Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.177
Filtrar
1.
J Cell Biol ; 223(3)2024 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-38329462

RESUMEN

TNF signaling does not result in cell death unless multiple inhibitory signals are overcome, which can be accomplished by simultaneous signaling through IFNγ. In this issue, Deng and colleagues (http://doi.org/10.1083/jcb.202305026) dissect the mechanisms by which IFNγ signaling combines with TNF to mediate cell death through caspase-8, discussed by James E. Vince.


Asunto(s)
Muerte Celular , Interferón gamma , Transducción de Señal , Interferón gamma/fisiología , Caspasa 8/fisiología , Factores de Necrosis Tumoral/fisiología
2.
Biochem J ; 479(3): 385-399, 2022 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-35084016

RESUMEN

The apelin receptor (APLNR) regulates many biological processes including metabolism, angiogenesis, circulating blood volume and cardiovascular function. Additionally, APLNR is overexpressed in various types of cancer and influences cancer progression. APLNR is reported to regulate tumor recognition during immune surveillance by modulating the IFN-γ response. However, the mechanism of APLNR cross-talk with intratumoral IFN-γ signaling remains unknown. Here, we show that activation of APLNR up-regulates IFN-γ signaling in melanoma cells through APLNR mediated ß-arrestin 1 but not ß-arrestin 2 recruitment. Our data suggests that ß-arrestin 1 directly interacts with STAT1 to inhibit STAT1 phosphorylation to attenuate IFN-γ signaling. The APLNR mutant receptor, I109A, which is deficient in ß-arrestins recruitment, is unable to enhance intratumoral IFN-γ signaling. While APLNR N112G, a constitutively active mutant receptor, increases intratumoral sensitivity to IFN-γ signaling by enhancing STAT1 phosphorylation upon IFN-γ exposure. We also demonstrate in a co-culture system that APLNR regulates tumor survival rate. Taken together, our findings reveal that APLNR modulates IFN-γ signaling in melanoma cells and suggest that APLNR may be a potential target to enhance the efficacy of immunotherapy.


Asunto(s)
Receptores de Apelina/fisiología , Interferón gamma/fisiología , Quinasas Janus/fisiología , Melanoma/metabolismo , Proteínas de Neoplasias/fisiología , Factor de Transcripción STAT1/fisiología , Transducción de Señal/fisiología , beta-Arrestina 1/fisiología , Receptores de Apelina/antagonistas & inhibidores , Receptores de Apelina/química , Receptores de Apelina/genética , Línea Celular Tumoral , Citotoxicidad Inmunológica , Células HEK293 , Humanos , Quinasas Janus/antagonistas & inhibidores , Melanoma/inmunología , Modelos Moleculares , Mapeo de Interacción de Proteínas , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Linfocitos T/inmunología , Arrestina beta 2/análisis
3.
Clin Transl Oncol ; 24(1): 57-65, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34275119

RESUMEN

PURPOSE: Ovarian cancer (OC) is a common malignancy, and IFN-γ, a multifunctional cytokine, is unveiled to impede the multiplication and enhance apoptosis in diverse tumor cells in previous research. Nonetheless, its function and mechanism in OC are blurred. METHODS: OC cell lines SKOV3 and OVCAR3 were dealt with different concentrations (0-40 ng/ml) of IFN-γ. CCK-8 experiment was utilized to examine cell multiplication; Flow cytometry was executed to detect apoptosis and cell cycle; Wound healing assay was utilized to detect cell migration; and Transwell experiment was implemented to examine cell invasion. qRT-PCR analysis was applied to detect STAT5, STAT3, JAK2 and JAK3 mRNA expression in OC cell lines. Western blot experiment was applied to detect the protein and phosphorylation levels of SOCS1, STAT5 and STAT3. RESULTS: IFN-γ suppressed OC cell multiplication in a concentration-dependent manner. Relative to the control group, IFN-γ restrained OC cell migration, invasion, enhanced apoptosis and prevented cell transformation from G0/G1 to S phase. Further analysis revealed that IFN-γ up-modulated SOCS1 expression and impeded STAT5 and STAT3 protein phosphorylation levels, and knockdown of SOCS1 partially counteracted the inhibitory effect of IFN-γ on STAT5 and STAT3 protein phosphorylation levels. CONCLUSION: IFN-γ represses OC progression by facilitating SOCS1 to suppress STAT3 and STAT5 protein phosphorylation.


Asunto(s)
Interferón gamma/fisiología , Janus Quinasa 2/fisiología , Neoplasias Ováricas/patología , Factor de Transcripción STAT5/fisiología , Transducción de Señal , Proteína 1 Supresora de la Señalización de Citocinas/fisiología , Progresión de la Enfermedad , Femenino , Humanos , Factor de Transcripción STAT3
4.
Life Sci ; 286: 120054, 2021 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-34662550

RESUMEN

AIMS: Type 2 diabetes mellitus (T2DM) is a risk factor for breast cancer initiation and progression. Glutamine (GLN) is a critical nutrient for cancer cells. The aim of this study was to investigate the effect of T2DM-associated compounds upon GLN uptake by breast cancer cells. MAIN METHODS: The in vitro uptake of 3H-GLN by breast cancer (MCF-7 and MDA-MB-231) and non-tumorigenic (MCF-12A) cell lines was measured. KEY FINDINGS: 3H-GLN uptake in the three cell lines is mainly Na+-dependent and sensitive to the ASCT2 inhibitor GPNA. IFN-γ increased total and Na+-dependent 3H-GLN uptake in the two breast cancer cell lines, and insulin increased total and Na+-dependent 3H-GLN uptake in the non-tumorigenic cell line. GPNA abolished the increase in 3H-GLN uptake promoted by these T2DM-associated compounds. ASCT2 knockdown confirmed that the increase in 3H-GLN uptake caused by IFN-γ (in breast cancer cells) and by insulin (in non-tumorigenic cells) is ASCT2-dependent. IFN-γ (in MDA-MB-231 cells) and insulin (in MCF-12A cells) increased ASCT2 transcript and protein levels. Importantly, the pro-proliferative effect of IFN-γ in breast cancer cell lines was associated with an increase in 3H-GLN uptake which was GPNA-sensitive, blocked by ASCT2 knockdown and mediated by activation of the PI3K-, STAT3- and STAT1 intracellular signalling pathways. SIGNIFICANCE: IFN-γ and insulin possess pro-proliferative effects in breast cancer and non-cancer cell lines, respectively, which are dependent on an increase in ASCT2-mediated glutamine transport. Thus, an effective inhibition of ASCT2-mediated glutamine uptake may be a therapeutic strategy against human breast cancer in T2DM patients.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC/metabolismo , Neoplasias de la Mama/metabolismo , Interferón gamma/metabolismo , Antígenos de Histocompatibilidad Menor/metabolismo , Sistema de Transporte de Aminoácidos ASC/fisiología , Apoptosis/efectos de los fármacos , Transporte Biológico , Neoplasias de la Mama/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Complicaciones de la Diabetes/fisiopatología , Diabetes Mellitus/fisiopatología , Femenino , Glutamina/metabolismo , Humanos , Interferón gamma/inmunología , Interferón gamma/fisiología , Antígenos de Histocompatibilidad Menor/fisiología , Factores de Riesgo , Transducción de Señal/efectos de los fármacos , Sodio
5.
Int J Mol Sci ; 22(17)2021 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-34502453

RESUMEN

Mesenchymal stem/stromal cells (MSCs) have an immunoregulatory capacity and have been used in different clinical protocols requiring control of the immune response. However, variable results have been obtained, mainly due to the effect of the microenvironment on the induction, increase, and maintenance of MSC immunoregulatory mechanisms. In addition, the importance of cell-cell contact for MSCs to efficiently modulate the immune response has recently been highlighted. Because these interactions would be difficult to achieve in the physiological context, the release of extracellular vesicles (EVs) and their participation as intermediaries of communication between MSCs and immune cells becomes relevant. Therefore, this article focuses on analyzing immunoregulatory mechanisms mediated by cell contact, highlighting the importance of intercellular adhesion molecule-1 (ICAM-1) and the participation of EVs. Moreover, the effects of tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ), the main cytokines involved in MSC activation, are examined. These cytokines, when used at the appropriate concentrations and times, would promote increases in the expression of immunoregulatory molecules in the cell and allow the acquisition of EVs enriched with these molecules. The establishment of certain in vitro activation guidelines will facilitate the design of conditioning protocols to obtain functional MSCs or EVs in different pathophysiological conditions.


Asunto(s)
Comunicación Celular , Inmunomodulación , Interferón gamma/fisiología , Células Madre Mesenquimatosas/inmunología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Citocinas/metabolismo , Vesículas Extracelulares/metabolismo , Humanos
6.
Biomed Res Int ; 2021: 4604856, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34527737

RESUMEN

IFN-γ licensing to mesenchymal stem cells (MSCs) is applied to enhance the therapeutic potential of MSCs. However, although the features of MSCs are affected by several stimuli, little information is available on changes to the therapeutic potential of IFN-γ-licensed differentiated MSCs during xenogeneic applications. Therefore, the present study is aimed at clarifying the effects of adipogenic/osteogenic differentiation and IFN-γ licensing on the in vitro immunomodulatory and migratory properties of porcine bone marrow-derived MSCs in xenogeneic applications using human peripheral blood mononuclear cells (PBMCs). IFN-γ licensing in differentiated MSCs lowered lineage-specific gene expression but did not affect MSC-specific cell surface molecules. Although indoleamine 2,3 deoxygenase (IDO) activity and expression were increased after IFN-γ licensing in undifferentiated MSCs, they were reduced after differentiation. IFN-γ licensing to differentiated MSCs elevated the reduced IDO expression in differentiated MSCs; however, the increase was not sufficient to reach to the level achieved by undifferentiated MSCs. During a mixed lymphocyte reaction with quantification of TNF-α concentration, proliferation and activation of xenogeneic PBMCs were suppressed by undifferentiated MSCs but inhibited to a lesser extent by differentiated MSCs. IFN-γ licensing increasingly suppressed proliferation of PBMCs in undifferentiated MSCs but it was incapable of elevating the reduced immunosuppressive ability of differentiated MSCs. Migratory ability through a scratch assay and gene expression study was reduced in differentiated MSCs than their undifferentiated counterparts; IFN-γ licensing was unable to enhance the reduced migratory ability in differentiated MSCs. Similar results were found in a Transwell system with differentiated MSCs in the upper chamber toward xenogeneic PBMCs in the lower chamber, despite IFN-γ licensing increased the migratory ability of undifferentiated MSCs. Overall, IFN-γ licensing did not enhance the reduced immunomodulatory and migratory properties of differentiated MSCs in a xenogeneic application. This study provides a better understanding of the ways in which MSC therapy can be applied.


Asunto(s)
Interferón gamma/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Animales , Médula Ósea/metabolismo , Células de la Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Citocinas/metabolismo , Xenoinjertos/metabolismo , Humanos , Inmunomodulación/efectos de los fármacos , Interferón gamma/fisiología , Células Madre Mesenquimatosas/fisiología , Osteogénesis/efectos de los fármacos , Porcinos , Factor de Necrosis Tumoral alfa/metabolismo
7.
Front Immunol ; 12: 642778, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33777039

RESUMEN

Systemic juvenile idiopathic arthritis (sJIA) is an immune disorder characterized by fever, skin rash, arthritis and splenomegaly. Recently, increasing number of sJIA patients were reported having lung disease. Here, we explored lung abnormalities in a mouse model for sJIA relying on injection of IFN-γ deficient (IFN-γ KO) mice with complete Freund's adjuvant (CFA). Monitoring of lung changes during development of sJIA using microcomputer tomography revealed a moderate enlargement of lungs, a decrease in aerated and increase in non-aerated lung density. When lung function and airway reactivity to methacholine was assessed, gender differences were seen. While male mice showed an increased tissue hysteresivity, female animals were characterized by an increased airway hyperactivity, mirroring ongoing inflammation. Histologically, lungs of sJIA-like mice showed subpleural and parenchymal cellular infiltrates and formation of small granulomas. Flow cytometric analysis identified immature and mature neutrophils, and activated macrophages as major cell infiltrates. Lung inflammation in sJIA-like mice was accompanied by augmented expression of IL-1ß and IL-6, two target cytokines in the treatment of sJIA. The increased expression of granulocyte colony stimulating factor, a potent inducer of granulopoiesis, in lungs of mice was striking considering the observed neutrophilia in patients. We conclude that development of sJIA in a mouse model is associated with lung inflammation which is distinct to the lung manifestations seen in sJIA patients. Our observations however underscore the importance of monitoring lung disease during systemic inflammation and the model provides a tool to explore the underlying mechanism of lung pathology in an autoinflammatory disease context.


Asunto(s)
Artritis Juvenil/complicaciones , Inflamación/etiología , Pulmón/fisiopatología , Animales , Artritis Juvenil/inmunología , Artritis Juvenil/patología , Artritis Juvenil/fisiopatología , Modelos Animales de Enfermedad , Femenino , Adyuvante de Freund/inmunología , Mediadores de Inflamación/análisis , Interferón gamma/fisiología , Pulmón/inmunología , Pulmón/patología , Activación de Macrófagos , Masculino , Ratones , Ratones Endogámicos BALB C
8.
Cell Immunol ; 362: 104303, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33611078

RESUMEN

We have previously identified a cystatin, TsCstN, derived from the L1 stage of Trichinella spiralis and have shown that this protein is internalised in macrophages. Here we sought to address if this macrophage-TsCstN interaction could alter downstream T-cell priming. Using LPS-primed macrophages to stimulate T-cells in a co-culture system with or without TsCstN we assessed the resultant T-cell outcomes. IFN-γ, both protein and mRNA, but not IL-17A was negatively regulated by inclusion of TsCstN during macrophage priming. We identified a cell-cell contact independent change in the levels of IL-12 that led to altered phosphorylated STAT4 levels and translocation. TsCstN also negatively regulated the autonomous response in the myotubule cell line, C2C12. This work identifies a potential pathyway for L1 larvae to evade protective Th1 based immune responses and establish muscle-stage T. spiralis infection.


Asunto(s)
Interferón gamma/metabolismo , Factor de Transcripción STAT4/metabolismo , Trichinella spiralis/metabolismo , Animales , Cistatinas/metabolismo , Cistatinas/farmacología , Citocinas/metabolismo , Femenino , Interferón gamma/fisiología , Interleucina-12/inmunología , Interleucina-12/metabolismo , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Factor de Transcripción STAT4/fisiología , Transducción de Señal , Linfocitos T/metabolismo , Trichinella spiralis/genética , Trichinella spiralis/inmunología
9.
Biomolecules ; 11(2)2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33572487

RESUMEN

There is a great deal of evidence pointing to interferons (IFNs) as being key cytokines in the pathogenesis of different systemic autoimmune diseases, including primary Sjögren's syndrome (pSS). In this disease, a large number of studies have shown that an overexpression of type I IFN, the 'so-called' type I IFN signature, is present in peripheral blood mononuclear cells, and that this finding is associated with the development of systemic extra-glandular manifestations, and a substantial production of autoantibodies and inflammatory cytokines. In contrast, the absence or a milder expression of type I IFN signature and low level of inflammatory cytokines characterizes patients with a different clinical phenotype, where the disease is limited to glandular involvement and often marked by the presence of widespread pain and depression. The role of type II (IFNγ) in this subset of pSS patients, together with the potentially related activation of completely different immunological and metabolic pathways, are emerging issues. Expression of both types of IFNs has also been shown in target tissues, namely in minor salivary glands where a predominance of type II IFN signature appeared to have a certain association with the development of lymphoma. In view of the role played by IFN overexpression in the development and progression of pSS, inhibition or modulation of IFN signaling has been regarded as a potential target for the therapeutic approach. A number of therapeutic compounds with variable mechanisms of action have been tested or are under consideration for the treatment of patients with pSS.


Asunto(s)
Interferón Tipo I/fisiología , Interferón gamma/fisiología , Síndrome de Sjögren/fisiopatología , Animales , Humanos , Interferón Tipo I/metabolismo , Interferón Tipo I/uso terapéutico , Interferón gamma/metabolismo , Interferón gamma/uso terapéutico , Glándulas Salivales/metabolismo , Transducción de Señal , Síndrome de Sjögren/tratamiento farmacológico , Síndrome de Sjögren/metabolismo
10.
Front Immunol ; 12: 696415, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34987496

RESUMEN

Disseminated infection with the high virulence strain of Mycobacterium avium 25291 leads to progressive thymic atrophy. We previously showed that M. avium-induced thymic atrophy results from increased glucocorticoid levels that synergize with nitric oxide (NO) produced by interferon gamma (IFNγ) activated macrophages. Where and how these mediators act is not understood. We hypothesized that IFNγ and NO promote thymic atrophy through their effects on bone marrow (BM) T cell precursors and T cell differentiation in the thymus. We show that M. avium infection cause a reduction in the percentage and number of common lymphoid progenitors (CLP). Additionally, BM precursors from infected mice show an overall impaired ability to reconstitute thymi of RAGKO mice, in part due to IFNγ. Thymi from infected mice present an IFNγ and NO-driven inflammation. When transplanted under the kidney capsule of uninfected mice, thymi from infected mice are unable to sustain T cell differentiation. Finally, we observed increased thymocyte death via apoptosis after infection, independent of both IFNγ and iNOS; and a decrease on active caspase-3 positive thymocytes, which is not observed in the absence of iNOS expression. Together our data suggests that M. avium-induced thymic atrophy results from a combination of defects mediated by IFNγ and NO, including alterations in the BM T cell precursors, the thymic structure and the thymocyte differentiation.


Asunto(s)
Médula Ósea/patología , Interferón gamma/fisiología , Células Progenitoras Linfoides/patología , Óxido Nítrico Sintasa de Tipo II/fisiología , Timo/patología , Tuberculosis/patología , Animales , Apoptosis , Atrofia , Trasplante de Médula Ósea , Diferenciación Celular , Proteínas de Unión al ADN/deficiencia , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mycobacterium avium , Óxido Nítrico/fisiología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/patología , Timocitos/patología , Timo/trasplante , Tuberculosis/inmunología
12.
Cell Rep ; 33(10): 108482, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33296659

RESUMEN

Regulatory T (Treg) cell infiltration of solid tumors often correlates with poor prognosis, but their tumor-suppressive function lacks mechanistic understanding. Through a combination of transgenic mice, cell fate mapping, adoptive transfer, and co-injection strategies, we demonstrate that Treg cell ablation-dependent anti-tumor effects in murine breast cancer require intratumoral recruitment of CCR2+ inflammatory monocytes, which primarily differentiate into tumor-associated macrophages (TAMs), and lead to reprogramming of their function in an IFN-γ-dependent manner. Furthermore, transcriptomic signatures from murine TAMs in Treg cell-ablated conditions correlate with increased overall survival in human breast cancer. Our studies highlight the strong myeloid dependency of breast cancer and provide the basis for the development of therapeutic strategies based on manipulation of the IFN-γ signaling pathway in monocytes.


Asunto(s)
Neoplasias de la Mama/metabolismo , Células Mieloides/metabolismo , Linfocitos T Reguladores/metabolismo , Traslado Adoptivo , Animales , Diferenciación Celular , Línea Celular Tumoral , Reprogramación Celular/fisiología , Modelos Animales de Enfermedad , Femenino , Humanos , Interferón gamma/metabolismo , Interferón gamma/fisiología , Macrófagos/metabolismo , Ratones , Ratones Desnudos , Ratones Transgénicos , Monocitos/metabolismo , Células Mieloides/fisiología , Linfocitos T Reguladores/fisiología , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Neurochem Int ; 141: 104860, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33022312

RESUMEN

Cerebral ischemia-reperfusion injury induces a secondary immune inflammatory reaction that exacerbates brain injury and clinical prognosis. Dendritic cells (DCs) and microglia are both important regulators of neuroinflammation. Studies have confirmed that a large number of cells express the DC surface marker CD11c in the ischemic area, and some of these cells also express microglial markers. However, the specific mechanism of transformation between microglia and DCs and their roles in the process of cerebral ischemia-reperfusion injury are still not clear. In this study, we established a mouse model and flow cytometry was used to detect the expression of mature DC surface molecules in activated microglia. IFN-γ knockout mice were used to determine the regulatory effect of IFN-γ on microglial transformation. We found that CD11c+ cells were derived from microglia after ischemia-reperfusion injury, and this group of cells highly expressed MHC-II molecules and other costimulatory molecules, such as CD80 and CD86, which were regulated by IFN-γ and its downstream signaling molecules ERK/c-myc. In summary, our results showed in cerebral ischemia-reperfusion injury, IFN-γ regulates the transformation of microglia to DC-like cells. Microglial-derived DC-like cells possess the ability to present antigens and activate naïve T cells which is regulated by the ERK/c-myc signaling pathway.


Asunto(s)
Dendritas/efectos de los fármacos , Interferón gamma/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Microglía/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/efectos de los fármacos , Daño por Reperfusión/patología , Animales , Antígenos CD11/metabolismo , Dendritas/patología , Genes MHC Clase II , Interferón gamma/fisiología , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/patología , Cultivo Primario de Células , Receptores de Interferón/biosíntesis , Linfocitos T
14.
J Vet Med Sci ; 82(10): 1410-1414, 2020 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-32759577

RESUMEN

Murine coronavirus (CoV) is a beta-CoV that infects mice by binding to carcinoembryonic antigen-related cell adhesion molecule 1. Intraperitoneal infection with the murine CoV strain JHM (JHMV) induces acute mild hepatitis in mice. While both innate and acquired immune responses play a significant role in the protection against murine CoV infection in mice, CD8+ cytotoxic T lymphocytes (CTLs) and interferon-γ are essential for viral clearance in JHMV-induced hepatitis. In addition, CoVs are characterized by high diversity, caused by mutations, recombination, and gene gain/loss. 25V16G is an immune-escape JHMV variant, which lacks a dominant CTL epitope. By evading immune responses, 25V16G establishes persistent infections, leading to granulomatous serositis in interferon-γ-deficient mice. These examples of CoV-associated pathogenesis in mice might provide useful information on other CoV infections, including coronavirus disease 2019 (COVID-19).


Asunto(s)
Infecciones por Coronavirus/veterinaria , Interferón gamma/fisiología , Virus de la Hepatitis Murina/patogenicidad , Linfocitos T Citotóxicos/fisiología , Animales , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Ratones
15.
Surgery ; 168(4): 610-616, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32631655

RESUMEN

BACKGROUND: Combining immune checkpoint blockade therapy with operative disruptive immunomodulation using irreversible electroporation may overcome the resistance to systemic therapy found in patients with locally advanced, unresectable pancreatic cancer. We describe the safety profile and efficacy of IRE with nivolumab. METHODS: In the preclinical phase of study, human pancreatic cell lines were cultured with interferon-γ (10 ng/mL) and murine models of pancreatic cancer were treated with irreversible electroporation and programmed death ligand-1 (PD-L1) expression was measured. In this phase 1b clinical trial (NCT03080974), surgical ablative irreversible electroporation was performed followed by nivolumab. The primary end point was dose-limiting toxicity. RESULTS: Human pancreatic cells express PD-L1 when cultured with interferon-γ: quantitative polymerase chain reaction MiaPaca (15.2 rel. fold ± 0.5; P < .01) and S20-13 (31.0 rel. fold ± 4.4; P < .01). Murine orthotopic tumors treated by irreversible electroporation had an increase in signal intensity score for the expression of PD-L1 in residual tumor (P < .01). Ten patients were included in the safety analysis with a 12-month median follow-up (interquartile range 6.0, 15.8). No dose-limiting toxicities occurred. Seven patients developed grade 3/4 treatment-related adverse events; none required a dose modification of nivolumab; nivolumab-related adverse events occurred in 1 patient. Mean time to progression was 6.3 months (confidence interval 3.5-10.0) with current median overall survival of 18.0 months (confidence interval 9.2-26.8). CONCLUSION: Irreversible electroporation induces expression of PD-L1 in vitro. Combination therapy with concurrent nivolumab is well tolerated. A multicenter, phase 2 adjuvant trial is underway using irreversible electroporation and nivolumab in patients with locally advanced pancreatic cancer.


Asunto(s)
Adenocarcinoma/terapia , Antineoplásicos Inmunológicos/uso terapéutico , Electroporación/métodos , Nivolumab/uso terapéutico , Neoplasias Pancreáticas/terapia , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Adulto , Anciano , Animales , Antineoplásicos Inmunológicos/efectos adversos , Antígeno B7-H1/genética , Línea Celular Tumoral , Terapia Combinada , Femenino , Expresión Génica , Humanos , Interferón gamma/fisiología , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Estadificación de Neoplasias , Nivolumab/efectos adversos , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Receptor de Muerte Celular Programada 1/genética , Subgrupos de Linfocitos T , Neoplasias Pancreáticas
16.
Int Immunopharmacol ; 84: 106515, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32311672

RESUMEN

Immunity-related GTPase family M1 protein (lRGM1) plays an important role in host resistance to infection, immune inflammation, and tumors, and it is expressed in various tissues and cells, including the central nervous system, cardiovascular system, bone marrow-derived cells, glioma, and melanoma. However, the effect of IRGM1 in the muscles has not been reported to date. In this study, Irgm1-/- mice were used to evaluate the effect of lrgm1 on regeneration after skeletal muscle injury. The tibialis anterior muscle in Irgm1-/- mice was poorly repaired after BaCl2-induced injury, whereas lrgm1 knockout itself had no significant effect on the differentiation of myoblasts. However, the microenvironment of Irgm1-/- mice with a high interferon-gamma level inhibited the differentiation of myoblasts in vivo. These results suggest that lrgm1 knockout indirectly inhibits skeletal muscle regeneration after injury, providing new insights into the biological function of IRGM1.


Asunto(s)
Proteínas de Unión al GTP/fisiología , Músculo Esquelético/fisiología , Animales , Compuestos de Bario , Diferenciación Celular , Células Cultivadas , Cloruros , Proteínas de Unión al GTP/genética , Interferón gamma/fisiología , Masculino , Ratones Noqueados , Músculo Esquelético/lesiones , Regeneración , Células Satélite del Músculo Esquelético/fisiología
17.
Neurosci Bull ; 36(7): 778-792, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32306216

RESUMEN

SOCS3, a feedback inhibitor of the JAK/STAT signal pathway, negatively regulates axonal regrowth and inflammation in the central nervous system (CNS). Here, we demonstrated a distinct role of SOCS3 in the injured spinal cord of the gecko following tail amputation. Severing the gecko spinal cord did not evoke an inflammatory cascade except for an injury-stimulated elevation of the granulocyte/macrophage colony-stimulating factor (GM-CSF) and interferon gamma (IFN-γ) cytokines. Simultaneously, the expression of SOCS3 was upregulated in microglia, and unexpectedly not in neurons. Enforced expression of SOCS3 was sufficient to suppress the GM-CSF/IFN-γ-driven inflammatory responses through its KIR domain by attenuating the activities of JAK1 and JAK2. SOCS3 was also linked to GM-CSF/IFN-γ-induced cross-tolerance. Transfection of adenovirus overexpressing SOCS3 in the injured cord resulted in a significant decrease of inflammatory cytokines. These results reveal a distinct role of SOCS3 in the regenerating spinal cord, and provide new hints for CNS repair in mammals.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos , Interferón gamma , Regeneración de la Medula Espinal , Proteína 3 Supresora de la Señalización de Citocinas/fisiología , Animales , Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Inflamación , Interferón gamma/fisiología , Lagartos , Microglía , Neuronas
18.
Front Immunol ; 11: 223, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32133005

RESUMEN

Immune checkpoint blockade (ICB) has revolutionized cancer treatment, providing remarkable clinical responses in some patients. However, the majority of patients do not respond. It is therefore crucial both to identify predictive biomarkers of response and to increase the response rates to immune checkpoint therapy. In this review we explore the current literature about the predictive characteristics of the tumor microenvironment and discuss therapeutic approaches that aim to change this toward a milieu that is conducive to response. We propose a personalized biomarker-based adaptive approach to immunotherapy, whereby a sensitizing therapy is tailored to the patient's specific tumor microenvironment, followed by on-treatment verification of a change in the targeted biomarker, followed by immune checkpoint therapy. By incorporating detailed knowledge of the immunological tumor microenvironment, we may be able to sensitize currently non-responsive tumors to respond to immune checkpoint therapy.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias/tratamiento farmacológico , Microambiente Tumoral , Animales , Biomarcadores de Tumor , Humanos , Interferón gamma/fisiología , Ratones , Microbiota , Neoplasias/irrigación sanguínea , Neoplasias/inmunología , Viroterapia Oncolítica , Medicina de Precisión , Linfocitos T/inmunología
19.
Front Immunol ; 11: 306, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32194558

RESUMEN

CCL3, a member of the CC-chemokine family, has been associated with macrophage recruitment to heart tissue and parasite control in the acute infection of mouse with Trypanosoma cruzi, the causative agent of Chagas disease. Here, we approached the participation of CCL3 in chronic chagasic cardiomyopathy (CCC), the main clinical form of Chagas disease. We induced CCC in C57BL/6 (ccl3+/+) and CCL3-deficient (ccl3-/-) mice by infection with the Colombian Type I strain. In ccl3+/+ mice, high levels of CCL3 mRNA and protein were detected in the heart tissue during the acute and chronic infection. Survival was not affected by CCL3 deficiency. In comparison with ccl3+/+, chronically infected ccl3-/- mice presented reduced cardiac parasitism and inflammation due to CD8+ cells and macrophages. Leukocytosis was decreased in infected ccl3-/- mice, paralleling the accumulation of CD8+ T cells devoid of activated CCR5+ LFA-1+ cells in the spleen. Further, T. cruzi-infected ccl3-/-mice presented reduced frequency of interferon-gamma (IFNγ)+ cells and numbers of parasite-specific IFNγ-producing cells, while the T. cruzi antigen-specific cytotoxic activity was increased. Stimulation of CCL3-deficient macrophages with IFNγ improved parasite control, in a milieu with reduced nitric oxide (NOx) and tumor necrosis factor (TNF), but similar interleukin-10 (IL-10), concentrations. In comparison with chronically T. cruzi-infected ccl3+/+ counterparts, ccl3-/- mice did not show enlarged heart, loss of left ventricular ejection fraction, QTc prolongation and elevated CK-MB activity. Compared with ccl3+/+, infected ccl3-/- mice showed reduced concentrations of TNF, while IL-10 levels were not affected, in the heart milieu. In spleen of ccl3+/+ NI controls, most of the CD8+ T-cells expressing the CCL3 receptors CCR1 or CCR5 were IL-10+, while in infected mice these cells were mainly TNF+. Lastly, selective blockage of CCR1/CCR5 (Met-RANTES therapy) in chronically infected ccl3+/+ mice reversed pivotal electrical abnormalities (bradycardia, prolonged PR, and QTc interval), in correlation with reduced TNF and, mainly, CCL3 levels in the heart tissue. Therefore, in the chronic T. cruzi infection CCL3 takes part in parasite persistence and contributes to form a CD8+ T-cell and macrophage-enriched cardiac inflammation. Further, increased levels of CCL3 create a scenario with abundant IFNγ and TNF, associated with cardiomyocyte injury, heart dysfunction and QTc prolongation, biomarkers of severity of Chagas' heart disease.


Asunto(s)
Cardiomiopatía Chagásica/fisiopatología , Quimiocina CCL3/fisiología , Interferón gamma/fisiología , Macrófagos Peritoneales/parasitología , Parasitemia/fisiopatología , Trypanosoma cruzi/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Células Cultivadas , Cardiomiopatía Chagásica/parasitología , Cardiomiopatía Chagásica/patología , Quimiocina CCL3/deficiencia , Quimiocina CCL3/farmacología , Quimiocina CCL5/farmacología , Quimiocina CCL5/uso terapéutico , Quimiotaxis de Leucocito/efectos de los fármacos , Citocinas/biosíntesis , Citocinas/genética , Citocinas/farmacología , Electrocardiografía/efectos de los fármacos , Femenino , Interferón gamma/farmacología , Macrófagos Peritoneales/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocarditis/etiología , Miocarditis/patología , Miocarditis/fisiopatología , ARN Mensajero/biosíntesis , Receptores de Quimiocina/antagonistas & inhibidores , Receptores de Quimiocina/biosíntesis , Receptores de Quimiocina/genética , Organismos Libres de Patógenos Específicos , Bazo/inmunología , Bazo/metabolismo , Volumen Sistólico , Trypanosoma cruzi/aislamiento & purificación , Factor de Necrosis Tumoral alfa/análisis
20.
J Gerontol A Biol Sci Med Sci ; 75(5): 826-834, 2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-32060555

RESUMEN

Bone loss in aging is linked with chronic low-grade inflammation and the accumulation of marrowfat in animals and humans. Peroxisome proliferator-activated receptor gamma (PPARγ), an adipogenic regulator, plays key roles in these biological processes. However, studies of the roles of PPARγ in age-related bone loss and inflammation are lacking. We hypothesized that deletion of PPARγ in bone marrow mesenchymal lineage cells would reduce bone loss with aging, potentially through a reduction in fat-generated inflammatory responses and an increase in osteoblastic activity. In the present study, we show that mice deficient of PPARγ in Dermo1-expressing mesenchymal lineage cells (Dermo1-Cre:PPARγ fl/fl) have reduced fat mass and increased cortical bone thickness but that deficiency of PPARγ had limited effect on protection of trabecular bone with aging as demonstrated by dual-energy X-ray absorptiometry, µCT, and histomorphometric analyses. Conditional knockout of PPARγ reduced serum concentrations of adipokines, including adiponectin, resistin, and leptin, and reduced marrow stromal cell expression levels of inflammation-related genes. Inflammation genes involved in the interferon signaling pathway were reduced the most. These results demonstrate that disruption of the master adipogenic regulator, PPARγ, has a certain protective effect on aging-induced bone loss, suggesting that regulation of adipose function and modulation of interferon signaling are among the key mechanisms by which PPARγ regulates bone homeostasis during aging process.


Asunto(s)
Envejecimiento/fisiología , Células Madre Mesenquimatosas/fisiología , Osteoporosis/etiología , PPAR gamma/fisiología , Adipoquinas/sangre , Animales , Linaje de la Célula , Células Cultivadas , Hueso Cortical/metabolismo , Femenino , Interferón gamma/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoporosis/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA