Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Exp Med ; 220(1)2023 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-36374226

RESUMEN

In this issue of JEM, Jenster et al. (2022. J. Exp. Med. https://doi.org/10.1084/jem.20220837) investigate how UVB radiation promotes activation of the inflammatory immune sensor NLRP1, and in doing so uncover how NLRP1 recognizes a diverse range of ribotoxic stresses.


Asunto(s)
Inflamasomas , Inflamasomas/efectos de la radiación , Rayos Ultravioleta , Humanos
2.
Science ; 377(6603): 328-335, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35857590

RESUMEN

Human NLRP1 (NACHT, LRR, and PYD domain-containing protein 1) is an innate immune sensor predominantly expressed in the skin and airway epithelium. Here, we report that human NLRP1 senses the ultraviolet B (UVB)- and toxin-induced ribotoxic stress response (RSR). Biochemically, RSR leads to the direct hyperphosphorylation of a human-specific disordered linker region of NLRP1 (NLRP1DR) by MAP3K20/ZAKα kinase and its downstream effector, p38. Mutating a single ZAKα phosphorylation site in NLRP1DR abrogates UVB- and ribotoxin-driven pyroptosis in human keratinocytes. Moreover, fusing NLRP1DR to CARD8, which is insensitive to RSR by itself, creates a minimal inflammasome sensor for UVB and ribotoxins. These results provide insight into UVB sensing by human skin keratinocytes, identify several ribotoxins as NLRP1 agonists, and establish inflammasome-driven pyroptosis as an integral component of the RSR.


Asunto(s)
Inflamasomas , Quinasas Quinasa Quinasa PAM , Proteínas NLR , Piroptosis , Ribosomas , Estrés Fisiológico , Anisomicina/toxicidad , Proteínas Adaptadoras de Señalización CARD/metabolismo , Humanos , Inflamasomas/efectos de los fármacos , Inflamasomas/metabolismo , Inflamasomas/efectos de la radiación , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Queratinocitos/efectos de la radiación , Quinasas Quinasa Quinasa PAM/metabolismo , Mutación , Proteínas NLR/genética , Proteínas NLR/metabolismo , Proteínas de Neoplasias/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/efectos de la radiación , Piroptosis/efectos de los fármacos , Piroptosis/efectos de la radiación , Ribosomas/efectos de los fármacos , Ribosomas/efectos de la radiación , Rayos Ultravioleta
3.
Radiat Res ; 196(6): 686-689, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34644390

RESUMEN

Exposure to high dose radiation causes life-threatening acute and delayed effects. Defining the mechanisms of lethal radiation-induced acute toxicity of gastrointestinal and hematopoietic tissues are critical steps to identify drug targets to mitigate and protect against the acute radiation syndrome (ARS). For example, one rational approach would be to design pharmaceuticals that block cell death pathways to preserve tissue integrity in radiation-sensitive organ systems including the gastrointestinal tract and hematopoietic compartment. A previous study reported that the inflammasome pathway, which mediates inflammatory cell death through pyroptosis, promotes ARS. However, we show that mice lacking the inflammatory executioner caspases, caspase-1 and caspase-11, are not protected from ARS when compared directly to littermates expressing caspase-1 and caspase-11. These results suggest that alternative pathways will need to be targeted by drugs that successfully mitigate and protect against the ARS.


Asunto(s)
Síndrome de Radiación Aguda/enzimología , Caspasa 1/metabolismo , Caspasas Iniciadoras/metabolismo , Inflamasomas/metabolismo , Animales , Sistema Hematopoyético/efectos de la radiación , Inflamasomas/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Piroptosis/efectos de la radiación
4.
Med Hypotheses ; 146: 110396, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33243665

RESUMEN

We have reviewed a considerable amount of recent scientific papers relating inflammation caused by air pollution with chronic and severe medical conditions. Furthermore, there are evidences relating organ inflammation caused by not only outdoor long-term but also short-term inhaled radioisotopes contained in high polluted air or in household natural radioactive background aerosols, in addition to SARS-COV-2 attached to bioaerosols, which are related with a worst evolution of severe acute respiratory syndrome patients. Reactive oxygen species (ROS) production induced by the interaction with environmental ionizing radiation contained in pollution is pointed out as a critical mechanism that predispose mainly to elder population, but not excluding young subjects, presenting previous chronic conditions of lung inflammation or neuroinflammation, which can lead to the most serious consequences.


Asunto(s)
Contaminación Radiactiva del Aire/efectos adversos , COVID-19/etiología , Cambio Climático , Inflamación/etiología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , SARS-CoV-2/patogenicidad , Aerosoles , Microbiología del Aire , COVID-19/mortalidad , Causalidad , Humanos , Inflamasomas/metabolismo , Inflamasomas/efectos de la radiación , Modelos Biológicos , Enfermedades del Sistema Nervioso/etiología , Pandemias , Tamaño de la Partícula , Material Particulado/efectos adversos , Neumonía/etiología
5.
PLoS Biol ; 18(8): e3000807, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32760056

RESUMEN

Radiotherapy is a commonly used conditioning regimen for bone marrow transplantation (BMT). Cytotoxicity limits the use of this life-saving therapy, but the underlying mechanisms remain poorly defined. Here, we use the syngeneic mouse BMT model to test the hypothesis that lethal radiation damages tissues, thereby unleashing signals that indiscriminately activate the inflammasome pathways in host and transplanted cells. We find that a clinically relevant high dose of radiation causes severe damage to bones and the spleen through mechanisms involving the NLRP3 and AIM2 inflammasomes but not the NLRC4 inflammasome. Downstream, we demonstrate that gasdermin D (GSDMD), the common effector of the inflammasomes, is also activated by radiation. Remarkably, protection against the injury induced by deadly ionizing radiation occurs only when NLRP3, AIM2, or GSDMD is lost simultaneously in both the donor and host cell compartments. Thus, this study reveals a continuum of the actions of lethal radiation relayed by the inflammasome-GSDMD axis, initially affecting recipient cells and ultimately harming transplanted cells as they grow in the severely injured and toxic environment. This study also suggests that therapeutic targeting of inflammasome-GSDMD signaling has the potential to prevent the collateral effects of intense radiation regimens.


Asunto(s)
Células de la Médula Ósea/efectos de la radiación , Trasplante de Médula Ósea , Proteínas de Unión al ADN/genética , Inflamasomas/efectos de la radiación , Péptidos y Proteínas de Señalización Intracelular/genética , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteínas de Unión a Fosfato/genética , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Proteínas de Unión al ADN/deficiencia , Femenino , Fémur/citología , Fémur/metabolismo , Regulación de la Expresión Génica , Inflamasomas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Proteínas de Unión a Fosfato/deficiencia , Piroptosis/genética , Piroptosis/efectos de la radiación , Transducción de Señal , Bazo/metabolismo , Bazo/patología , Bazo/efectos de la radiación , Trasplante Isogénico , Irradiación Corporal Total , Rayos X
6.
Radiat Oncol ; 15(1): 78, 2020 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-32276638

RESUMEN

PURPOSE: The aim of this study was to characterize changes in hippocampal inflammasomes, pyroptosis and apoptosis in juvenile rats after brain irradiation and to assess whether manganese-enhanced magnetic resonance imaging (MEMRI) reflected those changes. MATERIALS AND METHODS: Four-week-old male Sprague-Dawley rats received a whole-brain radiation dose of 15 Gy or 25 Gy. Hippocampal inflammasomes and apoptosis were measured using Western blot analysis at 4 days and 8 weeks after irradiation. MEMRI and magnetic resonance spectroscopy (MRS) were performed at the same time points. RESULTS: Neither the 15 Gy nor 25 Gy group showed changes in the expression of inflammasome proteins absent in melanoma 2 (AIM2), gasdermin-D (GSDMD), nucleotide oligomerization domain-like receptor protein 1 (NLRP1) and NLRP3 at 4 days or 8 weeks after radiation injury (P > 0.05). Furthermore, the expression levels of the inflammatory cytokines interleukin-1ß (IL-1ß) and IL-18 were not significantly different among the groups (P > 0.05). The expression levels of cleaved caspase-1 and -3, indicators of apoptosis, were higher in the irradiation groups than in the control group at 4 days post irradiation, especially for caspase-3 (P < 0.05), but this increase was slightly attenuated at 8 weeks after radiation injury. Four days post irradiation, the MEMRI signal intensity (SI) in the irradiation groups, especially the 25 Gy group, was significantly lower than that in the control group (P < 0.05). Eight weeks after radiation injury, the SI of the 15 Gy group and the 25 Gy group recovered by different degrees, but the SI of the 25 Gy group was still significantly lower than that of the control group (P < 0.05). On day 4 post irradiation, the metabolic ratio of N-acetylaspartate (NAA) to creatine (Cr) in the 15 Gy group and 25 Gy group was significantly lower than that in the control group (P < 0.05). The NAA/Cr ratio in the 15 Gy group recovered to control levels at 8 weeks (P > 0.05), but the NAA/Cr ratio in the 25 Gy group remained significantly lower than that in the control group (P < 0.05). CONCLUSION: Radiation-induced brain injury is dose-dependently associated with apoptosis but not inflammasomes or pyroptosis, and the change in apoptosis can be detected by MEMRI.


Asunto(s)
Apoptosis/efectos de la radiación , Lesiones Encefálicas/patología , Hipocampo/efectos de la radiación , Traumatismos Experimentales por Radiación/patología , Animales , Ácido Aspártico/análogos & derivados , Ácido Aspártico/metabolismo , Lesiones Encefálicas/diagnóstico por imagen , Lesiones Encefálicas/etiología , Lesiones Encefálicas/metabolismo , Caspasas/metabolismo , Creatina/metabolismo , Hipocampo/diagnóstico por imagen , Hipocampo/metabolismo , Hipocampo/patología , Inflamasomas/metabolismo , Inflamasomas/efectos de la radiación , Imagen por Resonancia Magnética , Masculino , Compuestos de Manganeso/administración & dosificación , Dosis de Radiación , Traumatismos Experimentales por Radiación/diagnóstico por imagen , Traumatismos Experimentales por Radiación/etiología , Traumatismos Experimentales por Radiación/metabolismo , Ratas , Ratas Sprague-Dawley
7.
FASEB J ; 34(5): 6437-6448, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32190930

RESUMEN

DNA damage accumulates in aged postmitotic retinal pigment epithelium (RPE) cells, a phenomenon associated with the development of age-related macular degeneration. In this study, we have experimentally induced DNA damage by ultraviolet B (UVB) irradiation in interleukin-1α (IL-1α)-primed ARPE-19 cells and examined inflammasome-mediated signaling. To reveal the mechanisms of inflammasome activation, cells were additionally exposed to high levels of extracellular potassium chloride, n-acetyl-cysteine, or mitochondria-targeted antioxidant MitoTEMPO, prior to UVB irradiation. Levels of interleukin-18 (IL-18) and IL-1ß mRNAs were detected with qRT-PCR and secreted amounts of IL-1ß, IL-18, and caspase-1 were measured with ELISA. The role of nucleotide-binding domain and leucine-rich repeat pyrin containing protein 3 (NLRP3) in UVB-induced inflammasome activation was verified by using the NLRP3-specific siRNA. Reactive oxygen species (ROS) levels were measured immediately after UVB exposure using the cell-permeant 2',7'-dichlorodihydrofluorescein diacetate (H2 DCFDA) indicator, the levels of cyclobutane pyrimidine dimers were assayed by cell-based ELISA, and the extracellular levels of adenosine triphosphate (ATP) determined using a commercial bioluminescence assay. We found that pro-IL-18 was constitutively expressed by ARPE-19 cells, whereas the expression of pro-IL-1ß was inducible by IL-1α priming. UVB induced the release of mature IL-18 and IL-1ß but NLRP3 contributed only to the secretion of IL-1ß. At the mechanistic level, the release of IL-1ß was regulated by K+ efflux, whereas the secretion of IL-18 was dependent on ROS production. As well as K+ efflux, the cells released ATP following UVB exposure. Collectively, our data suggest that UVB clearly stimulates the secretion of mature IL-18 as a result of ROS induction, and this response is associated with DNA damage. Moreover, in human RPE cells, K+ efflux mediates the UVB-activated NLRP3 inflammasome signaling, leading to the processing of IL-1ß.


Asunto(s)
Inflamasomas/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Rayos Ultravioleta , Daño del ADN , Reparación del ADN , Humanos , Inflamasomas/inmunología , Inflamasomas/efectos de la radiación , Especies Reactivas de Oxígeno/metabolismo , Epitelio Pigmentado de la Retina/inmunología , Epitelio Pigmentado de la Retina/efectos de la radiación , Transducción de Señal
8.
Curr Eye Res ; 44(1): 67-75, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30198786

RESUMEN

PURPOSE: Age-related macular degeneration (AMD) is a major cause of blindness in the elderly, and the activation of the NACHT, LRR, and PYD domain-containing protein 3 (NLRP3) inflammasome is involved in AMD pathogenesis. We investigated whether photooxidative blue-light stimulation in retinal pigment epithelium (RPE) cells promotes exosome secretion and modulates the activity of the NLRP3 inflammasome in vitro. METHODS: Exosomes were isolated from ARPE-19 cultures stimulated or not with blue-light photostimulation (488 nm). Isolated exosomes were characterized by transmission electron microscope and Western blot analyses. The contents of the NLRP3 inflammasome (IL-1ß, IL-18, and caspase-1 as markers of the inflammasome) in exosomes were analyzed by Western blotting. After culture, IL-1ß, IL-18, and caspase-1 in RPE cells were analyzed by both immunofluorescence and Western blotting. RT-PCR and Western blotting were conducted to assess the contents of NLRP3 in RPE cells. RESULTS: Exosomes exhibited a typical characteristic morphology (cup-shaped) and size (diameter between 50 and 150 nm) in both groups. The exosome markers CD9, CD63, and CD81 were strongly present. After blue-light photostimulation, ARPE-19 cells were noted to release exosomes with higher levels of IL-1ß, IL-18, and caspase-1 than those in the control group. The levels of IL-1ß, IL-18, and caspase-1 in ARPE-19 cells were significantly enhanced when treated with stressed RPE exosomes. Additionally, the NLRP3 mRNA and protein levels were found to be markedly higher in the treated group than in the control group. CONCLUSIONS: Under photooxidative blue-light stimulation, RPE-derived exosomes may aggravate a potentially harmful oxidative response through the upregulation of the NLRP3 inflammasome.


Asunto(s)
Exosomas/metabolismo , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Células Cultivadas , Color , Exosomas/efectos de la radiación , Humanos , Inflamasomas/efectos de la radiación , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Oxidación-Reducción , Epitelio Pigmentado de la Retina/efectos de la radiación
9.
Int J Radiat Oncol Biol Phys ; 101(1): 107-117, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29456024

RESUMEN

PURPOSE: To investigate changes induced in mouse intestines after irradiation and to explore the potential radioprotective effects of flagellin A N/C (FlaAN/C). METHODS AND MATERIALS: A mouse model of radiation-induced enteropathy was used in this study. A 10-Gy abdominal irradiation was performed on FlaAN/C- and vehicle-injected mice to explore the role of FlaAN/C in intestinal radiation injury and to study the molecular mechanism in this process. In the intestinal tissue, pathologic changes were investigated by hematoxylin and eosin staining, TUNEL staining, and immunohistochemistry; Western blotting and quantitative reverse transcription-polymerase chain reaction were used to determine the changes in protein and messenger RNA levels, respectively; and an enzyme-linked immunosorbent assay was performed to determine protein concentration in serum. The involvement of the reactive oxygen species pathway was investigated by determining superoxide dismutase, glutathione (GSH) peroxidase, GSH reductase, and GSH disulfide (GSSG) plus GSH activities. RESULTS: Flagellin A N/C inhibited radiation-induced reactive oxygen species production, decreased NLRP3 activity, and reduced the occurrence of caspase-1-dependent pyroptosis. The results revealed that oxidative stress, bioenergetic impairment, and subsequent NLRP3 inflammasome activation were involved in radiation-induced intestinal injury. Flagellin A N/C exerted a protective effect by blunting the activation of NLRP3 inflammasome signaling, thereby reducing the inflammatory response and occurrence of caspase-1-dependent pyroptosis in the intestine. Therefore, treatment of mice with FlaAN/C reduced radiation-induced intestinal injury. CONCLUSIONS: Reactive oxygen species-induced NLRP3 inflammasomes mediated radiation-induced pyroptosis of the intestinal cells, and FlaAN/C suppressed pyroptosis to protect the intestinal tissue. These results provide novel insights into the mechanisms underlying radiation-induced cytotoxicity, and FlaAN/C might be a potential preventive therapy for radiation-induced intestinal injury in patients with cancer.


Asunto(s)
Flagelina/farmacología , Inflamasomas , Intestino Delgado/efectos de la radiación , Proteínas NLR/efectos de la radiación , Dominio Pirina/efectos de los fármacos , Piroptosis/efectos de los fármacos , Traumatismos Experimentales por Radiación/prevención & control , Protectores contra Radiación/farmacología , Especies Reactivas de Oxígeno/metabolismo , Animales , Benzamidas/farmacología , Caspasa 1 , Inhibidores de Caspasas/farmacología , Dipéptidos/farmacología , Femenino , Indazoles/farmacología , Inflamasomas/efectos de los fármacos , Inflamasomas/efectos de la radiación , Interleucina-1beta/antagonistas & inhibidores , Interleucinas/metabolismo , Intestino Delgado/metabolismo , Ratones , Ratones Endogámicos BALB C , Proteínas NLR/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Dominio Pirina/efectos de la radiación , Distribución Aleatoria , para-Aminobenzoatos/farmacología
10.
Science ; 354(6313): 765-768, 2016 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-27846608

RESUMEN

Acute exposure to ionizing radiation induces massive cell death and severe damage to tissues containing actively proliferating cells, including bone marrow and the gastrointestinal tract. However, the cellular and molecular mechanisms underlying this pathology remain controversial. Here, we show that mice deficient in the double-stranded DNA sensor AIM2 are protected from both subtotal body irradiation-induced gastrointestinal syndrome and total body irradiation-induced hematopoietic failure. AIM2 mediates the caspase-1-dependent death of intestinal epithelial cells and bone marrow cells in response to double-strand DNA breaks caused by ionizing radiation and chemotherapeutic agents. Mechanistically, we found that AIM2 senses radiation-induced DNA damage in the nucleus to mediate inflammasome activation and cell death. Our results suggest that AIM2 may be a new therapeutic target for ionizing radiation exposure.


Asunto(s)
Apoptosis/efectos de la radiación , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/metabolismo , Inflamasomas/metabolismo , Traumatismos por Radiación/metabolismo , Animales , Médula Ósea/metabolismo , Médula Ósea/efectos de la radiación , Caspasa 1/genética , Caspasa 1/metabolismo , ADN/metabolismo , ADN/efectos de la radiación , Proteínas de Unión al ADN/genética , Hematopoyesis/efectos de los fármacos , Hematopoyesis/genética , Inflamasomas/genética , Inflamasomas/efectos de la radiación , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Traumatismos por Radiación/genética , Radiación Ionizante , Irradiación Corporal Total
11.
Inflammation ; 39(5): 1827-34, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27525422

RESUMEN

This study aims to investigate the influence of radiotherapy on absent in melanoma 2 (AIM2) inflammasome in radiation pneumonitis (RP). A rat model of RP was established. H&E staining was used to test radiation-induced lung tissue injury. Immunohistochemistry (IHC) was used to detect the expression of AIM2 and IL-1ß in rat lung tissues. Milliplex assay was used to test cytokine levels in rat serum. Comet assay was adopted to examine DNA breaks in THP1 cells. RT-PCR was used to detect the messenger RNA (mRNA) expression of AIM2, caspase-1, and IL-1ß in THP1 cells. As a result, the rat model indicated that irradiation induced obvious lung injury. A large amount of inflammatory cells infiltrated to the irradiated lung tissues. The structure of lung tissues collapsed. IHC revealed that AIM2 and IL-1ß expressions were significantly higher in irradiated lung tissues than in the control. IL-1ß level in rat serum significantly elevated on the 7th day post-irradiation, gradually decreased on the 15th day, and became minimal on the 30th day. Irradiation induced dsDNA break in a dose-dependent manner at 24 h after irradiation. Radiotherapy increased the mRNA expression level of AIM2 and IL-1ß in a time-dependent manner. In conclusion, radiotherapy triggered some critical components of AIM2 inflammasome in RP. The activation of AIM2 inflammasome by radiotherapy may contribute to the pathogenesis of RP.


Asunto(s)
Inflamasomas/efectos de la radiación , Neumonitis por Radiación/etiología , Radioterapia/efectos adversos , Animales , Proteínas de Unión al ADN/análisis , Interleucina-1beta/análisis , Pulmón/química , Lesión Pulmonar , Neumonitis por Radiación/patología , Ratas , Factores de Tiempo
13.
J Mol Med (Berl) ; 94(7): 809-19, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26864306

RESUMEN

UNLABELLED: Understanding the underlying molecular mechanisms in burn wound progression is crucial to providing appropriate diagnoses and designing therapeutic regimens for burn patients. When inflammation becomes unregulated, recurrent, or excessive, it interferes with burn wound healing. Autophagy, which is a homeostatic and catabolic degradation process, was found to protect against ischemic injury, inflammatory diseases, and apoptosis in some cases. In the present study, we investigated whether far-infrared (FIR) could ameliorate burn wound progression and promote wound healing both in vitro and in a rat model of deep second-degree burn. We found that FIR induced autophagy in differentiated THP-1 cells (human monocytic cells differentiated to macrophages). Furthermore, FIR inhibited both the NLRP3 inflammasome and the production of IL-1ß in lipopolysaccharide-activated THP-1 macrophages. In addition, FIR induced the ubiquitination of ASC, which is the adaptor protein of the inflammasome, by increasing tumor necrosis factor receptor-associated factor 6 (TRAF6), which is a ubiquitin E3 ligase. Furthermore, the exposure to FIR then promoted the delivery of inflammasome to autophagosomes for degradation. In a rat burn model, FIR ameliorated burn-induced epidermal thickening, inflammatory cell infiltration, and loss of distinct collagen fibers. Moreover, FIR enhanced autophagy and suppressed the activity of the NLRP3 inflammasome in the rat skin tissue of the burn model. Based on these results, we suggest that FIR-regulated autophagy and inflammasomes will be important for the discovery of novel therapeutics to promote the healing of burn wounds. KEY MESSAGES: Far-infrared (FIR) induced autophagy in THP-1 macrophages. FIR suppressed the NLRP3 inflammasome through the activation of autophagy. FIR induced the ubiquitination of ASC by increasing TRAF6. FIR ameliorated burn wound progression and promoted wound healing in a rat burn model.


Asunto(s)
Autofagia/efectos de la radiación , Quemaduras/terapia , Inflamasomas/efectos de la radiación , Rayos Infrarrojos/uso terapéutico , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Cicatrización de Heridas/efectos de la radiación , Animales , Autofagia/efectos de los fármacos , Autofagia/genética , Quemaduras/genética , Quemaduras/inmunología , Quemaduras/patología , Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Adaptadoras de Señalización CARD/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Inflamasomas/efectos de los fármacos , Inflamasomas/genética , Inflamasomas/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Péptidos y Proteínas de Señalización Intracelular , Lipopolisacáridos/farmacología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/efectos de la radiación , Masculino , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Ratas , Ratas Wistar , Factor 6 Asociado a Receptor de TNF/agonistas , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/inmunología , Ubiquitinación/efectos de la radiación , Cicatrización de Heridas/genética
14.
J Immunol ; 194(6): 2569-77, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25681340

RESUMEN

Extracorporeal photopheresis (ECP) is a widely used clinical cell-based therapy exhibiting efficacy in heterogenous immune-mediated diseases such as cutaneous T cell lymphoma, graft-versus-host disease, and organ allograft rejection. Despite its documented efficacy in cancer immunotherapy, little is known regarding the induction of immunostimulatory mediators by ECP. In this article, we show that ECP promotes marked release of the prototypic immunostimulatory cytokine IL-1ß. ECP primes IL-1ß production and activates IL-1ß maturation and release in the context of caspase-1 activation in monocytes and myeloid dendritic cells. Of interest, IL-1ß maturation by ECP was fully intact in murine cells deficient in caspase-1, suggesting the predominance of an inflammasome-independent pathway for ECP-dependent IL-1ß maturation. Clinically, patient analysis revealed significantly increased IL-1ß production in stimulated leukapheresis concentrates and peripheral blood samples after ECP. Collectively, these results provide evidence for promotion of IL-1ß production by ECP and offer new insight into the immunostimulatory capacity of ECP.


Asunto(s)
Interleucina-1beta/biosíntesis , Interleucina-1beta/sangre , Leucocitos Mononucleares/metabolismo , Fotoféresis/métodos , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/farmacología , Animales , Caspasa 1/genética , Caspasa 1/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Células Dendríticas/efectos de la radiación , Activación Enzimática/efectos de los fármacos , Activación Enzimática/efectos de la radiación , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Inflamasomas/efectos de los fármacos , Inflamasomas/metabolismo , Inflamasomas/efectos de la radiación , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/efectos de la radiación , Receptores de Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Metoxaleno/farmacología , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Monocitos/efectos de la radiación , Fármacos Fotosensibilizantes/farmacología , Rayos Ultravioleta
15.
J Dermatol Sci ; 76(3): 173-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25306296

RESUMEN

BACKGROUND: In patients with rosacea, environmental stressors, especially UVB radiation, trigger disease flares that are characterized by inflammation and vascular hyperactivity. An altered innate immune detection and response system, modulated to a large extent by the aberrant production and processing of human cathelicidin LL-37, is thought to play a central role in disease pathogenesis. OBJECTIVE: To investigate whether the proinflammatory and proangiogenic effects of UV radiation are enhanced in the presence of cathelicidin LL-37. METHODS: Human skin ex vivo and epidermal keratinocytes in vitro were exposed to UVB irradiation. The proinflammatory effects of UVB exposure in the presence and absence of LL-37 were characterized using immunoblot, transfection, qPCR, and a cell-based second messenger assay. ELISA was used to assess cytokine release and the angiogenic potential of endothelial cells was evaluated using an in vitro angiogenesis assay. RESULTS: UVB irradiation triggered the inflammasome-mediated processing and release of IL-1ß. LL-37 augmented this UV-induced IL-1ß secretion by acting on the P2X7 receptor on keratinocytes. P2X7 receptor activation by UVB and LL-37 resulted in an increase in intracellular calcium concentrations, which enhances inflammasome activation and subsequent IL-1ß release. Furthermore, IL-1ß and LL-37 worked synergistically to increase the angiogenic potential of endothelial cells. CONCLUSION: Cathelicidin LL-37 modulates the proinflammatory and proangiogenic effects of UV radiation and thereby contributes to enhanced sensitivity to sun exposure in rosacea.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/metabolismo , Inflamasomas/metabolismo , Inflamasomas/efectos de la radiación , Rosácea/etiología , Rosácea/metabolismo , Rayos Ultravioleta/efectos adversos , Calcio/metabolismo , Células Cultivadas , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-1beta/metabolismo , Queratinocitos/metabolismo , Queratinocitos/efectos de la radiación , Neovascularización Patológica/etiología , Tolerancia a Radiación/fisiología , Receptores Purinérgicos P2X7/metabolismo , Catelicidinas
16.
Photochem Photobiol ; 88(5): 1111-25, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22631445

RESUMEN

Although keratinocytes are relatively resistant to ultraviolet radiation (UVR) induced damage, repeated UVR exposure result in accumulated DNA mutations that can lead to epidermal malignancies. Keratinocytes play a central role in elaborating innate responses that lead to inflammation and influence the generation of adaptive immune responses in skin. Apart from the minor cellular constituents of the epidermis, specifically Langerhans cells and melanocytes, keratinocytes are the major source of cytokines. UVR exposure stimulates keratinocytes to secrete abundant pro-inflammatory IL-1-family proteins, IL-1α, IL-1ß, IL-18, and IL-33. Normal skin contains only low levels of inactive precursor forms of IL-1ß and IL-18, which require caspase 1-mediated proteolysis for their maturation and secretion. However, caspase-1 activation is not constitutive, but dependents on the UV-induced formation of an active inflammasome complex. IL-1 family cytokines can induce a secondary cascade of mediators and cytokines from keratinocytes and other cells resulting in wide range of innate processes including infiltration of inflammatory leukocytes, induction of immunosuppression, DNA repair or apoptosis. Thus, the ability of keratinocytes to produce a wide repertoire of proinflammatory cytokines can influence the immune response locally as well as systematically, and alter the host response to photodamaged cells. We will highlight differential roles played by each IL-1 family molecule generated by UV-damaged keratinocytes, and reveal their complementary influences in modulating acute inflammatory and immunological events that follow cutaneous UV exposure.


Asunto(s)
Epidermis/efectos de la radiación , Inflamasomas/efectos de la radiación , Interleucina-18/inmunología , Interleucina-1alfa/inmunología , Interleucina-1beta/inmunología , Interleucinas/inmunología , Queratinocitos/efectos de la radiación , Apoptosis/inmunología , Apoptosis/efectos de la radiación , ADN/metabolismo , Reparación del ADN/inmunología , Reparación del ADN/efectos de la radiación , Células Epidérmicas , Epidermis/inmunología , Humanos , Inmunidad Innata/efectos de la radiación , Inflamasomas/inmunología , Inflamación/inmunología , Interleucina-18/metabolismo , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Interleucina-33 , Interleucinas/metabolismo , Queratinocitos/citología , Transducción de Señal/inmunología , Transducción de Señal/efectos de la radiación , Rayos Ultravioleta/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...