Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 52(5): 782-793.e5, 2020 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-32272082

RESUMEN

Splenic red pulp macrophages (RPMs) contribute to erythrocyte homeostasis and are required for iron recycling. Heme induces the expression of SPIC transcription factor in monocyte-derived macrophages and promotes their differentiation into RPM precursors, pre-RPMs. However, the requirements for differentiation into mature RPMs remain unknown. Here, we have demonstrated that interleukin (IL)-33 associated with erythrocytes and co-cooperated with heme to promote the generation of mature RPMs through activation of the MyD88 adaptor protein and ERK1/2 kinases downstream of the IL-33 receptor, IL1RL1. IL-33- and IL1RL1-deficient mice showed defective iron recycling and increased splenic iron deposition. Gene expression and chromatin accessibility studies revealed a role for GATA transcription factors downstream of IL-33 signaling during the development of pre-RPMs that retained full potential to differentiate into RPMs. Thus, IL-33 instructs the development of RPMs as a response to physiological erythrocyte damage with important implications to iron recycling and iron homeostasis.


Asunto(s)
Proteína 1 Similar al Receptor de Interleucina-1/inmunología , Interleucina-33/inmunología , Hierro/metabolismo , Macrófagos/inmunología , Transducción de Señal/inmunología , Bazo/metabolismo , Animales , Eritrocitos/inmunología , Eritrocitos/metabolismo , Hemo/inmunología , Hemo/metabolismo , Homeostasis/inmunología , Proteína 1 Similar al Receptor de Interleucina-1/genética , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/genética , Interleucina-33/metabolismo , Macrófagos/metabolismo , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/inmunología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/inmunología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Factor 88 de Diferenciación Mieloide/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Bazo/citología
2.
Exp Dermatol ; 33(1): e14977, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38060347

RESUMEN

Notch signalling has generated considerable interest as a pathogenetic factor and a drug target in a range of human diseases. The gamma-secretase complex is crucial in the activation of Notch receptors by cleaving the intracellular domain allowing nuclear translocation. In recent years several mutations in gamma-secretase components have been discovered in patients with familial hidradenitis suppurativa (HS). This has led to hypotheses that impaired Notch signalling could be an important driver for HS in general, not only in the monogenic variants. However, no study has examined in situ Notch activation per se in HS, and some reports with conflicting results have instead been based on expression of Notch receptors or indirect measures of Notch target gene expression. In this study we established immunostaining protocols to identify native, activated Notch receptors in human skin tissue. The ability to detect changes in Notch activation was confirmed with an ex vivo skin organ model in which signal was reduced or obliterated in tissue exposed to a gamma-secretase inhibitor. Using these methods on skin biopsies from healthy volunteers and a general HS cohort we demonstrated for the first time the distribution of active Notch signalling in human apocrine-bearing skin. Quantification of activated NOTCH1 & NOTCH2 revealed similar levels in non-lesional and peri-lesional HS to that of healthy controls, thus ruling out a general defect in Notch activation in HS patients. We did find a variable but significant reduction of activated Notch in epidermis of lesional HS with a distribution that appeared related to the extent of surrounding tissue inflammation.


Asunto(s)
Hidradenitis Supurativa , Humanos , Hidradenitis Supurativa/metabolismo , Receptores Notch/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Piel/metabolismo , Inflamación/metabolismo
3.
Hum Mol Genet ; 30(21): 1919-1931, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34124757

RESUMEN

Sturge-Weber syndrome (SWS) is a neurocutaneous disorder characterized by vascular malformations affecting skin, eyes and leptomeninges of the brain, which can lead to glaucoma, seizures and intellectual disability. The discovery of a disease-causing somatic missense mutation in the GNAQ gene, encoding an alpha chain of heterotrimeric G-proteins, has initiated efforts to understand how G-proteins contribute to SWS pathogenesis. The mutation is predominantly detected in endothelial cells and is currently believed to affect downstream MAPK signalling. In this study of six Norwegian patients with classical SWS, we aimed to identify somatic mutations through deep sequencing of DNA from skin biopsies. Surprisingly, one patient was negative for the GNAQ mutation, but instead harbored a somatic mutation in GNB2 (NM_005273.3:c.232A>G, p.Lys78Glu), which encodes a beta chain of the same G-protein complex. The positions of the mutant amino acids in the G-protein are essential for complex reassembly. Therefore, failure of reassembly and continuous signalling is a likely consequence of both mutations. Ectopic expression of mutant proteins in endothelial cells revealed that expression of either mutant reduced cellular proliferation, yet regulated MAPK signalling differently, suggesting that dysregulated MAPK signalling cannot fully explain the SWS phenotype. Instead, both mutants reduced synthesis of Yes-associated protein (YAP), a transcriptional co-activator of the Hippo signalling pathway, suggesting a key role for this pathway in the vascular pathogenesis of SWS. The discovery of the GNB2 mutation sheds novel light on the pathogenesis of SWS and suggests that future research on targets of treatment should be directed towards the YAP, rather than the MAPK, signalling pathway.


Asunto(s)
Proteínas de Unión al GTP/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Mutación , Síndrome de Sturge-Weber/diagnóstico , Síndrome de Sturge-Weber/genética , Adolescente , Adulto , Niño , Análisis Mutacional de ADN , Proteínas de Unión al GTP/química , Frecuencia de los Genes , Estudios de Asociación Genética/métodos , Humanos , Persona de Mediana Edad , Modelos Moleculares , Nortriptilina , Fenotipo , Conformación Proteica , Subunidades de Proteína/genética , Relación Estructura-Actividad , Secuenciación del Exoma , Adulto Joven
4.
Dermatology ; 238(1): 109-120, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-33887725

RESUMEN

BACKGROUND: The pathophysiology in atopic dermatitis (AD) is not fully understood, but immune dysfunction, skin barrier defects, and alterations of the skin microbiota are thought to play important roles. AD skin is frequently colonized with Staphylococcus aureus (S. aureus) and microbial diversity on lesional skin (LS) is reduced compared to on healthy skin. Treatment with narrow-band ultraviolet B (nb-UVB) leads to clinical improvement of the eczema and reduced abundance of S. aureus. However, in-depth knowledge of the temporal dynamics of the skin microbiota in AD in response to nb-UVB treatment is lacking and could provide important clues to decipher whether the microbial changes are primary drivers of the disease, or secondary to the inflammatory process. OBJECTIVES: To map the temporal shifts in the microbiota of the skin, nose, and throat in adult AD patients after nb-UVB treatment. METHODS: Skin swabs were taken from lesional AD skin (n = 16) before and after 3 treatments of nb-UVB, and after 6-8 weeks of full-body treatment. We also obtained samples from non-lesional skin (NLS) and from the nose and throat. All samples were characterized by 16S rRNA gene sequencing. RESULTS: We observed shifts towards higher diversity in the microbiota of lesional AD skin after 6-8 weeks of treatment, while the microbiota of NLS and of the nose/throat remained unchanged. After only 3 treatments with nb-UVB, there were no significant changes in the microbiota. CONCLUSION: Nb-UVB induces changes in the skin microbiota towards higher diversity, but the microbiota of the nose and throat are not altered.


Asunto(s)
Dermatitis Atópica/microbiología , Dermatitis Atópica/radioterapia , Microbiota/efectos de la radiación , Piel/microbiología , Terapia Ultravioleta , Adulto , Anciano , Biodiversidad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Nariz/microbiología , Faringe/microbiología , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus aureus/efectos de la radiación , Resultado del Tratamiento , Adulto Joven
5.
Mol Med ; 27(1): 29, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33771098

RESUMEN

BACKGROUND: Alarmins are considered proximal mediators of the immune response after tissue injury. Understanding their biology could pave the way for development of new therapeutic targets and biomarkers in human disease, including multiple trauma. In this study we explored high-resolution concentration kinetics of the alarmin interleukin-33 (IL-33) early after human trauma. METHODS: Plasma samples were serially collected from 136 trauma patients immediately after hospital admission, 2, 4, 6, and 8 h thereafter, and every morning in the ICU. Levels of IL-33 and its decoy receptor sST2 were measured by immunoassays. RESULTS: We observed a rapid and transient surge of IL-33 in a subset of critically injured patients. These patients had more widespread tissue injuries and a greater degree of early coagulopathy. IL-33 half-life (t1/2) was 1.4 h (95% CI 1.2-1.6). sST2 displayed a distinctly different pattern with low initial levels but massive increase at later time points. CONCLUSIONS: We describe for the first time early high-resolution IL-33 concentration kinetics in individual patients after trauma and correlate systemic IL-33 release to clinical data. These findings provide insight into a potentially important axis of danger signaling in humans.


Asunto(s)
Interleucina-33/sangre , Heridas y Lesiones/sangre , Adulto , Biomarcadores/sangre , Femenino , Humanos , Proteína 1 Similar al Receptor de Interleucina-1/sangre , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Adulto Joven
6.
Exp Dermatol ; 30(2): 249-261, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33067891

RESUMEN

Phototherapy with narrow-band Ultraviolet B (nb-UVB) is a major therapeutic option in atopic dermatitis (AD), yet knowledge of the early molecular responses to this treatment is lacking. The objective of this study was to map the early transcriptional changes in AD skin in response to nb-UVB treatment. Adult patients (n = 16) with AD were included in the study and scored with validated scoring tools. AD skin was irradiated with local nb-UVB on day 0, 2 and 4. Skin biopsies were taken before and after treatment (day 0 and 7) and analysed for genome-wide modulation of transcription. When examining the early response after three local UVB treatments, gene expression analysis revealed 77 significantly modulated transcripts (30 down- and 47 upregulated). Among them were transcripts related to the inflammatory response, melanin synthesis, keratinization and epidermal structure. Interestingly, the pro-inflammatory cytokine IL-36γ was reduced after treatment, while the anti-inflammatory cytokine IL-37 increased after treatment with nb-UVB. There was also a modulation of several other mediators involved in inflammation, among them defensins and S100 proteins. This is the first study of early transcriptomic changes in AD skin in response to nb-UVB. We reveal robust modulation of a small group of inflammatory and anti-inflammatory targets, including the IL-1 family members IL36γ and IL-37, which is evident before any detectable changes in skin morphology or immune cell infiltrates. These findings provide important clues to the molecular mechanisms behind the treatment response and shed light on new potential treatment targets.


Asunto(s)
Dermatitis Atópica/genética , Dermatitis Atópica/radioterapia , Interleucina-1/genética , Transcripción Genética/efectos de la radiación , Terapia Ultravioleta , Adulto , Anciano , Defensinas/genética , Dermatitis Atópica/patología , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Proteínas S100/genética , Factores de Tiempo , Rayos Ultravioleta , Adulto Joven
7.
Mol Med ; 26(1): 5, 2019 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-31892315

RESUMEN

BACKGROUND: HMGB1 is a mediator of systemic inflammation in sepsis and trauma, and a promising biomarker in many diseases. There is currently no standard operating procedure for pre-analytical handling of HMGB1 samples, despite that pre-analytical conditions account for a substantial part of the overall error rate in laboratory testing. We hypothesized that the considerable variations in reported HMGB1 concentrations and kinetics in trauma patients could be partly explained by differences in pre-analytical conditions and choice of sample material. METHODS: Trauma patients (n = 21) admitted to a Norwegian Level I trauma center were prospectively included. Blood was drawn in K2EDTA coated tubes and serum tubes. The effects of delayed centrifugation were evaluated in samples stored at room temperature for 15 min, 3, 6, 12, and 24 h respectively. Plasma samples subjected to long-term storage in - 80 °C and to repeated freeze/thaw cycles were compared with previously analyzed samples. HMGB1 concentrations in simultaneously acquired arterial and venous samples were also compared. HMGB1 was assessed by standard ELISA technique, additionally we investigated the suitability of western blot in both serum and plasma samples. RESULTS: Arterial HMGB1 concentrations were consistently lower than venous concentrations in simultaneously obtained samples (arterial = 0.60 x venous; 95% CI 0.30-0.90). Concentrations in plasma and serum showed a strong linear correlation, however wide limits of agreement. Storage of blood samples at room temperature prior to centrifugation resulted in an exponential increase in plasma concentrations after ≈6 h. HMGB1 concentrations were fairly stable in centrifuged plasma samples subjected to long-term storage and freeze/thaw cycles. We were not able to detect HMGB1 in either serum or plasma from our trauma patients using western blotting. CONCLUSIONS: Arterial and venous HMGB1 concentrations cannot be directly compared, and concentration values in plasma and serum must be compared with caution due to wide limits of agreement. Although HMGB1 levels in clinical samples from trauma patients are fairly stable, strict adherence to a pre-analytical protocol is advisable in order to protect sample integrity. Surprisingly, we were unable to detect HMGB1 utilizing standard western blot analysis.


Asunto(s)
Arterias/metabolismo , Recolección de Muestras de Sangre/métodos , Proteína HMGB1/sangre , Venas/metabolismo , Heridas y Lesiones/metabolismo , Adolescente , Adulto , Anciano , Biomarcadores/sangre , Recolección de Muestras de Sangre/instrumentación , Femenino , Congelación , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Temperatura , Factores de Tiempo , Adulto Joven
8.
Arterioscler Thromb Vasc Biol ; 38(4): 854-869, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29449332

RESUMEN

OBJECTIVE: Endothelial upregulation of adhesion molecules serves to recruit leukocytes to inflammatory sites and appears to be promoted by NOTCH1; however, current models based on interactions between active NOTCH1 and NF-κB components cannot explain the transcriptional selectivity exerted by NOTCH1 in this context. APPROACH AND RESULTS: Observing that Cre/Lox-induced conditional mutations of endothelial Notch modulated inflammation in murine contact hypersensitivity, we found that IL (interleukin)-1ß stimulation induced rapid recruitment of RELA (v-rel avian reticuloendotheliosis viral oncogene homolog A) to genomic sites occupied by NOTCH1-RBPJ (recombination signal-binding protein for immunoglobulin kappa J region) and that NOTCH1 knockdown reduced histone H3K27 acetylation at a subset of NF-κB-directed inflammatory enhancers. CONCLUSIONS: Our findings reveal that NOTCH1 signaling supports the expression of a subset of inflammatory genes at the enhancer level and demonstrate how key signaling pathways converge on chromatin to coordinate the transition to an infla mmatory endothelial phenotype.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Histonas/metabolismo , Inflamación/prevención & control , Interleucina-1beta/farmacología , Receptor Notch1/antagonistas & inhibidores , Receptor Notch1/metabolismo , Acetilación , Animales , Apendicitis/metabolismo , Apendicitis/patología , Células Cultivadas , Dermatitis por Contacto/genética , Dermatitis por Contacto/metabolismo , Dermatitis por Contacto/patología , Dipéptidos/farmacología , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Receptor Notch1/genética , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo
9.
J Immunol ; 198(8): 3318-3325, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28258201

RESUMEN

IL-33, required for viral clearance by cytotoxic T cells, is generally expressed in vascular endothelial cells in healthy human tissues. We discovered that endothelial IL-33 expression was stimulated as a response to adenoviral transduction. This response was dependent on MRE11, a sensor of DNA damage that can also be activated by adenoviral DNA, and on IRF1, a transcriptional regulator of cellular responses to viral invasion and DNA damage. Accordingly, we observed that endothelial cells responded to adenoviral DNA by phosphorylation of ATM and CHK2 and that depletion or inhibition of MRE11, but not depletion of ATM, abrogated IL-33 stimulation. In conclusion, we show that adenoviral transduction stimulates IL-33 expression in endothelial cells in a manner that is dependent on the DNA-binding protein MRE11 and the antiviral factor IRF1 but not on downstream DNA damage response signaling.


Asunto(s)
Infecciones por Adenoviridae/inmunología , Daño del ADN/inmunología , Células Endoteliales de la Vena Umbilical Humana/inmunología , Interleucina-33/inmunología , Adenoviridae , Infecciones por Adenoviridae/metabolismo , Proteínas de Unión al ADN/inmunología , Proteínas de Unión al ADN/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Immunoblotting , Factor 1 Regulador del Interferón/inmunología , Factor 1 Regulador del Interferón/metabolismo , Interleucina-33/biosíntesis , Proteína Homóloga de MRE11 , Reacción en Cadena de la Polimerasa , Transfección
10.
Transpl Int ; 30(8): 827-840, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28436117

RESUMEN

In stable renal transplant recipients with hyperparathyroidism, previous studies have indicated that vitamin D agonist treatment might have anti-proteinuric effects. Animal studies indicate possible anti-fibrotic and anti-inflammatory effects. Early introduction of paricalcitol in de novo renal transplant recipients might reduce proteinuria and prevent progressive allograft fibrosis. We performed a single-center, prospective, randomized, open-label trial investigating effects of paricalcitol 2 µg/day added to standard care. Participants were included 8 weeks after engraftment and followed for 44 weeks. Primary end point was change in spot urine albumin/creatinine ratio. Exploratory microarray analyses of kidney biopsies at study end investigated potential effects on gene expression. Secondary end points included change in glomerular filtration rate (GFR), pulse wave velocity (PWV), and endothelial function measured by peripheral arterial tonometry as reactive hyperemia index (RHI). Seventy-seven de novo transplanted kidney allograft recipients were included, 37 receiving paricalcitol. Paricalcitol treatment lowered PTH levels (P = 0.01) but did not significantly reduce albuminuria (P = 0.76), change vascular parameters (PWV; P = 0.98, RHI; P = 0.33), or influence GFR (P = 0.57). Allograft gene expression was not influenced. To summarize, in newly transplanted renal allograft recipients, paricalcitol reduced PTH and was well tolerated without negatively affecting kidney function. Paricalcitol did not significantly reduce/prevent albuminuria, improve parameters of vascular health, or influence allograft gene expression.


Asunto(s)
Ergocalciferoles/administración & dosificación , Trasplante de Riñón/métodos , Administración Oral , Animales , Ergocalciferoles/efectos adversos , Expresión Génica/efectos de los fármacos , Tasa de Filtración Glomerular/efectos de los fármacos , Humanos , Hiperemia/fisiopatología , Hiperparatiroidismo/tratamiento farmacológico , Hiperparatiroidismo/prevención & control , Riñón/efectos de los fármacos , Riñón/patología , Riñón/fisiopatología , Hormona Paratiroidea/sangre , Estudios Prospectivos , Proteinuria/prevención & control , Análisis de la Onda del Pulso , Receptores de Calcitriol/agonistas
11.
Hepatology ; 62(4): 1249-59, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25855031

RESUMEN

UNLABELLED: Cholangiocytes express antigen-presenting molecules, but it has been unclear whether they can present antigens. Natural killer T (NKT) cells respond to lipid antigens presented by the major histocompatibility complex class I-like molecule CD1d and are abundant in the liver. We investigated whether cholangiocytes express CD1d and present lipid antigens to NKT cells and how CD1d expression varies in healthy and diseased bile ducts. Murine and human cholangiocyte cell lines as well as human primary cholangiocytes expressed CD1d as determined by flow cytometry and western blotting. Murine cholangiocyte cell lines were able to present both exogenous and endogenous lipid antigens to invariant and noninvariant NKT cell hybridomas and primary NKT cells in a CD1d-dependent manner. A human cholangiocyte cell line, cholangiocarcinoma cell lines, and human primary cholangiocytes also presented exogenous CD1d-restricted antigens to invariant NKT cell clones. CD1d expression was down-regulated in the biliary epithelium of patients with late primary sclerosing cholangitis, primary biliary cirrhosis, and alcoholic cirrhosis compared to healthy controls. CONCLUSIONS: Cholangiocytes express CD1d and present antigens to NKT cells and CD1d expression is down-regulated in diseased biliary epithelium, findings which show that the biliary epithelium can activate an important lymphocyte subset of the liver. This is a potentially important immune pathway in the biliary system, which may be capable of regulating inflammation in the context of biliary disease.


Asunto(s)
Conductos Biliares/inmunología , Activación de Linfocitos , Células T Asesinas Naturales/inmunología , Animales , Antígenos CD1d/fisiología , Células Cultivadas , Células Epiteliales/inmunología , Epitelio/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL
12.
Eur J Immunol ; 44(9): 2625-37, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24846412

RESUMEN

Tumor-specific Th1 cells can activate tumor-infiltrating macrophages that eliminate MHC class II negative (MHC II(NEG)) tumor cells. Activated M1-like macrophages lack antigen (Ag) receptors, and are presumably unable to discriminate and thus kill both Ag-positive (Ag(POS)) and Ag-negative (Ag(NEG)) tumor cells (bystander killing). The lack of specificity of macrophage-mediated cytotoxicity might be of clinical importance as it could provide a means of avoiding tumor escape. Here, we have tested this idea using mixed populations of Ag(POS) and Ag(NEG) tumor cells in a TCR-transgenic model in which CD4(+) T cells recognize a secreted tumor-specific antigen. Surprisingly, while Ag(POS) tumor cells were recognized and rejected, Ag(NEG) cells grew unimpeded and formed tumors. We further demonstrated that macrophage-mediated cytotoxicity was spatially restricted to areas dominated by Ag(POS) tumor cells, sparing Ag(NEG) tumor cells in the vicinity. As a consequence, macrophage tumoricidal activity did not confer bystander killing in vivo. The present results offer novel insight into the mechanisms of indirect Th1-mediated elimination of MHC II(NEG) tumor cells.


Asunto(s)
Antígenos de Histocompatibilidad Clase II/inmunología , Inmunidad Celular/fisiología , Macrófagos/inmunología , Neoplasias Experimentales/inmunología , Células TH1/inmunología , Animales , Antígenos de Histocompatibilidad Clase II/genética , Ratones , Ratones SCID , Ratones Transgénicos , Neoplasias Experimentales/genética
13.
Arterioscler Thromb Vasc Biol ; 33(2): e47-55, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23162017

RESUMEN

OBJECTIVE: Interleukin (IL)-33 is a nuclear protein that is released from stressed or damaged cells to act as an alarmin. We investigated the effects of IL-33 on endothelial cells, using the prototype IL-1 family member, IL-1ß, as a reference. METHODS AND RESULTS: Human umbilical vein endothelial cells were stimulated with IL-33 or IL-1ß, showing highly similar phosphorylation of signaling molecules, induction of adhesion molecules, and transcription profiles. However, intradermally injected IL-33 elicited significantly less proinflammatory endothelial activation when compared with IL-1ß and led us to observe that quiescent endothelial cells (ppRb(low)p27(high)) were strikingly resistant to IL-33. Accordingly, the IL-33 receptor was preferentially expressed in nonquiescent cells of low-density cultures, corresponding to selective induction of adhesion molecules and chemokines. Multiparameter phosphoflow cytometry confirmed that signaling driven by IL-33 was stronger in nonquiescent cells. Manipulation of nuclear IL-33 expression by siRNA or adenoviral transduction revealed no functional link between nuclear, endogenous IL-33, and exogenous IL-33 responsiveness. CONCLUSIONS: In contrast to other inflammatory cytokines, IL-33 selectively targets nonquiescent endothelial cells. By this novel concept, quiescent cells may remain nonresponsive to a proinflammatory stimulus that concomitantly triggers a powerful response in cells that have been released from contact inhibition.


Asunto(s)
Proliferación Celular , Dermatitis/inmunología , Células Endoteliales/inmunología , Mediadores de Inflamación/metabolismo , Interleucinas/metabolismo , Piel/irrigación sanguínea , Adenoviridae/genética , Animales , Biopsia , Células Cultivadas , Senescencia Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Dermatitis/patología , Selectina E/metabolismo , Células Endoteliales/patología , Femenino , Citometría de Flujo , Vectores Genéticos , Células Endoteliales de la Vena Umbilical Humana/inmunología , Humanos , Interleucina-1beta/metabolismo , Interleucina-33 , Interleucinas/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Neovascularización Fisiológica , Fosforilación , Interferencia de ARN , Receptores de Interleucina/metabolismo , Proteína de Retinoblastoma/metabolismo , Transducción de Señal , Transcripción Genética , Transducción Genética , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Am J Pathol ; 181(3): 1099-111, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22809957

RESUMEN

The molecular mechanisms that drive expression of the alarmin interleukin-33 (IL-33) in endothelial cells are unknown. Because nuclear IL-33 is a marker of endothelial cell quiescence (corroborated in this study by coexpression of cyclin-dependent kinase inhibitor p27(Kip1)), we hypothesized that Notch signaling might be involved in regulating IL-33 expression. Activation of Notch1 by immobilized Notch ligands was sufficient to induce nuclear IL-33 expression in cultured endothelial cells. Conversely, IL-33 expression was inhibited by the γ-secretase inhibitor DAPT or by inhibiting the function of Dll4, Jagged1, Notch1, or the canonical Notch transcription factor RBP-Jκ. Insensitivity to cycloheximide indicated that IL-33 was a direct target of Notch signaling, well in line with the identification of several conserved RBP-Jκ binding sites in the IL33 gene. The in vivo expression of Dll4 but not of Jagged1 was well correlated with expression of IL-33 in quiescent vessels, and subcutaneous injection of DAPT in healthy skin reduced IL-33 expression, indicating that Notch signaling was involved. On the other hand, loss of IL-33 during angiogenesis occurred despite sustained Dll4 and Notch1 expression, suggesting that other signals may override the IL-33-driving signal in this context. Taken together, our data demonstrate that endothelial nuclear IL-33 is induced by Notch and that Dll4 may be the dominant ligand responsible for this signaling in vivo.


Asunto(s)
Células Endoteliales/citología , Células Endoteliales/metabolismo , Interleucinas/metabolismo , Receptor Notch1/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Sitios de Unión , Biomarcadores/metabolismo , Proteínas de Unión al Calcio/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Dipéptidos/farmacología , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Femenino , Sitios Genéticos/genética , Genoma Humano/genética , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interleucina-33 , Interleucinas/genética , Proteína Jagged-1 , Masculino , Proteínas de la Membrana/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/genética , Unión Proteica/efectos de los fármacos , Ratas , Ratas Wistar , Receptor Notch1/antagonistas & inhibidores , Proteínas Serrate-Jagged , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Cicatrización de Heridas/efectos de los fármacos
15.
Nat Commun ; 14(1): 3109, 2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-37253747

RESUMEN

Antibody-based blocking of vascular endothelial growth factor (VEGF) reduces choroidal neovascularization (CNV) and retinal edema, rescuing vision in patients with neovascular age-related macular degeneration (nAMD). However, poor response and resistance to anti-VEGF treatment occurs. We report that targeting the Notch ligand Jagged1 by a monoclonal antibody reduces neovascular lesion size, number of activated phagocytes and inflammatory markers and vascular leakage in an experimental CNV mouse model. Additionally, we demonstrate that Jagged1 is expressed in mouse and human eyes, and that Jagged1 expression is independent of VEGF signaling in human endothelial cells. When anti-Jagged1 was combined with anti-VEGF in mice, the decrease in lesion size exceeded that of either antibody alone. The therapeutic effect was solely dependent on blocking, as engineering antibodies to abolish effector functions did not impair the therapeutic effect. Targeting of Jagged1 alone or in combination with anti-VEGF may thus be an attractive strategy to attenuate CNV-bearing diseases.


Asunto(s)
Neovascularización Coroidal , Factor A de Crecimiento Endotelial Vascular , Humanos , Ratones , Animales , Factor A de Crecimiento Endotelial Vascular/metabolismo , Células Endoteliales/metabolismo , Neovascularización Coroidal/patología , Anticuerpos Bloqueadores/uso terapéutico , Transducción de Señal/fisiología , Modelos Animales de Enfermedad , Inhibidores de la Angiogénesis/uso terapéutico
16.
Blood ; 115(4): 906-9, 2010 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-19965622

RESUMEN

The lymphatic vasculature is important for the regulation of tissue fluid homeostasis, immune response, and lipid absorption, and the development of in vitro models should allow for a better understanding of the mechanisms regulating lymphatic vascular growth, repair, and function. Here we report isolation and characterization of lymphatic endothelial cells from human intestine and show that intestinal lymphatic endothelial cells have a related but distinct gene expression profile from human dermal lymphatic endothelial cells. Furthermore, we identify liprin beta1, a member of the family of LAR transmembrane tyrosine phosphatase-interacting proteins, as highly expressed in intestinal lymphatic endothelial cells in vitro and lymphatic vasculature in vivo, and show that it plays an important role in the maintenance of lymphatic vessel integrity in Xenopus tadpoles.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Portadoras/metabolismo , Células Endoteliales/citología , Mucosa Intestinal/citología , Vasos Linfáticos/citología , Proteínas de Xenopus/metabolismo , Xenopus laevis/fisiología , Animales , Proteínas Portadoras/genética , Células Cultivadas , Dermis/citología , Células Endoteliales/fisiología , Técnicas de Silenciamiento del Gen , Proteínas Fluorescentes Verdes/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular , Larva/fisiología , Linfangiogénesis/fisiología , Vasos Linfáticos/fisiología , Modelos Animales , Organismos Modificados Genéticamente , Proteínas de Xenopus/genética , Xenopus laevis/crecimiento & desarrollo
17.
Sci Rep ; 12(1): 11550, 2022 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-35798804

RESUMEN

Epithelial alarmins are gaining interest as therapeutic targets for chronic inflammation. The nuclear alarmin interleukin-33 (IL-33) is upregulated in the colonic mucosa of acute ulcerative colitis (UC) and may represent an early instigator of the inflammatory cascade. However, it is not clear what signals drive the expression of IL-33 in the colonic mucosa, nor is the exact role of IL-33 elucidated. We established an ex vivo model using endoscopic colonic biopsies from healthy controls and UC patients. Colonic biopsies exposed to hypo-osmotic medium induced a strong nuclear IL-33 expression in colonic crypts in both healthy controls and UC biopsies. Mucosal IL33 mRNA was also significantly increased following hypo-osmotic stress in healthy controls compared to non-stimulated biopsies (fold change 3.9, p-value < 0.02). We observed a modest induction of IL-33 in response to TGF-beta-1 stimulation, whereas responsiveness to inflammatory cytokines TNF and IFN-gamma was negligible. In conclusion our findings indicate that epithelial IL-33 is induced by hypo-osmotic stress, rather than prototypic proinflammatory cytokines in colonic ex vivo biopsies. This is a novel finding, linking a potent cytokine and alarmin of the innate immune system with cellular stress mechanisms and mucosal inflammation.


Asunto(s)
Alarminas , Colitis Ulcerosa , Interleucina-33 , Presión Osmótica , Alarminas/metabolismo , Colitis Ulcerosa/patología , Colon/patología , Citocinas/metabolismo , Humanos , Inflamación/patología , Interleucina-33/metabolismo , Mucosa Intestinal/metabolismo
18.
J Exp Med ; 201(7): 1045-51, 2005 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-15795234

RESUMEN

Expression of the chemokine receptor CCR4 is strongly associated with trafficking of specialized cutaneous memory T helper (Th) lymphocytes to the skin. However, it is unknown whether CCR4 itself participates in the development of cutaneous Th populations. We have addressed this issue via competitive bone marrow (BM) reconstitution assays; equal numbers of BM cells from CCR4(+/+) and CCR4(-/-) donors were allowed to develop side-by-side within RAG-1(-/-) hosts. Cells from both donor types developed equally well into B cells, naive CD8 T cells, naive CD4 T cells, interferon-gamma(+) Th1 cells, and interleukin-4(+) Th2 cells. In marked contrast, circulating cutaneous memory Th cells (i.e., E-selectin ligand(+) [E-lig(+)]) were more than fourfold more likely to be derived from CCR4(+/+) donors than from CCR4(-/-) donors. Most of this effect resides within the CD103(+) subset of the E-lig(+) Th population, in which donor CCR4(+/+) cells can outnumber CCR4(-/-) cells by >12-fold. No similar effect was observed for alpha4beta7(+) intestinal memory Th cells or CD103(+)/E-lig(-) Th cells. We conclude that CCR4 expression provides a competitive advantage to cutaneous Th cells, either by participating in their development from naive Th cells, or by preferentially maintaining them within the memory population over time.


Asunto(s)
Memoria Inmunológica/inmunología , Piel/inmunología , Subgrupos de Linfocitos T/inmunología , Factores de Transcripción/metabolismo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Diferenciación Celular/inmunología , Linaje de la Célula/inmunología , Dermatitis por Contacto/inmunología , Citometría de Flujo , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores CCR4 , Receptores de Quimiocina , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Sialoglicoproteínas , Subgrupos de Linfocitos T/citología , Linfocitos T Colaboradores-Inductores
19.
Am J Pathol ; 177(6): 2804-15, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21037074

RESUMEN

Interleukin-33 (IL-33) is a novel member of the interleukin-1 family that induces mucosal pathology in vivo and may drive fibrosis development and angiogenesis. To address its potential role in inflammatory bowel disease, we explored its tissue expression in biopsy specimens from untreated ulcerative colitis patients, observing a 2.6-fold up-regulation of IL-33 mRNA levels, compared to controls. Immunohistochemical analyses of surgical specimens showed that a prominent source of IL-33 in ulcerative colitis lesions were ulceration-associated myofibroblasts that co-expressed the fibroblast marker heat shock protein 47, platelet-derived growth factor receptor (PDGFR)ß, and, in part, the myofibroblast marker α-smooth muscle actin (SMA). In contrast, IL-33-positive myofibroblasts were almost absent near the deep fissures seen in Crohn's disease. A screen of known and putative activators of IL-33 in cultured fibroblasts revealed that the Toll-like receptor-3 agonist poly (I:C) was among the strongest inducers of IL-33 and that it synergized with transforming growth factor-ß, a combination also known to boost myofibroblast differentiation. Experimental wound healing in rat skin revealed that the de novo induction of IL-33 in pericytes and the possible activation of scattered, tissue-resident IL-33(+)PDGFRß(+)αSMA(-) fibroblast-like cells were early events that preceded the later appearance of IL-33(+)PDGFRß(+)αSMA(+) cells. In conclusion, our data point to a novel role for IL-33 in mucosal healing and wound repair and to an interesting difference between ulcerative colitis and Crohn's disease.


Asunto(s)
Colitis Ulcerosa/genética , Enfermedades Inflamatorias del Intestino/genética , Interleucinas/genética , Miofibroblastos/metabolismo , Animales , Biopsia , Estudios de Casos y Controles , Células Cultivadas , Colitis Ulcerosa/metabolismo , Colitis Ulcerosa/patología , Colon/efectos de los fármacos , Colon/metabolismo , Colon/patología , Enfermedad de Crohn/genética , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/patología , Expresión Génica/efectos de los fármacos , Humanos , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Interleucina-33 , Interleucinas/metabolismo , Miofibroblastos/efectos de los fármacos , Miofibroblastos/patología , Poli I-C/farmacología , Ratas , Receptor Toll-Like 3/agonistas , Factor de Crecimiento Transformador beta/farmacología , Cicatrización de Heridas/genética , Cicatrización de Heridas/fisiología
20.
Front Cardiovasc Med ; 8: 619690, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33718448

RESUMEN

There are several reasons to consider the role of endothelial cells in COVID-19 and other emerging viral infections. First, severe cases of COVID-19 show a common breakdown of central vascular functions. Second, SARS-CoV-2 replicates in endothelial cells. Third, prior deterioration of vascular function exacerbates disease, as the most common comorbidities of COVID-19 (obesity, hypertension, and diabetes) are all associated with endothelial dysfunction. Importantly, SARS-CoV-2's ability to infect endothelium is shared by many emerging viruses, including henipaviruses, hantavirus, and highly pathogenic avian influenza virus, all specifically targeting endothelial cells. The ability to infect endothelium appears to support generalised dissemination of infection and facilitate the access to certain tissues. The disturbed vascular function observed in severe COVID-19 is also a prominent feature of many other life-threatening viral diseases, underscoring the need to understand how viruses modulate endothelial function. We here review the role of vascular endothelial cells in emerging viral infections, starting with a summary of endothelial cells as key mediators and regulators of vascular and immune responses in health and infection. Next, we discuss endotheliotropism as a possible virulence factor and detail features that regulate viruses' ability to attach to and enter endothelial cells. We move on to review how endothelial cells detect invading viruses and respond to infection, with particular focus on pathways that may influence vascular function and the host immune system. Finally, we discuss how endothelial cell function can be dysregulated in viral disease, either by viral components or as bystander victims of overshooting or detrimental inflammatory and immune responses. Many aspects of how viruses interact with the endothelium remain poorly understood. Considering the diversity of such mechanisms among different emerging viruses allows us to highlight common features that may be of general validity and point out important challenges.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA