Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Cell Rep ; 43(3): 113824, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38386557

RESUMEN

Adipose tissue homeostasis relies on the interplay between several regulatory lineages, such as type 2 innate lymphoid cells (ILC2s), T helper 2 (Th2) cells, regulatory T cells, eosinophils, and type 2 macrophages. Among them, ILC2s are numerically the dominant source of type 2 cytokines and are considered as major regulators of adiposity. Despite the overlap in immune effector molecules and sensitivity to alarmins (thymic stromal lymphopoietin and interleukin-33) between ILC2s and resident memory Th2 lymphocytes, the role of the adaptive axis of type 2 immunity remains unclear. We show that mice deficient in CD27, a member of the tumor necrosis factor receptor superfamily, are more resistant to obesity and associated disorders. A comparative analysis of the CD4 compartment of both strains revealed higher numbers of fat-resident memory Th2 cells in the adipose tissue of CD27 knockout mice, which correlated with decreased programmed cell death protein 1-induced apoptosis. Our data point to a non-redundant role for Th2 lymphocytes in obesogenic conditions.


Asunto(s)
Inmunidad Innata , Linfocitos , Animales , Ratones , Citocinas/metabolismo , Homeostasis , Interleucina-33 , Grasa Intraabdominal/metabolismo , Linfocitos/metabolismo , Células Th2 , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral
2.
Cell Rep ; 42(8): 112876, 2023 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-37543948

RESUMEN

The co-inhibitory programmed death (PD)-1 signaling pathway plays a major role in the context of tumor-specific T cell responses. Conversely, it also contributes to the maintenance of peripheral tolerance, as patients receiving anti-PD-1 treatment are prone to developing immune-related adverse events. Yet, the physiological role of the PD-1/PDL-1 axis in T cell homeostasis is still poorly understood. Herein, we show that under steady-state conditions, the absence of PD-1 signaling led to a preferential expansion of CD8+ T cells in the liver. These cells exhibit an oligoclonal T cell receptor (TCR) repertoire and a terminally differentiated exhaustion profile. The transcription factor EOMES is required for the clonal expansion and acquisition of this differentiation program. Finally, single-cell transcriptomics coupled with TCR repertoire analysis support the notion that these cells arise locally from liver-resident memory CD8+ T cells. Overall, we show a role for PD-1 signaling in liver memory T cell homeostasis.


Asunto(s)
Linfocitos T CD8-positivos , Regulación de la Expresión Génica , Humanos , Linfocitos T CD8-positivos/metabolismo , Hígado/metabolismo , Transducción de Señal , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo
3.
Front Immunol ; 14: 1023064, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36993956

RESUMEN

Introduction: Most T lymphocytes, including regulatory T cells, express the CD27 costimulatory receptor in steady state conditions. There is evidence that CD27 engagement on conventional T lymphocytes favors the development of Th1 and cytotoxic responses in mice and humans, but the impact on the regulatory lineage is unknown. Methods: In this report, we examined the effect of constitutive CD27 engagement on both regulatory and conventional CD4+ T cells in vivo, in the absence of intentional antigenic stimulation. Results: Our data show that both T cell subsets polarize into type 1 Tconvs or Tregs, characterized by cell activation, cytokine production, response to IFN-γ and CXCR3-dependent migration to inflammatory sites. Transfer experiments suggest that CD27 engagement triggers Treg activation in a cell autonomous fashion. Conclusion: We conclude that CD27 may regulate the development of Th1 immunity in peripheral tissues as well as the subsequent switch of the effector response into long-term memory.


Asunto(s)
Subgrupos de Linfocitos T , Linfocitos T Reguladores , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral , Animales , Humanos , Ratones , Antígenos/metabolismo , Ligando CD27/metabolismo , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/metabolismo , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo
4.
Cancer Immunol Res ; 11(3): 339-350, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36603132

RESUMEN

The prolyl hydroxylase domain/hypoxia-inducible factor (PHD/HIF) pathway has been implicated in a wide range of immune and inflammatory processes, including in the oxygen-deprived tumor microenvironment. To examine the effect of HIF stabilization in antitumor immunity, we deleted Phd2 selectively in T lymphocytes using the cre/lox system. We show that the deletion of PHD2 in lymphocytes resulted in enhanced regression of EG7-OVA tumors, in a HIF-1α-dependent manner. The enhanced control of neoplastic growth correlated with increased polyfunctionality of CD8+ tumor-infiltrating lymphocytes, as indicated by enhanced expression of IFNγ, TNFα, and granzyme B. Phenotypic and transcriptomic analyses pointed to a key role of glycolysis in sustaining CTL activity in the tumor bed and identified the PHD2/HIF-1 pathway as a potential target for cancer immunotherapy.


Asunto(s)
Prolina Dioxigenasas del Factor Inducible por Hipoxia , Neoplasias , Humanos , Procolágeno-Prolina Dioxigenasa/metabolismo , Oxígeno , Linfocitos T CD8-positivos/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Microambiente Tumoral
5.
JCI Insight ; 6(5)2021 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-33497366

RESUMEN

Cancer is caused primarily by genomic alterations resulting in deregulation of gene regulatory circuits in key growth, apoptosis, or DNA repair pathways. Multiple genes associated with the initiation and development of tumors are also regulated at the level of mRNA decay, through the recruitment of RNA-binding proteins to AU-rich elements (AREs) located in their 3'-untranslated regions. One of these ARE-binding proteins, tristetraprolin (TTP; encoded by Zfp36), is consistently dysregulated in many human malignancies. Herein, using regulated overexpression or conditional ablation in the context of cutaneous chemical carcinogenesis, we show that TTP represents a critical regulator of skin tumorigenesis. We provide evidence that TTP controlled both tumor-associated inflammation and key oncogenic pathways in neoplastic epidermal cells. We identify Areg as a direct target of TTP in keratinocytes and show that EGFR signaling potentially contributed to exacerbated tumor formation. Finally, single-cell RNA-Seq analysis indicated that ZFP36 was downregulated in human malignant keratinocytes. We conclude that TTP expression by epidermal cells played a major role in the control of skin tumorigenesis.


Asunto(s)
Carcinogénesis/metabolismo , Queratinocitos/metabolismo , Neoplasias Cutáneas/metabolismo , Piel/metabolismo , Tristetraprolina/metabolismo , Regiones no Traducidas 3' , Elementos Ricos en Adenilato y Uridilato , Animales , Carcinogénesis/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo , Receptores ErbB/metabolismo , Redes Reguladoras de Genes , Humanos , Inflamación/metabolismo , Ratones Endogámicos C57BL , Estabilidad del ARN , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Neoplasias Cutáneas/genética
6.
JCI Insight ; 5(11)2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32369450

RESUMEN

Tumor-associated macrophages (TAMs) contribute to the maintenance of a strong immunosuppressive environment, supporting tumor progression and resistance to treatment. To date, the mechanisms that drive acquisition of these immunosuppressive features are still poorly defined. Heme oxygenase-1 (HO-1) is the rate-limiting enzyme that catabolizes free heme. It displays important cytoprotective, antiinflammatory, and antioxidant properties. A growing body of evidence suggests that HO-1 may also promote tumor development. Herein, we show that HO-1 is highly expressed in monocytic cells in the tumor microenvironment (TME) once they differentiate into TAMs. Deletion of HO-1 in the myeloid compartment enhances the beneficial effects of a therapeutic antitumor vaccine by restoring CD8+ T cell proliferation and cytotoxicity. We further show that induction of HO-1 plays a major role in monocyte education by tumor cells by modulating their transcriptional and epigenetic programs. These results identify HO-1 as a valuable therapeutic target to reprogram the TME and synergize with current cancer therapies to facilitate antitumor response.


Asunto(s)
Hemo-Oxigenasa 1/inmunología , Tolerancia Inmunológica , Proteínas de la Membrana/inmunología , Neoplasias/inmunología , Microambiente Tumoral/inmunología , Macrófagos Asociados a Tumores/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Hemo-Oxigenasa 1/genética , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Neoplasias/genética , Microambiente Tumoral/genética , Macrófagos Asociados a Tumores/patología
7.
J Hepatol ; 73(2): 303-314, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32145259

RESUMEN

BACKGROUND & AIMS: Severe forms of alcohol-related liver disease are associated with increased susceptibility to infections which are associated with poor prognosis. The cellular and molecular mechanisms responsible for this altered host defense are incompletely understood. METHODS: We performed whole blood phenotypic analysis and ex vivo stimulation with various pathogen-associated molecular patterns (PAMPs). We included 34 patients with alcohol-related cirrhosis (18 of whom had biopsy-proven severe alcoholic hepatitis [sAH]), 12 healthy controls and 11 patients with chronic alcohol consumption without significant liver disease. We also evaluated the transcriptomic (RNA-seq) and chromatin accessibility (ATAC-seq) profiles of CD14+ monocytes from a subset of patients. RESULTS: Circulating monocytes and conventional dendritic cells (DCs) from patients with sAH displayed complex alterations characterized by increased expression of both activating and inhibitory surface markers and an impaired pro-inflammatory response upon stimulation with PAMPs representative of gram-negative bacteria (lipopolysaccharide, Pam3CSK4) or fungal pathogens (Zymosan). Their decreased ability to produce more than 1 cytokine (polyfunctionality) upon PAMP stimulation correlated with the risk of developing infection at 28 days or mortality at 90 days. The presence of acute-on-chronic liver failure in patients with sAH did not significantly modify the immune profile of monocytes and DCs. Moreover, CD14+ monocytes of patients with sAH displayed altered transcriptional and epigenomic profiles characterized by downregulation of key innate immune and metabolic pathways and upregulation of important immunomodulatory factors. CONCLUSIONS: In patients with sAH, the altered transcriptional program and functional properties of monocytes that contribute to patients' susceptibility to infection have strong epigenetic determinants. LAY SUMMARY: Patients with severe alcoholic hepatitis are at increased risk of infections, which contribute to the poor prognosis associated with the disease. Herein, we show that epigenetic determinants underly the immune cell dysfunction and inappropriate responses to pathogens that are associated with severe alcoholic hepatitis.


Asunto(s)
Citocinas/metabolismo , Epigénesis Genética , Hepatitis Alcohólica , Infecciones , Receptores de Lipopolisacáridos/análisis , Monocitos/inmunología , Biopsia/métodos , Células Dendríticas/inmunología , Progresión de la Enfermedad , Susceptibilidad a Enfermedades/epidemiología , Regulación hacia Abajo , Femenino , Bacterias Gramnegativas/aislamiento & purificación , Hepatitis Alcohólica/sangre , Hepatitis Alcohólica/inmunología , Hepatitis Alcohólica/mortalidad , Hepatitis Alcohólica/patología , Humanos , Infecciones/epidemiología , Infecciones/microbiología , Hígado/patología , Masculino , Pronóstico , Medición de Riesgo/métodos
8.
Front Immunol ; 11: 604785, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33613525

RESUMEN

c-Jun N-terminal protein kinase 1 (JNK1) is involved in multiple biological processes but its implication in inflammatory skin diseases is still poorly defined. Herein, we studied the role of JNK1 in the context of Aldara®-induced skin inflammation. We observed that constitutive ablation of JNK1 reduced Aldara®-induced acanthosis and expression of inflammatory markers. Conditional deletion of JNK1 in myeloid cells led to reduced skin inflammation, a finding that was associated with impaired Aldara®-induced inflammasome activation in vitro. Next, we evaluated the specific role of JNK1 in epidermal cells. We observed reduced Aldara®-induced acanthosis despite similar levels of inflammatory markers. Transcriptomic and epigenomic analysis of keratinocytes revealed the potential involvement of JNK1 in the EGFR signaling pathway. Finally, we show that inhibition of the EGFR pathway reduced Aldara®-induced acanthosis. Taken together, these data indicate that JNK1 plays a dual role in the context of psoriasis by regulating the production of inflammatory cytokines by myeloid cells and the sensitivity of keratinocytes to EGFR ligands. These results suggest that JNK1 could represent a valuable therapeutic target in the context of psoriasis.


Asunto(s)
Receptores ErbB/metabolismo , Queratinocitos/enzimología , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Células Mieloides/enzimología , Psoriasis/enzimología , Piel/enzimología , Animales , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Epigenoma , Receptores ErbB/genética , Femenino , Imiquimod , Mediadores de Inflamación/metabolismo , Queratinocitos/inmunología , Queratinocitos/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 8 Activada por Mitógenos/genética , Células Mieloides/inmunología , Psoriasis/inducido químicamente , Psoriasis/inmunología , Psoriasis/patología , Transducción de Señal , Piel/inmunología , Piel/patología , Transcriptoma
9.
Commun Biol ; 2: 472, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31872076

RESUMEN

Monocytes play a major role in the defense against pathogens. They are rapidly mobilized to inflamed sites where they exert both proinflammatory and regulatory effector functions. It is still poorly understood how this dynamic and exceptionally plastic system is controlled at the molecular level. Herein, we evaluated the differentiation process that occurs in Ly6Chi monocytes during oral infection by Toxoplasma gondii. Flow cytometry and single-cell analysis revealed distinct activation status and gene expression profiles in the bone marrow, the spleen and the lamina propria of infected mice. We provide further evidence that acquisition of effector functions, such as the capacity to produce interleukin-27, is accompanied by distinct waves of epigenetic programming, highlighting a role for STAT1/IRF1 in the bone marrow and AP-1/NF-κB in the periphery. This work broadens our understanding of the molecular events that occur in vivo during monocyte differentiation in response to inflammatory cues.


Asunto(s)
Diferenciación Celular/inmunología , Monocitos/inmunología , Toxoplasma/inmunología , Toxoplasmosis/inmunología , Toxoplasmosis/parasitología , Animales , Reprogramación Celular/genética , Biología Computacional/métodos , Epigénesis Genética , Perfilación de la Expresión Génica , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Monocitos/citología , Monocitos/metabolismo , Análisis de la Célula Individual , Toxoplasmosis/genética , Toxoplasmosis/metabolismo
10.
Nat Commun ; 10(1): 3306, 2019 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-31341159

RESUMEN

Memory CD8+ T cells have the ability to provide lifelong immunity against pathogens. Although memory features generally arise after challenge with a foreign antigen, naïve CD8 single positive (SP) thymocytes may acquire phenotypic and functional characteristics of memory cells in response to cytokines such as interleukin-4. This process is associated with the induction of the T-box transcription factor Eomesodermin (EOMES). However, the underlying molecular mechanisms remain ill-defined. Using epigenomic profiling, we show that these innate memory CD8SP cells acquire only a portion of the active enhancer repertoire of conventional memory cells. This reprograming is secondary to EOMES recruitment, mostly to RUNX3-bound enhancers. Furthermore, EOMES is found within chromatin-associated complexes containing BRG1 and promotes the recruitment of this chromatin remodelling factor. Also, the in vivo acquisition of EOMES-dependent program is BRG1-dependent. In conclusion, our results support a strong epigenetic basis for the EOMES-driven establishment of CD8+ T cell innate memory program.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/fisiología , ADN Helicasas/fisiología , Epigénesis Genética , Memoria Inmunológica , Proteínas Nucleares/fisiología , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/fisiología , Animales , Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , ADN Helicasas/inmunología , ADN Helicasas/metabolismo , Femenino , Perfilación de la Expresión Génica , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas Nucleares/inmunología , Proteínas Nucleares/metabolismo , Proteínas de Dominio T Box/genética , Factores de Transcripción/inmunología , Factores de Transcripción/metabolismo
11.
Front Immunol ; 10: 3007, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31998303

RESUMEN

RORγt-expressing Tregs form a specialized subset of intestinal CD4+ Foxp3+ cells which is essential to maintain gut homeostasis and tolerance to commensal microbiota. Recently, c-Maf emerged as a critical factor in the regulation of RORγt expression in Tregs. However, aside from c-Maf signaling, the signaling pathways involved in the differentiation of RORγt+ Tregs and their possible interplay with c-Maf in this process are largely unknown. We show that RORγt+ Treg development is controled by positive as well as negative signals. Along with c-Maf signaling, signals derived from a complex microbiota, as well as IL-6/STAT3- and TGF-ß-derived signals act in favor of RORγt+ Treg development. Ectopic expression of c-Maf did not rescue RORγt expression in STAT3-deficient Tregs, indicating the presence of additional effectors downstream of STAT3. Moreover, we show that an inflammatory IFN-γ/STAT1 signaling pathway acts as a negative regulator of RORγt+ Treg differentiation in a c-Maf independent fashion. These data thus argue for a complex integrative signaling network that finely tunes RORγt expression in Tregs. The finding that type 1 inflammation impedes RORγt+ Treg development even in the presence of an active IL-6/STAT3 pathway further suggests a dominant negative effect of STAT1 over STAT3 in this process.


Asunto(s)
Diferenciación Celular/genética , Diferenciación Celular/inmunología , Expresión Génica , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Transducción de Señal , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Inmunofenotipificación , Ratones , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-maf/genética , Proteínas Proto-Oncogénicas c-maf/metabolismo , Factor de Transcripción STAT3/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/citología , Factor de Crecimiento Transformador beta/metabolismo
12.
Elife ; 72018 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-29488879

RESUMEN

Cytotoxic CD4 (CD4CTX) T cells are emerging as an important component of antiviral and antitumor immunity, but the molecular basis of their development remains poorly understood. In the context of human cytomegalovirus infection, a significant proportion of CD4 T cells displays cytotoxic functions. We observed that the transcriptional program of these cells was enriched in CD8 T cell lineage genes despite the absence of ThPOK downregulation. We further show that establishment of CD4CTX-specific transcriptional and epigenetic programs occurred in a stepwise fashion along the Th1-differentiation pathway. In vitro, prolonged activation of naive CD4 T cells in presence of Th1 polarizing cytokines led to the acquisition of perforin-dependent cytotoxic activity. This process was dependent on the Th1 transcription factor Runx3 and was limited by the sustained expression of ThPOK. This work elucidates the molecular program of human CD4CTX T cells and identifies potential targets for immunotherapy against viral infections and cancer.


Asunto(s)
Diferenciación Celular , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Infecciones por Citomegalovirus/inmunología , Proteínas de Unión al ADN/metabolismo , Subgrupos de Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología , Células TH1/inmunología , Factores de Transcripción/metabolismo , Adulto , Animales , Células Cultivadas , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Persona de Mediana Edad
13.
Blood ; 113(14): 3172-81, 2009 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-19182210

RESUMEN

Retinoids triggers differentiation of acute promyelocytic leukemia (APL) blasts by transcriptional regulation of myeloid regulatory genes. Using a microarray approach, we have identified a novel retinoid-responsive gene (CXXC5) encoding a nuclear factor, retinoid-inducible nuclear factor (RINF), that contains a CXXC-type zinc-finger motif. RINF expression correlates with retinoid-induced differentiation of leukemic cells and with cytokine-induced myelopoiesis of normal CD34(+) progenitors. Furthermore, short hairpin RNA (shRNA) interference suggests for this gene a regulatory function in both normal and tumoral myelopoiesis. Interestingly, RINF localizes to 5q31.3, a small region often deleted in myeloid leukemia (acute myeloid leukemia [AML]/myelodysplasia [MDS]) and suspected to harbor one or several tumor suppressor gene.


Asunto(s)
Proteínas Portadoras/fisiología , Neoplasias Hematológicas/genética , Péptidos y Proteínas de Señalización Intracelular/fisiología , Mielopoyesis/genética , Secuencia de Aminoácidos , Proteínas Portadoras/genética , Proteínas de Unión al ADN , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Células Precursoras de Granulocitos/efectos de los fármacos , Células Precursoras de Granulocitos/fisiología , Células HL-60 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Células K562 , Modelos Biológicos , Datos de Secuencia Molecular , Mielopoyesis/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos , Homología de Secuencia de Aminoácido , Factores de Transcripción , Tretinoina/farmacología , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA