Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nutr Res ; 94: 10-24, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34571215

RESUMEN

Topical 17-beta-estradiol (E2) regulates the hair cycle, hair shaft differentiation, and sebum production. Vitamin A also regulates sebum production. Vitamin A metabolism proteins localized to the pilosebaceous unit (PSU; hair follicle and sebaceous gland); and were regulated by E2 in other tissues. This study tests the hypothesis that E2 also regulates vitamin A metabolism in the PSU. First, aromatase and estrogen receptors localized to similar sites as retinoid metabolism proteins during mid-anagen. Next, female and male wax stripped C57BL/6J mice were topically treated with E2, the estrogen receptor antagonist ICI 182,780 (ICI), letrozole, E2 plus letrozole, or vehicle control (acetone) during mid-anagen. E2 or one of its inhibitors regulated most of the vitamin A metabolism genes and proteins examined in a sex-dependent manner. Most components were higher in females and reduced with ICI in females. ICI reductions occurred in the premedulla, sebaceous gland, and epidermis. Reduced E2 also reduced RA receptors in the sebaceous gland and bulge in females. However, reduced E2 increased the number of retinal dehydrogenase 2 positive hair follicle associated dermal dendritic cells in males. These results suggest that estrogen regulates vitamin A metabolism in the skin. Interactions between E2 and vitamin A have implications in acne treatment, hair loss, and skin immunity.


Asunto(s)
Proteínas Portadoras/metabolismo , Estradiol/metabolismo , Estrógenos/metabolismo , Piel/metabolismo , Tretinoina/metabolismo , Animales , Aromatasa/metabolismo , Células Dendríticas/metabolismo , Epidermis , Antagonistas del Receptor de Estrógeno/farmacología , Femenino , Fulvestrant/farmacología , Cabello , Folículo Piloso/metabolismo , Masculino , Ratones Endogámicos C57BL , Oxidorreductasas/metabolismo , Receptores de Estrógenos/metabolismo , Glándulas Sebáceas/metabolismo , Factores Sexuales
2.
Hum Gene Ther Clin Dev ; 26(4): 228-42, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26684447

RESUMEN

No treatment is currently available for mucopolysaccharidosis (MPS) IIIB, a neuropathic lysosomal storage disease due to defect in α-N-acetylglucosaminidase (NAGLU). In preparation for a clinical trial, we performed an IND-enabling GLP-toxicology study to assess systemic rAAV9-CMV-hNAGLU gene delivery in WT C57BL/6 mice at 1 × 10(14) vg/kg and 2 × 10(14) vg/kg (n = 30/group, M:F = 1:1), and non-GLP testing in MPS IIIB mice at 2 × 10(14) vg/kg. Importantly, no adverse clinical signs or chronic toxicity were observed through the 6 month study duration. The rAAV9-mediated rNAGLU expression was rapid and persistent in virtually all tested CNS and somatic tissues. However, acute liver toxicity occurred in 33% (5/15) WT males in the 2 × 10(14) vg/kg cohort, which was dose-dependent, sex-associated, and genotype-specific, likely due to hepatic rNAGLU overexpression. Interestingly, a significant dose response was observed only in the brain and spinal cord, whereas in the liver at 24 weeks postinfection (pi), NAGLU activity was reduced to endogenous levels in the high dose cohort but remained at supranormal levels in the low dose group. The possibility of rAAV9 germline transmission appears to be minimal. The vector delivery resulted in transient T-cell responses and characteristic acute antibody responses to both AAV9 and rNAGLU in all rAAV9-treated animals, with no detectable impacts on tissue transgene expression. This study demonstrates a generally safe and effective profile, and may have identified the upper dosing limit of rAAV9-CMV-hNAGLU via systemic delivery for the treatment of MPS IIIB.


