citología , Ganglios Linfáticos/metabolismo , Miofibroblastos/fisiología , Linfocitos T/inmunología , Animales , Diferenciación Celular , Células Cultivadas , Fibroblastos/citología , Fibroblastos/inmunología , Interleucina-7/biosíntesis , Ganglios Linfáticos/inmunología , Receptor beta de Linfotoxina/metabolismo , Linfotoxina beta/biosíntesis , Linfotoxina beta/metabolismo , Glicoproteínas de Membrana/biosíntesis , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Virus de la Hepatitis Murina/inmunología , Miofibroblastos/citología , Transducción de Señal
8.
PLoS Biol ; 14(7): e1002515, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27415420

RESUMEN

Fibroblastic reticular cells (FRCs) form the cellular scaffold of lymph nodes (LNs) and establish distinct microenvironmental niches to provide key molecules that drive innate and adaptive immune responses and control immune regulatory processes. Here, we have used a graph theory-based systems biology approach to determine topological properties and robustness of the LN FRC network in mice. We found that the FRC network exhibits an imprinted small-world topology that is fully regenerated within 4 wk after complete FRC ablation. Moreover, in silico perturbation analysis and in vivo validation revealed that LNs can tolerate a loss of approximately 50% of their FRCs without substantial impairment of immune cell recruitment, intranodal T cell migration, and dendritic cell-mediated activation of antiviral CD8+ T cells. Overall, our study reveals the high topological robustness of the FRC network and the critical role of the network integrity for the activation of adaptive immune responses.


Asunto(s)
Comunicación Celular/inmunología , Movimiento Celular/inmunología , Fibroblastos/inmunología , Ganglios Linfáticos/inmunología , Linfocitos T/inmunología , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Recuento de Células , Movimiento Celular/genética , Quimiocina CCL19/genética , Quimiocina CCL19/inmunología , Quimiocina CCL19/metabolismo , Células Dendríticas/citología , Células Dendríticas/inmunología , Fibroblastos/citología , Fibroblastos/metabolismo , Ganglios Linfáticos/citología , Ganglios Linfáticos/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Modelos Inmunológicos , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo
9.
J Immunol ; 198(5): 1775-1781, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28223405

RESUMEN

Inflammation in the CNS must be tightly regulated to respond efficiently to infection with neurotropic pathogens. Access of immune cells to the CNS and their positioning within the tissue are controlled by stromal cells that construct the barriers of the CNS. Although the role of the endothelium in regulating the passage of leukocytes and small molecules into the CNS has been studied extensively, the contribution of fibroblastic stromal cells as portals of entry into the CNS was only recently uncovered. We review the critical immune-stimulating role of meningeal fibroblasts in promoting recruitment and retention of lymphocytes during CNS inflammation. Activated meningeal fibroblastic stromal cells have the capacity to rapidly elaborate an immune-competent niche that sustains protective immune cells entering the CNS from the draining cervical lymph node. Such stromal cell niches can ultimately foster the establishment of tertiary lymphoid tissues during chronic neuroinflammatory conditions.


Asunto(s)
Movimiento Celular/inmunología , Sistema Nervioso Central/inmunología , Inflamación , Células del Estroma/inmunología , Animales , Barrera Hematoencefálica , Sistema Nervioso Central/citología , Encefalomielitis Autoinmune Experimental , Fibroblastos/inmunología , Humanos , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Linfocitos/inmunología , Linfocitos/fisiología , Células del Estroma/fisiología
10.
J Allergy Clin Immunol ; 142(4): 1257-1271.e4, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29391257

RESUMEN

BACKGROUND: A particular characteristic of non-small cell lung cancer is the composition of the tumor microenvironment with a very high proportion of fibroblastic stromal cells (FSCs). OBJECTIVE: Lapses in our basic knowledge of fibroblast phenotype and function in the tumor microenvironment make it difficult to define whether FSC subsets exist that exhibit either tumor-promoting or tumor-suppressive properties. METHODS: We used gene expression profiling of lung versus tumor FSCs from patients with non-small cell lung cancer. Moreover, CCL19-expressing FSCs were studied in transgenic mouse models by using a lung cancer metastasis model. RESULTS: CCL19 mRNA expression in human tumor FSCs correlates with immune cell infiltration and intratumoral accumulation of CD8+ T cells. Mechanistic dissection in murine lung carcinoma models revealed that CCL19-expressing FSCs form perivascular niches to promote accumulation of CD8+ T cells in the tumor. Targeted ablation of CCL19-expressing tumor FSCs reduced immune cell recruitment and resulted in unleashed tumor growth. CONCLUSION: These data suggest that a distinct population of CCL19-producing FSCs fosters the development of an immune-stimulating intratumoral niche for immune cells to control cancer growth.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/inmunología , Quimiocina CCL19/inmunología , Fibroblastos/inmunología , Neoplasias Pulmonares/inmunología , Células del Estroma/inmunología , Animales , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Quimiocina CCL19/genética , Humanos , Neoplasias Pulmonares/genética , Ratones Endogámicos C57BL , Ratones Transgénicos , Linfocitos T/trasplante , Transcriptoma , Microambiente Tumoral/inmunología
11.
J Math Biol ; 69(6-7): 1547-83, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24337680

RESUMEN

Since their invention in 1994, fluorescent dyes such as carboxyfluorescein diacetate succinimidyl ester (CFSE) are used for cell proliferation analysis in flow cytometry. Importantly, the interpretation of such assays relies on the assumption that the label is divided equally between the daughter cells upon cell division. However, recent experimental studies indicate that division of cells is not perfectly symmetric and there is unequal distribution of protein between sister cell pairs. The uneven partition of protein or mass to daughter cells can lead to an overlap in the generations of CFSE-labelled cells with straightforward consequences for the resolution of individual generations. Numerous mathematical models developed so far for the analysis of CFSE proliferation assay incorporate the premise that the CFSE fluorescence intensity is halved in the two daughter cells. Here, we propose a novel modelling approach for the analysis of the CFSE cell proliferation assays which are characterized by poorly resolved peaks of cell generations in flow cytometric histograms. We formulate a mathematical model in the form of a system of delay hyperbolic partial differential equations which provides a good agreement with the CFSE histograms time-series data and allows an analytical treatment. The model is a further generalization of the recently proposed class of division- and label-structured models as it considers an asymmetric cell division. In addition, the basic structure of the cell cycle, i.e. the resting and cycling cell compartments, is taken into account. The model is used to estimate fundamental parameters such as activation rate, duration of the cell cycle, apoptosis rate, CFSE decay rate and asymmetry factor in cell division of monoclonal T cells during cognate interaction with dendritic cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Ciclo Celular/inmunología , División Celular/inmunología , Citometría de Flujo/métodos , Fluoresceínas/química , Modelos Teóricos , Succinimidas/química , Animales , Linfocitos T CD8-positivos/química , Ratones , Análisis Numérico Asistido por Computador
12.
Nat Commun ; 15(1): 451, 2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-38200005

RESUMEN

Immune cells must adapt to different environments during the course of an immune response. Here we study the adaptation of CD8+ T cells to the intestinal microenvironment and how this process shapes the establishment of the CD8+ T cell pool. CD8+ T cells progressively remodel their transcriptome and surface phenotype as they enter the gut wall, and downregulate expression of mitochondrial genes. Human and mouse intestinal CD8+ T cells have reduced mitochondrial mass, but maintain a viable energy balance to sustain their function. We find that the intestinal microenvironment is rich in prostaglandin E2 (PGE2), which drives mitochondrial depolarization in CD8+ T cells. Consequently, these cells engage autophagy to clear depolarized mitochondria, and enhance glutathione synthesis to scavenge reactive oxygen species (ROS) that result from mitochondrial depolarization. Impairing PGE2 sensing promotes CD8+ T cell accumulation in the gut, while tampering with autophagy and glutathione negatively impacts the T cell pool. Thus, a PGE2-autophagy-glutathione axis defines the metabolic adaptation of CD8+ T cells to the intestinal microenvironment, to ultimately influence the T cell pool.


Asunto(s)
Autofagia , Linfocitos T CD8-positivos , Humanos , Animales , Ratones , Dinoprostona , Genes Mitocondriales , Glutatión
13.
bioRxiv ; 2023 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-36993703

RESUMEN

Immune cells must adapt to different environments during the course of an immune response. We studied the adaptation of CD8 + T cells to the intestinal microenvironment and how this process shapes their residency in the gut. CD8 + T cells progressively remodel their transcriptome and surface phenotype as they acquire gut residency, and downregulate expression of mitochondrial genes. Human and mouse gut-resident CD8 + T cells have reduced mitochondrial mass, but maintain a viable energy balance to sustain their function. We found that the intestinal microenvironment is rich in prostaglandin E 2 (PGE 2 ), which drives mitochondrial depolarization in CD8 + T cells. Consequently, these cells engage autophagy to clear depolarized mitochondria, and enhance glutathione synthesis to scavenge reactive oxygen species (ROS) that result from mitochondrial depolarization. Impairing PGE 2 sensing promotes CD8 + T cell accumulation in the gut, while tampering with autophagy and glutathione negatively impacts the T cell population. Thus, a PGE 2 -autophagy-glutathione axis defines the metabolic adaptation of CD8 + T cells to the intestinal microenvironment, to ultimately influence the T cell pool.

14.
Cell Metab ; 34(5): 747-760.e6, 2022 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-35508110

RESUMEN

Adipose tissue (AT) plays a central role in systemic metabolic homeostasis, but its function during bacterial infection remains unclear. Following subcutaneous bacterial infection, adipocytes surrounding draining lymph nodes initiated a transcriptional response indicative of stimulation with IFN-γ and a shift away from lipid metabolism toward an immunologic function. Natural killer (NK) and invariant NK T (iNKT) cells were identified as sources of infection-induced IFN-γ in perinodal AT (PAT). IFN-γ induced Nos2 expression in adipocytes through a process dependent on nuclear-binding oligomerization domain 1 (NOD1) sensing of live intracellular bacteria. iNOS expression was coupled to metabolic rewiring, inducing increased diversion of extracellular L-arginine through the arginosuccinate shunt and urea cycle to produce nitric oxide (NO), directly mediating bacterial clearance. In vivo, control of infection in adipocytes was dependent on adipocyte-intrinsic sensing of IFN-γ and expression of iNOS. Thus, adipocytes are licensed by innate lymphocytes to acquire anti-bacterial functions during infection.


Asunto(s)
Señales (Psicología) , Células Asesinas Naturales , Adipocitos/metabolismo , Inmunidad , Interferón gamma/metabolismo
15.
Sci Immunol ; 7(76): eadd3263, 2022 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-36240286

RESUMEN

Type 2 immunity is associated with adipose tissue (AT) homeostasis and infection with parasitic helminths, but whether AT participates in immunity to these parasites is unknown. We found that the fat content of mesenteric AT (mAT) declined in mice during infection with a gut-restricted helminth. This was associated with the accumulation of metabolically activated, interleukin-33 (IL-33), thymic stromal lymphopoietin (TSLP), and extracellular matrix (ECM)-producing stromal cells. These cells shared transcriptional features, including the expression of Dpp4 and Pi16, with multipotent progenitor cells (MPC) that have been identified in numerous tissues and are reported to be capable of differentiating into fibroblasts and adipocytes. Concomitantly, mAT became infiltrated with resident T helper 2 (TH2) cells that responded to TSLP and IL-33 by producing stromal cell-stimulating cytokines, including transforming growth factor ß1 (TGFß1) and amphiregulin. These TH2 cells expressed genes previously associated with type 2 innate lymphoid cells (ILC2), including Nmur1, Calca, Klrg1, and Arg1, and persisted in mAT for at least 11 months after anthelmintic drug-mediated clearance of infection. We found that MPC and TH2 cells localized to ECM-rich interstitial spaces that appeared shared between mesenteric lymph node, mAT, and intestine. Stromal cell expression of epidermal growth factor receptor (EGFR), the receptor for amphiregulin, was required for immunity to infection. Our findings point to the importance of MPC and TH2 cell interactions within the interstitium in orchestrating AT remodeling and immunity to an intestinal infection.


Asunto(s)
Inmunidad Innata , Interleucina-33 , Tejido Adiposo/metabolismo , Anfirregulina , Animales , Citocinas/metabolismo , Dipeptidil Peptidasa 4 , Receptores ErbB , Linfocitos , Ratones , Células Th2 , Factor de Crecimiento Transformador beta1
16.
Oncoimmunology ; 10(1): 1945202, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34367729

RESUMEN

Squamous cell carcinoma of the tonsil is one of the most frequent cancers of the oropharynx. The escalating rate of tonsil cancer during the last decades is associated with the increase of high risk-human papilloma virus (HR-HPV) infections. While the microbiome in oropharyngeal malignant diseases has been characterized to some extent, the microbial colonization of HR-HPV-associated tonsil cancer remains largely unknown. Using 16S rRNA gene amplicon sequencing, we have characterized the microbiome of human palatine tonsil crypts in patients suffering from HR-HPV-associated tonsil cancer in comparison to a control cohort of adult sleep apnea patients. We found an increased abundance of the phyla Firmicutes and Actinobacteria in tumor patients, whereas the abundance of Spirochetes and Synergistetes was significantly higher in the control cohort. Furthermore, the accumulation of several genera such as Veillonella, Streptococcus and Prevotella_7 in tonsillar crypts was associated with tonsil cancer. In contrast, Fusobacterium, Prevotella and Treponema_2 were enriched in sleep apnea patients. Machine learning-based bacterial species analysis indicated that a particular bacterial composition in tonsillar crypts is tumor-predictive. Species-specific PCR-based validation in extended patient cohorts confirmed that differential abundance of Filifactor alocis and Prevotella melaninogenica is a distinct trait of tonsil cancer. This study shows that tonsil cancer patients harbor a characteristic microbiome in the crypt environment that differs from the microbiome of sleep apnea patients on all phylogenetic levels. Moreover, our analysis indicates that profiling of microbial communities in distinct tonsillar niches provides microbiome-based avenues for the diagnosis of tonsil cancer.


Asunto(s)
Carcinoma de Células Escamosas , Microbiota , Neoplasias Tonsilares , Clostridiales , Humanos , Microbiota/genética , Filogenia , ARN Ribosómico 16S/genética
17.
Nat Commun ; 12(1): 4734, 2021 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-34354077

RESUMEN

The tumor microenvironment (TME) is a complex amalgam of tumor cells, immune cells, endothelial cells and fibroblastic stromal cells (FSC). Cancer-associated fibroblasts are generally seen as tumor-promoting entity. However, it is conceivable that particular FSC populations within the TME contribute to immune-mediated tumor control. Here, we show that intratumoral treatment of mice with a recombinant lymphocytic choriomeningitis virus-based vaccine vector expressing a melanocyte differentiation antigen resulted in T cell-dependent long-term control of melanomas. Using single-cell RNA-seq analysis, we demonstrate that viral vector-mediated transduction reprogrammed and activated a Cxcl13-expressing FSC subset that show a pronounced immunostimulatory signature and increased expression of the inflammatory cytokine IL-33. Ablation of Il33 gene expression in Cxcl13-Cre-positive FSCs reduces the functionality of intratumoral T cells and unleashes tumor growth. Thus, reprogramming of FSCs by a self-antigen-expressing viral vector in the TME is critical for curative melanoma treatment by locally sustaining the activity of tumor-specific T cells.


Asunto(s)
Melanoma Experimental/terapia , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Fibroblastos Asociados al Cáncer/inmunología , Fibroblastos Asociados al Cáncer/patología , Técnicas de Reprogramación Celular/métodos , Quimiocina CXCL13/genética , Quimiocina CXCL13/inmunología , Femenino , Vectores Genéticos , Interleucina-33/deficiencia , Interleucina-33/genética , Interleucina-33/inmunología , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/inmunología , Virus de la Coriomeningitis Linfocítica/genética , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células del Estroma/inmunología , Células del Estroma/patología , Linfocitos T/inmunología , Linfocitos T/patología , Microambiente Tumoral/inmunología
18.
Life Sci Alliance ; 3(1)2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31822557

RESUMEN

Infectious complications in patients with cirrhosis frequently initiate episodes of decompensation and substantially contribute to the high mortality. Mechanisms of the underlying immuneparesis remain underexplored. TAM receptors (TYRO3/AXL/MERTK) are important inhibitors of innate immune responses. To understand the pathophysiology of immuneparesis in cirrhosis, we detailed TAM receptor expression in relation to monocyte function and disease severity prior to the onset of acute decompensation. TNF-α/IL-6 responses to lipopolysaccharide were attenuated in monocytes from patients with cirrhosis (n = 96) compared with controls (n = 27) and decreased in parallel with disease severity. Concurrently, an AXL-expressing (AXL+) monocyte population expanded. AXL+ cells (CD14+CD16highHLA-DRhigh) were characterised by attenuated TNF-α/IL-6 responses and T cell activation but enhanced efferocytosis and preserved phagocytosis of Escherichia coli Their expansion correlated with disease severity, complications, infection, and 1-yr mortality. AXL+ monocytes were generated in response to microbial products and efferocytosis in vitro. AXL kinase inhibition and down-regulation reversed attenuated monocyte inflammatory responses in cirrhosis ex vivo. AXL may thus serve as prognostic marker and deserves evaluation as immunotherapeutic target in cirrhosis.


