Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
J Am Acad Dermatol ; 83(2): 447-454, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31786163

RESUMEN

BACKGROUND: Recessive dystrophic epidermolysis bullosa (RDEB) is a hereditary blistering disorder due to a lack of type VII collagen. At present, treatment is mainly supportive. OBJECTIVES: To determine whether intravenous allogeneic bone marrow-derived mesenchymal stromal/stem cells (BM-MSCs) are safe in RDEB adults and if the cells improve wound healing and quality of life. METHODS: We conducted a prospective, phase I/II, open-label study recruiting 10 RDEB adults to receive 2 intravenous infusions of BM-MSCs (on day 0 and day 14; each dose 2-4 × 106 cells/kg). RESULTS: BM-MSCs were well tolerated with no serious adverse events to 12 months. Regarding efficacy, there was a transient reduction in disease activity scores (8/10 subjects) and a significant reduction in itch. One individual showed a transient increase in type VII collagen. LIMITATIONS: Open-label trial with no placebo. CONCLUSIONS: MSC infusion is safe in RDEB adults and can have clinical benefits for at least 2 months.


Asunto(s)
Epidermólisis Ampollosa Distrófica/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Prurito/terapia , Adolescente , Adulto , Anciano , Epidermólisis Ampollosa Distrófica/complicaciones , Epidermólisis Ampollosa Distrófica/diagnóstico , Femenino , Humanos , Infusiones Intravenosas , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Prurito/diagnóstico , Prurito/etiología , Calidad de Vida , Índice de Severidad de la Enfermedad , Trasplante Homólogo/métodos , Resultado del Tratamiento , Cicatrización de Heridas , Adulto Joven
2.
JCI Insight ; 4(11)2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-31167965

RESUMEN

BACKGROUNDRecessive dystrophic epidermolysis bullosa (RDEB) is a severe form of skin fragility disorder due to mutations in COL7A1 encoding basement membrane type VII collagen (C7), the main constituent of anchoring fibrils (AFs) in skin. We developed a self-inactivating lentiviral platform encoding a codon-optimized COL7A1 cDNA under the control of a human phosphoglycerate kinase promoter for phase I evaluation.METHODSIn this single-center, open-label phase I trial, 4 adults with RDEB each received 3 intradermal injections (~1 × 106 cells/cm2 of intact skin) of COL7A1-modified autologous fibroblasts and were followed up for 12 months. The primary outcome was safety, including autoimmune reactions against recombinant C7. Secondary outcomes included C7 expression, AF morphology, and presence of transgene in the injected skin.RESULTSGene-modified fibroblasts were well tolerated, without serious adverse reactions or autoimmune reactions against recombinant C7. Regarding efficacy, there was a significant (P < 0.05) 1.26-fold to 26.10-fold increase in C7 mean fluorescence intensity in the injected skin compared with noninjected skin in 3 of 4 subjects, with a sustained increase up to 12 months in 2 of 4 subjects. The presence of transgene (codon-optimized COL7A1 cDNA) was demonstrated in the injected skin at month 12 in 1 subject, but no new mature AFs were detected.CONCLUSIONTo our knowledge, this is the first human study demonstrating safety and potential efficacy of lentiviral fibroblast gene therapy with the presence of COL7A1 transgene and subsequent C7 restoration in vivo in treated skin at 1 year after gene therapy. These data provide a rationale for phase II studies for further clinical evaluation.TRIAL REGISTRATIONClincalTrials.gov NCT02493816.FUNDINGCure EB, Dystrophic Epidermolysis Bullosa Research Association (UK), UK NIHR Biomedical Research Centre at Guy's and St Thomas' NHS Foundation Trust and King's College London, and Fondation René Touraine Short-Exchange Award.


Asunto(s)
Epidermólisis Ampollosa Distrófica/terapia , Fibroblastos , Terapia Genética , Lentivirus/genética , Adulto , Colágeno Tipo VII/genética , Femenino , Fibroblastos/metabolismo , Fibroblastos/trasplante , Terapia Genética/efectos adversos , Terapia Genética/métodos , Humanos , Masculino , Persona de Mediana Edad , Resultado del Tratamiento
3.
Am J Hum Genet ; 100(2): 364-370, 2017 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-28157540

RESUMEN

SPG23 is an autosomal-recessive neurodegenerative subtype of lower limb spastic paraparesis with additional diffuse skin and hair dyspigmentation at birth followed by further patchy pigment loss during childhood. Previously, genome-wide linkage in an Arab-Israeli pedigree mapped the gene to an approximately 25 cM locus on chromosome 1q24-q32. By using whole-exome sequencing in a further Palestinian-Jordanian SPG23 pedigree, we identified a complex homozygous 4-kb deletion/20-bp insertion in DSTYK (dual serine-threonine and tyrosine protein kinase) in all four affected family members. DSTYK is located within the established linkage region and we also found the same mutation in the previously reported pedigree and another Israeli pedigree (total of ten affected individuals from three different families). The mutation removes the last two exons and part of the 3' UTR of DSTYK. Skin biopsies revealed reduced DSTYK protein levels along with focal loss of melanocytes. Ultrastructurally, swollen mitochondria and cytoplasmic vacuoles were also noted in remaining melanocytes and some keratinocytes and fibroblasts. Cultured keratinocytes and fibroblasts from an affected individual, as well as knockdown of Dstyk in mouse melanocytes, keratinocytes, and fibroblasts, were associated with increased cell death after ultraviolet irradiation. Keratinocytes from an affected individual showed loss of kinase activity upon stimulation with fibroblast growth factor. Previously, dominant mutations in DSTYK were implicated in congenital urological developmental disorders, but our study identifies different phenotypic consequences for a recurrent autosomal-recessive deletion mutation in revealing the genetic basis of SPG23.


Asunto(s)
Trastornos de la Pigmentación/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Eliminación de Secuencia , Paraplejía Espástica Hereditaria/genética , Vitíligo/genética , Secuencia de Aminoácidos , Animales , Apoptosis/genética , Pueblo Asiatico/genética , Cromosomas Humanos Par 1/genética , Exones , Facies , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Ligamiento Genético , Sitios Genéticos , Estudio de Asociación del Genoma Completo , Homocigoto , Humanos , Queratinocitos/citología , Queratinocitos/metabolismo , Masculino , Melanocitos/citología , Melanocitos/metabolismo , Ratones , Células 3T3 NIH , Linaje , Trastornos de la Pigmentación/diagnóstico , Paraplejía Espástica Hereditaria/diagnóstico , Vitíligo/diagnóstico , Adulto Joven
7.
J Invest Dermatol ; 136(1): 284-92, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26763448

RESUMEN

Cells therapies, engineered to secrete replacement proteins, are being developed to ameliorate otherwise debilitating diseases. Recessive dystrophic epidermolysis bullosa (RDEB) is caused by defects of type VII collagen, a protein essential for anchoring fibril formation at the dermal-epidermal junction. Whereas allogeneic fibroblasts injected directly into the dermis can mediate transient disease modulation, autologous gene-modified fibroblasts should evade immunological rejection and support sustained delivery of type VII collagen at the dermal-epidermal junction. We demonstrate the feasibility of such an approach using a therapeutic grade, self-inactivating-lentiviral vector, encoding codon-optimized COL7A1, to transduce RDEB fibroblasts under conditions suitable for clinical application. Expression and secretion of type VII collagen was confirmed with transduced cells exhibiting supranormal levels of protein expression, and ex vivo migration of fibroblasts was restored in functional assays. Gene-modified RDEB fibroblasts also deposited type VII collagen at the dermal-epidermal junction of human RDEB skin xenografts placed on NOD-scid IL2Rgamma(null) recipients, with reconstruction of human epidermal structure and regeneration of anchoring fibrils at the dermal-epidermal junction. Fibroblast-mediated restoration of protein and structural defects in this RDEB model strongly supports proposed therapeutic applications in man.


Asunto(s)
Colágeno Tipo VII/genética , Epidermólisis Ampollosa Distrófica/genética , Epidermólisis Ampollosa Distrófica/terapia , Fibroblastos/trasplante , Animales , Codón , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Vectores Genéticos , Xenoinjertos , Humanos , Lentivirus/genética , Masculino , Ratones , Ratones SCID , Distribución Aleatoria , Trasplante de Piel/métodos , Ingeniería de Tejidos , Cicatrización de Heridas/fisiología
8.
J Cell Sci ; 128(24): 4475-86, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26527396

RESUMEN

The transmission of mechanical forces to the nucleus is important for intracellular positioning, mitosis and cell motility, yet the contribution of specific components of the cytoskeleton to nuclear mechanotransduction remains unclear. In this study, we examine how crosstalk between the cytolinker plectin and F-actin controls keratin network organisation and the 3D nuclear morphology of keratinocytes. Using micro-patterned surfaces to precisely manipulate cell shape, we find that cell adhesion and spreading regulate the size and shape of the nucleus. Disruption of the keratin cytoskeleton through loss of plectin facilitated greater nuclear deformation, which depended on acto-myosin contractility. Nuclear morphology did not depend on direct linkage of the keratin cytoskeleton with the nuclear membrane, rather loss of plectin reduced keratin filament density around the nucleus. We further demonstrate that keratinocytes have abnormal nuclear morphologies in the epidermis of plectin-deficient, epidermolysis bullosa simplex patients. Taken together, our data demonstrate that plectin is an essential regulator of nuclear morphology in vitro and in vivo and protects the nucleus from mechanical deformation.


Asunto(s)
Núcleo Celular/metabolismo , Mecanotransducción Celular/fisiología , Plectina/metabolismo , Células 3T3 , Animales , Núcleo Celular/genética , Humanos , Masculino , Ratones , Ratones Noqueados , Plectina/genética
9.
PLoS One ; 10(9): e0137639, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26380979

RESUMEN

Recessive dystrophic epidermolysis bullosa (RDEB) is caused by mutations in COL7A1 resulting in reduced or absent type VII collagen, aberrant anchoring fibril formation and subsequent dermal-epidermal fragility. Here, we identify a significant decrease in PLOD3 expression and its encoded protein, the collagen modifying enzyme lysyl hydroxylase 3 (LH3), in RDEB. We show abundant LH3 localising to the basement membrane in normal skin which is severely depleted in RDEB patient skin. We demonstrate expression is in-part regulated by endogenous type VII collagen and that, in agreement with previous studies, even small reductions in LH3 expression lead to significantly less secreted LH3 protein. Exogenous type VII collagen did not alter LH3 expression in cultured RDEB keratinocytes and we show that RDEB patients receiving bone marrow transplantation who demonstrate significant increase in type VII collagen do not show increased levels of LH3 at the basement membrane. Our data report a direct link between LH3 and endogenous type VII collagen expression concluding that reduction of LH3 at the basement membrane in patients with RDEB will likely have significant implications for disease progression and therapeutic intervention.


Asunto(s)
Membrana Basal/enzimología , Membrana Basal/patología , Epidermólisis Ampollosa Distrófica/enzimología , Epidermólisis Ampollosa Distrófica/patología , Procolágeno-Lisina 2-Oxoglutarato 5-Dioxigenasa/análisis , Membrana Basal/metabolismo , Trasplante de Médula Ósea , Células Cultivadas , Colágeno Tipo VII/análisis , Colágeno Tipo VII/metabolismo , Epidermólisis Ampollosa Distrófica/metabolismo , Epidermólisis Ampollosa Distrófica/terapia , Humanos , Queratinocitos/enzimología , Queratinocitos/metabolismo , Queratinocitos/patología , Procolágeno-Lisina 2-Oxoglutarato 5-Dioxigenasa/metabolismo , Mapas de Interacción de Proteínas , Piel/enzimología , Piel/metabolismo , Piel/patología
10.
Am J Hum Genet ; 95(3): 308-14, 2014 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-25152456

RESUMEN

Grainyhead-like 2, encoded by GRHL2, is a member of a highly conserved family of transcription factors that play essential roles during epithelial development. Haploinsufficiency for GRHL2 has been implicated in autosomal-dominant deafness, but mutations have not yet been associated with any skin pathology. We investigated two unrelated Kuwaiti families in which a total of six individuals have had lifelong ectodermal defects. The clinical features comprised nail dystrophy or nail loss, marginal palmoplantar keratoderma, hypodontia, enamel hypoplasia, oral hyperpigmentation, and dysphagia. In addition, three individuals had sensorineural deafness, and three had bronchial asthma. Taken together, the features were consistent with an unusual autosomal-recessive ectodermal dysplasia syndrome. Because of consanguinity in both families, we used whole-exome sequencing to search for novel homozygous DNA variants and found GRHL2 mutations common to both families: affected subjects in one family were homozygous for c.1192T>C (p.Tyr398His) in exon 9, and subjects in the other family were homozygous for c.1445T>A (p.Ile482Lys) in exon 11. Immortalized keratinocytes (p.Ile482Lys) showed altered cell morphology, impaired tight junctions, adhesion defects, and cytoplasmic translocation of GRHL2. Whole-skin transcriptomic analysis (p.Ile482Lys) disclosed changes in genes implicated in networks of cell-cell and cell-matrix adhesion. Our clinical findings of an autosomal-recessive ectodermal dysplasia syndrome provide insight into the role of GRHL2 in skin development, homeostasis, and human disease.


Asunto(s)
Fisura del Paladar/genética , Proteínas de Unión al ADN/genética , Displasia Ectodérmica/genética , Genes Recesivos/genética , Discapacidad Intelectual/genética , Mutación/genética , Piel/patología , Sindactilia/genética , Factores de Transcripción/genética , Western Blotting , Niño , Proteínas de Unión al ADN/metabolismo , Exones/genética , Femenino , Humanos , Masculino , Linaje , Fenotipo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Piel/metabolismo , Síndrome , Factores de Transcripción/metabolismo
11.
J Invest Dermatol ; 134(10): 2570-2578, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24691054

RESUMEN

Epidermal growth factor receptor (EGFR) signaling is fundamentally important for tissue homeostasis through EGFR/ligand interactions that stimulate numerous signal transduction pathways. Aberrant EGFR signaling has been reported in inflammatory and malignant diseases, but thus far no primary inherited defects in EGFR have been recorded. Using whole-exome sequencing, we identified a homozygous loss-of-function missense mutation in EGFR (c.1283 G>A; p.Gly428Asp) in a male infant with lifelong inflammation affecting the skin, bowel, and lungs. During the first year of life, his skin showed erosions, dry scale, and alopecia. Subsequently, there were numerous papules and pustules--similar to the rash seen in patients receiving EGFR inhibitor drugs. Skin biopsy demonstrated an altered cellular distribution of EGFR in the epidermis with reduced cell membrane labeling, and in vitro analysis of the mutant receptor revealed abrogated EGFR phosphorylation and EGF-stimulated downstream signaling. Microarray analysis on the patient's skin highlighted disturbed differentiation/premature terminal differentiation of keratinocytes and upregulation of several inflammatory/innate immune response networks. The boy died at the age of 2.5 years from extensive skin and chest infections as well as electrolyte imbalance. This case highlights the major mechanism of epithelial dysfunction following EGFR signaling ablation and illustrates the broader impact of EGFR inhibition on other tissues.


Asunto(s)
Dermatitis/genética , Receptores ErbB/genética , Homocigoto , Inflamación/genética , Mutación Missense/genética , Piel/patología , Biopsia , Diferenciación Celular/fisiología , Preescolar , Dermatitis/patología , Dermatitis/fisiopatología , Epitelio/metabolismo , Epitelio/patología , Receptores ErbB/metabolismo , Resultado Fatal , Humanos , Técnicas In Vitro , Inflamación/patología , Inflamación/fisiopatología , Queratinocitos/metabolismo , Queratinocitos/patología , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Proteínas Proto-Oncogénicas c-akt/fisiología , Transducción de Señal/fisiología , Piel/metabolismo
12.
J Am Acad Dermatol ; 68(3): 482-8, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23219556

RESUMEN

BACKGROUND: Diffuse melanosis cutis (DMC) is a rare presentation of metastatic melanoma characterized by a progressive blue-gray discoloration of the skin and mucous membranes. OBJECTIVE: To foster a better understanding of the clinical presentation, histological findings, and pathophysiology underlying DMC. METHODS: A systematic review of the literature was completed utilizing MEDLINE, CINAHL, Embase, and Google. Data were extracted using a protocol-driven spread sheet with all statistical analyses completed using SPSS. RESULTS: The review identified 68 original cases of DMC. The mean time from diagnosis of melanoma until development of DMC was 11.48 months (95% confidence interval [CI]: 0-48.16). The mean time to death following the onset of DMC was 4.43 months (95% CI: 0.00-11.11). Histological findings were relatively consistent demonstrating intracellular and extracellular melanin deposition in the dermis, with a pronounced perivascular distribution. The pathophysiological mechanisms underlying DMC could not be definitively elucidated; however, it is hypothesized that the melanin precursors, melanin, and melanosomes liberated by cytolytic metastatic melanoma deposits are phagocytosed by dermal histiocytes, manifesting clinically as diffuse melanosis. LIMITATIONS: The cross-sectional nature of case reports, paucity of cases of DMC, and heterogeneity in reporting limit any conclusions being drawn regarding the pathophysiology of DMC definitively. CONCLUSION: DMC heralds a poor prognosis for patients with metastatic melanoma and affected patients should be made aware of the implications of this condition on survival.


Asunto(s)
Melanoma/secundario , Melanosis/patología , Neoplasias Cutáneas/patología , Pigmentación de la Piel , Piel/patología , Dermis/patología , Humanos , Melaninas/metabolismo , Melanoma/patología , Melanosomas/patología , Pronóstico
13.
Am J Hum Genet ; 91(6): 1115-21, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-23176819

RESUMEN

The Rab GTPase Rab27B and one of its effector proteins, Slac2-b (also known as EXPH5, exophilin-5), have putative roles in intracellular vesicle trafficking but their relevance to human disease is not known. By using whole-exome sequencing, we identified a homozygous frameshift mutation in EXPH5 in three siblings with inherited skin fragility born to consanguineous Iraqi parents. All three individuals harbor the mutation c.5786delC (p.Pro1929Leufs(∗)8) in EXPH5, which truncates the 1,989 amino acid Slac2-b protein by 52 residues. The clinical features comprised generalized scale-crusts and occasional blisters, mostly induced by trauma, as well as mild diffuse pigmentary mottling on the trunk and proximal limbs. There was no increased bleeding tendency, no neurologic abnormalities, and no increased incidence of infection. Analysis of an affected person's skin showed loss of Slac2-b immunostaining (C-terminal antibody), disruption of keratinocyte adhesion within the lower epidermis, and an increased number of perinuclear vesicles. A role for Slac2-b in keratinocyte biology was supported by findings of cytoskeletal disruption (mainly keratin intermediate filaments) and decreased keratinocyte adhesion in both keratinocytes from an affected subject and after shRNA knockdown of Slac2-b in normal keratinocytes. Slac2-b was also shown to colocalize with Rab27B and ß4 integrin to early adhesion initiation sites in spreading normal keratinocytes. Collectively, our findings identify an unexpected role for Slac2-b in inherited skin fragility and expand the clinical spectrum of human disorders of GTPase effector proteins.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Mutación de Línea Germinal , Enfermedades del Cabello/congénito , Enfermedades del Cabello/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Bases , Femenino , Enfermedades del Cabello/diagnóstico , Humanos , Queratinocitos/metabolismo , Queratinocitos/patología , Masculino , Linaje , Piel/patología , Piel/ultraestructura , Proteínas de Unión al GTP rab/metabolismo
14.
Clin Dermatol ; 30(1): 103-7, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22137233

RESUMEN

Genetic and acquired bullous dermatoses can severely affect multiple domains of a patient's quality of life (QOL). Integrating formal evaluation of QOL into the clinical evaluation of patients facilitates an objective assessment of disease severity, mapping of disease trajectory, and captures therapeutic intervention outcomes. There have been 5 studies evaluating QOL in autoimmune dermatoses and 4 studies reviewing QOL in the genodermatoses. All literature to date indicates a significant disease burden in this setting. The development of formal QOL instruments has facilitated quantification of QOL deficits in this arena and offers promising tools for patient assessment in the future.


Asunto(s)
Ansiedad/epidemiología , Depresión/epidemiología , Calidad de Vida/psicología , Enfermedades Cutáneas Vesiculoampollosas/epidemiología , Enfermedades Cutáneas Vesiculoampollosas/psicología , Estrés Psicológico/epidemiología , Adaptación Psicológica , Actitud Frente a la Salud , Causalidad , Comorbilidad , Humanos , Índice de Severidad de la Enfermedad , Percepción Social
15.
Clin Dermatol ; 29(4): 412-9, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21679868

RESUMEN

Epidermolysis bullosa (EB) is classified into the three major subtypes depending on the level of skin cleavage within the epidermal keratinocyte or basement membrane zone. Tissue separation occurs within the intraepidermal cytoplasm of the basal keratinocyte, through the lamina lucida, or in sublamina densa regions of the basal lamina (basement membrane) in EB simplex, junctional EB, and dystrophic EB, respectively. Transmission electron microscopy (TEM) is an effective method for determining the level of tissue separation and hemidesmosome (HD) and anchoring fibril morphology if performed by experienced operators, and has proven to be a powerful technique for the diagnosis of new EB patients. Recent advances in genetic and immunofluorescence studies have enabled us to diagnose EB more easily and with greater accuracy. This contribution reviews TEM findings in the EB subtypes and discusses the importance of observations in the molecular morphology of HD and basement membrane associated structures.


Asunto(s)
Membrana Basal/ultraestructura , Epidermólisis Ampollosa/patología , Hemidesmosomas/ultraestructura , Epidermis/ultraestructura , Epidermólisis Ampollosa/clasificación , Epidermólisis Ampollosa/metabolismo , Técnica del Anticuerpo Fluorescente/métodos , Humanos , Microscopía Electrónica de Transmisión
16.
Burns ; 37(6): 1001-9, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21501931

RESUMEN

High-resolution, high-contrast, three-dimensional images of live cell and tissue architecture can be obtained using second harmonic generation (SHG), which comprises non-absorptive frequency changes in an excitation laser line. SHG does not require any exogenous antibody or fluorophore labeling, and can generate images from unstained sections of several key endogenous biomolecules, in a wide variety of species and from different types of processed tissue. Here, we examined normal control human skin sections and human burn scar tissues using SHG on a multi-photon microscope (MPM). Examination and comparison of normal human skin and burn scar tissue demonstrated a clear arrangement of fibers in the dermis, similar to dermal collagen fiber signals. Fluorescence-staining confirmed the MPM-SHG collagen colocalization with antibody staining for dermal collagen type-I but not fibronectin or elastin. Furthermore, we were able to detect collagen MPM-SHG signal in human frozen sections as well as in unstained paraffin embedded tissue sections that were then compared with hematoxylin and eosin staining in the identical sections. This same approach was also successful in localizing collagen in porcine and ovine skin samples, and may be particularly important when species-specific antibodies may not be available. Collectively, our results demonstrate that MPM SHG-detection is a useful tool for high resolution examination of collagen architecture in both normal and wounded human, porcine and ovine dermal tissue.


Asunto(s)
Quemaduras , Cicatriz , Colágeno Tipo I/análisis , Epidermis/química , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Animales , Quemaduras/patología , Niño , Cicatriz/patología , Elastina/análisis , Epidermis/patología , Femenino , Feto , Fibronectinas/análisis , Humanos , Ovinos , Porcinos
17.
Biomaterials ; 32(21): 4782-92, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21477857

RESUMEN

Biomaterial scaffolds are categorized into artificial or natural polymers, or combinations of the two. Artificial polymers often undergo serum protein adsorption, elicit foreign body and encapsulation immune responses post-implantation. Large pore bovine electrospun collagen I was therefore screened as a candidate for human keratinocyte and fibroblast cell scaffolds. Human HaCaT keratinocyte and dermal fibroblasts were seeded on electrospun denatured collagen I microfiber (DCM) scaffolds and after 72 h Livedead(®) assays performed to determine adhesive cell, survival and scaffold penetration. Both keratinocytes and fibroblasts attached to and survived on DCM scaffolds, however only fibroblasts migrated over and into this biomaterial. HaCaT keratinocytes remained largely stationary on the scaffold surface in discrete islands of monolayered cells. For this reason, normal human epidermal keratinocyte (NHEK) scaffold interactions were assessed using scanning and transmission electron microscopy (EM) that demonstrated DCM scaffolds comprised networks of interlocking and protruding collagen fibers with a mean diameter of 2-5 µm, with a mean inter-fiber pore size of 6.7 µm (range 3-10 µm) and scaffold thickness 50-70 µm. After 72 h the keratinocytes and fibroblasts on DCM scaffolds had attached, flattened and spread over the entire scaffold with assembly of lamellapodia and focal adhesion (FA)-like junctions. Using transmission EM, NHEKs and HaCaT keratinocytes assembled desmosomes, lamellapodia and FA junctions, however, neither hemidesmosomes nor basal lamina were present. In long term (21 day) co-culture fibroblasts migrated throughout the scaffold and primary keratinocytes (and to a lesser extend HaCaTs) stratified on the scaffold surface forming a human skin equivalent (HSE). In vivo testing of these HSEs on immunocompetent (BalbC) and immunodeficient (SCID) excisionally wounded model mice demonstrated scaffold wound biocompatibility and ability to deliver human cells after scaffold biodegradation.


Asunto(s)
Materiales Biocompatibles/química , Colágeno/química , Fibroblastos/citología , Queratinocitos/citología , Microfibrillas/ultraestructura , Trasplante de Piel/métodos , Andamios del Tejido/química , Animales , Bovinos , Células Cultivadas , Técnicas de Cocultivo , Fibroblastos/fisiología , Fibroblastos/trasplante , Humanos , Queratinocitos/fisiología , Queratinocitos/trasplante , Ensayo de Materiales , Ratones , Ratones SCID , Desnaturalización Proteica , Piel/ultraestructura , Ingeniería de Tejidos/métodos
18.
Int J Dermatol ; 50(4): 439-42, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21413955

RESUMEN

BACKGROUND: Mutations in the plectin gene (PLEC) generally lead to epidermolysis bullosa simplex (EBS) associated with muscular dystrophy. It has been recently demonstrated that PLEC mutations can also cause a different clinical subtype, EBS associated with pyloric atresia (EBS-PA), which shows early lethality. Prenatal diagnosis (PND) of EBS-PA using mutation screening of PLEC has not been described. OBJECTIVE: This study aimed to perform DNA-based PND for an EBS-PA family. MATERIALS AND METHODS: The EBS-PA proband was compound-heterozygous for a paternal c.1350G>A splice-site mutation and a maternal p.Q305X nonsense mutation. Genomic DNA was obtained from amniocytes taken from an at-risk fetus of the proband's family. Direct sequencing and restriction enzyme digestion of polymerase chain reaction products from the genomic DNA were performed. RESULTS: Mutational analysis showed that the fetus harbored both pathogenic mutations, suggesting that the fetus was a compound-heterozygote and therefore affected with EBS-PA. The skin sample obtained by autopsy from the abortus confirmed the absence of plectin expression at the dermal-epidermal junction. CONCLUSIONS: This is the first successful DNA-based PND for an EBA-PA family.


Asunto(s)
Amniocentesis/métodos , Enfermedades Fetales/genética , Pruebas Genéticas/métodos , Plectina/genética , Aborto Terapéutico , Proteínas Bacterianas , Displasia Ectodérmica/genética , Epidermólisis Ampollosa Simple/genética , Salud de la Familia , Femenino , Humanos , Masculino , Linaje , Embarazo
19.
Cell Stem Cell ; 8(2): 177-87, 2011 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-21295274

RESUMEN

In most stem cell systems, the organization of the stem cell niche and the anchoring matrix required for stem cell maintenance are largely unknown. We report here that collagen XVII (COL17A1/BP180/BPAG2), a hemidesmosomal transmembrane collagen, is highly expressed in hair follicle stem cells (HFSCs) and is required for the maintenance not only of HFSCs but also of melanocyte stem cells (MSCs), which do not express Col17a1 but directly adhere to HFSCs. Mice lacking Col17a1 show premature hair graying and hair loss. Analysis of Col17a1-null mice revealed that COL17A1 is critical for the self-renewal of HFSCs through maintaining their quiescence and immaturity, potentially explaining the mechanism underlying hair loss in human COL17A1 deficiency. Moreover, forced expression of COL17A1 in basal keratinocytes, including HFSCs, in Col17a1-null mice rescues MSCs from premature differentiation and restores TGF-ß signaling, demonstrating that HFSCs function as a critical regulatory component of the MSC niche.


Asunto(s)
Folículo Piloso/citología , Melanocitos/citología , Células Madre/citología , Animales , Autoantígenos/genética , Autoantígenos/metabolismo , Células Cultivadas , Citometría de Flujo , Folículo Piloso/metabolismo , Folículo Piloso/ultraestructura , Inmunohistoquímica , Melanocitos/metabolismo , Melanocitos/ultraestructura , Ratones , Ratones Noqueados , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Colágenos no Fibrilares/genética , Colágenos no Fibrilares/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo , Células Madre/ultraestructura , Colágeno Tipo XVII
20.
Proc Natl Acad Sci U S A ; 107(32): 14345-50, 2010 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-20660747

RESUMEN

Attempts to treat congenital protein deficiencies using bone marrow-derived cells have been reported. These efforts have been based on the concepts of stem cell plasticity. However, it is considered more difficult to restore structural proteins than to restore secretory enzymes. This study aims to clarify whether bone marrow transplantation (BMT) treatment can rescue epidermolysis bullosa (EB) caused by defects in keratinocyte structural proteins. BMT treatment of adult collagen XVII (Col17) knockout mice induced donor-derived keratinocytes and Col17 expression associated with the recovery of hemidesmosomal structure and better skin manifestations, as well improving the survival rate. Both hematopoietic and mesenchymal stem cells have the potential to produce Col17 in the BMT treatment model. Furthermore, human cord blood CD34(+) cells also differentiated into keratinocytes and expressed human skin component proteins in transplanted immunocompromised (NOD/SCID/gamma(c)(null)) mice. The current conventional BMT techniques have significant potential as a systemic therapeutic approach for the treatment of human EB.


Asunto(s)
Membrana Basal/metabolismo , Trasplante de Médula Ósea/fisiología , Epidermólisis Ampollosa/terapia , Proteínas de la Membrana/biosíntesis , Animales , Autoantígenos/biosíntesis , Membrana Basal/química , Epidermis , Humanos , Queratinocitos/citología , Ratones , Ratones SCID , Colágenos no Fibrilares/biosíntesis , Colágenos no Fibrilares/deficiencia , Trasplante de Células Madre , Tasa de Supervivencia , Resultado del Tratamiento , Colágeno Tipo XVII
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...