Asunto(s)
Encéfalo/metabolismo , Terapia Genética/efectos adversos , Vectores Genéticos/efectos adversos , Hígado/metabolismo , Mucopolisacaridosis III/terapia , Guías de Práctica Clínica como Asunto , Médula Espinal/metabolismo , Acetilglucosaminidasa/genética , Acetilglucosaminidasa/metabolismo , Animales , Dependovirus/genética , Dependovirus/metabolismo , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos
3.
Photochem Photobiol ; 91(4): 901-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25715879

RESUMEN

Ultraviolet light B (UVB) exposure induces cutaneous squamous cell carcinoma (cSCC), one of the most prevalent human cancers. Reoccurrence of cSCC in high-risk patients is prevented by oral retinoids. But oral retinoid treatment causes significant side effects; and patients develop retinoid resistance. Exactly how retinoids prevent UVB-induced cSCC is currently not well understood. Retinoid resistance blocks mechanistic studies in the leading mouse model of cSCC, the UVB-exposed SKH-1 hairless mouse. To begin to understand the role of retinoids in UVB-induced cSCC we first examined the localization pattern of key retinoid metabolism proteins by immunohistochemistry 48 h after UVB treatment of female SKH-1 mice. We next inhibited retinoic acid (RA) synthesis immediately after UVB exposure. Acute UVB increased RA synthesis, signaling and degradation proteins in the stratum granulosum. Some of these proteins changed their localization; while other proteins just increased in intensity. In contrast, acute UVB reduced the retinoid storage protein lectin:retinol acyltransferase (LRAT) in the epidermis. Inhibiting RA synthesis disrupted the epidermis and impaired differentiation. These data suggest that repair of the epidermis after acute UVB exposure requires endogenous RA synthesis.


Asunto(s)
Epidermis/patología , Tretinoina/fisiología , Rayos Ultravioleta , Animales , Epidermis/efectos de la radiación , Femenino , Ratones , Ratones Pelados
4.
Mol Ther ; 23(4): 638-47, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25592334

RESUMEN

Mucopolysaccharidosis (MPS) IIIA is a neuropathic lysosomal storage disease caused by deficiency in N-sulfoglucosamine sulfohydrolase (SGSH). Genome-wide gene expression microarrays in MPS IIIA mice detected broad molecular abnormalities (greater than or equal to twofold, false discovery rate ≤10) in numerous transcripts (314) in the brain and blood (397). Importantly, 22 dysregulated blood transcripts are known to be enriched in the brain and linked to broad neuronal functions. To target the root cause, we used a self-complementary AAVrh74 vector to deliver the human SGSH gene into 4-6 weeks old MPS IIIA mice by an intravenous injection. The treatment resulted in global central nervous system (CNS) and widespread somatic restoration of SGSH activity, clearance of CNS and somatic glycosaminoglycan storage, improved behavior performance, and significantly extended survival. The scAAVrh74-hSGSH treatment also led to the correction of the majority of the transcriptional abnormalities in the brain (95.9%) and blood (97.7%), of which 182 and 290 transcripts were normalized in the brain and blood, respectively. These results demonstrate that a single systemic scAAVrh74-hSGSH delivery mediated efficient restoration of SGSH activity and resulted in a near complete correction of MPS IIIA molecular pathology. This study also demonstrates that blood transcriptional profiles reflect the biopathological status of MPS IIIA, and also respond well to effective treatments.


Asunto(s)
Dependovirus/genética , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Hidrolasas/genética , Mucopolisacaridosis III/terapia , Animales , Terapia Genética , Humanos , Ratones , Ratones Endogámicos C57BL
5.
J Alzheimers Dis ; 43(1): 93-108, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25079797

RESUMEN

To date, little is known regarding the etiology and disease mechanisms of Alzheimer's disease (AD). There is a general urgency for novel approaches to advance AD research. In this study, we analyzed blood RNA from female patients with advanced AD and matched healthy controls using genome-wide gene expression microarrays. Our data showed significant alterations in 3,944 genes (≥2-fold, FDR ≤1%) in AD whole blood, including 2,932 genes that are involved in broad biological functions. Importantly, we observed abnormal transcripts of numerous tissue-specific genes in AD blood involving virtually all tissues, especially the brain. Of altered genes, 157 are known to be essential in neurological functions, such as neuronal plasticity, synaptic transmission and neurogenesis. More importantly, 205 dysregulated genes in AD blood have been linked to neurological disease, including AD/dementia and Parkinson's disease, and 43 are known to be the causative genes of 42 inherited mental retardation and neurodegenerative diseases. The detected transcriptional abnormalities also support robust inflammation, profound extracellular matrix impairments, broad metabolic dysfunction, aberrant oxidative stress, DNA damage, and cell death. While the mechanisms are currently unclear, this study demonstrates strong blood-brain correlations in AD. The blood transcriptional profiles reflect the complex neuropathological status in AD, including neuropathological changes and broad somatic impairments. The majority of genes altered in AD blood have not previously been linked to AD. We believe that blood genome-wide transcriptional profiling may provide a powerful and minimally invasive tool for the identification of novel targets beyond Aß and tauopathy for AD research.


Asunto(s)
Enfermedad de Alzheimer/sangre , Anciano , Femenino , Perfilación de la Expresión Génica , Humanos , Análisis por Micromatrices , Reacción en Cadena en Tiempo Real de la Polimerasa
6.
Hum Gene Ther Clin Dev ; 25(2): 72-84, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24720466

RESUMEN

No treatment is currently available for mucopolysaccharidosis (MPS) IIIB, a neuropathic lysosomal storage disease caused by autosomal recessive defect in α-N-acetylglucosaminidase (NAGLU). In anticipation of a clinical gene therapy treatment for MPS IIIB in humans, we tested the rAAV9-CMV-hNAGLU vector administration to cynomolgus monkeys (n=8) at 1E13 vg/kg or 2E13 vg/kg via intravenous injection. No adverse events or detectable toxicity occurred over a 6-month period. Gene delivery resulted in persistent global central nervous system and broad somatic transduction, with NAGLU activity detected at 2.9-12-fold above endogenous levels in somatic tissues and 1.3-3-fold above endogenous levels in the brain. Secreted rNAGLU was detected in serum. Low levels of preexisting anti-AAV9 antibodies (Abs) did not diminish vector transduction. Importantly, high-level preexisting anti-AAV9 Abs lead to reduced transduction in liver and other somatic tissues, but had no detectable impact on transgene expression in the brain. Enzyme-linked immunoabsorbent assay showed Ab responses to both AAV9 and rNAGLU in treated animals. Serum anti-hNAGLU Abs, but not anti-AAV9 Abs, correlated with the loss of circulating rNAGLU enzyme. However, serum Abs did not affect tissue rNAGLU activity levels. Weekly or monthly peripheral blood interferon-γ enzyme-linked immunospot assays detected a CD4(+) T-cell (Th-1) response to rNAGLU only at 4 weeks postinjection in one treated subject, without observable correlation to tissue transduction levels. The treatment did not result in detectable CTL responses to either AAV9 or rNAGLU. Our data demonstrate an effective and safe profile for systemic rAAV9-hNAGLU vector delivery in nonhuman primates, supporting its clinical potential in humans.


Asunto(s)
Acetilglucosaminidasa/genética , Dependovirus/genética , Vectores Genéticos/metabolismo , Mucopolisacaridosis III/terapia , Acetilglucosaminidasa/inmunología , Acetilglucosaminidasa/metabolismo , Animales , Anticuerpos/sangre , Anticuerpos/inmunología , Encéfalo/metabolismo , Sistema Nervioso Central/metabolismo , Dependovirus/inmunología , Ensayo de Immunospot Ligado a Enzimas , Terapia Genética , Vectores Genéticos/efectos adversos , Vectores Genéticos/genética , Humanos , Macaca fascicularis , Proteínas Recombinantes/sangre , Proteínas Recombinantes/líquido cefalorraquídeo , Células TH1/citología , Células TH1/inmunología , Distribución Tisular
7.
PLoS One ; 8(11): e80142, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24278249

RESUMEN

Mucopolysaccharidosis (MPS) IIIB is a devastating neuropathic lysosomal storage disease with complex pathology. This study identifies molecular signatures in peripheral blood that may be relevant to MPS IIIB pathogenesis using a mouse model. Genome-wide gene expression microarrays on pooled RNAs showed dysregulation of 2,802 transcripts in blood from MPS IIIB mice, reflecting pathological complexity of MPS IIIB, encompassing virtually all previously reported and as yet unexplored disease aspects. Importantly, many of the dysregulated genes are reported to be tissue-specific. Further analyses of multiple genes linked to major pathways of neurodegeneration demonstrated a strong brain-blood correlation in amyloidosis and synucleinopathy in MPS IIIB. We also detected prion protein (Prnp) deposition in the CNS and Prnp dysregulation in the blood in MPS IIIB mice, suggesting the involvement of Prnp aggregation in neuropathology. Systemic delivery of trans-BBB-neurotropic rAAV9-hNAGLU vector mediated not only efficient restoration of functional α-N-acetylglucosaminidase and clearance of lysosomal storage pathology in the central nervous system (CNS) and periphery, but also the correction of impaired neurodegenerative molecular pathways in the brain and blood. Our data suggest that molecular changes in blood may reflect pathological status in the CNS and provide a useful tool for identifying potential CNS-specific biomarkers for MPS IIIB and possibly other neurological diseases.


Asunto(s)
Amiloidosis/complicaciones , Biomarcadores/sangre , Encefalopatías/complicaciones , Mucopolisacaridosis III/complicaciones , Enfermedades por Prión/complicaciones , Sinucleínas/metabolismo , Acetilglucosaminidasa/genética , Animales , Sistema Nervioso Central/metabolismo , Ensayo de Inmunoadsorción Enzimática , Expresión Génica , Terapia Genética , Humanos , Aprendizaje por Laberinto , Ratones , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa
8.
J Invest Dermatol ; 133(2): 334-43, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23014334

RESUMEN

Alopecia areata (AA) is an autoimmune disease that attacks anagen hair follicles. Gene array in graft-induced C3H/HeJ mice revealed that genes involved in retinoic acid (RA) synthesis were increased, whereas RA degradation genes were decreased in AA compared with sham controls. This was confirmed by immunohistochemistry in biopsies from patients with AA and both mouse and rat AA models. RA levels were also increased in C3H/HeJ mice with AA. C3H/HeJ mice were fed a purified diet containing one of the four levels of dietary vitamin A or an unpurified diet 2 weeks before grafting and disease progression followed. High vitamin A accelerated AA, whereas mice that were not fed vitamin A had more severe disease by the end of the study. More hair follicles were in anagen in mice fed high vitamin A. Both the number and localization of granzyme B-positive cells were altered by vitamin A. IFNγ was also the lowest and IL13 highest in mice fed high vitamin A. Other cytokines were reduced and chemokines increased as the disease progressed, but no additional effects of vitamin A were seen. Combined, these results suggest that vitamin A regulates both the hair cycle and immune response to alter the progression of AA.


Asunto(s)
Alopecia Areata/etiología , Alopecia Areata/patología , Folículo Piloso/patología , Retinoides/metabolismo , Alopecia Areata/inmunología , Alimentación Animal , Animales , Biopsia , Quimiocina CCL5/metabolismo , Quimiocina CXCL9/metabolismo , Progresión de la Enfermedad , Granzimas/metabolismo , Folículo Piloso/crecimiento & desarrollo , Folículo Piloso/metabolismo , Humanos , Interferón gamma/metabolismo , Interleucina-13/metabolismo , Ratones , Ratones Endogámicos C3H , Ratas , Retinoides/biosíntesis , Retinoides/inmunología , Bancos de Tejidos , Vitamina A/farmacología
10.
Genes Chromosomes Cancer ; 48(6): 490-501, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19296524

RESUMEN

In immunocompromised individuals, such as organ transplant recipients, the risk of cutaneous squamous cell carcinoma (SCC) is increased 60-250 fold, and there is an increased likelihood to develop aggressive, metastatic SCC. An understanding of the genes involved in SCC tumorigenesis is critical to prevent SCC-associated morbidity and mortality. Mouse models show that different immunosuppressive drugs lead to SCCs varying in size, number, and malignant potential. In this study, we used mouse models that mimic adult transplant recipients to study the effect of immunosuppressive drugs and UV light on SCC development. UV-induced tumors from six treatment groups, control, tacrolimus (Tac), rapamycin (Rap), cyclosporin (CsA), mycophenolate mofetil (MMF), and Rap plus CsA, were evaluated by array comparative genomic hybridization. Mouse SCCs appear to show similar genomic aberrations as those reported in human SCCs and offer the ability to identify genomic changes associated with specific and combinatorial effects of drugs. Fewer aberrations were seen in tumors of mice treated with MMF or Rap. Tumors from Tac-treated animals showed the highest number of changes. Calcineurin inhibitors (Tac and CsA) did not cluster together by their genomic aberrations, indicating their contribution to UV mediated carcinogenesis may be through different pathways. The combination treatment (Rap plus CsA) did not cluster with either treatment individually, suggesting it may influence SCC tumorigenesis via a different mechanism. Future studies will identify specific genes mapping to regions of aberration that are different between treatment groups to identify target pathways that may be affected by these drugs.


Asunto(s)
Carcinoma de Células Escamosas/genética , Aberraciones Cromosómicas , Inmunosupresores/toxicidad , Neoplasias Inducidas por Radiación/genética , Neoplasias Cutáneas/genética , Rayos Ultravioleta , Análisis de Varianza , Animales , Carcinoma de Células Escamosas/inducido químicamente , Carcinoma de Células Escamosas/metabolismo , Hibridación Genómica Comparativa , Modelos Animales de Enfermedad , Femenino , Dosificación de Gen , Humanos , Huésped Inmunocomprometido , Ratones , Ratones Pelados , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Inducidas por Radiación/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/metabolismo
11.
FASEB J ; 23(3): 720-30, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18952710

RESUMEN

Mounting evidence suggests that macrophage migration inhibitory factor (MIF) may serve as an important link between chronic inflammation and cancer development. The proinflammatory and proangiogenic activities of MIF position it as a potentially important player in the development and progression of nonmelanoma skin cancer (NMSC). To assess the role of MIF in the development and progression of NMSC, we exposed MIF(-/-) BALB/c mice to acute and chronic ultraviolet B (UVB) irradiation. Our studies demonstrate that MIF(-/-) BALB/c mice have a significantly diminished acute inflammatory response to UVB exposure compared to wild-type mice, as measured by myeloperoxidase activity, dermal neutrophil infiltration, and edematous response. Relative to wild-type mice, MIF(-/-) mice also show significantly lower vascular endothelial growth factor (VEGF) concentrations in whole skin and significantly lower 8-oxo-dG adduct concentrations in epidermal DNA following UVB exposure. Furthermore, MIF(-/-) mice showed significant increases in p53 activity, epidermal thickness, and epidermal cell proliferation following acute UVB insult. In response to chronic UVB exposure, MIF(-/-) mice showed a 45% reduction in tumor incidence, significantly less angiogenesis, and delayed tumor progression when compared to their wild-type counterparts. These data indicate that MIF plays an important role in UVB-induced NMSC development and progression.


Asunto(s)
Factores Inhibidores de la Migración de Macrófagos/genética , Macrófagos/metabolismo , Neoplasias Inducidas por Radiación/genética , Neoplasias Cutáneas/genética , Rayos Ultravioleta/efectos adversos , Animales , Relación Dosis-Respuesta en la Radiación , Edema , Femenino , Regulación de la Expresión Génica , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Neoplasias Inducidas por Radiación/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Neoplasias Cutáneas/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
12.
Lab Invest ; 88(8): 831-41, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18458671

RESUMEN

We previously reported ultraviolet radiation (UVR) induction of Slug, a Snail family zinc-finger transcription factor, in the epidermis of mice; we now report that Slug-knockout mice are, unexpectedly, more resistant to sunburn than wild-type mice. There was a marked difference between the cutaneous inflammatory response in the skin of Slug-knockout and wild-type mice from 12 h to 1 week following a single exposure to 3 minimal erythemal doses of UVR. Slug-knockout mice showed a much reduced immediate increase in skin thickness and neutrophil infiltration compared to wild-type mice. However, there were as many or more intraepidermal T cells, dermal mast cells, and dermal blood vessels in the UVR-exposed skin of Slug-knockout mice as in the skin of wild-type mice. Differences in cytokine and chemokine expression following UVR appeared to account for at least some differences between the genotypes in cutaneous inflammatory response. Despite the reported antiapoptotic and antiproliferative role for Slug in some cell types, we observed little difference between the genotypes in UVR-induced keratinocyte apoptosis or proliferation. Our findings indicate an unexpected but important role for Slug in the acute cutaneous inflammatory response to UVR.


Asunto(s)
Factores Inmunológicos/metabolismo , Traumatismos Experimentales por Radiación/metabolismo , Piel/efectos de la radiación , Factores de Transcripción/metabolismo , Rayos Ultravioleta , Animales , Apoptosis/efectos de la radiación , Daño del ADN , Dermatitis/inmunología , Dermatitis/metabolismo , Dermatitis/patología , Femenino , Expresión Génica , Factores Inmunológicos/genética , Masculino , Ratones , Ratones Noqueados , Traumatismos Experimentales por Radiación/inmunología , Traumatismos Experimentales por Radiación/patología , Piel/inmunología , Piel/metabolismo , Piel/patología , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética
13.
J Invest Dermatol ; 128(10): 2467-73, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18463679

RESUMEN

Transplant immunosuppressants have been implicated in the increased incidence of non-melanoma skin cancer in transplant recipients, most of whom harbor considerable UVB-induced DNA damage in their skin prior to transplantation. This study was designed to evaluate the effects of two commonly used immunosuppressive drugs, cyclosporine A (CsA) and sirolimus (SRL), on the development and progression of UVB-induced non-melanoma skin cancer. SKH-1 hairless mice were exposed to UVB alone for 15 weeks, and then were treated with CsA, SRL, or CsA+SRL for 9 weeks following cessation of UVB treatment. Compared with vehicle, CsA treatment resulted in enhanced tumor size and progression. In contrast, mice treated with SRL or CsA+SRL had decreased tumor multiplicity, size, and progression compared with vehicle-treated mice. CsA, but not SRL or combined treatment, increased dermal mast cell numbers and TGF-beta1 levels in the skin. These findings demonstrate that specific immunosuppressive agents differentially alter the cutaneous tumor microenvironment, which in turn may contribute to enhanced development of UVB-induced skin cancer in transplant recipients. Furthermore, these results suggest that CsA alone causes enhanced growth and progression of skin cancer, whereas co-administration of SRL with CsA causes the opposite effect. JID JOURNAL CLUB ARTICLE: For questions, answers, and open discussion about this article please go to http://network.nature.com/group/jidclub


Asunto(s)
Antineoplásicos/farmacología , Ciclosporina/farmacología , Inmunosupresores/farmacología , Sirolimus/farmacología , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/prevención & control , Rayos Ultravioleta , Animales , Recuento de Células , Progresión de la Enfermedad , Femenino , Incidencia , Ganglios Linfáticos/patología , Mastocitos/patología , Ratones , Ratones Pelados , Neoplasias Inducidas por Radiación/patología , Neoplasias Inducidas por Radiación/prevención & control , Neoplasias Primarias Secundarias/epidemiología , Neoplasias Primarias Secundarias/etiología , Neoplasias Primarias Secundarias/prevención & control , Neutrófilos/patología , Piel/efectos de los fármacos , Piel/metabolismo , Piel/patología , Neoplasias Cutáneas/epidemiología , Neoplasias Cutáneas/etiología , Factor de Crecimiento Transformador beta1/metabolismo , Carga Tumoral/efectos de los fármacos
14.
J Invest Dermatol ; 127(6): 1507-15, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17363918

RESUMEN

Solid organ transplant recipients have a 60-250-fold increased likelihood of developing sunlight-induced squamous cell carcinoma (SCC) compared with the general population. This increased risk is linked to the immunosuppressive drugs taken by these patients to modulate T cell function, thus preventing organ rejection. To determine the importance of T cells in the development of cutaneous SCC, we examined the effects of selectively depleting Skh-1 mice of systemic CD4+ or CD8+ T cells, using monoclonal antibodies, on ultraviolet B (UVB) radiation-induced inflammation and tumor development. Decreases in systemic CD4+ but not CD8+ T cells significantly increased and prolonged the acute UVB-induced cutaneous inflammatory response, as measured by neutrophil influx, myeloperoxidase activity, and prostaglandin E2 levels. Significantly more p53+ keratinocytes were observed in UVB-exposed CD4-depleted than in CD4-replete mice, and this difference was abrogated in mice depleted of neutrophils before UVB exposure. Increased acute inflammation was associated with significantly increased tumor numbers in CD4-depleted mice chronically exposed to UVB. Furthermore, topical treatment with the anti-inflammatory drug celecoxib significantly decreased tumor numbers in both CD4-replete and CD4-depleted mice. Our findings suggest that CD4+ T cells play an important role in modulating both the acute inflammatory and the chronic carcinogenic response of the skin to UVB.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/efectos de la radiación , Dermatitis/inmunología , Piel/inmunología , Piel/efectos de la radiación , Rayos Ultravioleta/efectos adversos , Enfermedad Aguda , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/efectos de la radiación , Enfermedad Crónica , Dermatitis/epidemiología , Dinoprostona/metabolismo , Femenino , Huésped Inmunocomprometido/inmunología , Queratinocitos/citología , Ratones , Ratones Pelados , Neutrófilos/citología , Neutrófilos/efectos de la radiación , Peroxidasa/metabolismo , Factores de Riesgo , Neoplasias Cutáneas/epidemiología , Proteína p53 Supresora de Tumor/metabolismo
15.
Kidney Int ; 66(5): 1907-17, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15496162

RESUMEN

BACKGROUND: We investigated the expression and function of CC chemokine receptors (CCR) on highly-purified kidney and blood dendritic cells isolated from mice in which dendritic cells were mobilized with fms-like tyrosine 3 kinase ligand (Flt3L). METHODS: CCR and CC chemokine expression were determined by RNase protection assay or flow cytometry, and dendritic cell migratory responses assayed using Transwell chambers. Chemokine production in renal tissue was detected by immunofluorescence staining. Trafficking of fluorochrome-labeled dendritic cells was monitored in vivo. RESULTS: Freshly-isolated renal dendritic cells expressed mRNA for CCR1, 2, 5, and 7 and CCR1 and 5 protein. They did not migrate to inducible chemokines--CCL3 [macrophage inflammatory protein (MIP)-1alpha], CCL5 [regulated upon activation, normal T cell expressed and secreted (RANTES)], or CCL20 (MIP-3alpha). Following lipopolysaccharide (LPS) stimulation, the dendritic cells down-regulated CCR1, 2, and 5 expression, up-regulated or sustained signals for CCR7, and migrated to the constitutively expressed ligands CCL19 (MIP-3beta) and CCL21 (secondary lymphoid tissue chemokine). Normal kidneys expressed weak message for CCL2, 3, and 4, with stronger signals for CCL5 and 19. Intrarenal CCL5 production was enhanced by Flt3L administration, in association with marked increases in interstitial CD45+ mononuclear cells. Mobilized blood dendritic cells migrated to CCR2 and CCR5 ligands and trafficked to renal intertubular sites following adoptive (intravenous) transfer. Their migration to the CCR5 ligand MIP-1beta (CCL4) and homing to kidneys of Flt3L-treated recipients were inhibited by CCR5 antagonism. CONCLUSION: These data implicate specific CCR and their ligands in regulation of the dendritic cell constituency of the kidney. CCR5 antagonism inhibits their directed migration and intrarenal accumulation.


Asunto(s)
Células Sanguíneas/fisiología , Quimiocinas CC/metabolismo , Quimiotaxis/efectos de los fármacos , Células Dendríticas/fisiología , Riñón/citología , Proteínas de la Membrana/farmacología , Receptores de Quimiocina/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Células Sanguíneas/metabolismo , Senescencia Celular/fisiología , Quimiocina CCL5/metabolismo , Células Dendríticas/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Riñón/efectos de los fármacos , Riñón/metabolismo , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Transporte de Proteínas/efectos de los fármacos , ARN Mensajero/metabolismo , Receptores CCR1 , Receptores CCR2 , Receptores CCR5/genética , Receptores CCR5/inmunología , Receptores de Quimiocina/genética , Bazo , Factores de Tiempo , Regulación hacia Arriba
16.
Transplantation ; 77(7): 1080-9, 2004 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15087775

RESUMEN

BACKGROUND: Migratory antigen-presenting cells resident in kidneys may have tolerogenic potential. Difficulties inherent in their isolation have limited their characterization. The authors examined the phenotype and function of murine kidney dendritic cells (DC) mobilized in vivo by systemic administration of fms-like tyrosine 3 kinase ligand (Flt3L). METHODS: Monoclonal antibody staining was used to characterize DC subsets in situ, immediately after their isolation, and after lipopolysaccharide stimulation. Cytokine and CC chemokine receptor (CCR) gene expression was analyzed by RNase protection assay. Mixed leukocyte reactions were performed to assess DC allostimulatory ability and also the function of putative T-regulatory cells. In vivo DC trafficking was monitored by fluorescence imaging of dye-labeled cells and the influence of renal DC on vascularized heart allograft survival was determined. RESULTS: Flt3L induced a marked increase both in CD11cCD8alpha and in CD11cCD8alpha DC within the renal cortex and medulla. Rarer, CD11cB220 (precursor plasmacytoid) DC were also detected. Bulk freshly isolated DC exhibited no interleukin (IL)-12p35 mRNA, low surface co-stimulatory molecule expression, and CCR transcripts, consistent with immaturity. They elicited only weak allogeneic T-cell proliferative responses, and repeated stimulation induced CD4CD25 IL-10 T cells. In vivo, the freshly isolated DC failed to prime T cells of naive allogeneic hosts for anti-donor cytotoxic T-cell responses. When infused systemically, 1 week before organ transplantation, they prolonged graft survival without immunosuppressive therapy. CONCLUSIONS: Hematopoietin-mobilized renal DC are functionally immature and exhibit tolerogenic potential. Mobilization of DC within kidneys is likely to affect their antigen-handling capacity, immunogenicity, and tolerogenic ability.


Asunto(s)
Células Dendríticas/fisiología , Supervivencia de Injerto , Riñón/inmunología , Linfocitos T/inmunología , Animales , Movimiento Celular , Inmunofenotipificación , Interleucina-12/genética , Subunidad p35 de la Interleucina-12 , Subunidad p40 de la Interleucina-12 , Lipopolisacáridos/farmacología , Masculino , Proteínas de la Membrana/farmacología , Ratones , Ratones Endogámicos , Subunidades de Proteína/genética , Transcripción Genética , Trasplante Homólogo
17.
Blood ; 102(7): 2513-21, 2003 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-12829599

RESUMEN

We provide phenotypic and functional evidence of premonocytoid dendritic cells (DCs) and preplasmacytoid DCs in blood and of corresponding DC subsets in secondary lymphoid tissue of rhesus monkeys. Subsets were identified and sorted by 4-color flow cytometry using antihuman monoclonal antibodies cross-reactive with rhesus monkey. To mobilize pre-DC subsets, fms-like tyrosine 3 kinase ligand (Flt3L; 100 microg/kg subcutaneously) was administered for 10 days. Presumptive pre-DC subsets were identified within the lineage- (Lin-) major histocompatibility complex (MHC) class II+ fraction of blood mononuclear cells. Premonocytoid DCs were CD11c+CD123- (interleukin-3Ralpha- [IL-3Ralpha-]). Preplasmacytoid DCs were characterized as CD11c-CD123++ Flt3L increased the CD11c+ pre-DC (7-fold) and CD123++ pre-DC subsets (3-fold) in blood. The freshly isolated CD11c+ pre-DC subset induced modest proliferation of naive allogeneic T cells. After overnight culture with granulocyte macro-phage-colony-stimulating factor (GMCSF) and CD40L, both subsets up-regulated surface costimulatory molecules, and CD11c+ pre-DCs became potent allostimulators. Freshly isolated CD123++ pre-DCs showed typical plasmacytoid morphology and, when cultured with IL-3 and CD40L for 72 hours, developed mature DC morphology. Following stimulation with CD40L, CD11c+ pre-DCs secreted increased levels of IL-12p40. Importantly, herpes simplex virus-stimulated CD123++ pre-DCs, but not CD11c+ pre-DCs, secreted interferon-alpha (IFN-alpha). Corresponding DC subsets were identified by flow analysis and immunohistochemistry in lymph nodes wherein both populations were increased 2- to 3-fold by Flt3L administration. CD123+ pre-DCs produced IFN-alpha in response to in vivo viral infection. Thus, rhesus monkeys exhibit 2 distinct DC precursor populations that closely resemble those of humans. Both are mobilized into blood and lymphoid tissue by Flt3L, offering potential for their further characterization and possible therapeutic application.


Asunto(s)
Separación Celular/métodos , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Ganglios Linfáticos/citología , Proteínas de la Membrana/farmacología , Animales , Anticuerpos Monoclonales , Antígeno CD11c/metabolismo , Antígenos CD40/metabolismo , Color , Células Dendríticas/metabolismo , Citometría de Flujo/métodos , Herpes Simple/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Inmunofenotipificación , Virus de la Influenza A/inmunología , Interferón gamma/metabolismo , Interleucina-12/metabolismo , Subunidad alfa del Receptor de Interleucina-3 , Leucaféresis , Macaca mulatta , Infecciones por Orthomyxoviridae/inmunología , Receptores de Interleucina-3/metabolismo , Simplexvirus/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...