Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(2)2024 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-38255836

RESUMO

Antisense oligonucleotides (ASOs) represent an emerging therapeutic platform for targeting genetic diseases by influencing various aspects of (pre-)mRNA biology, such as splicing, stability, and translation. In this study, we investigated the potential of modulating the splicing pattern in recessive dystrophic epidermolysis bullosa (RDEB) patient cells carrying a frequent genomic variant (c.425A > G) that disrupts splicing in the COL7A1 gene by using short 2'-O-(2-Methoxyethyl) oligoribo-nucleotides (2'-MOE ASOs). COL7A1-encoded type VII collagen (C7) forms the anchoring fibrils within the skin that are essential for the attachment of the epidermis to the underlying dermis. As such, gene variants of COL7A1 leading to functionally impaired or absent C7 manifest in the form of extensive blistering and wounding. The severity of the disease pattern warrants the development of novel therapies for patients. The c.425A > G variant at the COL7A1 exon 3/intron 3 junction lowers the efficiency of splicing at this junction, resulting in non-functional C7 transcripts. However, we found that correct splicing still occurs, albeit at a very low level, highlighting an opportunity for intervention by modulating the splicing reaction. We therefore screened 2'-MOE ASOs that bind along the COL7A1 target region ranging from exon 3 to the intron 3/exon 4 junction for their ability to modulate splicing. We identified ASOs capable of increasing the relative levels of correctly spliced COL7A1 transcripts by RT-PCR, sqRT-PCR, and ddPCR. Furthermore, RDEB-derived skin equivalents treated with one of the most promising ASOs exhibited an increase in full-length C7 expression and its accurate deposition along the basement membrane zone (BMZ).


Assuntos
Epidermólise Bolhosa Distrófica , Humanos , Epidermólise Bolhosa Distrófica/genética , Epidermólise Bolhosa Distrófica/terapia , Splicing de RNA , Pele , Íntrons , Precursores de RNA , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/uso terapêutico , Colágeno Tipo VII/genética
2.
Br J Dermatol ; 190(1): 80-93, 2023 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-37681509

RESUMO

BACKGROUND: Cutaneous squamous cell carcinoma (SCC) is the leading cause of death in patients with recessive dystrophic epidermolysis bullosa (RDEB). However, the survival time from first diagnosis differs between patients; some tumours spread particularly fast, while others may remain localized for years. As treatment options are limited, there is an urgent need for further insights into the pathomechanisms of RDEB tumours, to foster therapy development and support clinical decision-making. OBJECTIVES: To investigate differences in RDEB tumours of diverging aggressiveness at the molecular and phenotypic level, with a particular focus on epithelial-to-mesenchymal (EMT) transition states and thus microRNA-200b (miR-200b) as a regulator. METHODS: Primary RDEB-SCC keratinocyte lines were characterized with respect to their EMT state. For this purpose, cell morphology was classified and the expression of EMT markers analysed using immunofluorescence, flow cytometry, semi-quantitative reverse transcriptase polymerase chain reaction and Western blotting. The motility of RDEB-SCC cells was determined and conditioned medium of RDEB-SCC cells was used to treat endothelial cells in an angiogenesis assay. In addition, we mined previously generated microRNA (miRNA) profiling data to identify a candidate with potential therapeutic relevance and performed transient miRNA transfection studies to investigate the candidate's ability to reverse EMT characteristics. RESULTS: We observed high variability in EMT state in the RDEB-SCC cell lines, which correlated with in situ analysis of two available patient biopsies and respective clinical disease course. Furthermore, we identified miR-200b-3p to be downregulated in RDEB-SCCs, and the extent of deregulation significantly correlated with the EMT features of the various tumour lines. miR-200b-3p was reintroduced into RDEB-SCC cell lines with pronounced EMT features, which resulted in a significant increase in epithelial characteristics, including cell morphology, EMT marker expression, migration and angiogenic potential. CONCLUSIONS: RDEB-SCCs exist in different EMT states and the level of miR-200b is indicative of how far an RDEB-SCC has gone down the EMT path. Moreover, the reintroduction of miR-200b significantly reduced mesenchymal features.


Assuntos
Carcinoma de Células Escamosas , Epidermólise Bolhosa Distrófica , Transição Epitelial-Mesenquimal , MicroRNAs , Neoplasias Cutâneas , Humanos , Carcinoma de Células Escamosas/etiologia , Células Endoteliais/patologia , Epidermólise Bolhosa Distrófica/genética , Epidermólise Bolhosa Distrófica/complicações , Transição Epitelial-Mesenquimal/genética , MicroRNAs/genética , Neoplasias Cutâneas/patologia
3.
Mol Ther ; 30(8): 2680-2692, 2022 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-35490295

RESUMO

Junctional epidermolysis bullosa (JEB) is a debilitating hereditary skin disorder caused by mutations in genes encoding laminin-332, type XVII collagen (C17), and integrin-α6ß4, which maintain stability between the dermis and epidermis. We designed patient-specific Cas9-nuclease- and -nickase-based targeting strategies for reframing a common homozygous deletion in exon 52 of COL17A1 associated with a lack of full-length C17 expression. Subsequent characterization of protein restoration, indel composition, and divergence of DNA and mRNA outcomes after treatment revealed auspicious efficiency, safety, and precision profiles for paired nicking-based COL17A1 editing. Almost 46% of treated primary JEB keratinocytes expressed reframed C17. Reframed COL17A1 transcripts predominantly featured 25- and 37-nt deletions, accounting for >42% of all edits and encoding C17 protein variants that localized accurately to the cell membrane. Furthermore, corrected cells showed accurate shedding of the extracellular 120-kDa C17 domain and improved adhesion capabilities to laminin-332 compared with untreated JEB cells. Three-dimensional (3D) skin equivalents demonstrated accurate and continuous deposition of C17 within the basal membrane zone between epidermis and dermis. Our findings constitute, for the first time, gene-editing-based correction of a COL17A1 mutation and demonstrate the superiority of proximal paired nicking strategies based on Cas9 D10A nickase over wild-type Cas9-based strategies for gene reframing in a clinical context.


Assuntos
Autoantígenos , Epidermólise Bolhosa Juncional , Epidermólise Bolhosa , Colágenos não Fibrilares , Autoantígenos/genética , Desoxirribonuclease I/genética , Epidermólise Bolhosa/metabolismo , Epidermólise Bolhosa Juncional/genética , Epidermólise Bolhosa Juncional/terapia , Homozigoto , Humanos , Laminina/genética , Mutação , Colágenos não Fibrilares/genética , Deleção de Sequência , Colágeno Tipo XVII
4.
Int J Mol Sci ; 24(6)2023 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-36982270

RESUMO

Junctional epidermolysis bullosa (JEB) is a severe blistering skin disease caused by mutations in genes encoding structural proteins essential for skin integrity. In this study, we developed a cell line suitable for gene expression studies of the JEB-associated COL17A1 encoding type XVII collagen (C17), a transmembrane protein involved in connecting basal keratinocytes to the underlying dermis of the skin. Using the CRISPR/Cas9 system of Streptococcus pyogenes we fused the coding sequence of GFP to COL17A1 leading to the constitutive expression of GFP-C17 fusion proteins under the control of the endogenous promoter in human wild-type and JEB keratinocytes. We confirmed the accurate full-length expression and localization of GFP-C17 to the plasma membrane via fluorescence microscopy and Western blot analysis. As expected, the expression of GFP-C17mut fusion proteins in JEB keratinocytes generated no specific GFP signal. However, the CRISPR/Cas9-mediated repair of a JEB-associated frameshift mutation in GFP-COL17A1mut-expressing JEB cells led to the restoration of GFP-C17, apparent in the full-length expression of the fusion protein, its accurate localization within the plasma membrane of keratinocyte monolayers as well as within the basement membrane zone of 3D-skin equivalents. Thus, this fluorescence-based JEB cell line provides the potential to serve as a platform to screen for personalized gene editing molecules and applications in vitro and in appropriate animal models in vivo.


Assuntos
Epidermólise Bolhosa Juncional , Epidermólise Bolhosa , Animais , Humanos , Epidermólise Bolhosa Juncional/genética , Edição de Genes , Pele , Mutação , Queratinócitos , Epidermólise Bolhosa/genética
5.
Int J Mol Sci ; 24(5)2023 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-36901775

RESUMO

Mutations in the COL7A1 gene lead to malfunction, reduction or complete absence of type VII collagen (C7) in the skin's basement membrane zone (BMZ), impairing skin integrity. In epidermolysis bullosa (EB), more than 800 mutations in COL7A1 have been reported, leading to the dystrophic form of EB (DEB), a severe and rare skin blistering disease associated with a high risk of developing an aggressive form of squamous cell carcinoma. Here, we leveraged a previously described 3'-RTMS6m repair molecule to develop a non-viral, non-invasive and efficient RNA therapy to correct mutations within COL7A1 via spliceosome-mediated RNA trans-splicing (SMaRT). RTM-S6m, cloned into a non-viral minicircle-GFP vector, is capable of correcting all mutations occurring between exon 65 and exon 118 of COL7A1 via SMaRT. Transfection of the RTM into recessive dystrophic EB (RDEB) keratinocytes resulted in a trans-splicing efficiency of ~1.5% in keratinocytes and ~0.6% in fibroblasts, as confirmed on mRNA level via next-generation sequencing (NGS). Full-length C7 protein expression was primarily confirmed in vitro via immunofluorescence (IF) staining and Western blot analysis of transfected cells. Additionally, we complexed 3'-RTMS6m with a DDC642 liposomal carrier to deliver the RTM topically onto RDEB skin equivalents and were subsequently able to detect an accumulation of restored C7 within the basement membrane zone (BMZ). In summary, we transiently corrected COL7A1 mutations in vitro in RDEB keratinocytes and skin equivalents derived from RDEB keratinocytes and fibroblasts using a non-viral 3'-RTMS6m repair molecule.


Assuntos
Epidermólise Bolhosa Distrófica , Epidermólise Bolhosa , Humanos , Trans-Splicing , Pele/metabolismo , Epidermólise Bolhosa Distrófica/genética , Epidermólise Bolhosa/genética , Queratinócitos/metabolismo , Colágeno Tipo VII/genética , Mutação
6.
Int J Mol Sci ; 23(2)2022 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-35055192

RESUMO

Despite a significant rise in the incidence of cutaneous squamous cell carcinoma (SCC) in recent years, most SCCs are well treatable. However, against the background of pre-existing risk factors such as immunosuppression upon organ transplantation, or conditions such as recessive dystrophic epidermolysis bullosa (RDEB), SCCs arise more frequently and follow a particularly aggressive course. Notably, such SCC types display molecular similarities, despite their differing etiologies. We leveraged the similarities in transcriptomes between tumors from organ transplant recipients and RDEB-patients, augmented with data from more common head and neck (HN)-SCCs, to identify drugs that can be repurposed to treat these SCCs. The in silico approach used is based on the assumption that SCC-derived transcriptome profiles reflect critical tumor pathways that, if reversed towards healthy tissue, will attenuate the malignant phenotype. We determined tumor-specific signatures based on differentially expressed genes, which were then used to mine drug-perturbation data. By leveraging recent efforts in the systematic profiling and cataloguing of thousands of small molecule compounds, we identified drugs including selumetinib that specifically target key molecules within the MEK signaling cascade, representing candidates with the potential to be effective in the treatment of these rare and aggressive SCCs.


Assuntos
Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/genética , Biologia Computacional/métodos , Epidermólise Bolhosa Distrófica/complicações , Transplante de Órgãos/efeitos adversos , Neoplasias Cutâneas/genética , Antineoplásicos/uso terapêutico , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/etiologia , Mineração de Dados , Reposicionamento de Medicamentos , Epidermólise Bolhosa Distrófica/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Redes Reguladoras de Genes/efeitos dos fármacos , Humanos , RNA-Seq , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/etiologia
7.
Int J Mol Sci ; 23(3)2022 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-35163654

RESUMO

Mutations within the COL7A1 gene underlie the inherited recessive subtype of the blistering skin disease dystrophic epidermolysis bullosa (RDEB). Although gene replacement approaches for genodermatoses are clinically advanced, their implementation for RDEB is challenging and requires endogenous regulation of transgene expression. Thus, we are using spliceosome-mediated RNA trans-splicing (SMaRT) to repair mutations in COL7A1 at the mRNA level. Here, we demonstrate the capability of a COL7A1-specific RNA trans-splicing molecule (RTM), initially selected using a fluorescence-based screening procedure, to accurately replace COL7A1 exons 1 to 64 in an endogenous setting. Retroviral RTM transduction into patient-derived, immortalized keratinocytes resulted in an increase in wild-type transcript and protein levels, respectively. Furthermore, we revealed accurate deposition of recovered type VII collagen protein within the basement membrane zone of expanded skin equivalents using immunofluorescence staining. In summary, we showed for the first time the potential of endogenous 5' trans-splicing to correct pathogenic mutations within the COL7A1 gene. Therefore, we consider 5' RNA trans-splicing a suitable tool to beneficially modulate the RDEB-phenotype, thus targeting an urgent need of this patient population.


Assuntos
Colágeno Tipo VII/genética , Epidermólise Bolhosa/genética , RNA/metabolismo , Humanos , Splicing de RNA , Trans-Splicing
8.
Int J Mol Sci ; 23(1)2022 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-35008999

RESUMO

Conventional anti-cancer therapies based on chemo- and/or radiotherapy represent highly effective means to kill cancer cells but lack tumor specificity and, therefore, result in a wide range of iatrogenic effects. A promising approach to overcome this obstacle is spliceosome-mediated RNA trans-splicing (SMaRT), which can be leveraged to target tumor cells while leaving normal cells unharmed. Notably, a previously established RNA trans-splicing molecule (RTM44) showed efficacy and specificity in exchanging the coding sequence of a cancer target gene (Ct-SLCO1B3) with the suicide gene HSV1-thymidine kinase in a colorectal cancer model, thereby rendering tumor cells sensitive to the prodrug ganciclovir (GCV). In the present work, we expand the application of this approach, using the same RTM44 in aggressive skin cancer arising in the rare genetic skin disease recessive dystrophic epidermolysis bullosa (RDEB). Stable expression of RTM44, but not a splicing-deficient control (NC), in RDEB-SCC cells resulted in expression of the expected fusion product at the mRNA and protein level. Importantly, systemic GCV treatment of mice bearing RTM44-expressing cancer cells resulted in a significant reduction in tumor volume and weight compared with controls. Thus, our results demonstrate the applicability of RTM44-mediated targeting of the cancer gene Ct-SLCO1B3 in a different malignancy.


Assuntos
Epidermólise Bolhosa Distrófica/complicações , Epidermólise Bolhosa/complicações , Terapia Genética/métodos , Splicing de RNA , Neoplasias Cutâneas/etiologia , Neoplasias Cutâneas/terapia , Trans-Splicing , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Gerenciamento Clínico , Modelos Animais de Doenças , Suscetibilidade a Doenças , Epidermólise Bolhosa/genética , Epidermólise Bolhosa Distrófica/genética , Ganciclovir/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Loci Gênicos , Terapia Genética/efeitos adversos , Humanos , Camundongos , Neoplasias Cutâneas/diagnóstico , Neoplasias Cutâneas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Cell Commun Signal ; 18(1): 61, 2020 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-32276641

RESUMO

BACKGROUND: Cutaneous squamous cell carcinomas (cSCC) are the primary cause of premature deaths in patients suffering from the rare skin-fragility disorder recessive dystrophic epidermolysis bullosa (RDEB), which is in marked contrast to the rarely metastasizing nature of these carcinomas in the general population. This remarkable difference is attributed to the frequent development of chronic wounds caused by impaired skin integrity. However, the specific molecular and cellular changes to malignancy, and whether there are common players in different types of aggressive cSCCs, remain relatively undefined. METHODS: MiRNA expression profiling was performed across various cell types isolated from skin and cSCCs. Microarray results were confirmed by qPCR and by an optimized in situ hybridization protocol. Functional impact of overexpression or knock-out of a dysregulated miRNA was assessed in migration and 3D-spheroid assays. Sample-matched transcriptome data was generated to support the identification of disease relevant miRNA targets. RESULTS: Several miRNAs were identified as dysregulated in cSCCs compared to control skin. These included the metastasis-linked miR-10b, which was significantly upregulated in primary cell cultures and in archival biopsies. At the functional level, overexpression of miR-10b conferred the stem cell-characteristic of 3D-spheroid formation capacity to keratinocytes. Analysis of miR-10b downstream effects identified a novel putative target of miR-10b, the actin- and tubulin cytoskeleton-associated protein DIAPH2. CONCLUSION: The discovery that miR-10b mediates an aspect of cancer stemness - that of enhanced tumor cell adhesion, known to facilitate metastatic colonization - provides an important avenue for future development of novel therapies targeting this metastasis-linked miRNA.


Assuntos
Carcinoma de Células Escamosas , Epidermólise Bolhosa Distrófica/patologia , MicroRNAs/fisiologia , Células-Tronco Neoplásicas , Neoplasias Cutâneas , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Células Cultivadas , Regulação Neoplásica da Expressão Gênica , Humanos , Queratinócitos/metabolismo , Queratinócitos/patologia , Invasividade Neoplásica , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/metabolismo , Cultura Primária de Células , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia
10.
Biol Pharm Bull ; 41(3): 445-449, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29491222

RESUMO

Cancer-type organic anion transporting polypeptide 1B3 (Ct-OATP1B3) mRNA is a variant isoform of the liver-type OATP1B3. Because Ct-OATP1B3 mRNA shows an excellent cancer-specific expression profile in colorectal cancer (CRC), and that its expression levels are associated with CRC prognosis, it holds the potential to become a useful CRC detection and diagnosis biomarker. While the potential is currently justified only at the tissue level, if existence of Ct-OATP1B3 mRNA in CRC-derived extracellular vesicles (EVs) is validated, the findings could enhance its translational potential as a CRC detection and diagnosis biomarker. Therefore, this study aims at proving that Ct-OATP1B3 mRNA exists in CRC-derived EVs, and can be detected using serum specimens. To examine the possibility of Ct-OATP1B3 mRNA being existed in extracellular milieu, we isolated EVs from the human CRC (HCT116, HT-29, and SW480) cell lines, and prepared their cDNAs. The RT-PCR results showed that Ct-OATP1B3 mRNA was clearly present in EVs derived from the human CRC cell lines. Then, in order to further explore the possibility that Ct-OATP1B3 mRNA in CRC-derived EVs can be detected in serum, we isolated serum EVs derived from human CRC xenograft mice, and then performed RT-PCR. The results showed that Ct-OATP1B3 mRNA could be found in all serum EV and CRC tissue samples of the mice examined. Collectively, our findings, which show that Ct-OATP1B3 mRNA exists in EVs and can be detected in (at least) mouse serum, strengthen the potential use of Ct-OATP1B3 mRNA as a serum-based CRC biomarker.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Colorretais/sangue , Vesículas Extracelulares/metabolismo , RNA Mensageiro/sangue , RNA Mensageiro/genética , RNA Neoplásico/sangue , RNA Neoplásico/genética , Membro 1B3 da Família de Transportadores de Ânion Orgânico Carreador de Soluto/genética , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/diagnóstico , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Prognóstico , Reação em Cadeia da Polimerase em Tempo Real , Membro 1B3 da Família de Transportadores de Ânion Orgânico Carreador de Soluto/biossíntese
11.
Int J Mol Sci ; 16(1): 1179-91, 2015 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-25569093

RESUMO

Spliceosome-mediated RNA trans-splicing has become an emergent tool for the repair of mutated pre-mRNAs in the treatment of genetic diseases. RNA trans-splicing molecules (RTMs) are designed to induce a specific trans-splicing reaction via a binding domain for a respective target pre-mRNA region. A previously established reporter-based screening system allows us to analyze the impact of various factors on the RTM trans-splicing efficiency in vitro. Using this system, we are further able to investigate the potential of antisense RNAs (AS RNAs), presuming to improve the trans-splicing efficiency of a selected RTM, specific for intron 102 of COL7A1. Mutations in the COL7A1 gene underlie the dystrophic subtype of the skin blistering disease epidermolysis bullosa (DEB). We have shown that co-transfections of the RTM and a selected AS RNA, interfering with competitive splicing elements on a COL7A1-minigene (COL7A1-MG), lead to a significant increase of the RNA trans-splicing efficiency. Thereby, accurate trans-splicing between the RTM and the COL7A1-MG is represented by the restoration of full-length green fluorescent protein GFP on mRNA and protein level. This mechanism can be crucial for the improvement of an RTM-mediated correction, especially in cases where a high trans-splicing efficiency is required.


Assuntos
Oligonucleotídeos Antissenso/metabolismo , Trans-Splicing , Colágeno Tipo VII/genética , Colágeno Tipo VII/metabolismo , Epidermólise Bolhosa/genética , Epidermólise Bolhosa/patologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Íntrons , Mutação , Transfecção
12.
J Dtsch Dermatol Ges ; 13(12): 1257-66, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26612796

RESUMO

BACKGROUND: A genetic blistering skin disease, recessive dystrophic epidermolysis bullosa (RDEB), is marked by severe wound healing defects and finger contractures. The purpose of this investigation was to elucidate the mechanisms of impaired wound healing and pseudosyndactyly occurring in RDEB patients by studying the role of known inflammation and fibrosis markers in RDEB pseudosyndactyly tissue. PATIENTS AND METHODS: We studied the expression of the fibrosis and/or inflammation markers tenascin-C, α-smooth muscle actin, transforming growth factor-ß1, interleukin-1ß, and interleukin-6 in scarring and nonscarring tissue from healthy donors and RDEB patients by semiquantitative real time-PCR and, where applicable, by immunoblots. Furthermore, the distribution pattern of α-smooth muscle actin and tenascin-C were assessed by immunofluorescence microscopy. RESULTS: Based on mRNA and protein analysis, we found upregulation of tenascin-C, interleukin-1ß, and interleukin-6 - but not of transforming growth factor-ß1 - in recessive dystrophic epidermolysis bullosa scar samples taken from pseudosyndactyly hands. Unexpectedly, α-smooth muscle actin was not upregulated. CONCLUSIONS: Our results confirm inflammation and fibrosis in recessive dystrophic epidermolysis bullosa, especially in scars, suggesting major roles for these processes in pseudosyndactyly. Our data therefore suggests the potential use of antiinflammatory and antifibrotic drugs in the prevention of pseudosyndactyly.


Assuntos
Dermatite/imunologia , Epidermólise Bolhosa Distrófica/imunologia , Deformidades Adquiridas da Mão/imunologia , Pele/imunologia , Pele/patologia , Cicatrização/imunologia , Adolescente , Adulto , Idoso , Citocinas/imunologia , Dermatite/patologia , Epidermólise Bolhosa Distrófica/patologia , Feminino , Fibrose/imunologia , Fibrose/patologia , Humanos , Fatores Imunológicos/imunologia , Masculino , Pessoa de Meia-Idade , Adulto Jovem
14.
Adv Drug Deliv Rev ; 208: 115294, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38527624

RESUMO

Genodermatoses represent a large group of inherited skin disorders encompassing clinically-heterogeneous conditions that manifest in the skin and other organs. Depending on disease variant, associated clinical manifestations and secondary complications can severely impact patients' quality of life and currently available treatments are transient and not curative. Multiple emerging approaches using CRISPR-based technologies offer promising prospects for therapy. Here, we explore current advances and challenges related to gene editing in rare skin diseases, including different strategies tailored to mutation type and structural organization of the affected gene, considerations for in vivo and ex vivo applications, the critical issue of delivery into the skin, and immune aspects of therapy. Against the backdrop of a landmark FDA approval for the first re-dosable gene replacement therapy for a rare genetic skin disorder, gene editing approaches are inching closer to the clinics and the possibility of a local permanent cure for patients affected by these disorders.


Assuntos
Edição de Genes , Dermatopatias , Humanos , Sistemas CRISPR-Cas/genética , Qualidade de Vida , Pele , Dermatopatias/genética , Dermatopatias/terapia
15.
Stem Cell Res ; 75: 103306, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38271763

RESUMO

We have generated MLi005-A, a new induced pluripotent stem cell (iPSC) line derived from skin fibroblasts of a male patient with dominant dystrophic epidermolysis bullosa (DDEB). This iPSC line may be used as a model system for studies on skin integrity, the extracellular matrix and skin barrier function. The characterization of the MLi005-A cell line consisted of molecular karyotyping, next-generation sequencing of the COL7A1 alleles, pluripotency and differentiation potentials testing by immunofluorescence of associated markers in vitro. The MLi-005A line has been also tested for ability to differentiate into fibroblasts and keratinocytes and markers associated with these cell types.


Assuntos
Epidermólise Bolhosa Distrófica , Células-Tronco Pluripotentes Induzidas , Humanos , Masculino , Epidermólise Bolhosa Distrófica/genética , Epidermólise Bolhosa Distrófica/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Colágeno Tipo VII/genética , Colágeno Tipo VII/metabolismo , Pele/metabolismo , Queratinócitos/metabolismo
16.
Nucleic Acids Res ; 39(16): e108, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21685452

RESUMO

Trans-splicing is a powerful approach to reprogram the genome. It can be used to replace 5', 3' or internal exons. The latter approach has been characterized by low efficiency, as the requirements to promote internal trans-splicing are largely uncharacterized. The trans-splicing process is induced by engineered 'RNA trans-splicing molecules' (RTMs), which target a selected pre-mRNA to be reprogrammed via two complementary binding domains. To facilitate the development of more efficient RTMs for therapeutic applications we constructed a novel fluorescence based screening system. We incorporated exon 52 of the COL17A1 gene into a GFP-based cassette system as the target exon. This exon is mutated in many patients with the devastating skin blistering disease epidermolysis bullosa. In a double transfection assay we were able to rapidly identify optimal binding domains targeted to sequences in the surrounding introns 51 and 52. The ability to replace exon 52 was then evaluated in a more endogenous context using a target containing COL17A1 exon 51-intron 51-exon 52-intron 52-exon 53. Two selected RTMs produced significantly higher levels of GFP expression in up to 61% assayed cells. This novel approach allows for rapid identification of efficient RTMs for internal exon replacement.


Assuntos
Éxons , Trans-Splicing , Autoantígenos/genética , Western Blotting , Linhagem Celular , Citometria de Fluxo , Corantes Fluorescentes , Genes Reporter , Técnicas Genéticas , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Células HEK293 , Humanos , Colágenos não Fibrilares/genética , RNA Mensageiro/metabolismo , Colágeno Tipo XVII
17.
J Anim Sci ; 1012023 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-37220904

RESUMO

Animal feeds are often contaminated with ochratoxin A (OTA), a potent natural mycotoxin hazardous to animal and human health that accumulates in blood and tissues. To the best of our knowledge, this study is the first to investigate the in vivo application of an enzyme (OTA amidohydrolase; OAH) that degrades OTA into the nontoxic molecules phenylalanine and ochratoxin α (OTα) in the gastrointestinal tract (GIT) of pigs. Piglets were fed six experimental diets over 14 days, varying in OTA contamination level (50 or 500 µg/kg; OTA50 and OTA500) and presence of OAH; a negative control diet (no OTA added) and a diet containing OTα at 318 µg/kg (OTα318). The absorption of OTA and OTα into the systemic circulation (plasma and dried blood spots, DBS), their accumulation in kidney, liver, and muscle tissues, and excretion through feces and urine were assessed. The efficiency of OTA degradation in the digesta content of the GIT was also estimated. At the end of the trial, accumulation of OTA in blood was significantly higher in OTA groups (OTA50 and OTA500) in comparison to enzyme groups (OAH50 and OAH500, respectively). The supplementation of OAH explicitly reduced the absorption of OTA (P < 0.005) into plasma by 54% and 59% (from 40.53 ± 3.53 to 18.66 ± 2.28 ng/mL in piglets fed the 50 µg OTA/kg diets and from 413.50 ± 71.88 to 168.35 ± 41.02 ng/mL in piglets fed the 500 µg OTA/kg diets, respectively) and in DBS by 50% and 53% (from 22.79 ± 2.63 to 10.67 ± 1.93 ng/mL in piglets fed the 50 µg OTA/kg diets and from 232.85 ± 35.16 to 105.71 ± 24.18 ng/mL in piglets fed the 500 µg OTA/kg diets, respectively). The OTA concentrations in plasma were positively associated with the OTA levels detected in all tissues analyzed; adding OAH reduced OTA levels in the kidney, liver, and muscle (P < 0.005) by 52%, 67%, and 59%, respectively. The analysis of GIT digesta content showed that OAH supplementation led to OTA degradation in the proximal GIT where natural hydrolysis is inefficient. Overall, the data of present in vivo study demonstrated that supplementation of swine feeds with OAH successfully reduced OTA levels in blood (plasma and DBS) as well as in kidney, liver, and muscle tissues. Therefore, an approach to use enzymes as feed additives might be most promising to mitigate the harmful effects of OTA on the productivity and welfare of pigs and at the same time improving the safety of pig-derived food products.


Ochratoxin A (OTA) is a potent toxin frequently present in animal feeds, which accumulates in the animal tissues for human consumption. This results in critical animal welfare issues, as well as food safety issues. To the best of our knowledge, the present study is the first to show that OTA can be degraded by an enzyme supplemented with pigs' feed. In the pig gut, this enzyme successfully breaks down the toxic OTA molecule into a nontoxic form that is excreted through urine and feces, which is applicable in supporting the pig's health, welfare, and productivity as well as assuring the safety of the pig meat for human consumption.


Assuntos
Micotoxinas , Humanos , Animais , Suínos , Micotoxinas/toxicidade , Dieta/veterinária , Rim/metabolismo , Trato Gastrointestinal/metabolismo , Ração Animal/análise
18.
Animals (Basel) ; 13(3)2023 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-36766220

RESUMO

Probiotic feed additives can support the gut health of shrimp and thereby improve performance, production efficiency and disease resistance. Two experiments in white leg shrimp aimed to investigate the effects of a multi-species probiotic feed supplement (AquaStar®, 3 g/kg feed, Biomin GmbH, Getzersdorf, Austria) in feed formulations with different marine meal levels (32% and 15%) on growth performance and resistance against Vibrio parahaemolyticus. Juvenile shrimp were stocked in a recirculating aquaculture tank system at a density of 20 shrimp/46.8 L and were fed diets with and without the probiotic supplementation for 8 weeks. Afterwards, a bath immersion with V. parahaemolyticus was performed and mortality was observed over a period of 14 days. Independent of the diet formulation, probiotic supplementation significantly improved the survival rate of the shrimp and the specific growth rate while decreasing feed consumption and feed conversion ratio when compared to the control (p ≤ 0.042). After the Vibrio immersion challenge, mortality was significantly decreased by 13.33% with probiotic supplementation in the high marine meal diet experiment (p = 0.042) and numerically decreased by 11.67% in the low marine meal diet experiment (p = 0.133). Overall, the results suggest that the beneficial effects of the probiotic can occur independently of the diet formulation.

19.
Cancers (Basel) ; 15(13)2023 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-37444397

RESUMO

Machine learning has been proven to be a powerful tool in the identification of diagnostic tumor biomarkers but is often impeded in rare cancers due to small patient numbers. In patients suffering from recessive dystrophic epidermolysis bullosa (RDEB), early-in-life development of particularly aggressive cutaneous squamous-cell carcinomas (cSCCs) represents a major threat and timely detection is crucial to facilitate prompt tumor excision. As miRNAs have been shown to hold great potential as liquid biopsy markers, we characterized miRNA signatures derived from cultured primary cells specific for the potential detection of tumors in RDEB patients. To address the limitation in RDEB-sample accessibility, we analyzed the similarity of RDEB miRNA profiles with other tumor entities derived from the Cancer Genome Atlas (TCGA) repository. Due to the similarity in miRNA expression with RDEB-SCC, we used HN-SCC data to train a tumor prediction model. Three models with varying complexity using 33, 10 and 3 miRNAs were derived from the elastic net logistic regression model. The predictive performance of all three models was determined on an independent HN-SCC test dataset (AUC-ROC: 100%, 83% and 96%), as well as on cell-based RDEB miRNA-Seq data (AUC-ROC: 100%, 100% and 91%). In addition, the ability of the models to predict tumor samples based on RDEB exosomes (AUC-ROC: 100%, 93% and 100%) demonstrated the potential feasibility in a clinical setting. Our results support the feasibility of this approach to identify a diagnostic miRNA signature, by exploiting publicly available data and will lay the base for an improvement of early RDEB-SCC detection.

20.
Exp Dermatol ; 21(7): 526-30, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22716248

RESUMO

Epidermolysis bullosa (EB) is a group of hereditary skin disorders. Although each subtype is caused by mutations in genes encoding differentially located components of the skin, the resulting phenotype is similar. In this study, we investigated similarities in the gene expression profiles of each subtype on mRNA level. Type XVI collagen (COL16A1), G0/G1 switch 2 (G0S2), fibronectin (FN1), ribosomal protein S27A (RPS27A) and low density lipoprotein receptor (LDLR) were shown to exhibit corresponding changes in gene expression in all three EB subtypes. While COL16A1, G0S2 and FN1 are up-regulated, LDLR and RPS27A mRNA levels are decreased. These data indicate that EB cells seem to take measures increasing their mechanical stability. Apoptosis is likely to be exacerbated, and migratory potential appears to be elevated. Protein degradation is hampered, and the release of fatty acids and glycerol is restricted, probably to save energy. These commonalities might benefit existing EB treatment strategies or could help to reveal new starting points for the treatment of EB in the future.


Assuntos
Epidermólise Bolhosa Distrófica/genética , Epidermólise Bolhosa Simples/genética , Epidermólise Bolhosa Juncional/genética , Expressão Gênica , Proteínas de Ciclo Celular/genética , Células Cultivadas , Colágeno/genética , Regulação para Baixo , Fibronectinas/genética , Humanos , Queratinócitos , Distrofias Musculares/genética , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptores de LDL/genética , Proteínas Ribossômicas/genética , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA