Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
EMBO Rep ; 23(8): e54558, 2022 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-35856334

RESUMEN

Diabetic foot ulcers (DFU) are a serious complication of diabetes mellitus and associated with reduced quality of life and high mortality rate. DFUs are characterized by a deregulated immune response with decreased neutrophils due to loss of the transcription factor, FOXM1. Diabetes primes neutrophils to form neutrophil extracellular traps (NETs), contributing to tissue damage and impaired healing. However, the role of FOXM1 in priming diabetic neutrophils to undergo NET formation remains unknown. Here, we found that FOXM1 regulates reactive oxygen species (ROS) levels in neutrophils and inhibition of FOXM1 results in increased ROS leading to NET formation. Next generation sequencing revealed that TREM1 promoted the recruitment of FOXM1+ neutrophils and reversed effects of diabetes and promoted wound healing in vivo. Moreover, we found that TREM1 expression correlated with clinical healing outcomes of DFUs, indicating TREM1 may serve as a useful biomarker or a potential therapeutic target. Our findings highlight the clinical relevance of TREM1, and indicates FOXM1 pathway as a novel regulator of NET formation during diabetic wound healing, revealing new therapeutic strategies to promote healing in DFUs.


Asunto(s)
Diabetes Mellitus , Pie Diabético , Trampas Extracelulares , Diabetes Mellitus/metabolismo , Pie Diabético/genética , Pie Diabético/metabolismo , Trampas Extracelulares/genética , Trampas Extracelulares/metabolismo , Proteína Forkhead Box M1/genética , Proteína Forkhead Box M1/metabolismo , Proteína Forkhead Box M1/farmacología , Humanos , Calidad de Vida , Especies Reactivas de Oxígeno/metabolismo , Receptor Activador Expresado en Células Mieloides 1/genética , Receptor Activador Expresado en Células Mieloides 1/metabolismo
2.
Exp Eye Res ; 227: 109353, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36539051

RESUMEN

In this paper, we use RNAseq to identify senescence and phagocytosis as key factors to understanding how mitomyin C (MMC) stimulates regenerative wound repair. We use conditioned media (CM) from untreated (CMC) and MMC treated (CMM) human and mouse corneal epithelial cells to show that corneal epithelial cells indirectly exposed to MMC secrete elevated levels of immunomodulatory proteins including IL-1α and TGFß1 compared to cells exposed to CMC. These factors increase epithelial and macrophage phagocytosis and promote ECM turnover. IL-1α supplementation can increase phagocytosis in control epithelial cells and attenuate TGFß1 induced αSMA expression by corneal fibroblasts. Yet, we show that epithelial cell CM contains factors besides IL-1α that regulate phagocytosis and αSMA expression by fibroblasts. Exposure to CMM also impacts the activation of bone marrow derived dendritic cells and their ability to present antigen. These in vitro studies show how a brief exposure to MMC induces corneal epithelial cells to release proteins and other factors that function in a paracrine way to enhance debris removal and enlist resident epithelial and immune cells as well as stromal fibroblasts to support regenerative and not fibrotic wound healing.


Asunto(s)
Mitomicina , Comunicación Paracrina , Humanos , Animales , Ratones , Mitomicina/farmacología , Células Cultivadas , Fibroblastos/metabolismo , Cicatrización de Heridas , Células Epiteliales/metabolismo
3.
Proc Natl Acad Sci U S A ; 117(10): 5409-5419, 2020 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-32094169

RESUMEN

Type III IFN lambdas (IFN-λ) have recently been described as important mediators of immune responses at barrier surfaces. However, their role in autoimmune diseases such as systemic lupus erythematosus (SLE), a condition characterized by aberrant type I IFN signaling, has not been determined. Here, we identify a nonredundant role for IFN-λ in immune dysregulation and tissue inflammation in a model of TLR7-induced lupus. IFN-λ protein is increased in murine lupus and IFN-λ receptor (Ifnlr1) deficiency significantly reduces immune cell activation and associated organ damage in the skin and kidneys without effects on autoantibody production. Single-cell RNA sequencing in mouse spleen and human peripheral blood revealed that only mouse neutrophils and human B cells are directly responsive to this cytokine. Rather, IFN-λ activates keratinocytes and mesangial cells to produce chemokines that induce immune cell recruitment and promote tissue inflammation. These data provide insights into the immunobiology of SLE and identify type III IFNs as important factors for tissue-specific pathology in this disease.


Asunto(s)
Interferones/fisiología , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Animales , Linfocitos B/inmunología , Línea Celular , Eliminación de Gen , Humanos , Imiquimod/farmacología , Inflamación/inmunología , Inflamación/patología , Inductores de Interferón/farmacología , Interferón Tipo I/fisiología , Interferones/farmacología , Queratinocitos/efectos de los fármacos , Queratinocitos/inmunología , Queratinocitos/patología , Células Mesangiales/efectos de los fármacos , Células Mesangiales/inmunología , Células Mesangiales/patología , Ratones Endogámicos C57BL , Ratones Mutantes , Receptores de Interferón/genética , Transducción de Señal , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 7/fisiología , Interferón lambda
5.
PLoS Genet ; 14(1): e1007168, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29357356

RESUMEN

Pachyonychia congenita (PC) is a cutaneous disorder primarily characterized by nail dystrophy and painful palmoplantar keratoderma. PC is caused by mutations in KRT6A, KRT6B, KRT6C, KRT16, and KRT17, a set of keratin genes expressed in the nail bed, palmoplantar epidermis, oral mucosal epithelium, hair follicle and sweat gland. RNA-seq analysis revealed that all PC-associated keratins (except for Krt6c that does exist in the mouse genome) are expressed in the mouse enamel organ. We further demonstrated that these keratins are produced by ameloblasts and are incorporated into mature human enamel. Using genetic and intraoral examination data from 573 adults and 449 children, we identified several missense polymorphisms in KRT6A, KRT6B and KRT6C that lead to a higher risk for dental caries. Structural analysis of teeth from a PC patient carrying a p.Asn171Lys substitution in keratin-6a (K6a) revealed disruption of enamel rod sheaths resulting in altered rod shape and distribution. Finally, this PC-associated substitution as well as more frequent caries-associated SNPs, found in two of the KRT6 genes, that result in p.Ser143Asn substitution (rs28538343 in KRT6B and rs151117600 in KRT6C), alter the assembly of K6 filaments in ameloblast-like cells. These results identify a new set of keratins involved in tooth enamel formation, distinguish novel susceptibility loci for tooth decay and reveal additional clinical features of pachyonychia congenita.


Asunto(s)
Queratinas/genética , Paquioniquia Congénita/genética , Polimorfismo de Nucleótido Simple , Erosión de los Dientes/genética , Adulto , Sustitución de Aminoácidos , Animales , Células Cultivadas , Niño , Caries Dental/genética , Esmalte Dental/crecimiento & desarrollo , Esmalte Dental/metabolismo , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Queratina-6/genética , Masculino , Ratones , Persona de Mediana Edad , Paquioniquia Congénita/complicaciones , Ratas
6.
J Biol Chem ; 294(48): 18475-18487, 2019 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-31628189

RESUMEN

A highly specialized cytoskeletal protein, keratin 75 (K75), expressed primarily in hair follicles, nail beds, and lingual papillae, was recently discovered in dental enamel, the most highly mineralized hard tissue in the human body. Among many questions this discovery poses, the fundamental question regarding the trafficking and secretion of this protein, which lacks a signal peptide, is of an utmost importance. Here, we present evidence that K75 is expressed during the secretory stage of enamel formation and is present in the forming enamel matrix. We further show that K75 is secreted together with major enamel matrix proteins amelogenin and ameloblastin, and it was detected in Golgi and the endoplasmic reticulum (ER)-Golgi intermediate compartment (ERGIC) but not in rough ER (rER). Inhibition of ER-Golgi transport by brefeldin A did not affect the association of K75 with Golgi, whereas ameloblastin accumulated in rER, and its transport from rER into Golgi was disrupted. Together, these results indicate that K75, a cytosolic protein lacking a signal sequence, is secreted into the forming enamel matrix utilizing portions of the conventional ER-Golgi secretory pathway. To the best of our knowledge, this is the first study providing insights into mechanisms of keratin secretion.


Asunto(s)
Ameloblastos/metabolismo , Esmalte Dental/metabolismo , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Queratina-6/metabolismo , Amelogenina/genética , Amelogenina/metabolismo , Animales , Antibacterianos , Brefeldino A/farmacología , Proteínas del Esmalte Dental/genética , Proteínas del Esmalte Dental/metabolismo , Expresión Génica , Humanos , Queratina-6/genética , Ratones Endogámicos C57BL , Transporte de Proteínas/efectos de los fármacos , Ratas Sprague-Dawley
7.
Int J Mol Sci ; 21(4)2020 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-32059344

RESUMEN

Cutaneous squamous cell carcinoma (cSCC) represents the second most frequent skin cancer,recently showing a rapid increase in incidence worldwide, with around >1 million cases/year in theUnited States and 2500 deaths [1] [...].


Asunto(s)
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/terapia , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/terapia , Animales , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Modelos Animales , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Estados Unidos
8.
Am J Med Genet A ; 179(3): 442-447, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30703280

RESUMEN

An international advisory group met at the National Institutes of Health in Bethesda, Maryland in 2017, to discuss a new classification system for the ectodermal dysplasias (EDs) that would integrate both clinical and molecular information. We propose the following, a working definition of the EDs building on previous classification systems and incorporating current approaches to diagnosis: EDs are genetic conditions affecting the development and/or homeostasis of two or more ectodermal derivatives, including hair, teeth, nails, and certain glands. Genetic variations in genes known to be associated with EDs that affect only one derivative of the ectoderm (attenuated phenotype) will be grouped as non-syndromic traits of the causative gene (e.g., non-syndromic hypodontia or missing teeth associated with pathogenic variants of EDA "ectodysplasin"). Information for categorization and cataloging includes the phenotypic features, Online Mendelian Inheritance in Man number, mode of inheritance, genetic alteration, major developmental pathways involved (e.g., EDA, WNT "wingless-type," TP63 "tumor protein p63") or the components of complex molecular structures (e.g., connexins, keratins, cadherins).


Asunto(s)
Displasia Ectodérmica/diagnóstico , Displasia Ectodérmica/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Genotipo , Fenotipo , Alelos , Biomarcadores , Bases de Datos Genéticas , Displasia Ectodérmica/metabolismo , Humanos , Transducción de Señal
9.
Int J Mol Sci ; 20(14)2019 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-31331058

RESUMEN

Well-regulated epidermal homeostasis depends on the function of different classes of factors, such as transcription regulators and receptors. Alterations in this homeostatic balance may lead to the development of cutaneous squamous tumorigenesis. The homeobox transcription factor DLX3 is determinant for a p53-dependent regulation of epidermal differentiation and modulates skin carcinogenesis. The maintenance of skin homeostasis also involves the action of neurotrophins (NTs) and their receptors, Trk and CD271. While Trk receptor overexpression is a hallmark of cancer, there are conflicting data on CD271 expression and function in cutaneous SCC (cSCC). Previous studies have reported NT receptors expression in head and neck SSC (HNSCC). We show that CD271 is expressed at low levels in primary cSCC cells and the number of CD271+ cells correlates with cell cohesion in SCC spheroids. In normal epidermis, CD271 is expressed in proliferative progenitor cells and DLX3 in terminally differentiated keratinocytes. Brain-derived neurotrophic factor (BDNF) and neurotrophin 3 (NT3) increase DLX3 expression. In the absence of a functional BDNF receptor TrkB in keratinocytes, we hypothesize that the BDNF-dependent DLX3 response could be mediated via CD271. Altogether, our results support a putative CD271-DLX3 connection in keratinocytes, which might be crucial to preventing squamous skin cancer.


Asunto(s)
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Proteínas de Homeodominio/genética , Queratinocitos/metabolismo , Proteínas del Tejido Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/genética , Factores de Transcripción/genética , Biomarcadores , Línea Celular Tumoral , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Humanos , Modelos Biológicos , Proteínas del Tejido Nervioso/metabolismo , Unión Proteica , Receptores de Factor de Crecimiento Nervioso/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo
10.
Exp Dermatol ; 27(3): 289-292, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29380438

RESUMEN

Dlx4 is a member of a family of homeobox genes with homology to Drosophila distal-less (dll) gene. We show that Dlx4 expression pattern partially overlaps with its cis-linked gene Dlx3 during mouse development as well as in neonatal and adult skin. In mice, Dlx4 is expressed in the branchial arches, embryonic limbs, digits, nose, hair follicle and in the basal and suprabasal layers of mouse interfollicular epidermis. We show that inactivation of Dlx4 in mice did not result in any overtly gross pathology. Skin development, homeostasis and response to TPA treatment were similar in mice with loss of Dlx4 compared to wild-type counterparts.


Asunto(s)
Epidermis/fisiología , Proteínas de Homeodominio/genética , Homeostasis/genética , Fenómenos Fisiológicos de la Piel/genética , Factores de Transcripción/genética , Animales , Diferenciación Celular , Epidermis/efectos de los fármacos , Epidermis/crecimiento & desarrollo , Queratinocitos/fisiología , Ratones , Ratones Noqueados , Acetato de Tetradecanoilforbol/farmacología , Transcriptoma/genética
11.
Connect Tissue Res ; 59(sup1): 30-34, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29745813

RESUMEN

DLX3 is essential for tooth enamel development and is so far the only transcription factor known to be mutated in a syndromic form of amelogenesis imperfecta. Through conditional deletion of Dlx3 in the dental epithelium in mouse, we have previously established the involvement of DLX3 in enamel pH regulation, as well as in controlling the expression of sets of keratins that contribute to enamel rod sheath formation. Here, we show that the decussation pattern of enamel rods was lost in conditional knockout animals, suggesting that DLX3 controls the coordinated migration of ameloblasts during enamel secretion. We further demonstrate that DLX3 regulates the expression of some components of myosin II complexes potentially involved in driving the movement of ameloblasts that leads to enamel rod decussation.


Asunto(s)
Ameloblastos/metabolismo , Esmalte Dental/metabolismo , Proteínas de Homeodominio/metabolismo , Queratinas/biosíntesis , Factores de Transcripción/metabolismo , Ameloblastos/citología , Animales , Movimiento Celular , Proteínas de Homeodominio/genética , Concentración de Iones de Hidrógeno , Queratinas/genética , Ratones , Ratones Transgénicos , Factores de Transcripción/genética
12.
Proc Natl Acad Sci U S A ; 111(33): E3422-31, 2014 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-25092341

RESUMEN

Psoriasis is a chronic inflammatory skin disease characterized by abnormal keratinocyte proliferation and differentiation and by an influx of inflammatory cells. The mechanisms underlying psoriasis in humans and in mouse models are poorly understood, although evidence strongly points to crucial contributions of IL-17 cytokines, which signal via the obligatory adaptor CIKS/Act1. Here we identify critical roles of CIKS/Act1-mediated signaling in imiquimod-induced psoriatic inflammation, a mouse model that shares features with the human disease. We found that IL-17 cytokines/CIKS-mediated signaling into keratinocytes is essential for neutrophilic microabscess formation and contributes to hyperproliferation and markedly attenuated differentiation of keratinocytes, at least in part via direct effects. In contrast, IL-17 cytokines/CIKS-mediated signaling into nonkeratinocytes, particularly into dermal fibroblasts, promotes cellular infiltration and, importantly, leads to enhanced the accumulation of IL-17-producing γδT cells in skin, comprising a positive feed-forward mechanism. Thus, CIKS-mediated signaling is central in the development of both dermal and epidermal hallmarks of psoriasis, inducing distinct pathologies via target cell-specific effects. CIKS-mediated signaling represents a potential therapeutic target in psoriasis.


Asunto(s)
Interleucina-17/inmunología , Psoriasis/inmunología , Aminoquinolinas/administración & dosificación , Animales , Diferenciación Celular/inmunología , Proliferación Celular , Modelos Animales de Enfermedad , Epidermis/efectos de los fármacos , Epidermis/patología , Imiquimod , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Psoriasis/inducido químicamente , Reacción en Cadena en Tiempo Real de la Polimerasa
13.
Semin Cell Dev Biol ; 25-26: 22-33, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24361867

RESUMEN

Mouse models have greatly helped in elucidating the molecular mechanisms involved in hair formation and regeneration. Recent publications have reviewed the genes involved in mouse hair development based on the phenotype of transgenic, knockout and mutant animal models. While much of this information has been instrumental in determining molecular aspects of human hair development and cycling, mice exhibit a specific pattern of hair morphogenesis and hair distribution throughout the body that cannot be directly correlated to human hair. In this mini-review, we discuss specific aspects of human hair follicle development and present an up-to-date summary of human genetic disorders associated with abnormalities in hair follicle morphogenesis, structure or regeneration.


Asunto(s)
Enfermedades del Cabello/genética , Enfermedades del Cabello/patología , Folículo Piloso/anomalías , Folículo Piloso/crecimiento & desarrollo , Cabello/anomalías , Cabello/crecimiento & desarrollo , Alopecia/genética , Alopecia/patología , Animales , Modelos Animales de Enfermedad , Cabello/patología , Folículo Piloso/patología , Humanos , Ratones , Morfogénesis
14.
Exp Dermatol ; 24(1): 55-7, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25316000

RESUMEN

Calcium-mediated signals play important roles in epidermal barrier formation, skin homoeostasis and wound repair. Calmodulin 4 (Calm4) is a small, Ca2+ -binding protein with strong expression in suprabasal keratinocytes. In mice, Calm4 first appears in the skin at the time of barrier formation, and its expression increases in response to epidermal barrier challenges. In this study, we report the generation of Calm4 knockout mice and provide evidence that Calm4 is dispensable for epidermal barrier formation, maintenance and repair.


Asunto(s)
Calmodulina/fisiología , Calpaína/fisiología , Epidermis/metabolismo , Animales , Proteínas de Unión al Calcio/química , Calmodulina/genética , Calpaína/genética , Movimiento Celular , Queratinocitos/citología , Operón Lac , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Transducción de Señal , Piel/metabolismo , Cicatrización de Heridas
15.
J Cell Sci ; 125(Pt 7): 1827-36, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22366455

RESUMEN

The process by which the periderm transitions to stratified epidermis with the establishment of the skin barrier is unknown. Understanding the cellular and molecular processes involved is crucial for the treatment of human pathologies, where abnormal skin development and barrier dysfunction are associated with hypothermia and perinatal dehydration. For the first time, we demonstrate that retinoic acid (RA) levels are important for periderm desquamation, embryonic skin differentiation and barrier formation. Although excess exogenous RA has been known to have teratogenic effects, little is known about the consequences of elevated endogenous retinoids in skin during embryogenesis. Absence of cytochrome P450, family 26, subfamily b, polypeptide 1 (Cyp26b1), a retinoic-acid-degrading enzyme, results in aberrant epidermal differentiation and filaggrin expression, defective cornified envelopes and skin barrier formation, in conjunction with peridermal retention. We show that these alterations are RA dependent because administration of exogenous RA in vivo and to organotypic skin cultures phenocopy Cyp26b1(-/-) skin abnormalities. Furthermore, utilizing the Flaky tail (Ft/Ft) mice, a mouse model for human ichthyosis, characterized by mutations in the filaggrin gene, we establish that proper differentiation and barrier formation is a prerequisite for periderm sloughing. These results are important in understanding pathologies associated with abnormal embryonic skin development and barrier dysfunction.


Asunto(s)
Sistema Enzimático del Citocromo P-450/deficiencia , Piel/embriología , Piel/metabolismo , Tretinoina/metabolismo , Animales , Sistema Enzimático del Citocromo P-450/metabolismo , Modelos Animales de Enfermedad , Proteínas Filagrina , Proteínas de Filamentos Intermediarios/genética , Proteínas de Filamentos Intermediarios/metabolismo , Ratones , Ratones Noqueados , Ácido Retinoico 4-Hidroxilasa , Piel/efectos de los fármacos , Piel/patología , Tretinoina/farmacología
16.
Proc Natl Acad Sci U S A ; 108(28): 11566-71, 2011 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-21709238

RESUMEN

In an effort to understand the role of Distal-less 3 (Dlx3) in cutaneous biology and pathophysiology, we generated and characterized a mouse model with epidermal ablation of Dlx3. K14cre;Dlx3(Kin/f) mice exhibited epidermal hyperproliferation and abnormal differentiation of keratinocytes. Results from subsequent analyses revealed cutaneous inflammation that featured accumulation of IL-17-producing CD4(+) T, CD8(+) T, and γδ T cells in the skin and lymph nodes of K14cre;Dlx3(Kin/f) mice. The gene expression signature of K14cre;Dlx3(Kin/f) skin shared features with lesional psoriatic skin, and Dlx3 expression was markedly and selectively decreased in psoriatic skin. Interestingly, cultured Dlx3 null keratinocytes triggered cytokine production that is potentially linked to inflammatory responses in K14cre;Dlx3(Kin/f) mice. Thus, Dlx3 ablation in epidermis is linked to altered epidermal differentiation, barrier development, and IL-17-associated skin inflammation. This model provides a platform that will allow the systematic exploration of the contributions of keratinocytes to cutaneous inflammation.


Asunto(s)
Dermatitis/etiología , Proteínas de Homeodominio/inmunología , Interleucina-17/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/inmunología , Animales , Diferenciación Celular , Citocinas/biosíntesis , Dermatitis/genética , Dermatitis/inmunología , Dermatitis/patología , Modelos Animales de Enfermedad , Epidermis/inmunología , Epidermis/patología , Femenino , Proteínas de Homeodominio/genética , Humanos , Hiperplasia , Mediadores de Inflamación/metabolismo , Queratinocitos/inmunología , Queratinocitos/patología , Leucocitos/inmunología , Leucocitos/patología , Ratones , Ratones Noqueados , Embarazo , Células Th17/inmunología , Células Th17/patología , Factores de Transcripción/genética
17.
J Invest Dermatol ; 144(1): 142-151.e5, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37516309

RESUMEN

Ischemia-reperfusion (I/R) injury is a key player in the pathogeneses of pressure ulcer formation. Our previous work demonstrated that inducing the transcription factor SOX2 promotes cutaneous wound healing through EGFR signaling pathway enhancement. However, its protective effect on cutaneous I/R injury was not well-characterized. We aimed to assess the role of SOX2 in cutaneous I/R injury and the tissue-protective effect of SOX2 induction in keratinocytes (KCs) in cutaneous I/R injury. SOX2 was transiently expressed in KCs after cutaneous I/R injury. Ulcer formation was significantly suppressed in KC-specific SOX2-overexpressing mice. SOX2 in skin KCs significantly suppressed the infiltrating inflammatory cells, apoptotic cells, vascular damage, and hypoxic areas in cutaneous I/R injury. Oxidative stress-induced mRNA levels of inflammatory cytokine expression were suppressed, and antioxidant stress factors and amphiregulin were elevated by SOX2 induction in skin KCs. Recombinant amphiregulin administration suppressed pressure ulcer development after cutaneous I/R injury in mice and suppressed oxidative stress-induced ROS production and apoptosis in vitro. These findings support that SOX2 in KCs might regulate cutaneous I/R injury through amphiregulin production, resulting in oxidative stress suppression. Recombinant amphiregulin can be a potential therapeutic agent for cutaneous I/R injury.


Asunto(s)
Úlcera por Presión , Daño por Reperfusión , Animales , Ratones , Anfirregulina/genética , Anfirregulina/metabolismo , Apoptosis , Queratinocitos/metabolismo , Daño por Reperfusión/complicaciones , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo , Piel/metabolismo
18.
J Biol Chem ; 287(47): 39304-15, 2012 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-23007396

RESUMEN

Retinoic acid (RA) is essential during embryogenesis and for tissue homeostasis, whereas excess RA is well known as a teratogen. In humans, excess RA is associated with hair loss. In the present study, we demonstrate that specific levels of RA, regulated by Cyp26b1, one of the RA-degrading enzymes, are required for hair follicle (hf) morphogenesis. Mice with embryonic ablation of Cyp26b1 (Cyp26b1(-/-)) have excessive endogenous RA, resulting in arrest of hf growth at the hair germ stage. The altered hf development is rescued by grafting the mutant skin on immunodeficient mice. Our results show that normalization of RA levels is associated with reinitiation of hf development. Conditional deficiency of Cyp26b1 in the dermis (En1Cre;Cyp26b1f/-) results in decreased hair follicle density and specific effect on hair type, indicating that RA levels also influence regulators of hair bending. Our results support the model of RA-dependent dermal signals regulating hf downgrowth and bending. To elucidate target gene pathways of RA, we performed microarray and RNA-Seq profiling of genes differentially expressed in Cyp26b1(-/-) skin and En1Cre;Cyp26b1f/- tissues. We show specific effects on the Wnt-catenin pathway and on members of the Runx, Fox, and Sox transcription factor families, indicating that RA modulates pathways and factors implicated in hf downgrowth and bending. Our results establish that proper RA distribution is essential for morphogenesis, development, and differentiation of hfs.


Asunto(s)
Sistema Enzimático del Citocromo P-450/biosíntesis , Dermis/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Folículo Piloso/enzimología , Queratolíticos/farmacología , Tretinoina/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Animales , Sistema Enzimático del Citocromo P-450/genética , Dermis/citología , Folículo Piloso/citología , Humanos , Ratones , Ratones Noqueados , Ratones Desnudos , Ácido Retinoico 4-Hidroxilasa , Trasplante de Piel , Trasplante Homólogo
19.
J Biol Chem ; 287(15): 12230-40, 2012 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-22351765

RESUMEN

During development, Dlx3 is expressed in ectodermal appendages such as hair and teeth. Thus far, the evidence that Dlx3 plays a crucial role in tooth development comes from reports showing that autosomal dominant mutations in DLX3 result in severe enamel and dentin defects leading to abscesses and infections. However, the normal function of DLX3 in odontogenesis remains unknown. Here, we use a mouse model to demonstrate that the absence of Dlx3 in the neural crest results in major impairment of odontoblast differentiation and dentin production. Mutant mice develop brittle teeth with hypoplastic dentin and molars with an enlarged pulp chamber and underdeveloped roots. Using this mouse model, we found that dentin sialophosphoprotein (Dspp), a major component of the dentin matrix, is strongly down-regulated in odontoblasts lacking Dlx3. Using ChIP-seq, we further demonstrate the direct binding of Dlx3 to the Dspp promoter in vivo. Luciferase reporter assays determined that Dlx3 positively regulates Dspp expression. This establishes a regulatory pathway where the transcription factor Dlx3 is essential in dentin formation by directly regulating a crucial matrix protein.


Asunto(s)
Dentina/patología , Proteínas de la Matriz Extracelular/genética , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Cresta Neural/metabolismo , Fosfoproteínas/genética , Sialoglicoproteínas/genética , Factores de Transcripción/genética , Ameloblastos/metabolismo , Ameloblastos/fisiología , Animales , Secuencia de Bases , Diferenciación Celular , Línea Celular , Esmalte Dental/crecimiento & desarrollo , Esmalte Dental/metabolismo , Dentina/crecimiento & desarrollo , Dentina/metabolismo , Displasia de la Dentina/genética , Displasia de la Dentina/patología , Regulación hacia Abajo , Proteínas de la Matriz Extracelular/metabolismo , Genes Reporteros , Proteínas de Homeodominio/metabolismo , Luciferasas de Renilla/biosíntesis , Luciferasas de Renilla/genética , Mandíbula/metabolismo , Mesodermo/metabolismo , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Odontoblastos/metabolismo , Odontoblastos/fisiología , Fosfoproteínas/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Sialoglicoproteínas/metabolismo , Diente/crecimiento & desarrollo , Diente/metabolismo , Diente/patología , Factores de Transcripción/metabolismo
20.
J Cell Physiol ; 228(3): 654-64, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22886599

RESUMEN

Mutations in DLX3 in humans lead to defects in craniofacial and appendicular bones, yet the in vivo activities related to Dlx3 function during normal skeletal development have not been fully elucidated. Here we used a conditional knockout approach to analyze the effects of neural crest deletion of Dlx3 on craniofacial bones development. At birth, mutant mice exhibit a normal overall positioning of the skull bones, but a change in the shape of the calvaria was observed. Molecular analysis of the genes affected in the frontal bones and mandibles from these mice identified several bone markers known to affect bone development, with a strong prediction for increased bone formation and mineralization in vivo. Interestingly, while a subset of these genes were similarly affected in frontal bones and mandibles (Sost, Mepe, Bglap, Alp, Ibsp, Agt), several genes, including Lect1 and Calca, were specifically affected in frontal bones. Consistent with these molecular alterations, cells isolated from the frontal bone of mutant mice exhibited increased differentiation and mineralization capacities ex vivo, supporting cell autonomous defects in neural crest cells. However, adult mutant animals exhibited decreased bone mineral density in both mandibles and calvaria, as well as a significant increase in bone porosity. Together, these observations suggest that mature osteoblasts in the adult respond to signals that regulate adult bone mass and remodeling. This study provides new downstream targets for Dlx3 in craniofacial bone, and gives additional evidence of the complex regulation of bone formation and homeostasis in the adult skeleton.


Asunto(s)
Huesos Faciales/anomalías , Proteínas de Homeodominio/genética , Cresta Neural/anomalías , Cráneo/anomalías , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Animales , Secuencia de Bases , Densidad Ósea/genética , Densidad Ósea/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/fisiología , Humanos , Masculino , Mandíbula/anomalías , Ratones , Ratones Noqueados , Osteogénesis/genética , Osteogénesis/fisiología , Embarazo , Factores de Transcripción/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA