Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Mol Ther Methods Clin Dev ; 27: 96-108, 2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36212909

ABSTRACT

Recessive dystrophic epidermolysis bullosa, a devastating skin fragility disease characterized by recurrent skin blistering, scarring, and a high risk of developing squamous cell carcinoma is caused by mutations in COL7A1, the gene encoding type VII collagen, which is the major component of the anchoring fibrils that bind the dermis and epidermis. Ex vivo correction of COL7A1 by gene editing in patients' cells has been achieved before. However, in vivo editing approaches are necessary to address the direct treatment of the blistering lesions characteristic of this disease. We have now generated adenoviral vectors for CRISPR-Cas9 delivery to remove exon 80 of COL7A1, which contains a highly prevalent frameshift mutation in Spanish patients. For in vivo testing, a humanized skin mouse model was used. Efficient viral transduction of skin was observed after excisional wounds generated with a surgical punch on regenerated patient skin grafts were filled with the adenoviral vectors embedded in a fibrin gel. Type VII collagen deposition in the basement membrane zone of the wounded areas treated with the vectors correlated with restoration of dermal-epidermal adhesion, demonstrating that recessive dystrophic epidermolysis bullosa (RDEB) patient skin lesions can be directly treated by CRISPR-Cas9 delivery in vivo.

2.
Gene Ther ; 29(3-4): 157-170, 2022 04.
Article in English | MEDLINE | ID: mdl-34363036

ABSTRACT

Recent advances in molecular biology have led to the CRISPR revolution, but the lack of an efficient and safe delivery system into cells and tissues continues to hinder clinical translation of CRISPR approaches. Polymeric vectors offer an attractive alternative to viruses as delivery vectors due to their large packaging capacity and safety profile. In this paper, we have demonstrated the potential use of a highly branched poly(ß-amino ester) polymer, HPAE-EB, to enable genomic editing via CRISPRCas9-targeted genomic excision of exon 80 in the COL7A1 gene, through a dual-guide RNA sequence system. The biophysical properties of HPAE-EB were screened in a human embryonic 293 cell line (HEK293), to elucidate optimal conditions for efficient and cytocompatible delivery of a DNA construct encoding Cas9 along with two RNA guides, obtaining 15-20% target genomic excision. When translated to human recessive dystrophic epidermolysis bullosa (RDEB) keratinocytes, transfection efficiency and targeted genomic excision dropped. However, upon delivery of CRISPR-Cas9 as a ribonucleoprotein complex, targeted genomic deletion of exon 80 was increased to over 40%. Our study provides renewed perspective for the further development of polymer delivery systems for application in the gene editing field in general, and specifically for the treatment of RDEB.


Subject(s)
CRISPR-Cas Systems , Epidermolysis Bullosa Dystrophica , Collagen Type VII/genetics , Epidermolysis Bullosa Dystrophica/genetics , Epidermolysis Bullosa Dystrophica/metabolism , Epidermolysis Bullosa Dystrophica/therapy , Gene Editing , HEK293 Cells , Humans , Polymers/metabolism
3.
Mol Ther ; 29(6): 2008-2018, 2021 06 02.
Article in English | MEDLINE | ID: mdl-33609734

ABSTRACT

Genome-editing technologies that enable the introduction of precise changes in DNA sequences have the potential to lead to a new class of treatments for genetic diseases. Epidermolysis bullosa (EB) is a group of rare genetic disorders characterized by extreme skin fragility. The recessive dystrophic subtype of EB (RDEB), which has one of the most severe phenotypes, is caused by mutations in COL7A1. In this study, we report a gene-editing approach for ex vivo homology-directed repair (HDR)-based gene correction that uses the CRISPR-Cas9 system delivered as a ribonucleoprotein (RNP) complex in combination with donor DNA templates delivered by adeno-associated viral vectors (AAVs). We demonstrate sufficient mutation correction frequencies to achieve therapeutic benefit in primary RDEB keratinocytes containing different COL7A1 mutations as well as efficient HDR-mediated COL7A1 modification in healthy cord blood-derived CD34+ cells and mesenchymal stem cells (MSCs). These results are a proof of concept for HDR-mediated gene correction in different cell types with therapeutic potential for RDEB.


Subject(s)
Epidermolysis Bullosa Dystrophica/genetics , Gene Editing/methods , Genes, Recessive , Genetic Therapy/methods , Mutation , Recombinational DNA Repair , CRISPR-Cas Systems , Cell Line , Collagen Type VII/genetics , Dependovirus/genetics , Epidermolysis Bullosa Dystrophica/therapy , Gene Expression , Gene Transfer Techniques , Genetic Vectors/genetics , Humans , Keratinocytes/metabolism
4.
Mol Ther Methods Clin Dev ; 18: 280-290, 2020 Sep 11.
Article in English | MEDLINE | ID: mdl-32637457

ABSTRACT

Current efforts to find specific genodermatoses treatments and define precise pathogenesis mechanisms require appropriate surrogate models with human cells. Although transgenic and gene knockout mouse models for several of these disorders exist, they often fail to faithfully replicate the clinical and histopathological features of the human skin condition. We have established a highly efficient method for precise deletion of critical gene sequences in primary human keratinocytes, based on CRISPR-Cas9-mediated gene editing. Using this methodology, in the present study we generated a model of Netherton syndrome by disruption of SPINK5. Gene-edited cells showed absence of LEKTI expression and were able to recapitulate a hyperkeratotic phenotype with most of the molecular hallmarks of Netherton syndrome, after grafting to immunodeficient mice and in organotypic cultures. To validate the model as a platform for therapeutic intervention, we tested an ex vivo gene therapy approach using a lentiviral vector expressing SPINK5. Re-expression of SPINK5 in an immortalized clone of SPINK5-knockout keratinocytes was capable of reverting from Netherton syndrome to a normal skin phenotype in vivo and in vitro. Our results demonstrate the feasibility of modeling genodermatoses, such as Netherton syndrome, by efficiently disrupting the causative gene to better understand its pathogenesis and to develop novel therapeutic approaches.

5.
Mol Ther ; 27(5): 986-998, 2019 05 08.
Article in English | MEDLINE | ID: mdl-30930113

ABSTRACT

Gene editing constitutes a novel approach for precisely correcting disease-causing gene mutations. Frameshift mutations in COL7A1 causing recessive dystrophic epidermolysis bullosa are amenable to open reading frame restoration by non-homologous end joining repair-based approaches. Efficient targeted deletion of faulty COL7A1 exons in polyclonal patient keratinocytes would enable the translation of this therapeutic strategy to the clinic. In this study, using a dual single-guide RNA (sgRNA)-guided Cas9 nuclease delivered as a ribonucleoprotein complex through electroporation, we have achieved very efficient targeted deletion of COL7A1 exon 80 in recessive dystrophic epidermolysis bullosa (RDEB) patient keratinocytes carrying a highly prevalent frameshift mutation. This ex vivo non-viral approach rendered a large proportion of corrected cells producing a functional collagen VII variant. The effective targeting of the epidermal stem cell population enabled long-term regeneration of a properly adhesive skin upon grafting onto immunodeficient mice. A safety assessment by next-generation sequencing (NGS) analysis of potential off-target sites did not reveal any unintended nuclease activity. Our strategy could potentially be extended to a large number of COL7A1 mutation-bearing exons within the long collagenous domain of this gene, opening the way to precision medicine for RDEB.


Subject(s)
CRISPR-Cas Systems/genetics , Collagen Type VII/genetics , Epidermolysis Bullosa Dystrophica/therapy , Gene Editing , Animals , Disease Models, Animal , Epidermolysis Bullosa Dystrophica/genetics , Epidermolysis Bullosa Dystrophica/pathology , Exons/genetics , Frameshift Mutation/genetics , High-Throughput Nucleotide Sequencing , Humans , Keratinocytes/metabolism , Mice , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/therapeutic use
6.
Mol Ther Nucleic Acids ; 11: 68-78, 2018 Jun 01.
Article in English | MEDLINE | ID: mdl-29858091

ABSTRACT

Recessive dystrophic epidermolysis bullosa is a severe skin fragility disease caused by loss of functional type VII collagen at the dermal-epidermal junction. A frameshift mutation in exon 80 of COL7A1 gene, c.6527insC, is highly prevalent in the Spanish patient population. We have implemented gene-editing strategies for COL7A1 frame restoration by NHEJ-induced indels in epidermal stem cells from patients carrying this mutation. TALEN nucleases designed to cut within the COL7A1 exon 80 sequence were delivered to primary patient keratinocyte cultures by non-integrating viral vectors. After genotyping a large collection of vector-transduced patient keratinocyte clones with high proliferative potential, we identified a significant percentage of clones with COL7A1 reading frame recovery and Collagen VII protein expression. Skin equivalents generated with cells from a clone lacking exon 80 entirely were able to regenerate phenotypically normal human skin upon their grafting onto immunodeficient mice. These patient-derived human skin grafts showed Collagen VII deposition at the basement membrane zone, formation of anchoring fibrils, and structural integrity when analyzed 12 weeks after grafting. Our data provide a proof-of-principle for recessive dystrophic epidermolysis bullosa treatment through ex vivo gene editing based on removal of pathogenic mutation-containing, functionally expendable COL7A1 exons in patient epidermal stem cells.

7.
Pediatr Dermatol ; 35(2): e94-e98, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29272047

ABSTRACT

Dystrophic epidermolysis bullosa is a rare blistering condition caused by mutations in the COL7A1 gene. Different clinical variants have been described, with dominant and recessive inheritance, but no consistent findings have been elucidated to establish a genotype-phenotype correlation. We present three unrelated patients with two identical pathogenic compound heterozygous mutations in the COL7A1 gene that developed different clinical forms of dystrophic epidermolysis bullosa-epidermolysis bullosa pruriginosa and mild recessive non-Hallopeau-Siemens-raising the possibility of other genetic or environmental modifying factors responsible for the phenotype of the disease.


Subject(s)
Collagen Type VII/genetics , Epidermolysis Bullosa Dystrophica/genetics , Adult , Child , Child, Preschool , Female , Genetic Association Studies , Heterozygote , Humans , Male , Mutation , Pedigree , Phenotype , Skin/pathology
8.
Mol Ther Nucleic Acids ; 5: e307, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-27045209

ABSTRACT

Clonal gene therapy protocols based on the precise manipulation of epidermal stem cells require highly efficient gene-editing molecular tools. We have combined adeno-associated virus (AAV)-mediated delivery of donor template DNA with transcription activator-like nucleases (TALE) expressed by adenoviral vectors to address the correction of the c.6527insC mutation in the COL7A1 gene, causing recessive dystrophic epidermolysis bullosa in a high percentage of Spanish patients. After transduction with these viral vectors, high frequencies of homology-directed repair were found in clones of keratinocytes derived from a recessive dystrophic epidermolysis bullosa (RDEB) patient homozygous for the c.6527insC mutation. Gene-edited clones recovered the expression of the COL7A1 transcript and collagen VII protein at physiological levels. In addition, treatment of patient keratinocytes with TALE nucleases in the absence of a donor template DNA resulted in nonhomologous end joining (NHEJ)-mediated indel generation in the vicinity of the c.6527insC mutation site in a large proportion of keratinocyte clones. A subset of these indels restored the reading frame of COL7A1 and resulted in abundant, supraphysiological expression levels of mutant or truncated collagen VII protein. Keratinocyte clones corrected both by homology-directed repair (HDR) or NHEJ were used to regenerate skin displaying collagen VII in the dermo-epidermal junction.

9.
Exp Dermatol ; 25(4): 269-74, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26739954

ABSTRACT

Epidermolysis bullosa with pyloric atresia (EB-PA) is a rare autosomal recessive hereditary disease with a variable prognosis from lethal to very mild. EB-PA is classified into Simplex form (EBS-PA: OMIM #612138) and Junctional form (JEB-PA: OMIM #226730), and it is caused by mutations in ITGA6, ITGB4 and PLEC genes. We report the analysis of six patients with EB-PA, including two dizygotic twins. Skin immunofluorescence epitope mapping was performed followed by PCR and direct sequencing of the ITGB4 gene. Two of the patients presented with non-lethal EB-PA associated with missense ITGB4 gene mutations. For the other four, early postnatal demise was associated with complete lack of ß4 integrin due to a variety of ITGB4 novel mutations (2 large deletions, 1 splice-site mutation and 3 missense mutations). One of the deletions spanned 278 bp, being one of the largest reported to date for this gene. Remarkably, we also found for the first time a founder effect for one novel mutation in the ITGB4 gene. We have identified 6 novel mutations in the ITGB4 gene to be added to the mutation database. Our results reveal genotype-phenotype correlations that contribute to the molecular understanding of this heterogeneous disease, a pivotal issue for prognosis and for the development of novel evidence-based therapeutic options for EB management.


Subject(s)
Ectodermal Dysplasia/genetics , Integrin beta4/genetics , Sequence Deletion , Biopsy , Child, Preschool , DNA Mutational Analysis , Ectodermal Dysplasia/diagnosis , Epitope Mapping , Epitopes/chemistry , Female , Genetic Association Studies , Humans , Infant , Infant, Newborn , Keratinocytes/cytology , Male , Microsatellite Repeats/genetics , Microscopy, Fluorescence , Mutation, Missense , Polymerase Chain Reaction , Prognosis , Sequence Analysis, DNA , Twins, Dizygotic
10.
Hum Mol Genet ; 23(10): 2551-68, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24363064

ABSTRACT

Tecta is a modular, non-collagenous protein of the tectorial membrane (TM), an extracellular matrix of the cochlea essential for normal hearing. Missense mutations in Tecta cause dominant forms of non-syndromic deafness and a genotype-phenotype correlation has been reported in humans, with mutations in different Tecta domains causing mid- or high-frequency hearing impairments that are either stable or progressive. Three mutant mice were created as models for human Tecta mutations; the Tecta(L1820F,G1824D/+) mouse for zona pellucida (ZP) domain mutations causing stable mid-frequency hearing loss in a Belgian family, the Tecta(C1837G/+) mouse for a ZP-domain mutation underlying progressive mid-frequency hearing loss in a Spanish family and the Tecta(C1619S/+) mouse for a zonadhesin-like (ZA) domain mutation responsible for progressive, high-frequency hearing loss in a French family. Mutations in the ZP and ZA domains generate distinctly different changes in the structure of the TM. Auditory brainstem response thresholds in the 8-40 kHz range are elevated by 30-40 dB in the ZP-domain mutants, whilst those in the ZA-domain mutant are elevated by 20-30 dB. The phenotypes are stable and no evidence has been found for a progressive deterioration in TM structure or auditory function. Despite elevated auditory thresholds, the Tecta mutant mice all exhibit an enhanced tendency to have audiogenic seizures in response to white noise stimuli at low sound pressure levels (≤84 dB SPL), revealing a previously unrecognised consequence of Tecta mutations. These results, together with those from previous studies, establish an allelic series for Tecta unequivocally demonstrating an association between genotype and phenotype.


Subject(s)
Deafness/genetics , Extracellular Matrix Proteins/genetics , Tectorial Membrane/pathology , Acoustic Stimulation , Animals , Deafness/pathology , Deafness/physiopathology , Disease Models, Animal , Epilepsy, Reflex/genetics , Female , GPI-Linked Proteins/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Hair Cells, Auditory, Inner/pathology , Homozygote , Humans , Male , Mice, 129 Strain , Mice, Transgenic , Molecular Motor Proteins/metabolism , Mutation, Missense , Organ of Corti/pathology , Phenotype , Tectorial Membrane/metabolism
11.
Exp Dermatol ; 22(9): 601-3, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23947675

ABSTRACT

Recessive dystrophic epidermolysis bullosa (RDEB) is caused by deficiency of type VII collagen due to COL7A1 mutations such as c.6527insC, recurrently found in the Spanish RDEB population. Assessment of clonal correction-based therapeutic approaches for RDEB requires large expansions of cells, exceeding the replication capacity of human primary keratinocytes. Thus, immortalized RDEB cells with enhanced proliferative abilities would be valuable. Using either the SV40 large T antigen or papillomavirus HPV16-derived E6-E7 proteins, we immortalized and cloned RDEB keratinocytes carrying the c.6527insC mutation. Clones exhibited high proliferative and colony-forming features. Cytogenetic analysis revealed important differences between T antigen-driven and E6-E7-driven immortalization. Immortalized cells responded to differentiation stimuli and were competent for epidermal regeneration and recapitulation of the blistering RDEB phenotype in vivo. These features make these cell lines useful to test novel therapeutic approaches including those aimed at editing mutant COL7A1.


Subject(s)
Collagen Type VII/genetics , Epidermolysis Bullosa Dystrophica/genetics , Epidermolysis Bullosa Dystrophica/therapy , Keratinocytes/metabolism , Mutation , Animals , Cell Line , Cell- and Tissue-Based Therapy , Epidermolysis Bullosa Dystrophica/pathology , Genetic Therapy , Heterografts , Homozygote , Humans , Keratinocytes/transplantation , Mice , Models, Genetic , Regeneration
12.
J Clin Invest ; 121(10): 3872-6, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21926461

ABSTRACT

T cells recognize antigens via their cell surface TCR and are classified as either αß or γδ depending on the variable chains in their TCR, α and ß or γ and δ, respectively. Both αß and γδ TCRs also contain several invariant chains, including CD3δ, which support surface TCR expression and transduce the TCR signal. Mutations in variable chains would be expected to affect a single T cell lineage, while mutations in the invariant chains would affect all T cells. Consistent with this, all CD3δ-deficient patients described to date showed a complete block in T cell development. However, CD3δ-KO mice have an αß T cell-specific defect. Here, we report 2 unrelated cases of SCID with a selective block in αß but not in γδ T cell development, associated with a new splicing mutation in the CD3D gene. The patients' T cells showed reduced CD3D transcripts, CD3δ proteins, surface TCR, and early TCR signaling. Their lymph nodes showed severe T cell depletion, recent thymus emigrants in peripheral blood were strongly decreased, and the scant αß T cells were oligoclonal. T cell-dependent B cell functions were also impaired, despite the presence of normal B cell numbers. Strikingly, despite the specific loss of αß T cells, surface TCR expression was more reduced in γδ than in αß T cells. Analysis of individuals with this CD3D mutation thus demonstrates the contrasting CD3δ requirements for αß versus γδ T cell development and TCR expression in humans and highlights the diagnostic and clinical relevance of studying both TCR isotypes when a T cell defect is suspected.


Subject(s)
CD3 Complex/genetics , Mutation , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/immunology , T-Lymphocyte Subsets/immunology , Animals , B-Lymphocytes/immunology , Base Sequence , DNA Mutational Analysis , Female , Humans , Infant , Killer Cells, Natural/immunology , Male , Mice , Pedigree , RNA Splice Sites/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Severe Combined Immunodeficiency/etiology
13.
Hum Mutat ; 32(7): 825-34, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21520338

ABSTRACT

The prevalence of DFNA8/DFNA12 (DFNA8/12), a type of autosomal dominant nonsyndromic hearing loss (ADNSHL), is unknown as comprehensive population-based genetic screening has not been conducted. We therefore completed unbiased screening for TECTA mutations in a Spanish cohort of 372 probands from ADNSHL families. Three additional families (Spanish, Belgian, and English) known to be linked to DFNA8/12 were also included in the screening. In an additional cohort of 835 American ADNSHL families, we preselected 73 probands for TECTA screening based on audiometric data. In aggregate, we identified 23 TECTA mutations in this process. Remarkably, 20 of these mutations are novel, more than doubling the number of reported TECTA ADNSHL mutations from 13 to 33. Mutations lie in all domains of the α-tectorin protein, including those for the first time identified in the entactin domain, as well as the vWFD1, vWFD2, and vWFD3 repeats, and the D1-D2 and TIL2 connectors. Although the majority are private mutations, four of them-p.Cys1036Tyr, p.Cys1837Gly, p.Thr1866Met, and p.Arg1890Cys-were observed in more than one unrelated family. For two of these mutations founder effects were also confirmed. Our data validate previously observed genotype-phenotype correlations in DFNA8/12 and introduce new correlations. Specifically, mutations in the N-terminal region of α-tectorin (entactin domain, vWFD1, and vWFD2) lead to mid-frequency NSHL, a phenotype previously associated only with mutations in the ZP domain. Collectively, our results indicate that DFNA8/12 hearing loss is a frequent type of ADNSHL.


Subject(s)
Extracellular Matrix Proteins/genetics , Hearing Loss, Sensorineural/genetics , Adolescent , Adult , Aged , Audiometry/methods , Child , Child, Preschool , Female , Founder Effect , GPI-Linked Proteins/genetics , Genetic Association Studies , Genetic Linkage , Haplotypes , Humans , Male , Middle Aged , Mutation , Pedigree , Protein Structure, Tertiary/genetics
14.
Hum Mol Genet ; 18(16): 3075-89, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19477959

ABSTRACT

Here we report the functional assessment of two novel deafness-associated gamma-actin mutants, K118N and E241K, in a spectrum of different situations with increasing biological complexity by combining biochemical and cell biological analysis in yeast and mammalian cells. Our in vivo experiments showed that while the K118N had a very mild effect on yeast behaviour, the phenotype caused by the E241K mutation was very severe and characterized by a highly compromised ability to grow on glycerol as a carbon source, an aberrant multi-vacuolar pattern and the deposition of thick F-actin bundles randomly in the cell. The latter feature is consistent with the highly unusual spontaneous tendency of the E241K mutant to form bundles in vitro, although this propensity to bundle was neutralized by tropomyosin and the E241K filament bundles were hypersensitive to severing in the presence of cofilin. In transiently transfected NIH3T3 cells both mutant actins were normally incorporated into cytoskeleton structures, although cytoplasmic aggregates were also observed indicating an element of abnormality caused by the mutations in vivo. Interestingly, gene-gun mediated expression of these mutants in cochlear hair cells results in no gross alteration in cytoskeletal structures or the morphology of stereocilia. Our results provide a more complete picture of the biological consequences of deafness-associated gamma-actin mutants and support the hypothesis that the post-lingual and progressive nature of the DFNA20/26 hearing loss is the result of a progressive deterioration of the hair cell cytoskeleton over time.


Subject(s)
Actins/genetics , Hearing Loss/genetics , Mutation, Missense , Actins/chemistry , Actins/metabolism , Animals , Base Sequence , Cells, Cultured , Cytoskeleton/metabolism , Hair Cells, Auditory/metabolism , Hearing Loss/metabolism , Humans , Mice , Molecular Conformation , Molecular Sequence Data , NIH 3T3 Cells , Pedigree , Yeasts/genetics , Yeasts/metabolism
15.
Nat Genet ; 41(5): 609-13, 2009 May.
Article in English | MEDLINE | ID: mdl-19363479

ABSTRACT

MicroRNAs (miRNAs) bind to complementary sites in their target mRNAs to mediate post-transcriptional repression, with the specificity of target recognition being crucially dependent on the miRNA seed region. Impaired miRNA target binding resulting from SNPs within mRNA target sites has been shown to lead to pathologies associated with dysregulated gene expression. However, no pathogenic mutations within the mature sequence of a miRNA have been reported so far. Here we show that point mutations in the seed region of miR-96, a miRNA expressed in hair cells of the inner ear, result in autosomal dominant, progressive hearing loss. This is the first study implicating a miRNA in a mendelian disorder. The identified mutations have a strong impact on miR-96 biogenesis and result in a significant reduction of mRNA targeting. We propose that these mutations alter the regulatory role of miR-96 in maintaining gene expression profiles in hair cells required for their normal function.


Subject(s)
Hearing Loss/genetics , MicroRNAs/genetics , Mutation , Adolescent , Adult , Aged , Base Sequence , Child , Female , Hair Cells, Auditory/metabolism , Humans , Male , MicroRNAs/chemistry , Middle Aged , Molecular Sequence Data , Pedigree , RNA, Messenger/metabolism
16.
J Assoc Res Otolaryngol ; 9(2): 202-14, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18452040

ABSTRACT

The TECTA gene encodes alpha-tectorin (TECTA), a major noncollagenous component of the tectorial membrane (TM). In humans, mutations in TECTA lead to either dominant (DFNA8/A12) or recessive (DFNB21) forms of nonsyndromic hearing loss. All missense mutations in TECTA that have been reported thus far are associated with the dominant subtype, whereas those leading to recessive deafness are all inactivating mutations. In this paper, we characterize a spontaneous missense mutation (c.1046C > A, p.A349D) arising in the mouse Tecta gene that is, unlike all previously reported missense mutations in TECTA, recessive. The morphological phenotype of the Tecta (A349D/A349D) mouse resembles but is not identical to that previously described for the Tecta(deltaENT)/(deltaENT) mouse. As in the Tecta(deltaENT/(deltaENT) mouse, the TM is completely detached from the surface of the organ of Corti and spiral limbus, lacks a striated-sheet matrix, and is deficient in both beta-tectorin (Tectb) and otogelin. A significant amount of Tecta is, however, detected in the TM of the Tecta (A349D/A349D) mouse, and numerous, electron-dense matrix granules are seen interspersed among the disorganized collagen fibrils. Mutated Tecta (A349D) is therefore incorporated into the TM but presumably unable to interact with either Tectb or otogelin. The Tecta (A349D/A349D) mouse reveals that missense mutations in Tecta can be recessive and lead to TM detachment and suggests that should similar mutations arise in the human population, they would likely cause deafness.


Subject(s)
Extracellular Matrix Proteins/genetics , Genes, Recessive , Hearing Loss, Sensorineural/genetics , Membrane Glycoproteins/genetics , Mutation, Missense , Animals , Base Sequence , Disease Models, Animal , Extracellular Matrix Proteins/metabolism , Female , GPI-Linked Proteins , Hearing Loss, Sensorineural/pathology , Male , Membrane Glycoproteins/metabolism , Mice , Molecular Sequence Data , Pedigree , Phenotype , Pregnancy , Tectorial Membrane/pathology , Tectorial Membrane/physiology
17.
Immunol Cell Biol ; 86(3): 289-91, 2008.
Article in English | MEDLINE | ID: mdl-18283295

ABSTRACT

To dissect the phenotypic and functional features of mucosal T lymphocytes in patients with gastric adenocarcinoma, we have used the Herpesvirus saimiri transformation procedure to achieve T-cell lines from gastric origin. Once achieved, cell function was assessed by in vitro stimulation with mitogens. CD2-specific monoclonal antibodies (alpha-CD2), alone or in combination with interleukin (IL)-2, rendered fewer counts in patients (34 408+/-3965 and 52 157+/-6473 c.p.m., respectively) than in controls (67 471+/-11 755 c.p.m., P<0.01 and 77 864+/-12 545 c.p.m., P<0.05, respectively). Likewise, CD3-based responses were defective in cancer cell lines: alpha-CD3 (54 794+/-9269 vs 86 104+/-10 341 c.p.m., P<0.01), alpha-CD3+IL-2 (57 789+/-8590 vs 88855+/-8516 c.p.m., P<0.01) and alpha-CD3+alpha-CD2 (52 130+/-7559 vs 120 852+/-16 552 c.p.m., P<0.01). Finally, IL-2 failed to adequately stimulate patient cell lines (39 310+/-4023 vs 60 945+/-9463 c.p.m., P<0.05). These results suggest that mucosal T lymphocytes in cancer patients are inherently impaired in their proliferative ability. This may be crucial in the control of tumour growth.


Subject(s)
Adenocarcinoma/immunology , Cell Line, Transformed , Gastric Mucosa/pathology , Herpesvirus 2, Saimiriine , Stomach Neoplasms/immunology , T-Lymphocytes/pathology , Adenocarcinoma/pathology , Aged , Antigens, CD/immunology , Cell Proliferation , Cell Transformation, Viral , Female , Gastric Mucosa/immunology , Humans , Immunity, Cellular , Immunophenotyping , Lymphocyte Activation , Male , Middle Aged , Stomach Neoplasms/pathology , T-Lymphocytes/immunology , T-Lymphocytes/virology
18.
Mol Immunol ; 45(2): 479-84, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17658607

ABSTRACT

We describe the second case of CD8 immunodeficiency. It confirms the pathogenic effect of p.Gly111Ser, leading to complete deficit of CD8+ lymphocytes, although the clinical manifestations may vary in severity. Similarly to the first case reported, our patient is also from Spanish Gypsy origin and homozygous for the p.Gly111Ser mutation in CD8alpha chain. The patient has suffered repeated respiratory infections from childhood but with conservation of her pulmonary parenchyma, on the contrary to the first patient, who died because of his respiratory injury. We developed an AluI-PCR-RFLP assay to screen a total of 1127 unrelated control individuals: 734 subjects of Gypsy ancestry from different sub-isolates and geographic locations in Europe, and 393 of Spanish (non-Gypsy) ethnicity. The results indicate that p.Gly111Ser is confined to the Spanish Gypsy population, where it occurs at a carrier rate of 0.4%. Analysis of microsatellite markers flanking the CD8A mutated gene revealed a shared polymorphic haplotype suggesting a common founder for p.Gly111Ser mutation that causes CD8 deficiency in the Spanish Gypsy population. CD8 immunodeficiency should be given diagnostic consideration in Spanish Gypsies with recurrent infections. Our findings may also have implications for these patients in terms of specific recommendations in vaccination and healthy habits and for genetic counseling of affected families.


Subject(s)
CD8 Antigens/genetics , Glycine/genetics , Immunologic Deficiency Syndromes/genetics , Mutation/genetics , Roma/genetics , Serine/genetics , Adolescent , DNA Mutational Analysis , Female , Haplotypes , Humans , Male , Pedigree , Spain
19.
Hum Genet ; 123(1): 41-53, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18030493

ABSTRACT

Mutations in the potassium channel gene KCNQ4 underlie DFNA2, a subtype of autosomal dominant progressive, high-frequency hearing loss. Based on a phenotype-guided mutational screening we have identified a novel mutation c.886G>A, leading to the p.G296S substitution in the pore region of KCNQ4 channel. The possible impact of this mutation on total KCNQ4 protein expression, relative surface expression and channel function was investigated. When the G296S mutant was expressed in Xenopus oocytes, electrophysiological recordings did not show voltage-activated K(+) currents. The p.G296S mutation impaired KCNQ4 channel activity in two manners. It greatly reduced surface expression and, secondarily, abolished channel function. The deficient expression at the cell surface membrane was further confirmed in non-permeabilized NIH-3T3 cells transfected with the mutant KCNQ4 tagged with the hemagglutinin epitope in the extracellular S1-S2 linker. Co-expression of mutant and wild type KCNQ4 in oocytes was performed to mimic the heterozygous condition of the p.G296S mutation in the patients. The results showed that the G296S mutant exerts a strong dominant-negative effect on potassium currents by reducing the wild type KCNQ4 channel expression at the cell surface. This is the first study to identify a trafficking-dependent dominant mechanism for the loss of KCNQ4 channel function in DFNA2.


Subject(s)
Deafness/genetics , KCNQ Potassium Channels/genetics , Mutation , 3T3 Cells , Amino Acid Sequence , Animals , Blotting, Western , Cell Membrane/metabolism , Female , Humans , Ion Channel Gating , Male , Mice , Molecular Sequence Data , Patch-Clamp Techniques , Pedigree , Sequence Homology, Amino Acid , Xenopus laevis
20.
Am J Hum Genet ; 80(6): 1076-89, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17503326

ABSTRACT

We previously mapped a novel autosomal dominant deafness locus, DFNA44, by studying a family with postlingual, progressive, nonsyndromic hearing loss. We report here on the identification of a mutation in CCDC50 as the cause of hearing loss in the family. CCDC50 encodes Ymer, an effector of epidermal growth factor (EGF)-mediated cell signaling that is ubiquitously expressed in different organs and has been suggested to inhibit down-regulation of the EGF receptor. We have examined its expression pattern in mouse inner ear. Western blotting and cell transfection results indicate that Ymer is a soluble, cytoplasmic protein, and immunostaining shows that Ymer is expressed in a complex spatiotemporal pattern during inner ear development. In adult inner ear, the expression of Ymer is restricted to the pillar cells of the cochlea, the stria vascularis, and the vestibular sensory epithelia, where it shows spatial overlap with the microtubule-based cytoskeleton. In dividing cells, Ymer colocalizes with microtubules of the mitotic apparatus. We suggest that DFNA44 hearing loss may result from a time-dependent disorganization of the microtubule-based cytoskeleton in the pillar cells and stria vascularis of the adult auditory system.


Subject(s)
Epidermal Growth Factor/metabolism , Hearing Loss/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mutation , Signal Transduction , Amino Acid Sequence , Animals , Cochlea/cytology , Cytoplasm/metabolism , Ear, Inner/metabolism , Hair Cells, Vestibular/metabolism , HeLa Cells , Humans , Immunohistochemistry , Mice , Microtubules/metabolism , Molecular Sequence Data , NIH 3T3 Cells , Open Reading Frames , Sequence Homology, Amino Acid , Solubility , Stria Vascularis/metabolism , Transfection , Vestibule, Labyrinth/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...