Asunto(s)
Cirrosis Hepática/sangre , Cirrosis Hepática/mortalidad , Monocitos/inmunología , Proteínas Proto-Oncogénicas/sangre , Proteínas Tirosina Quinasas Receptoras/sangre , Índice de Severidad de la Enfermedad , Adulto , Anciano , Biomarcadores/sangre , Femenino , Estudios de Seguimiento , Humanos , Inmunidad Innata , Interleucina-6/metabolismo , Activación de Linfocitos/genética , Masculino , Persona de Mediana Edad , Monocitos/metabolismo , Fagocitosis/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Transducción de Señal/genética , Células THP-1 , Transducción Genética , Factor de Necrosis Tumoral alfa/metabolismo , Tirosina Quinasa del Receptor Axl
19.
Nat Commun ; 11(1): 3677, 2020 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-32699279

RESUMEN

Through the formation of concentration gradients, morphogens drive graded responses to extracellular signals, thereby fine-tuning cell behaviors in complex tissues. Here we show that the chemokine CXCL13 forms both soluble and immobilized gradients. Specifically, CXCL13+ follicular reticular cells form a small-world network of guidance structures, with computer simulations and optimization analysis predicting that immobilized gradients created by this network promote B cell trafficking. Consistent with this prediction, imaging analysis show that CXCL13 binds to extracellular matrix components in situ, constraining its diffusion. CXCL13 solubilization requires the protease cathepsin B that cleaves CXCL13 into a stable product. Mice lacking cathepsin B display aberrant follicular architecture, a phenotype associated with effective B cell homing to but not within lymph nodes. Our data thus suggest that reticular cells of the B cell zone generate microenvironments that shape both immobilized and soluble CXCL13 gradients.


Asunto(s)
Linfocitos B/inmunología , Microambiente Celular/inmunología , Quimiocina CXCL13/metabolismo , Células Dendríticas Foliculares/inmunología , Inmunidad Adaptativa , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Catepsina B/genética , Catepsina B/metabolismo , Línea Celular , Quimiocina CXCL13/inmunología , Simulación por Computador , Células Dendríticas Foliculares/citología , Células Dendríticas Foliculares/metabolismo , Matriz Extracelular/metabolismo , Humanos , Ratones , Ratones Noqueados , Microscopía Fluorescente , Modelos Biológicos , Tonsila Palatina/citología , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Células del Estroma/inmunología , Células del Estroma/metabolismo
20.
J Exp Med ; 211(13): 2507-17, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25488981

RESUMEN

Interactions between dendritic cells (DCs) and T cells control the decision between activation and tolerance induction. Thromboxane A2 (TXA2) and its receptor TP have been suggested to regulate adaptive immune responses through control of T cell-DC interactions. Here, we show that this control is achieved by selectively reducing expansion of low-avidity CD4(+) T cells. During inflammation, weak tetramer-binding TP-deficient CD4(+) T cells were preferentially expanded compared with TP-proficient CD4(+) T cells. Using intravital imaging of cellular interactions in reactive peripheral lymph nodes (PLNs), we found that TXA2 led to disruption of low- but not high-avidity interactions between DCs and CD4(+) T cells. Lack of TP correlated with higher expression of activation markers on stimulated CD4(+) T cells and with augmented accumulation of follicular helper T cells (TFH), which correlated with increased low-avidity IgG responses. In sum, our data suggest that tonic suppression of weak CD4(+) T cell-DC interactions by TXA2-TP signaling improves the overall quality of adaptive immune responses.


Asunto(s)
Afinidad de Anticuerpos/efectos de los fármacos , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Comunicación Celular/efectos de los fármacos , Células Dendríticas/citología , Factores Inmunológicos/farmacología , Tromboxano A2/farmacología , Animales , Biomarcadores/metabolismo , Linfocitos T CD4-Positivos/efectos de los fármacos , Pollos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Antígenos de Histocompatibilidad/inmunología , Inmunoglobulina G/biosíntesis , Inflamación/patología , Activación de Linfocitos/efectos de los fármacos , Recuento de Linfocitos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía de Fluorescencia por Excitación Multifotónica , Ovalbúmina/inmunología , Receptores de Tromboxano A2 y Prostaglandina H2/deficiencia , Receptores de Tromboxano A2 y Prostaglandina H2/metabolismo , Transducción de Señal/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA