Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Cancers (Basel) ; 16(13)2024 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-39001538

RESUMEN

Recessive dystrophic epidermolysis bullosa (RDEB) is a rare severe hereditary skin disease characterized by skin and mucosa fragility, resulting in blister formation. The most severe complication in RDEB patients is the development of cutaneous squamous cell carcinoma (SCC), leading to premature death. There is a great deal of evidence suggesting a permissive tumor microenvironment (TME) as a driver of SCC development in RDEB patients. In a cohort of RDEB patients, we characterized the immune profiles of RDEB-SCCs and compared them with clinical, histopathological, and prognostic features. RDEB-SCCs were subdivided into four groups based on their occurrence (first onset or recurrences) and grading according to clinical, histopathological parameters of aggressiveness. Thirty-eight SCCs from 20 RDEB patients were analyzed. Five RDEB patients experienced an unfavorable course after the diagnosis of the first SCC, with early recurrence or metastasis, whereas 15 patients developed multiple SCCs without metastasis. High-risk primary RDEB-SCCs showed a higher neutrophil-to-lymphocyte ratio in the tumor microenvironment and an increased proportion of neutrophil extracellular traps (NETs). Additionally, citrullinated histone H3, a marker of NETs, was increased in the serum of RDEB patients with high-risk primary SCC, suggesting that this modified form of histone H3 may serve as a potential blood marker of unfavorable prognosis in RDEB-SCCs.

2.
J Invest Dermatol ; 144(4): 748-754, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38099888

RESUMEN

Pachyonychia congenita (PC) is a dominantly inherited genetic disorder of cornification. PC stands out among other genodermatoses because despite its rarity, it has been the focus of a very large number of pioneering translational research efforts over the past 2 decades, mostly driven by a patient support organization, the Pachyonychia Congenita Project. These efforts have laid the ground for innovative strategies that may broadly impact approaches to the management of other inherited cutaneous and noncutaneous diseases. This article outlines current avenues of research in PC, expected outcomes, and potential hurdles.


Asunto(s)
Queratodermia Palmoplantar , Paquioniquia Congénita , Humanos , Paquioniquia Congénita/diagnóstico , Paquioniquia Congénita/genética , Paquioniquia Congénita/terapia , Queratodermia Palmoplantar/genética , Administración Cutánea , Apoptosis , Diferenciación Celular , Mutación
3.
Nat Commun ; 12(1): 6446, 2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34750385

RESUMEN

The use of chimeric antigen receptor (CAR)-engineered regulatory T cells (Tregs) has emerged as a promising strategy to promote immune tolerance. However, in conventional T cells (Tconvs), CAR expression is often associated with tonic signaling, which can induce CAR-T cell dysfunction. The extent and effects of CAR tonic signaling vary greatly according to the expression intensity and intrinsic properties of the CAR. Here, we show that the 4-1BB CSD-associated tonic signal yields a more dramatic effect in CAR-Tregs than in CAR-Tconvs with respect to activation and proliferation. Compared to CD28 CAR-Tregs, 4-1BB CAR-Tregs exhibit decreased lineage stability and reduced in vivo suppressive capacities. Transient exposure of 4-1BB CAR-Tregs to a Treg stabilizing cocktail, including an mTOR inhibitor and vitamin C, during ex vivo expansion sharply improves their in vivo function and expansion after adoptive transfer. This study demonstrates that the negative effects of 4-1BB tonic signaling in Tregs can be mitigated by transient mTOR inhibition.


Asunto(s)
Receptores Quiméricos de Antígenos/inmunología , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Serina-Treonina Quinasas TOR/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Animales , Antígenos CD28/inmunología , Antígenos CD28/metabolismo , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/terapia , Antígeno HLA-A2/inmunología , Antígeno HLA-A2/metabolismo , Humanos , Inmunosupresores/farmacología , Inmunoterapia Adoptiva/métodos , Células Jurkat , Masculino , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Receptores Quiméricos de Antígenos/metabolismo , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Trasplante Heterólogo , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo
4.
Cell ; 184(14): 3812-3828.e30, 2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-34214472

RESUMEN

We study a patient with the human papilloma virus (HPV)-2-driven "tree-man" phenotype and two relatives with unusually severe HPV4-driven warts. The giant horns form an HPV-2-driven multifocal benign epithelial tumor overexpressing viral oncogenes in the epidermis basal layer. The patients are unexpectedly homozygous for a private CD28 variant. They have no detectable CD28 on their T cells, with the exception of a small contingent of revertant memory CD4+ T cells. T cell development is barely affected, and T cells respond to CD3 and CD2, but not CD28, costimulation. Although the patients do not display HPV-2- and HPV-4-reactive CD4+ T cells in vitro, they make antibodies specific for both viruses in vivo. CD28-deficient mice are susceptible to cutaneous infections with the mouse papillomavirus MmuPV1. The control of HPV-2 and HPV-4 in keratinocytes is dependent on the T cell CD28 co-activation pathway. Surprisingly, human CD28-dependent T cell responses are largely redundant for protective immunity.


Asunto(s)
Antígenos CD28/deficiencia , Patrón de Herencia/genética , Papillomaviridae/fisiología , Piel/virología , Linfocitos T/inmunología , Adulto , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Antígenos CD28/genética , Antígenos CD28/metabolismo , Linfocitos T CD4-Positivos/inmunología , Niño , Endopeptidasas/metabolismo , Femenino , Genes Recesivos , Células HEK293 , Homocigoto , Humanos , Inmunidad Humoral , Memoria Inmunológica , Células Jurkat , Queratinocitos/patología , Masculino , Ratones Endogámicos C57BL , Oncogenes , Papiloma/patología , Papiloma/virología , Linaje , Señales de Clasificación de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo
5.
Sci Transl Med ; 10(455)2018 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-30135250

RESUMEN

Recessive dystrophic epidermolysis bullosa (RDEB) is a rare inherited skin and mucous membrane fragility disorder complicated by early-onset, highly malignant cutaneous squamous cell carcinomas (SCCs). The molecular etiology of RDEB SCC, which arises at sites of sustained tissue damage, is unknown. We performed detailed molecular analysis using whole-exome, whole-genome, and RNA sequencing of 27 RDEB SCC tumors, including multiple tumors from the same patient and multiple regions from five individual tumors. We report that driver mutations were shared with spontaneous, ultraviolet (UV) light-induced cutaneous SCC (UV SCC) and head and neck SCC (HNSCC) and did not explain the early presentation or aggressive nature of RDEB SCC. Instead, endogenous mutation processes associated with apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like (APOBEC) deaminases dominated RDEB SCC. APOBEC mutation signatures were enhanced throughout RDEB SCC tumor evolution, relative to spontaneous UV SCC and HNSCC mutation profiles. Sixty-seven percent of RDEB SCC driver mutations was found to emerge as a result of APOBEC and other endogenous mutational processes previously associated with age, potentially explaining a >1000-fold increased incidence and the early onset of these SCCs. Human papillomavirus-negative basal and mesenchymal subtypes of HNSCC harbored enhanced APOBEC mutational signatures and transcriptomes similar to those of RDEB SCC, suggesting that APOBEC deaminases drive other subtypes of SCC. Collectively, these data establish specific mutagenic mechanisms associated with chronic tissue damage. Our findings reveal a cause for cancers arising at sites of persistent inflammation and identify potential therapeutic avenues to treat RDEB SCC.


Asunto(s)
Desaminasas APOBEC/genética , Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/genética , Citosina Desaminasa/genética , Epidermólisis Ampollosa Distrófica/enzimología , Epidermólisis Ampollosa Distrófica/genética , Mutación/genética , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/genética , Variaciones en el Número de Copia de ADN/genética , Reparación del ADN/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Mutagénesis/genética , Tasa de Mutación , Transcriptoma/genética
6.
Sci Immunol ; 3(24)2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29907691

RESUMEN

Heterozygosity for human signal transducer and activator of transcription 3 (STAT3) dominant-negative (DN) mutations underlies an autosomal dominant form of hyper-immunoglobulin E syndrome (HIES). We describe patients with an autosomal recessive form of HIES due to loss-of-function mutations of a previously uncharacterized gene, ZNF341 ZNF341 is a transcription factor that resides in the nucleus, where it binds a specific DNA motif present in various genes, including the STAT3 promoter. The patients' cells have low basal levels of STAT3 mRNA and protein. The autoinduction of STAT3 production, activation, and function by STAT3-activating cytokines is strongly impaired. Like patients with STAT3 DN mutations, ZNF341-deficient patients lack T helper 17 (TH17) cells, have an excess of TH2 cells, and have low memory B cells due to the tight dependence of STAT3 activity on ZNF341 in lymphocytes. Their milder extra-hematopoietic manifestations and stronger inflammatory responses reflect the lower ZNF341 dependence of STAT3 activity in other cell types. Human ZNF341 is essential for the STAT3 transcription-dependent autoinduction and sustained activity of STAT3.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Síndrome de Job/genética , Factor de Transcripción STAT3/genética , Factores de Transcripción/genética , Transcripción Genética/inmunología , Adolescente , Adulto , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Núcleo Celular/metabolismo , Consanguinidad , Citocinas/inmunología , Citocinas/metabolismo , Exones/genética , Femenino , Genes Recesivos/genética , Genes Recesivos/inmunología , Homocigoto , Humanos , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Síndrome de Job/sangre , Síndrome de Job/inmunología , Mutación con Pérdida de Función , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Linaje , Regiones Promotoras Genéticas/genética , ARN Mensajero/metabolismo , Factor de Transcripción STAT3/inmunología , Factor de Transcripción STAT3/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Células Th2/inmunología , Células Th2/metabolismo , Factores de Transcripción/inmunología , Factores de Transcripción/metabolismo , Secuenciación del Exoma , Adulto Joven , Dedos de Zinc/genética
8.
J Invest Dermatol ; 137(5): e123-e129, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28411843

RESUMEN

Stunning technological advances in genomics have led to spectacular breakthroughs in the understanding of the underlying defects, biological pathways and therapeutic targets of skin diseases leading to new therapeutic interventions. Next-generation sequencing has revolutionized the identification of disease-causing genes and has a profound impact in deciphering gene and protein signatures in rare and frequent skin diseases. Gene addition strategies have shown efficacy in junctional EB and in recessive dystrophic EB (RDEB). TALENs and Cripsr/Cas9 have emerged as highly efficient new tools to edit genomic sequences to creat new models and to correct or disrupt mutated genes to treat human diseases. Therapeutic approaches have not been limited to DNA modification and strategies at the mRNA, protein and cellular levels have also emerged, some of which have already proven clinical efficacy in RDEB. Improved understanding of the pathogenesis of skin disorders has led to the development of specific drugs or repurposing of existing medicines as in basal cell nevus syndrome, alopecia areata, melanoma and EB simplex. These discoveries pave the way for improved targeted personalized medicine for rare and frequent diseases. It is likely that a growing number of orphan skin diseases will benefit from combinatory new therapies in a near future.


Asunto(s)
Edición Génica/métodos , Genómica/métodos , Enfermedades de la Piel/genética , Animales , Dermatología/métodos , Diseño de Fármacos , Reposicionamiento de Medicamentos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Medicina de Precisión/métodos , Enfermedades Raras/genética , Enfermedades Raras/fisiopatología , Enfermedades Raras/terapia , Enfermedades de la Piel/fisiopatología , Enfermedades de la Piel/terapia
9.
J Invest Dermatol ; 136(7): 1346-1354, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26994967

RESUMEN

Patients with recessive dystrophic epidermolysis bullosa (RDEB) lack type VII collagen and therefore have severely impaired dermal-epidermal stability causing recurrent skin and mucosal blistering. There is currently no specific approved treatment for RDEB. We present preclinical data showing that intradermal injections of genetically corrected patient-derived RDEB fibroblasts using a Good Manufacturing Practices grade self-inactivating COL7A1 retroviral vector reverse the disease phenotype in a xenograft model in nude mice. We obtained 50% transduction efficiency in primary human RDEB fibroblasts with an average low copy number (range = 1-2) of integrated provirus. Transduced fibroblasts showed strong type VII collagen re-expression, improved adhesion properties, normal proliferative capabilities, and viability in vitro. We show that a single intradermal injection of 3 × 10(6) genetically corrected RDEB fibroblasts beneath RDEB skin equivalents grafted onto mice allows type VII collagen deposition, anchoring fibril formation at the dermal-epidermal junction, and improved dermal-epidermal adherence 2 months after treatment, supporting functional correction in vivo. Gene-corrected fibroblasts previously showed no tumorigenicity. These data show the efficacy and safety of gene-corrected fibroblast therapy using a self-inactivating vector that has now been good manufacturing grade-certified and pave the way for clinical translation to treat nonhealing wounds in RDEB patients.


Asunto(s)
Colágeno Tipo VII/genética , Epidermólisis Ampollosa Distrófica/genética , Epidermólisis Ampollosa Distrófica/terapia , Fibroblastos/metabolismo , Terapia Genética , Animales , Adhesión Celular , Proliferación Celular , Colágeno Tipo VII/metabolismo , Fibroblastos/citología , Genes Recesivos , Vectores Genéticos , Células HEK293 , Humanos , Queratinocitos/citología , Ratones , Ratones Desnudos , Mutación , Trasplante de Neoplasias , Proteínas Recombinantes/genética , Retroviridae , Piel/metabolismo , Temperatura
10.
Hum Gene Ther Clin Dev ; 25(4): 218-28, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25381930

RESUMEN

The clinical application of self-inactivating (SIN) retroviral vectors requires an efficient vector production technology. To enable production of γ-retroviral SIN vectors from stable producer cells, new targetable HEK293-based producer clones were selected, providing amphotropic, GALV, or RD114 pseudotyping. Viral vector expression constructs can reliably be inserted at a predefined genomic locus via Flp-recombinase-mediated cassette exchange. Introduction of a clean-up step, mediated by Cre-recombinase, allows the removal of residual sequences that were required for targeting and selection, but were dispensable for the final producer clones and eliminated homology-driven recombination between the tagging and the therapeutic vector. The system was used to establish GALV and RD114 pseudotyping producer cells (HG- and HR820) for a clinically relevant long terminal repeat-driven therapeutic vector, designed for the transfer of a recombinant TCR that delivered titers in the range of 2×10(7) infectious particles (IP)/ml. Production capacity of the amphotropic producer cell (HA820) was challenged by a therapeutic SIN vector transferring the large COL7A1 cDNA. The final producer clone delivered a titer of 4×10(6) IP/ml and the vector containing supernatant was used directly to functionally restore primary fibroblasts and keratinocytes isolated from recessive dystrophic epidermolysis bullosa patients. Thus, the combinatorial approach (fc-technology) to generate producer cells for therapeutic γ-retroviral (SIN) vectors is feasible, is highly efficient, and allows their safe production and application in clinical trials.


Asunto(s)
Colágeno Tipo VII/genética , ADN Recombinante/genética , Gammaretrovirus/genética , Ingeniería Genética/métodos , Vectores Genéticos/genética , Colágeno Tipo VII/metabolismo , ADN Recombinante/aislamiento & purificación , Gammaretrovirus/metabolismo , Marcación de Gen/métodos , Vectores Genéticos/aislamiento & purificación , Células HEK293 , Humanos
12.
J Invest Dermatol ; 131(4): 848-56, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21124339

RESUMEN

Recessive dystrophic epidermolysis bullosa (RDEB) is an inherited blistering skin disorder caused by mutations in the COL7A1 gene-encoding type VII collagen (Col7), the major component of anchoring fibrils at the dermal-epidermal junction. Individuals with RDEB develop painful blisters and mucosal erosions, and currently, there are no effective forms of therapy. Nevertheless, some advances in patient therapy are being made, and cell-based therapies with mesenchymal and hematopoietic cells have shown promise in early clinical trials. To establish a foundation for personalized, gene-corrected, patient-specific cell transfer, we generated induced pluripotent stem (iPS) cells from three subjects with RDEB (RDEB iPS cells). We found that Col7 was not required for stem cell renewal and that RDEB iPS cells could be differentiated into both hematopoietic and nonhematopoietic lineages. The specific epigenetic profile associated with de-differentiation of RDEB fibroblasts and keratinocytes into RDEB iPS cells was similar to that observed in wild-type (WT) iPS cells. Importantly, human WT and RDEB iPS cells differentiated in vivo into structures resembling the skin. Gene-corrected RDEB iPS cells expressed Col7. These data identify the potential of RDEB iPS cells to generate autologous hematopoietic grafts and skin cells with the inherent capacity to treat skin and mucosal erosions that typify this genodermatosis.


Asunto(s)
Epidermólisis Ampollosa Distrófica , Genes Recesivos , Trasplante de Células Madre Hematopoyéticas , Trasplante de Células Madre Mesenquimatosas , Células Madre Pluripotentes/citología , Diferenciación Celular/fisiología , Células Cultivadas , Colágeno Tipo VII/genética , Epidermólisis Ampollosa Distrófica/genética , Epidermólisis Ampollosa Distrófica/patología , Epidermólisis Ampollosa Distrófica/terapia , Epigénesis Genética/fisiología , Fibroblastos/citología , Humanos , Técnicas In Vitro , Queratinocitos/citología , Medicina de Precisión
13.
Mol Ther ; 18(8): 1509-18, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20485266

RESUMEN

Recessive dystrophic epidermolysis bullosa (RDEB) is caused by loss-of-function mutations in COL7A1 encoding type VII collagen which forms key structures (anchoring fibrils) for dermal-epidermal adherence. Patients suffer since birth from skin blistering, and develop severe local and systemic complications resulting in poor prognosis. We lack a specific treatment for RDEB, but ex vivo gene transfer to epidermal stem cells shows a therapeutic potential. To minimize the risk of oncogenic events, we have developed new minimal self-inactivating (SIN) retroviral vectors in which the COL7A1 complementary DNA (cDNA) is under the control of the human elongation factor 1alpha (EF1alpha) or COL7A1 promoters. We show efficient ex vivo genetic correction of primary RDEB keratinocytes and fibroblasts without antibiotic selection, and use either of these genetically corrected cells to generate human skin equivalents (SEs) which were grafted onto immunodeficient mice. We achieved long-term expression of recombinant type VII collagen with restored dermal-epidermal adherence and anchoring fibril formation, demonstrating in vivo functional correction. In few cases, rearranged proviruses were detected, which were probably generated during the retrotranscription process. Despite this observation which should be taken under consideration for clinical application, this preclinical study paves the way for a therapy based on grafting the most severely affected skin areas of patients with fully autologous SEs genetically corrected using a SIN COL7A1 retroviral vector.


Asunto(s)
Colágeno Tipo VII/metabolismo , Epidermólisis Ampollosa Distrófica/terapia , Vectores Genéticos/genética , Retroviridae/genética , Animales , Southern Blotting , Western Blotting , Células Cultivadas , Colágeno Tipo VII/genética , Epidermólisis Ampollosa Distrófica/metabolismo , Fibroblastos/metabolismo , Humanos , Inmunohistoquímica , Queratinocitos/metabolismo , Ratones , Ratones SCID , Microscopía Electrónica de Transmisión , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas/genética
14.
J Clin Immunol ; 30(4): 607-19, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20405178

RESUMEN

INTRODUCTION: Bone marrow mesenchymal stem cells (BM-MSCs) and adipose tissue-derived stem cells share immunosuppressive capacities, suggesting that the latter could be a general property of stromal cells. METHODS: To check this hypothesis, we compared human BM-MSC and fibroblasts for their in vitro multi-potentiality, expandability and their immunomodulatory properties under normalized optimized culture conditions. RESULTS: We report that, unlike BM-MSCs, fibroblasts cannot differentiate in vitro into adipocytes and osteoblasts and differ from BM-MSCs by the expression of membrane CD106, CD10 and CD26 and by the expression of collagen VII mRNA. Like BM-MSCs, fibroblasts are unable to provoke in vitro allogeneic reactions, but strongly suppress lymphocyte proliferation induced by allogeneic mixed lymphocyte culture (MLC) or mitogens. We show that fibroblasts' immunosuppressive capacity is independent from prostaglandin E2, IL-10 and the tryptophan catabolising enzyme indoleamine 2,3-dioxygenase and is not abrogated after the depletion of CD8+ T lymphocytes, NK cells and monocytes. CONCLUSION: Finally, fibroblasts and BM-MSCs act at an early stage through blockage of lymphocyte activation, as demonstrated by down-regulation of GZMB (granzyme B) and IL2RA (CD25) expression.


Asunto(s)
Fibroblastos/inmunología , Células Madre Mesenquimatosas/inmunología , Tejido Adiposo/citología , Células de la Médula Ósea , Células Cultivadas , Regulación hacia Abajo/genética , Fibroblastos/citología , Granzimas/genética , Humanos , Subunidad alfa del Receptor de Interleucina-2/genética , Activación de Linfocitos , Células Madre Mesenquimatosas/citología , Células del Estroma/inmunología
15.
Hum Mutat ; 29(2): 267-76, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18030675

RESUMEN

Recessive dystrophic epidermolysis bullosa (RDEB) is caused by mutations in the COL7A1 gene encoding type VII collagen. Variations in severity between the different clinical forms of RDEB likely depend on the nature and location of COL7A1 mutations, but observed intrafamilial phenotypic variations suggest additional genetic and/or environmental factors. Candidate modifier genes include MMP1, encoding matrix metalloproteinase 1, the first gene implicated in RDEB before its primary role in the disease was excluded. Type VII collagen is a substrate of MMP1 and an imbalance between its synthesis and degradation could conceivably worsen the RDEB phenotype. Here, we studied a previously described family with three affected siblings of identical COL7A1 genotype but displaying great sibling-to-sibling variations in disease severity. RDEB severity did not correlate with type VII collagen synthesis levels, but with protein levels at the dermal-epidermal junction, suggesting increased degradation by metalloproteinases. This was supported by the presence of increased transcript and active MMP1 levels in the most severely affected children, who carried a known SNP (1G/2G) in the MMP1 promoter. This SNP creates a functional Ets binding site resulting in transcriptional upregulation. We next studied a French cohort of 31 unrelated RDEB patients harboring at least one in-frame COL7A1 mutation, ranging from mild localized RDEB to the severe Hallopeau-Siemens form. We found a strong genetic association between the 2G variant and the Hallopeau-Siemens disease type (odds ratio: 73.6). This is the first example of a modifier gene in RDEB and has implications for its prognosis and possible new treatments.


Asunto(s)
Epidermólisis Ampollosa Distrófica/enzimología , Epidermólisis Ampollosa Distrófica/genética , Genes Recesivos , Metaloproteinasa 1 de la Matriz/genética , Polimorfismo de Nucleótido Simple/genética , Regiones Promotoras Genéticas/genética , Adolescente , Adulto , Sitios de Unión , Células Cultivadas , Estudios de Cohortes , Colágeno Tipo VII/metabolismo , Epidermólisis Ampollosa Distrófica/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Francia , Regulación Enzimológica de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Proteínas Proto-Oncogénicas c-ets/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Población Blanca/genética
16.
J Invest Dermatol ; 127(3): 568-73, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17082782

RESUMEN

Until the identification of ABCA12 as the causative gene, prenatal diagnosis (PD) for harlequin ichthyosis (HI) had been performed by electron microscopic observation of fetal skin biopsy samples. We report the first case of HI DNA-based PD. Direct sequence analysis of ABCA12 revealed that the deceased proband was a compound heterozygote for two novel mutations. The maternal nonsense mutation p.Ser1249Term likely leads to nonsense-mediated messenger RNA decay. The paternal mutation c.7436G>A affects the last codon of exon 50 and was expected to be a splice site mutation. For their third pregnancy, the parents requested PD. Direct sequence analysis of fetal genomic DNA from amniotic fluid cells at 17 weeks gestation revealed the fetus was a compound heterozygote for both mutations. The parents requested the pregnancy to be terminated. Analysis of ABCA12 transcripts of cultured keratinocytes from the abortus showed the presence of six abnormally spliced products from the allele carrying the splice site mutation. Four of them lead to premature termination codons whereas the two others produced shortened proteins missing 21 and 31 amino acids from the second ATP-binding cassette. This report provides evidence for residual ABCA12 expression in HI, and demonstrates the efficiency of early DNA-based PD of HI.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , ADN/genética , Ictiosis Lamelar/diagnóstico , Ictiosis Lamelar/genética , Mutación , Diagnóstico Prenatal/métodos , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Análisis Mutacional de ADN , Femenino , Humanos , Recién Nacido , Masculino , Ratones , Datos de Secuencia Molecular , Embarazo , Homología de Secuencia de Ácido Nucleico
17.
J Biol Chem ; 281(19): 13525-13532, 2006 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-16524880

RESUMEN

Ultraviolet (UV) radiation from the sun is widely considered as a major cause of human skin photoaging and skin cancer. Granzyme B (GrB) and perforin (PFN) are two proteins contained in granules and implicated in one of the mechanisms by which cytotoxic lymphocytes and natural killer cells exert their cytotoxicity against virus-infected, alloreactive, or transformed cells. The distribution of GrB and PFN in the skin has received little attention. However, Berthou and co-workers (Berthou, C., Michel, L., Soulie, A., Jean-Louis, F., Flageul, B., Dubertret, L., Sigaux, F., Zhang, Y., and Sasportes, M. (1997) J. Immunol. 159, 5293-5300) described that, whereas freshly isolated epidermal cells did not express GrB or PFN, keratinocyte growth to confluence was associated with GrB and PFN mRNA and protein synthesis. In this work, we have investigated the possible role of UV-B on GrB and PFN expression in keratinocytes. We found that UV-B induces GrB and PFN expression in these cells through redox-, epidermal growth factor receptor-, and mitogen-activated protein kinase-dependent signaling. Furthermore, under UV irradiation, keratinocytes acquire a significant cytotoxicity, which is GrB and PFN dependent, toward a variety of cellular targets including transformed T-lymphocytes, melanocytes, and keratinocytes. This phenomenon may have important functional consequences in the regulation of skin inflammatory response and in the emergence of cancer skin.


Asunto(s)
Queratinocitos/metabolismo , Queratinocitos/efectos de la radiación , Glicoproteínas de Membrana/metabolismo , Serina Endopeptidasas/metabolismo , Rayos Ultravioleta , Línea Celular , Receptores ErbB/metabolismo , Regulación de la Expresión Génica/efectos de la radiación , Granzimas , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Melanocitos , Glicoproteínas de Membrana/genética , Perforina , Proteínas Citotóxicas Formadoras de Poros , ARN Mensajero/metabolismo , Serina Endopeptidasas/genética , Linfocitos T
18.
Hum Mutat ; 27(3): 291-2, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16470588

RESUMEN

Loss-of-function mutations in the gene encoding type VII collagen, COL7A1, are the molecular basis of the blistering skin disorder, recessive dystrophic epidermolysis bullosa (RDEB). COL7A1 maps to a region of the short arm of chromosome 3 that has been found to be deleted in many types of malignancies. We have characterized the first case of a large genomic deletion in chromosome 3p21.31 that removes COL7A1 entirely in an RDEB patient. This interstitial deletion spans 255 to 520 kb and encompasses 9 to 15 genes, but seems to have no pathological consequences other than RDEB. We show that the second, hemizygous allele of COL7A1 in this patient bears a base substitution within exon 94, c.7245G>A. This translates into an amino acid substitution, p.M2415I, and leads to a complex splicing abnormality that allows marginal levels of functional mRNA and protein to be synthesized. We propose that the leakiness of the splicing defect enables the partial rescue of collagen VII deficiency. This is consistent with the diagnosis of the moderately severe form of RDEB in the proband, at variance with the most severe form, RDEB Hallopeau-Siemens, that would arise from complete collagen VII deficiency.


Asunto(s)
Colágeno Tipo VII/genética , Epidermólisis Ampollosa Distrófica/genética , Mutación Missense , Empalme del ARN , Alelos , Animales , Células Cultivadas , Preescolar , Epidermólisis Ampollosa Distrófica/etiología , Femenino , Genes Recesivos , Humanos , Hibridación Fluorescente in Situ , Masculino , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA