Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Nature ; 626(8000): 864-873, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38326607

RESUMEN

Macrophage activation is controlled by a balance between activating and inhibitory receptors1-7, which protect normal tissues from excessive damage during infection8,9 but promote tumour growth and metastasis in cancer7,10. Here we report that the Kupffer cell lineage-determining factor ID3 controls this balance and selectively endows Kupffer cells with the ability to phagocytose live tumour cells and orchestrate the recruitment, proliferation and activation of natural killer and CD8 T lymphoid effector cells in the liver to restrict the growth of a variety of tumours. ID3 shifts the macrophage inhibitory/activating receptor balance to promote the phagocytic and lymphoid response, at least in part by buffering the binding of the transcription factors ELK1 and E2A at the SIRPA locus. Furthermore, loss- and gain-of-function experiments demonstrate that ID3 is sufficient to confer this potent anti-tumour activity to mouse bone-marrow-derived macrophages and human induced pluripotent stem-cell-derived macrophages. Expression of ID3 is therefore necessary and sufficient to endow macrophages with the ability to form an efficient anti-tumour niche, which could be harnessed for cell therapy in cancer.


Asunto(s)
Proteínas Inhibidoras de la Diferenciación , Macrófagos del Hígado , Neoplasias , Animales , Humanos , Ratones , Células de la Médula Ósea/citología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linaje de la Célula , Células Madre Pluripotentes Inducidas/citología , Proteínas Inhibidoras de la Diferenciación/deficiencia , Proteínas Inhibidoras de la Diferenciación/genética , Proteínas Inhibidoras de la Diferenciación/metabolismo , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Macrófagos del Hígado/citología , Macrófagos del Hígado/inmunología , Macrófagos del Hígado/metabolismo , Hígado/inmunología , Hígado/patología , Activación de Macrófagos , Proteínas de Neoplasias , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/terapia , Fagocitosis
2.
Immunity ; 51(4): 655-670.e8, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31587991

RESUMEN

Tissue environment plays a powerful role in establishing and maintaining the distinct phenotypes of resident macrophages, but the underlying molecular mechanisms remain poorly understood. Here, we characterized transcriptomic and epigenetic changes in repopulating liver macrophages following acute Kupffer cell depletion as a means to infer signaling pathways and transcription factors that promote Kupffer cell differentiation. We obtained evidence that combinatorial interactions of the Notch ligand DLL4 and transforming growth factor-b (TGF-ß) family ligands produced by sinusoidal endothelial cells and endogenous LXR ligands were required for the induction and maintenance of Kupffer cell identity. DLL4 regulation of the Notch transcriptional effector RBPJ activated poised enhancers to rapidly induce LXRα and other Kupffer cell lineage-determining factors. These factors in turn reprogrammed the repopulating liver macrophage enhancer landscape to converge on that of the original resident Kupffer cells. Collectively, these findings provide a framework for understanding how macrophage progenitor cells acquire tissue-specific phenotypes.


Asunto(s)
Macrófagos del Hígado/fisiología , Hígado/metabolismo , Macrófagos/fisiología , Células Mieloides/fisiología , Animales , Diferenciación Celular , Células Cultivadas , Microambiente Celular , Reprogramación Celular , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hígado/citología , Receptores X del Hígado/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
3.
Nat Immunol ; 16(6): 642-52, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25915733

RESUMEN

Fungal infection stimulates the canonical C-type lectin receptor (CLR) signaling pathway via activation of the tyrosine kinase Syk. Here we identify a crucial role for the tyrosine phosphatase SHP-2 in mediating CLR-induced activation of Syk. Ablation of the gene encoding SHP-2 (Ptpn11; called 'Shp-2' here) in dendritic cells (DCs) and macrophages impaired Syk-mediated signaling and abrogated the expression of genes encoding pro-inflammatory molecules following fungal stimulation. Mechanistically, SHP-2 operated as a scaffold, facilitating the recruitment of Syk to the CLR dectin-1 or the adaptor FcRγ, through its N-SH2 domain and a previously unrecognized carboxy-terminal immunoreceptor tyrosine-based activation motif (ITAM). We found that DC-derived SHP-2 was crucial for the induction of interleukin 1ß (IL-1ß), IL-6 and IL-23 and anti-fungal responses of the TH17 subset of helper T cells in controlling infection with Candida albicans. Together our data reveal a mechanism by which SHP-2 mediates the activation of Syk in response to fungal infection.


Asunto(s)
Candidiasis/inmunología , Células Dendríticas/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Macrófagos/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Células Th17/inmunología , Secuencias de Aminoácidos/genética , Animales , Antígenos Fúngicos/inmunología , Células Cultivadas , Citocinas/metabolismo , Activación Enzimática , Mediadores de Inflamación/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Activación de Linfocitos , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Receptores de IgE/genética , Receptores de IgE/metabolismo , Transducción de Señal , Quinasa Syk
4.
Cell Commun Signal ; 18(1): 30, 2020 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-32093731

RESUMEN

BACKGROUND: Candida albicans is the most common opportunistic human fungal pathogen. The chemokine ligand CXCL1 plays a protective role in fungal infection through the recruitment of neutrophils. TRAF1 (tumor necrosis factor-associated factor 1) can be highly induced by proinflammatory stimuli such as LPS and TNF and has been implicated in septic shock. However, the role of TRAF1 in infection, especially fungal infection, remains elusive. Herein, we reveal that TRAF1 suppresses the antifungal immune response to Candida albicans intradermal infection through the regulation of CXCL1 induction and neutrophil recruitment. METHODS: A mouse model of C. albicans intradermal infection was established. The Traf1-/- mice and Traf1-/- immortalized human keratinocytes were generated. The p65 inhibitor triptolide, STAT1 inhibitor fludarabine, neutrophil-depletion antibody Ly6G, and neutralizing antibody for CXCL1 were utilized. The expression of proinflammatory cytokines and chemokines was assessed by real-time PCR and ELISA, and the activation of signaling molecules was analyzed by Western blotting. Hematoxylin and eosin staining and periodic acid Schiff staining were used for histology or fungal detection, respectively. The immunofluorescence and flow cytometry analyses were employed in the assessment of immune cell infiltration. Bone marrow transplantation and adoptive transfer experiments were conducted to establish a role for TRAF1 in the macrophage compartment in fungal skin infection. RESULTS: TRAF1-deficient mice demonstrated improved control of Candida albicans intradermal infection, and concomitant increase in neutrophil recruitment and reduction in fungal burden. The chemokine CXCL1 was upregulated in the TRAF1-deficient macrophages treated with heat-killed C. albicans. Mechanistically, TRAF1-deficient macrophages showed increased activation of transcription factor NFκB p65. The human CXCL8 was also highly induced in the TRAF1-deficient human keratinocytes upon TNF stimulation through decreasing the activation of transcription factor STAT1. TRAF1-deficient macrophages played a critical role in containing the C. albicans skin infection in vivo. CONCLUSION: TRAF1-deficient mice can better control fungal infection in the skin, a process attributable to the CXCL-neutrophil axis. Mechanistically, TRAF1 likely regulates CXCL1 expression in both macrophages and keratinocytes through the transcriptional factor NFκB and STAT1, respectively. Our finding offers new insight into the understanding of the immune regulatory mechanisms in host defense against C. albicans infection.


Asunto(s)
Candidiasis Cutánea/inmunología , Quimiocina CXCL1/inmunología , Neutrófilos , Piel/inmunología , Factor 1 Asociado a Receptor de TNF/inmunología , Animales , Células de la Médula Ósea , Femenino , Células HEK293 , Células HaCaT , Humanos , Macrófagos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/citología , Neutrófilos/inmunología , Piel/citología , Piel/patología
5.
Immunohorizons ; 5(2): 90-101, 2021 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-33597177

RESUMEN

Candida albicans is the most common, opportunistic human fungal pathogen whose complex interplay with the host innate immune system remains incompletely understood. In this study, we revealed that infection macrophages with C. albicans triggers prominent cell death, which is largely attributed to the RIPK3/MLKL-mediated necroptosis. Our results further demonstrated that the TSC1-mTOR pathway plays a pivotal role in the control of macrophage necroptosis upon engaging the Dectin-1/2 and TLR-2/4 pathways through fungal components ß-glucan/α-mannan or Sel1, respectively. Notably, the rapamycin-sensitive mTORC1 pathway, rather than the rapamycin-insensitive mTORC2 pathway, was responsible for elevated activation of RIPK1, RIPK3, and MLKL in TSC1-deficient macrophages. Following systemic infection with C. albicans, mice with macrophage/neutrophil-specific deletion of Tsc1 (Tsc1 M/N-/-) showed heightened fungal burden in multiple organs, such as the kidney, liver, and spleen, severe morbidity, and mortality. Notably, Tsc1 M/N-/- kidneys exhibited prominent cell death and concomitant loss of tissue-resident macrophages, which likely contributing to a dampened phagocytosis of fungal pathogens. Together, our data demonstrate a crucial role for the TSC1-mTOR pathway in the regulation of macrophage necroptosis and suggest that both Dectin- and TLRs-induced necroptosis may undermine the immune defense effector functions of these innate receptors during C. albicans infection.


Asunto(s)
Candidiasis/metabolismo , Macrófagos/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteína 1 del Complejo de la Esclerosis Tuberosa/metabolismo , Animales , Candida albicans , Candidiasis/patología , Candidiasis/prevención & control , Interacciones Huésped-Patógeno , Ratones , Ratones Endogámicos C57BL , Necroptosis , Fagocitosis , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo
6.
Nat Commun ; 10(1): 1015, 2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30833559

RESUMEN

Candida albicans can switch from commensal to pathogenic mode, causing mucosal or disseminated candidiasis. The host relies on pattern-recognition receptors including Toll-like receptors (TLRs) and C-type lectin receptors (CLRs) to sense invading fungal pathogens and launch immune defense mechanisms. However, the complex interplay between fungus and host innate immunity remains incompletely understood. Here we report that C. albicans upregulates expression of a small secreted cysteine-rich protein Sel1 upon encountering limited nitrogen and abundant serum. Sel1 activates NF-κB and MAPK signaling pathways, leading to expression of proinflammatory cytokines and chemokines. Comprehensive genetic and biochemical analyses reveal both TLR2 and TLR4 are required for the recognition of Sel1. Further, SEL1-deficient C. albicans display an impaired immune response in vivo, causing increased morbidity and mortality in a bloodstream infection model. We identify a critical component in the Candida-host interaction that opens a new avenue to tackle Candida infection and inflammation.


Asunto(s)
Candida albicans/patogenicidad , Candidiasis/inmunología , Proteínas Portadoras/inmunología , Proteínas Portadoras/metabolismo , Proteínas con Dominio LIM/inmunología , Proteínas con Dominio LIM/metabolismo , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 4/metabolismo , Animales , Proteínas Portadoras/genética , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inmunidad Innata , Inflamación/inmunología , Proteínas con Dominio LIM/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Células RAW 264.7 , Alineación de Secuencia , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética , Receptores Toll-Like/inmunología
7.
J Exp Med ; 215(10): 2536-2553, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30201786

RESUMEN

Tissue-resident macrophages can self-maintain without contribution of adult hematopoiesis. Herein we show that tissue-resident interstitial macrophages (Res-TAMs) in mouse lungs contribute to the pool of tumor-associated macrophages (TAMs) together with CCR2-dependent recruited macrophages (MoD-TAMs). Res-TAMs largely correlated with tumor cell growth in vivo, while MoD-TAMs accumulation was associated with enhanced tumor spreading. Both cell subsets were depleted after chemotherapy, but MoD-TAMs rapidly recovered and performed phagocytosis-mediated tumor clearance. Interestingly, anti-VEGF treatment combined with chemotherapy inhibited both Res and Mod-TAM reconstitution without affecting monocyte infiltration and improved its efficacy. Our results reveal that the developmental origin of TAMs dictates their relative distribution, function, and response to cancer therapies in lung tumors.


Asunto(s)
Neoplasias Pulmonares/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Fagocitosis , Animales , Neoplasias Pulmonares/patología , Macrófagos/patología , Ratones , Ratones Noqueados , Monocitos/patología , Receptores CCR2/inmunología
8.
J Exp Med ; 214(5): 1471-1491, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28416650

RESUMEN

TLR7/9 signals are capable of mounting massive interferon (IFN) response in plasmacytoid dendritic cells (pDCs) immediately after viral infection, yet the involvement of epigenetic regulation in this process has not been documented. Here, we report that zinc finger CXXC family epigenetic regulator CXXC5 is highly expressed in pDCs, where it plays a crucial role in TLR7/9- and virus-induced IFN response. Notably, genetic ablation of CXXC5 resulted in aberrant methylation of the CpG-containing island (CGI) within the Irf7 gene and impaired IRF7 expression in steady-state pDCs. Mechanistically, CXXC5 is responsible for the recruitment of DNA demethylase Tet2 to maintain the hypomethylation of a subset of CGIs, a process coincident with active histone modifications and constitutive transcription of these CGI-containing genes. Consequently, CXXC5-deficient mice had compromised early IFN response and became highly vulnerable to infection by herpes simplex virus and vesicular stomatitis virus. Together, our results identify CXXC5 as a novel epigenetic regulator for pDC-mediated antiviral response.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Células Dendríticas/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Glicoproteínas de Membrana/fisiología , Proteínas Proto-Oncogénicas/fisiología , Receptor Toll-Like 7/fisiología , Receptor Toll-Like 9/fisiología , Animales , Islas de CpG/fisiología , Metilación de ADN , Células Dendríticas/metabolismo , Dioxigenasas , Epigénesis Genética/fisiología , Herpes Simple/metabolismo , Interferones/fisiología , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción , Estomatitis Vesicular/metabolismo
9.
Int Immunopharmacol ; 11(5): 536-42, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21081189

RESUMEN

FOXP3(+)CD4(+)CD25(+) Regulatory T (Treg) cells and IL-17 producing helper T cells (Th17) are critical subsets of T cells which play essential roles in immune homeostasis. The Forkhead family transcription factor FOXP3 is predominantly expressed in Treg cells, where the FOXP3 ensemble is essential for Treg cell development and function. As FOXP3 is to Treg cells, the orphan retinoic acid nuclear receptor (ROR) family transcription factor RORγt is essential for Th17 development and function. In this review, we summarize recent progress of our understanding towards the molecular mechanisms underlying the differentiation and function of FOXP3(+) Treg cells and RORγt expressing Th17 cells. These may provide new insights into therapeutic intervention and targeting of human immune-deficient diseases.


Asunto(s)
Factores de Transcripción Forkhead/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Animales , Diferenciación Celular , Regulación de la Expresión Génica/inmunología , Humanos , Activación Transcripcional/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA