Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 754
Filtrar
1.
Cell Mol Life Sci ; 81(1): 380, 2024 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-39222120

RESUMEN

The cytokine IFNγ is a principal effector of macrophage activation and immune resistance to mycobacterial infection; however, pathogenic mycobacteria are capable of surviving in IFNγ-activated macrophages by largely unknown mechanisms. In this study, we find that pathogenic mycobacteria, including M. bovis BCG and M. tuberculosis can sense IFNγ to promote their proliferative activity and virulence phenotype. Moreover, interaction with the host intracellular environment increases the susceptibility of mycobacteria to IFNγ through upregulating expression of mmpL10, a mycobacterial IFNγ receptor, thereby facilitating IFNγ-dependent survival and growth of mycobacteria in macrophages. Transmission electron microscopy analysis reveals that IFNγ triggers the secretion of extracellular vesicles, an essential virulence strategy of intracellular mycobacteria, while proteomics identifies numerous pivotal IFNγ-induced effectors required for mycobacterial infection in macrophages. Our study suggests that sensing host IFNγ is a crucial virulence mechanism used by pathogenic mycobacteria to survive and proliferate inside macrophages.


Asunto(s)
Interferón gamma , Macrófagos , Ratones Endogámicos C57BL , Mycobacterium tuberculosis , Interferón gamma/metabolismo , Interferón gamma/inmunología , Macrófagos/microbiología , Macrófagos/metabolismo , Macrófagos/inmunología , Animales , Ratones , Mycobacterium tuberculosis/patogenicidad , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/metabolismo , Mycobacterium bovis/inmunología , Mycobacterium bovis/metabolismo , Humanos , Interacciones Huésped-Patógeno/inmunología , Virulencia , Receptores de Interferón/metabolismo , Receptores de Interferón/genética , Receptor de Interferón gamma , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/inmunología , Activación de Macrófagos , Infecciones por Mycobacterium/microbiología , Infecciones por Mycobacterium/inmunología , Infecciones por Mycobacterium/metabolismo , Infecciones por Mycobacterium/patología
2.
Nature ; 633(8029): 451-458, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39112706

RESUMEN

Cancer cells frequently alter their lipids to grow and adapt to their environment1-3. Despite the critical functions of lipid metabolism in membrane physiology, signalling and energy production, how specific lipids contribute to tumorigenesis remains incompletely understood. Here, using functional genomics and lipidomic approaches, we identified de novo sphingolipid synthesis as an essential pathway for cancer immune evasion. Synthesis of sphingolipids is surprisingly dispensable for cancer cell proliferation in culture or in immunodeficient mice but required for tumour growth in multiple syngeneic models. Blocking sphingolipid production in cancer cells enhances the anti-proliferative effects of natural killer and CD8+ T cells partly via interferon-γ (IFNγ) signalling. Mechanistically, depletion of glycosphingolipids increases surface levels of IFNγ receptor subunit 1 (IFNGR1), which mediates IFNγ-induced growth arrest and pro-inflammatory signalling. Finally, pharmacological inhibition of glycosphingolipid synthesis synergizes with checkpoint blockade therapy to enhance anti-tumour immune response. Altogether, our work identifies glycosphingolipids as necessary and limiting metabolites for cancer immune evasion.


Asunto(s)
Glicoesfingolípidos , Evasión Inmune , Neoplasias , Proteínas Proto-Oncogénicas p21(ras) , Escape del Tumor , Animales , Femenino , Ratones , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Proliferación Celular , Glicoesfingolípidos/biosíntesis , Glicoesfingolípidos/deficiencia , Glicoesfingolípidos/inmunología , Glicoesfingolípidos/metabolismo , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Receptor de Interferón gamma/metabolismo , Interferón gamma/inmunología , Células Asesinas Naturales/inmunología , Ratones Endogámicos C57BL , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Transducción de Señal , Lipidómica
3.
Nat Commun ; 15(1): 6613, 2024 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-39098861

RESUMEN

Tumour-host immune interactions lead to complex changes in the tumour microenvironment (TME), impacting progression, metastasis and response to therapy. While it is clear that cancer cells can have the capacity to alter immune landscapes, our understanding of this process is incomplete. Herein we show that endocytic trafficking at the plasma membrane, mediated by the small GTPase ARF6, enables melanoma cells to impose an immunosuppressive TME that accelerates tumour development. This ARF6-dependent TME is vulnerable to immune checkpoint blockade therapy (ICB) but in murine melanoma, loss of Arf6 causes resistance to ICB. Likewise, downregulation of ARF6 in patient tumours correlates with inferior overall survival after ICB. Mechanistically, these phenotypes are at least partially explained by ARF6-dependent recycling, which controls plasma membrane density of the interferon-gamma receptor. Collectively, our findings reveal the importance of endomembrane trafficking in outfitting tumour cells with the ability to shape their immune microenvironment and respond to immunotherapy.


Asunto(s)
Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP , Membrana Celular , Inhibidores de Puntos de Control Inmunológico , Melanoma , Microambiente Tumoral , Microambiente Tumoral/inmunología , Animales , Humanos , Ratones , Factores de Ribosilacion-ADP/metabolismo , Factores de Ribosilacion-ADP/genética , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Melanoma/genética , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Melanoma/patología , Melanoma/inmunología , Línea Celular Tumoral , Membrana Celular/metabolismo , Receptor de Interferón gamma , Receptores de Interferón/metabolismo , Receptores de Interferón/genética , Transporte de Proteínas , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Melanoma Experimental/genética , Ratones Endogámicos C57BL , Femenino
4.
J Autoimmun ; 148: 103279, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38972102

RESUMEN

B cells of people with multiple sclerosis (MS) are more responsive to IFN-γ, corresponding to their brain-homing potential. We studied how a coding single nucleotide polymorphism (SNP) in IFNGR2 (rs9808753) co-operates with Epstein-Barr virus (EBV) infection as MS risk factors to affect the IFN-γ signaling pathway in human B cells. In both cell lines and primary cells, EBV infection positively associated with IFN-γ receptor expression and STAT1 phosphorylation. The IFNGR2 risk SNP selectively promoted downstream signaling via STAT1, particularly in transitional B cells. Altogether, EBV and the IFNGR2 risk SNP independently amplify IFN-γ signaling, potentially driving B cells to enter the MS brain.


Asunto(s)
Linfocitos B , Infecciones por Virus de Epstein-Barr , Receptor de Interferón gamma , Interferón gamma , Esclerosis Múltiple , Polimorfismo de Nucleótido Simple , Receptores de Interferón , Transducción de Señal , Humanos , Esclerosis Múltiple/genética , Esclerosis Múltiple/inmunología , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Interferón gamma/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/genética , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT1/genética , Predisposición Genética a la Enfermedad , Herpesvirus Humano 4 , Femenino , Masculino , Fosforilación
5.
Nat Commun ; 15(1): 5506, 2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-38951527

RESUMEN

Obesity is a major cause of metabolic dysfunction-associated steatohepatitis (MASH) and is characterized by inflammation and insulin resistance. Interferon-γ (IFNγ) is a pro-inflammatory cytokine elevated in obesity and modulating macrophage functions. Here, we show that male mice with loss of IFNγ signaling in myeloid cells (Lyz-IFNγR2-/-) are protected from diet-induced insulin resistance despite fatty liver. Obesity-mediated liver inflammation is also attenuated with reduced interleukin (IL)-12, a cytokine primarily released by macrophages, and IL-12 treatment in vivo causes insulin resistance by impairing hepatic insulin signaling. Following MASH diets, Lyz-IFNγR2-/- mice are rescued from developing liver fibrosis, which is associated with reduced fibroblast growth factor (FGF) 21 levels. These results indicate critical roles for IFNγ signaling in macrophages and their release of IL-12 in modulating obesity-mediated insulin resistance and fatty liver progression to MASH. In this work, we identify the IFNγ-IL12 axis in regulating intercellular crosstalk in the liver and as potential therapeutic targets to treat MASH.


Asunto(s)
Hígado Graso , Resistencia a la Insulina , Interferón gamma , Interleucina-12 , Hígado , Macrófagos , Ratones Noqueados , Obesidad , Transducción de Señal , Animales , Interferón gamma/metabolismo , Interleucina-12/metabolismo , Masculino , Obesidad/metabolismo , Ratones , Hígado Graso/metabolismo , Hígado Graso/patología , Macrófagos/metabolismo , Hígado/metabolismo , Hígado/patología , Ratones Endogámicos C57BL , Dieta Alta en Grasa/efectos adversos , Receptores de Interferón/metabolismo , Receptores de Interferón/genética , Receptor de Interferón gamma , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Cirrosis Hepática/genética
6.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167314, 2024 10.
Artículo en Inglés | MEDLINE | ID: mdl-38936516

RESUMEN

The integrity of the lymphatic system is critical for preventing the dissemination of tumor cells, such as melanoma, to distant parts of the body. IFN-γ is well studied as a negative regulator for lymphangiogenesis, which is strongly associated with cancer metastasis. However, the exact mechanisms underlying this process remain unclear. In the present study, we investigated whether IFN-γ signaling in lymphatic endothelial cells (LECs) affects tumor cell dissemination by regulating the barrier function of tumor-associated lymphatic vessels. Using LEC-specific IFN-γ receptor (IFN-γR) knockout mice, we found that the loss of IFN-γR in LECs increased the dissemination of melanoma cells into the draining lymph nodes. Notably, IFN-γ signaling in LECs inhibited trans-lymphatic endothelial cell migration of melanoma cells, indicating its regulation of lymphatic barrier function. Further investigations revealed that IFN-γ upregulated the expression of the tight junction protein Claudin-3 in LECs, while knockdown of Claudin-3 in LECs abolished IFN-γ-induced inhibition of trans-lymphatic endothelial migration activity. Mechanistically, IFN-γ inhibits AMPK signaling activation, which is involved in the regulation of fatty acid metabolism. Modulating fatty acid metabolism and AMPK activation in LECs also affected the lymphatic dissemination of melanoma cells, further confirming that this process is involved in IFN-γ-induced regulation of lymphatic barrier function. These results provide novel insights into how IFN-γ modulates tight junctions in LECs, inhibiting the dissemination of melanoma cells via the lymphatic vessels.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Células Endoteliales , Interferón gamma , Melanoma , Ratones Noqueados , Animales , Interferón gamma/metabolismo , Ratones , Células Endoteliales/metabolismo , Células Endoteliales/patología , Proteínas Quinasas Activadas por AMP/metabolismo , Melanoma/patología , Melanoma/metabolismo , Movimiento Celular , Transducción de Señal , Receptor de Interferón gamma , Receptores de Interferón/metabolismo , Receptores de Interferón/genética , Metástasis Linfática , Línea Celular Tumoral , Linfangiogénesis , Humanos , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patología , Ratones Endogámicos C57BL
7.
J Am Chem Soc ; 146(26): 17691-17699, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38888290

RESUMEN

Nonproteinogenic amino acids, including d-α-, ß-, and γ-amino acids, present in bioactive peptides play pivotal roles in their biochemical activities and proteolytic stabilities. d-α-Amino acids (dαAA) are widely used building blocks that can enhance the proteolytic stability. Cyclic ß2,3-amino acids (cßAA), for instance, can fold peptides into rigid secondary structures, improving the binding affinity and proteolytic stability. Cyclic γ2,4-amino acids (cγAA) are recently highlighted as rigid residues capable of preventing the proteolysis of flanking residues. Simultaneous incorporation of all dαAA, cßAA, and cγAA into a peptide is expected to yield l-α/d-α/ß/γ-hybrid peptides with improved stability and potency. Despite challenges in the ribosomal incorporation of multiple nonproteinogenic amino acids, our engineered tRNAPro1E2 successfully reaches such a difficulty. Here, we report the ribosomal synthesis of macrocyclic l-α/d-α/ß/γ-hybrid peptide libraries and their application to in vitro selection against interferon gamma receptor 1 (IFNGR1). One of the resulting l-α/d-α/ß/γ-hybrid peptides, IB1, exhibited remarkable inhibitory activity against the IFN-γ/IFNGR1 protein-protein interaction (PPI) (IC50 = 12 nM), primarily attributed to the presence of a cßAA in the sequence. Additionally, cγAAs and dαAAs in the resulting peptides contributed to their serum stability. Furthermore, our peptides effectively inhibit IFN-γ/IFNGR1 PPI at the cellular level (best IC50 = 0.75 µM). Altogether, our platform expands the chemical space available for exploring peptides with high activity and stability, thereby enhancing their potential for drug discovery.


Asunto(s)
Receptor de Interferón gamma , Interferón gamma , Receptores de Interferón , Interferón gamma/metabolismo , Receptores de Interferón/metabolismo , Receptores de Interferón/química , Humanos , Unión Proteica , Compuestos Macrocíclicos/química , Compuestos Macrocíclicos/farmacología , Péptidos/química , Péptidos/farmacología , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacología , Péptidos Cíclicos/metabolismo
8.
PLoS Pathog ; 20(5): e1011820, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38718306

RESUMEN

The production of IFN-γ is crucial for control of multiple enteric infections, but its impact on intestinal epithelial cells (IEC) is not well understood. Cryptosporidium parasites exclusively infect epithelial cells and the ability of interferons to activate the transcription factor STAT1 in IEC is required for parasite clearance. Here, the use of single cell RNA sequencing to profile IEC during infection revealed an increased proportion of mid-villus enterocytes during infection and induction of IFN-γ-dependent gene signatures that was comparable between uninfected and infected cells. These analyses were complemented by in vivo studies, which demonstrated that IEC expression of the IFN-γ receptor was required for parasite control. Unexpectedly, treatment of Ifng-/- mice with IFN-γ showed the IEC response to this cytokine correlates with a delayed reduction in parasite burden but did not affect parasite development. These data sets provide insight into the impact of IFN-γ on IEC and suggest a model in which IFN-γ signalling to uninfected enterocytes is important for control of Cryptosporidium.


Asunto(s)
Criptosporidiosis , Interferón gamma , Mucosa Intestinal , Ratones Noqueados , Animales , Interferón gamma/metabolismo , Interferón gamma/inmunología , Criptosporidiosis/inmunología , Criptosporidiosis/parasitología , Ratones , Mucosa Intestinal/parasitología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/inmunología , Cryptosporidium , Células Epiteliales/parasitología , Células Epiteliales/metabolismo , Células Epiteliales/inmunología , Enterocitos/parasitología , Enterocitos/metabolismo , Enterocitos/inmunología , Ratones Endogámicos C57BL , Receptor de Interferón gamma , Factor de Transcripción STAT1/metabolismo , Receptores de Interferón/metabolismo , Receptores de Interferón/genética , Transducción de Señal
9.
Oral Oncol ; 145: 106524, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37482043

RESUMEN

OBJECTIVE: The aim of this study is to evaluate the role of FRCs regulated by cancer cell-derived extracellular vesicles (CEVs) played in pre-metastatic niche (PMN) formation of lymph node (LN). MATERIALS AND METHODS: The FRCs in sixty fresh cervical LNs from 20 patients were evaluated by flow cytometric analysis. Cells in LN with or without metastasis were analyzed by single-cell RNA sequencing (scRNA-seq). CEVs were isolated from the culture supernatant of primarily cultured cancer cells and cocultured with FRCs. Mass Spectrometry was used to identify LN metastasis related protein in CEVs. The activation of IFNGR1/JAK1/STAT1-activated-PD-L1 pathway in FRCs was detected by western blotting. FRCs were co-cultured with CD8+ T lymphocytes to confirm the cytotoxicity assay of FRCs. RESULTS: The proportion of fibroblastic reticular cells (FRCs) was significantly higher in micro-metastatic LN in head and neck squamous cell carcinoma patients (HNSCC, p < 0.05) and scRNA-seq analysis further showed a high focus of extracellular vesicles-related pathway on FRCs in LN with metastasis (p < 0.05). Interferon gamma receptor 1 (IFNGR1) in CEVs can be engulfed by FRCs and promote PD-L1 expression on FRCs via JAK1-STAT1 pathway, resulting in an increased CD8+ T cell exhaustion. CONCLUSION: IFNGR1, originated from cancer cell-derived extracellular vesicles, promote PD-L1 expression on FRCs and subsequent CD8+ T cell exhaustion via JAK1-STAT1 activation, which facilitate pre-metastatic niche formation and tumor metastasis in sentinel lymph node in HNSCC.


Asunto(s)
Vesículas Extracelulares , Neoplasias de Cabeza y Cuello , Humanos , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Antígeno B7-H1/metabolismo , Neoplasias de Cabeza y Cuello/patología , Ganglios Linfáticos/patología , Vesículas Extracelulares/metabolismo , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Janus Quinasa 1/metabolismo , Receptor de Interferón gamma
10.
J Biol Chem ; 299(8): 104990, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37392850

RESUMEN

Cycloartenyl ferulate (CF) is abundant in brown rice with multiple biologic functions. It has been reported to possess antitumor activity; however, the related mechanism of action of CF has not been clarified. Herein, we unexpectedly uncover the immunological regulation effects of CF and its molecular mechanism. We discovered that CF directly enhanced the killing capacity of natural killer (NK) cells for various cancer cells in vitro. In vivo, CF also improved cancer surveillance in mouse models of lymphoma clearance and metastatic melanoma dependent on NK cells. In addition, CF promoted anticancer efficacy of the anti-PD1 antibody with improvement of tumor immune microenvironment. Mechanistically, we first unveiled that CF acted on the canonical JAK1/2-STAT1 signaling pathway to enhance the immunity of the NK cells by selectively binding to interferon γ receptor 1. Collectively, our results indicate that CF is a promising immunoregulation agent worthy of attention in clinical application in the future. Due to broad biological significance of interferon γ, our findings also provide a capability to understand the diverse functions of CF.


Asunto(s)
Ácidos Cumáricos , Células Asesinas Naturales , Neoplasias , Receptores de Interferón , Animales , Ratones , Interferón gamma/inmunología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Neoplasias/inmunología , Microambiente Tumoral , Ácidos Cumáricos/farmacología , Receptores de Interferón/inmunología , Receptor de Interferón gamma
11.
BMC Pediatr ; 23(1): 278, 2023 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-37277724

RESUMEN

BACKGROUND: Interferon-gamma receptor deficiency is a heterogeneous spectrum of disease which involves mutations in IFNGR1, IFNGR2 genes, and the downstream signaling proteins such as STAT1. These mutations are associated with immunodeficiency 27 A and 27B, making the patient prone to mycobacterial infections. Patients with this condition are also at increased risk for affliction with viral and bacterial infections, such as with the Herpesviridae family, Listeria, and Salmonella. Moreover, SH2B3 mutation is associated with autoimmune and lymphoproliferative conditions. CASE PRESENTATION: the patient was a 19-month-old infant girl who presented with a two-week history of fever. She had near-normal flowcytometry with high IgM and IgE. She had pneumonic infiltration in her chest and right hilar and para-aortic lymphadenopathy. PCR of whole blood for Aspergillus fumigatus came back positive. In her Whole Exome Sequencing she had IFNGR1 and SH2B3 mutations. CONCLUSION: systemic fungal infections such as Aspergillosis can occur in patients with interferon-gamma receptor one deficiency. This type of immunodeficiency should be considered in treating patients with systemic Aspergillosis.


Asunto(s)
Aspergilosis , Síndromes de Inmunodeficiencia , Lactante , Femenino , Humanos , Interferón gamma/genética , Aspergilosis/diagnóstico , Aspergilosis/genética , Receptores de Interferón/genética , Síndromes de Inmunodeficiencia/complicaciones , Síndromes de Inmunodeficiencia/genética , Receptor de Interferón gamma
12.
Cytokine ; 169: 156274, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37327531

RESUMEN

Interferon-gamma (IFN-γ) is a pro-inflammatory cytokine playing essential role in immunity against tuberculosis (TB). IFN-γ performs function by binding to its receptor complex, consisting of two polypeptide chains viz. IFN-γ receptor 1 (IFN-γR1) and IFN-γ receptor 2 (IFN-γR2). Structural and functional deficiencies in IFN-γR1 can make individual vulnerable to even weak mycobacterial infections. Studies from different populations of the world have reported the association of single nucleotide polymorphisms (SNPs) present in IFNGR1 gene with TB, however, there are no such studies from India. Thus, the present study was designed to analyse the association of rs2234711 (C/T), rs7749390 (C/T) and rs1327475 (C/T) SNPs of IFNGR1 with TB in the population of North India. For the present study, 263 TB patients (at zero day of anti-tuberculosis therapy) and 256 healthy controls (HCs) were recruited. The genotyping of selected SNPs was done by high-resolution melting (HRM) curve analysis. The mRNA and surface expression data of IFNGR1 was extracted from our previous study and was grouped according to the genotypes of studied SNPs. The genotype 'TT' and 'T' allele of SNP rs2234711 (C/T) were found to be associated with TB in studied population ['T' vs 'C': OR (CI) = 1.79 (1.39-2.29); p-value < 0.0001]. The haplotypes 'C-C-C' of rs2234711-rs7749390-rs1327475 confers protection, while haplotype 'T-C-C' is a risk factor for TB in studied population. It was also found that 'TT' genotype of rs2234711 in HCs is associated with lower surface expression of IFNGR1 (p-value = 0.0078). In conclusion, 'TT' genotype is associated with lower surface expression of IFNGR1 and is increasing the susceptibility to TB in North Indian population.


Asunto(s)
Predisposición Genética a la Enfermedad , Polimorfismo de Nucleótido Simple , Receptores de Interferón , Tuberculosis Pulmonar , Humanos , Estudios de Casos y Controles , Genotipo , Interferón gamma/genética , Polimorfismo de Nucleótido Simple/genética , Receptores de Interferón/genética , Tuberculosis Pulmonar/genética , Receptor de Interferón gamma
13.
J Clin Invest ; 133(3)2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36445781

RESUMEN

The absence of IFN-γ receptor (IFN-γR) or STAT1 signaling in donor cells has been shown to result in reduced induction of acute graft-versus-host disease (GVHD). In this study, we unexpectedly observed increased activation and expansion of donor lymphocytes in both lymphohematopoietic organs and GVHD target tissues of IFN-γR/STAT1-deficient recipient mice, leading to rapid mortality following the induction of GVHD. LPS-matured, BM-derived Ifngr1-/- Stat1-/- DCs (BMDCs) were more potent allogeneic stimulators and expressed increased levels of MHC II and costimulatory molecules. Similar effects were observed in human antigen-presenting cells (APCs) with knockdown of Stat1 by CRISPR/Cas9 and treatment with a JAK1/2 inhibitor. Furthermore, we demonstrated that the absence of IFN-γR/STAT1 signaling in hematopoietic APCs impaired the presentation of exogenous antigens, while promoting the presentation of endogenous antigens. Thus, the indirect presentation of host antigens to donor lymphocytes was defective in IFN-γR/STAT1-deficient, donor-derived APCs in fully donor chimeric mice. The differential effects of IFN-γR/STAT1 signaling on endogenous and exogenous antigen presentation could provide further insight into the roles of the IFN-γ/STAT1 signaling pathway in the pathogenesis of GVHD, organ rejection, and autoimmune diseases.


Asunto(s)
Células Presentadoras de Antígenos , Enfermedad Injerto contra Huésped , Ratones , Humanos , Animales , Receptores de Interferón/genética , Enfermedad Injerto contra Huésped/genética , Transducción de Señal , Trasplante de Médula Ósea/efectos adversos , Ratones Endogámicos C57BL , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Receptor de Interferón gamma
14.
Biomed Pharmacother ; 155: 113683, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36095965

RESUMEN

IFN-γ, a soluble cytokine being produced by T lymphocytes, macrophages, mucosal epithelial cells, or natural killer cells, is able to bind to the IFN-γ receptor (IFNγR) and in turn activate the Janus kinase (JAK)-signal transducer and transcription protein (STAT) pathway and induce expression of IFN-γ-stimulated genes. IFN-γ is critical for innate and adaptive immunity and aberrant IFN-γ expression and functions have been associated with different human diseases. However, the IFN-γ/IFNγR signaling could be a double-edged sword in cancer development because the tissue microenvironments could determine its anti- or pro-tumorigenic activities. The IFNγR protein consists of two IFNγR1 and IFNγR2 chains, subunits of which play different roles under certain conditions. This review assessed IFNγR polymorphisms, expression and functions in development and progression of various human diseases in an IFN-γ-dependent or independent manner. This review also discussed tumor microenvironment, microbial infection, and vital molecules in the IFN-γ upstream signaling that might regulate IFNγR expression, drug resistance, and druggable strategy, to provide evidence for further application of IFNγR.


Asunto(s)
Interferón gamma , Neoplasias , Humanos , Interferón gamma/metabolismo , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Quinasas Janus/metabolismo , Neoplasias/genética , Factor de Transcripción STAT1/metabolismo , Microambiente Tumoral , Receptor de Interferón gamma
15.
Clin Breast Cancer ; 22(7): 674-680, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35918221

RESUMEN

BACKGROUND: A large number of breast cancer survivors suffer from psychological distress. The purpose of this study was to investigate the association between genetic variations in Chinese breast cancer patients and anxiety or depression, and to screen patients who are susceptible to psychological problems. METHODS: A total of 300 early-stage breast cancer patients were recruited in this prospective observational single-center cohort study. With reference to the previous literature and the mechanism concerning anxiety and depression, 9 candidate genes and 29 single-nucleotide polymorphisms (SNPs) loci were selected. The association between SNP variations and anxiety/depression were analyzed. RESULTS: After we incorporated meaningful clinicopathological and demographic factors, multivariate analysis showed that the A/G and G/G genotypes of IFNGR1 (rs2234711) and the T/C and T/T genotypes of BDNF (rs6265) were significantly associated with depression (HR 3.10, P = .008; HR 2.04, P = .03). The G/A and G/G genotypes of IL-10 (rs1554286) remained independent predictors of anxiety (HR 1.85, P = .019). CONCLUSIONS: These findings suggested that variations in IL-10, IFNGR1 and BDNF were associated with anxious/depressive symptoms in early-stage breast cancer patients in China, which could help identify patients at high risk for psychological problems.


Asunto(s)
Ansiedad , Neoplasias de la Mama , Depresión , Femenino , Humanos , Ansiedad/epidemiología , Ansiedad/genética , Factor Neurotrófico Derivado del Encéfalo/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/psicología , Estudios de Cohortes , Depresión/epidemiología , Depresión/genética , Interleucina-10/genética , Polimorfismo de Nucleótido Simple , Receptores de Interferón , Receptor de Interferón gamma
16.
Cytokine ; 157: 155956, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35785668

RESUMEN

IFN-γ, a type 2 interferon and a cytokine, is critical for both innate and adaptive immunity. IFN-γ binds to the IFN-γRs on the cell membrane of macrophages, signals through JAK1-STAT-1 pathway and induces IFN-γ-stimulated genes (ISGs). As Leishmania amastigotes reside and replicate within macrophages, IFN-γ mediated macrophage activation eventuate in Leishmania elimination. As befits the principle of parasitism, the impaired IFN-γ responsiveness in macrophages ensures Leishmania survival. IFN-γ responsiveness is a function of integrated molecular events at multiple levels in the cells that express IFN-γ receptors. In Leishmania-infected macrophages, reduced IFN-γRα expression, impaired IFN-γRα and IFN-γRß hetero-dimerization due to altered membrane lipid composition, reduced JAK-1 and STAT-1 phosphorylation but increased STAT-1 degradation and impaired ISGs induction collectively determine the IFN-γ responsiveness and the efficacy of IFN-γ induced antileishmanial function of macrophages. Therefore, parasite load is not only decided by the levels of IFN-γ produced but also by the IFN-γ responsiveness. Indeed, in Leishmania-infected patients, IFN-γ is produced but IFN-γ signalling is downregulated. However, the molecular mechanisms of IFN-γ responsiveness remain unclear. Therefore, we review the current understanding of IFN-γ responsiveness of Leishmania-infected macrophages.


Asunto(s)
Leishmania , Humanos , Interferón gamma , Macrófagos/metabolismo , Receptores de Interferón , Transducción de Señal , Receptor de Interferón gamma
17.
Cytokine ; 156: 155892, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35653895

RESUMEN

The intestinal mucosa protects the body from physical damage, pathogens, and antigens. However, inflammatory bowel diseases (IBDs) patients suffer from poor mucosal tissue function, including the lack of an effective cellular and/or mucus barrier. We investigated the mucus producing human colonic epithelial cell line HT29-MTX E12 to study its suitability as an in vitro model of cell/mucus barrier adaption during IBD. It was found that the proinflammatory cytokine interferon-gamma (IFN-γ), but not tumor necrosis factor-alpha (TNF-α), reduced cell viability. IFN-γ and TNF-α were found to synergize to decrease barrier function, as measured by trans-epithelial electric resistance (TER) and molecular flux assays. Cells cultured under an air-liquid interface produced an adherent mucus layer, and under these conditions reduced barrier function was found after cytokine exposure. Furthermore, IFN-γ, but not TNF-α treatment, upregulated the IFN-γ receptor 1 (IFNGR1) and TNF-α receptor super family 1A (TNFRSF1A) subunit mRNA in vitro. Co-stimulation resulted in increased mRNA expression of CLDN 2 and 5, two gene known to play a role in epithelial barrier integrity. Analysis of IBD patient samples revealed IFNGR1 and TNFRSF mRNA increased coincidently with guanylate binding protein 1 (GBP1) expression, an indicator of NFkB activity. Lastly, CLDN2 was found at higher levels in IBD patients while HNF4a was suppressed with disease. In conclusion, IFN-γ and TNF-α degrade epithelial/mucus barriers coincident with changes in CLDN gene and cytokine receptor subunit mRNA expression in HT29-MTX E12 cells. These changes largely reflect those observed in IBD patient samples.


Asunto(s)
Enfermedades Inflamatorias del Intestino , Interferón gamma , Citocinas/metabolismo , Células HT29 , Humanos , Interferón gamma/metabolismo , Interferón gamma/farmacología , Mucosa Intestinal/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Citocinas/metabolismo , Receptores de Interferón/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Receptor de Interferón gamma
18.
Nat Commun ; 13(1): 1923, 2022 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-35395848

RESUMEN

The cytokine IFNγ differentially impacts on tumors upon immune checkpoint blockade (ICB). Despite our understanding of downstream signaling events, less is known about regulation of its receptor (IFNγ-R1). With an unbiased genome-wide CRISPR/Cas9 screen for critical regulators of IFNγ-R1 cell surface abundance, we identify STUB1 as an E3 ubiquitin ligase for IFNγ-R1 in complex with its signal-relaying kinase JAK1. STUB1 mediates ubiquitination-dependent proteasomal degradation of IFNγ-R1/JAK1 complex through IFNγ-R1K285 and JAK1K249. Conversely, STUB1 inactivation amplifies IFNγ signaling, sensitizing tumor cells to cytotoxic T cells in vitro. This is corroborated by an anticorrelation between STUB1 expression and IFNγ response in ICB-treated patients. Consistent with the context-dependent effects of IFNγ in vivo, anti-PD-1 response is increased in heterogenous tumors comprising both wildtype and STUB1-deficient cells, but not full STUB1 knockout tumors. These results uncover STUB1 as a critical regulator of IFNγ-R1, and highlight the context-dependency of STUB1-regulated IFNγ signaling for ICB outcome.


Asunto(s)
Interferón gamma , Neoplasias , Receptores de Interferón , Ubiquitina-Proteína Ligasas , Humanos , Inhibidores de Puntos de Control Inmunológico , Interferón gamma/metabolismo , Neoplasias/inmunología , Receptores de Interferón/metabolismo , Transducción de Señal , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Receptor de Interferón gamma
19.
Adv Sci (Weinh) ; 9(11): e2105170, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35166455

RESUMEN

The cytokine interferon-gamma (IFN-γ) is a master regulator of innate and adaptive immunity involved in a broad array of human diseases that range from atherosclerosis to cancer. IFN-γ exerts it signaling action by binding to a specific cell surface receptor, the IFN-γ receptor (IFN-γR), whose activation critically depends on its partition into lipid nanodomains. However, little is known about the impact of specific lipids on IFN-γR signal transduction activity. Here, a new conserved cholesterol (chol) binding motif localized within its single transmembrane domain is identified. Through direct binding, chol drives the partition of IFN-γR2 chains into plasma membrane lipid nanodomains, orchestrating IFN-γR oligomerization and transmembrane signaling. Bioinformatics studies show that the signature sequence stands for a conserved chol-binding motif presented in many mammalian membrane proteins. The discovery of chol as the molecular switch governing IFN-γR transmembrane signaling represents a significant advance for understanding the mechanism of lipid selectivity by membrane proteins, but also for figuring out the role of lipids in modulating cell surface receptor function. Finally, this study suggests that inhibition of the chol-IFNγR2 interaction may represent a potential therapeutic strategy for various IFN-γ-dependent diseases.


Asunto(s)
Receptores de Interferón , Transducción de Señal , Animales , Sitios de Unión , Colesterol , Humanos , Interferón gamma/metabolismo , Interferón gamma/farmacología , Lípidos , Mamíferos/metabolismo , Receptores de Interferón/metabolismo , Receptor de Interferón gamma
20.
Oncoimmunology ; 11(1): 2029298, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35127252

RESUMEN

The oncogenic MUC1-C protein drives dedifferentiation of castrate resistant prostate cancer (CRPC) cells in association with chromatin remodeling. The present work demonstrates that MUC1-C is necessary for expression of IFNGR1 and activation of the type II interferon-gamma (IFN-γ) pathway. We show that MUC1-C→ARID1A/BAF signaling induces IFNGR1 transcription and that MUC1-C-induced activation of the NuRD complex suppresses FBXW7 in stabilizing the IFNGR1 protein. MUC1-C and NuRD were also necessary for expression of the downstream STAT1 and IRF1 transcription factors. We further demonstrate that MUC1-C and PBRM1/PBAF are necessary for IRF1-induced expression of (i) IDO1, WARS and PTGES, which metabolically suppress the immune tumor microenvironment (TME), and (ii) the ISG15 and SERPINB9 inhibitors of T cell function. Of translational relevance, we show that MUC1 associates with expression of IFNGR1, STAT1 and IRF1, as well as the downstream IDO1, WARS, PTGES, ISG15 and SERPINB9 immunosuppressive effectors in CRPC tumors. Analyses of scRNA-seq data further demonstrate that MUC1 correlates with cancer stem cell (CSC) and IFN gene signatures across CRPC cells. Consistent with these results, MUC1 associates with immune cell-depleted "cold" CRPC TMEs. These findings demonstrate that MUC1-C integrates chronic activation of the type II IFN-γ pathway and induction of chromatin remodeling complexes in linking the CSC state with immune evasion.


Asunto(s)
Ensamble y Desensamble de Cromatina , Interferón gamma , Mucina-1 , Neoplasias de la Próstata Resistentes a la Castración , Ensamble y Desensamble de Cromatina/inmunología , Humanos , Terapia de Inmunosupresión , Masculino , Mucina-1/inmunología , Neoplasias de la Próstata Resistentes a la Castración/inmunología , Neoplasias de la Próstata Resistentes a la Castración/patología , Receptores de Interferón/inmunología , Transducción de Señal/inmunología , Microambiente Tumoral , Receptor de Interferón gamma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA