Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
bioRxiv ; 2024 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-38496616

RESUMEN

Raf kinases play vital roles in normal mitogenic signaling and cancer, however, the identities of functionally important Raf-proximal proteins throughout the cell are not fully known. Raf1 proximity proteomics/BioID in Raf1-dependent cancer cells unexpectedly identified Raf1-adjacent proteins known to reside in the mitochondrial matrix. Inner-mitochondrial localization of Raf1 was confirmed by mitochondrial purification and super-resolution microscopy. Inside mitochondria, Raf1 associated with glutaminase (GLS) in diverse human cancers and enabled glutaminolysis, an important source of biosynthetic precursors in cancer. These impacts required Raf1 kinase activity and were independent of canonical MAP kinase pathway signaling. Kinase-dead mitochondrial matrix-localized Raf1 impaired glutaminolysis and tumorigenesis in vivo. These data indicate that Raf1 localizes inside mitochondria where it interacts with GLS to engage glutamine catabolism and support tumorigenesis.

2.
Orphanet J Rare Dis ; 17(1): 377, 2022 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-36253825

RESUMEN

BACKGROUND: Recessive dystrophic epidermolysis bullosa (RDEB) is a rare, devastating blistering genodermatosis caused by mutations in the COL7A1 gene, which encodes for type VII collagen and is necessary for dermal-epidermal adhesion and integrity. Disease manifestations include severe and debilitating wounds, aggressive squamous cell carcinomas, and premature death; however, there are currently no approved therapies. This Phase 1/2a, open-label study evaluated the long-term efficacy and safety of gene-corrected autologous keratinocyte grafts (EB-101) for chronic RDEB wounds. METHODS: Autologous keratinocytes were harvested from participants with severe RDEB, transduced with a retrovirus containing the full-length COL7A1 gene, and grown into 5 × 7 cm (35 cm2) sheets. Gene-corrected keratinocyte sheets were then transplanted onto chronic RDEB wounds present for ≥ 12 weeks. RESULTS: Seven adult participants with severe RDEB were grafted with six sheets each (42 total sheets) onto wounds and followed for a mean of 5.9 years (range 4-8 years). Long-term improvements in wound healing and symptoms were observed. At year five, 70% (21/30) of treated sites demonstrated ≥ 50% wound healing compared to baseline by investigator global assessment. No sites with ≥ 50% wound healing were painful or pruritic, compared to 67% (6/9) of sites with < 50% wound healing (p < 0.001) at year five. Grafts were well-tolerated throughout long-term follow-up. No serious adverse events related to treatment were reported over a mean of 5.9 years of follow-up. No persistent systemic autoimmunity against type VII collagen or replication-competent retrovirus infections were identified, and no participants developed squamous cell carcinomas related to treatment during long-term follow-up. CONCLUSIONS: Treatment with EB-101 appears safe and efficacious, and produces long-term improvements in wound healing, pain, and itch for RDEB patients. Results from the Phase 3 randomized controlled trial are forthcoming. TRIAL REGISTRATION: ClinicalTrials.gov, NCT01263379. Registered December 15, 2010. https://clinicaltrials.gov/ct2/show/NCT01263379.


Asunto(s)
Carcinoma de Células Escamosas , Epidermólisis Ampollosa Distrófica , Adulto , Humanos , Colágeno Tipo VII/genética , Colágeno Tipo VII/metabolismo , Epidermólisis Ampollosa Distrófica/genética , Epidermólisis Ampollosa Distrófica/patología , Queratinocitos/metabolismo , Cicatrización de Heridas/genética
3.
Nat Methods ; 19(8): 959-968, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35927480

RESUMEN

DNA-protein interactions mediate physiologic gene regulation and may be altered by DNA variants linked to polygenic disease. To enhance the speed and signal-to-noise ratio (SNR) in the identification and quantification of proteins associated with specific DNA sequences in living cells, we developed proximal biotinylation by episomal recruitment (PROBER). PROBER uses high-copy episomes to amplify SNR, and proximity proteomics (BioID) to identify the transcription factors and additional gene regulators associated with short DNA sequences of interest. PROBER quantified both constitutive and inducible association of transcription factors and corresponding chromatin regulators to target DNA sequences and binding quantitative trait loci due to single-nucleotide variants. PROBER identified alterations in regulator associations due to cancer hotspot mutations in the hTERT promoter, indicating that these mutations increase promoter association with specific gene activators. PROBER provides an approach to rapidly identify proteins associated with specific DNA sequences and their variants in living cells.


Asunto(s)
Cromatina , ADN , Biotinilación , Cromatina/genética , ADN/genética , ADN/metabolismo , Plásmidos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
4.
Oncogene ; 40(44): 6299-6307, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34584216

RESUMEN

Collagens are the most abundant proteins in the body and comprise the basement membranes and stroma through which cancerous invasion occurs; however, a pro-neoplastic function for mutant collagens is undefined. Here we identify COL11A1 mutations in 66 of 100 cutaneous squamous cell carcinomas (cSCCs), the second most common U.S. cancer, concentrated in a triple helical region known to produce trans-dominant collagens. Analysis of COL11A1 and other collagen genes found that they are mutated across common epithelial malignancies. Knockout of mutant COL11A1 impairs cSCC tumorigenesis in vivo. Compared to otherwise genetically identical COL11A1 wild-type tissue, gene-edited mutant COL11A1 skin is characterized by induction of ß1 integrin targets and accelerated neoplastic invasion. In mosaic tissue, mutant COL11A1 cells enhanced invasion by neighboring wild-type cells. These results suggest that specific collagens are commonly mutated in cancer and that mutant collagens may accelerate this process.


Asunto(s)
Carcinoma de Células Escamosas/patología , Colágeno Tipo XI/genética , Integrina beta1/metabolismo , Mutación , Neoplasias Cutáneas/patología , Animales , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Estudios de Casos y Controles , Colágeno Tipo XI/química , Femenino , Humanos , Ratones , Invasividad Neoplásica , Trasplante de Neoplasias , Estructura Secundaria de Proteína , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Secuenciación del Exoma
5.
Nat Commun ; 10(1): 5118, 2019 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-31712554

RESUMEN

KRAS receives and relays signals at the plasma membrane (PM) where it transmits extracellular growth factor signals to downstream effectors. SNORD50A/B were recently found to bind KRAS and inhibit its tumorigenic action by unknown mechanisms. KRAS proximity protein labeling was therefore undertaken in SNORD50A/B wild-type and knockout cells, revealing that SNORD50A/B RNAs shape the composition of proteins proximal to KRAS, notably by inhibiting KRAS proximity to the SNARE vesicular transport proteins SNAP23, SNAP29, and VAMP3. To remain enriched on the PM, KRAS undergoes cycles of endocytosis, solubilization, and vesicular transport to the PM. Here we report that SNAREs are essential for the final step of this process, with KRAS localization to the PM facilitated by SNAREs but antagonized by SNORD50A/B. Antagonism between SNORD50A/B RNAs and specific SNARE proteins thus controls KRAS localization, signaling, and tumorigenesis, and disrupting SNARE-enabled KRAS function represents a potential therapeutic opportunity in KRAS-driven cancer.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Proteínas Proto-Oncogénicas p21(ras)/genética , ARN Pequeño no Traducido/metabolismo , Proteínas SNARE/metabolismo , Animales , Línea Celular Tumoral , Membrana Celular/metabolismo , Endocitosis , Endosomas/metabolismo , Humanos , Ratones , Neoplasias/metabolismo , Neoplasias/patología , Unión Proteica , Transporte de Proteínas , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , ARN Pequeño no Traducido/genética , Transducción de Señal
6.
JCI Insight ; 4(19)2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31578311

RESUMEN

BACKGROUNDRecessive dystrophic epidermolysis bullosa (RDEB) patients have mutations in the COL7A1 gene and thus lack functional type VII collagen (C7) protein; they have marked skin fragility and blistering. This single-center phase 1/2a open-label study evaluated the long-term efficacy, safety, and patient-reported outcomes in RDEB patients treated with gene-corrected autologous cell therapy.METHODSAutologous keratinocytes were isolated from participant skin biopsies. Epidermal sheets were prepared from cells transduced with a retrovirus carrying the full-length human COL7A1 gene. These gene-corrected autologous epidermal sheets measured 5 × 7 cm (35 cm2) and were transplanted onto 6 wound sites in each of 7 adult participants (n = 42 sites total) from 2013 to 2017. Participants were followed for 2 to 5 years.RESULTSNo participants experienced any serious related adverse events. Wound healing of 50% or greater by Investigator Global Assessment was present in 95% (36 of 38) of treated wounds versus 0% (0 of 6) of untreated control wounds at 6 months (P < 0.0001). At year 1, 68% (26 of 38) of treated wounds had 50% or greater healing compared with 17% (1 of 6) of control wounds (P = 0.025). At year 2, 71% (27 of 38) of treated wounds had 50% or greater healing compared with 17% (1 of 6) of control wounds (P = 0.019).CONCLUSIONC7 expression persisted up to 2 years after treatment in 2 participants. Treated wounds with 50% or greater healing demonstrated improvement in patient-reported pain, itch, and wound durability. This study provides additional data to support the clinically meaningful benefit of treating chronic RDEB wounds with ex vivo, C7 gene-corrected autologous cell therapy. This approach was safe and promoted wound healing that was associated with improved patient-reported outcomes.TRIAL REGISTRATIONClinicaltrials.gov identifier: NCT01263379.FUNDINGEpidermolysis Bullosa Research Partnership, Epidermolysis Bullosa Medical Research Foundation, NIH R01 AR055914, Office of Research and Development at the Palo Alto Veteran's Affairs Medical Center, and the Dermatology Foundation.


Asunto(s)
Epidermólisis Ampollosa Distrófica/genética , Epidermólisis Ampollosa Distrófica/terapia , Terapia Genética/métodos , Adolescente , Biopsia , Tratamiento Basado en Trasplante de Células y Tejidos , Niño , Preescolar , Colágeno Tipo VII/genética , Epidermólisis Ampollosa Distrófica/patología , Femenino , Humanos , Queratinocitos , Masculino , Mutación , Piel/patología , Cicatrización de Heridas , Adulto Joven
7.
Nat Methods ; 15(3): 207-212, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29400715

RESUMEN

RNA-protein interactions play numerous roles in cellular function and disease. Here we describe RNA-protein interaction detection (RaPID), which uses proximity-dependent protein labeling, based on the BirA* biotin ligase, to rapidly identify the proteins that bind RNA sequences of interest in living cells. RaPID displays utility in multiple applications, including in evaluating protein binding to mutant RNA motifs in human genetic disorders, in uncovering potential post-transcriptional networks in breast cancer, and in discovering essential host proteins that interact with Zika virus RNA. To improve the BirA*-labeling component of RaPID, moreover, a new mutant BirA* was engineered from Bacillus subtilis, termed BASU, that enables >1,000-fold faster kinetics and >30-fold increased signal-to-noise ratio over the prior standard Escherichia coli BirA*, thereby enabling direct study of RNA-protein interactions in living cells on a timescale as short as 1 min.


Asunto(s)
Biotina/química , Proteínas de Unión al ARN/metabolismo , ARN/metabolismo , Proteínas Virales/metabolismo , Virus Zika/metabolismo , Bacillus subtilis/metabolismo , Ligasas de Carbono-Nitrógeno/genética , Ligasas de Carbono-Nitrógeno/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Células HEK293 , Humanos , Neuronas/citología , Neuronas/metabolismo , ARN/química , ARN/genética , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteínas Virales/química , Proteínas Virales/genética , Virus Zika/genética
8.
Oncotarget ; 8(12): 18657-18669, 2017 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-28423633

RESUMEN

Clear cell renal cell carcinomas (ccRCC) show a broad range of clinical behavior, and prognostic biomarkers are needed to stratify patients for appropriate management. We sought to determine whether long intergenic non-coding RNAs (lincRNAs) might predict patient survival. Candidate prognostic lincRNAs were identified by mining The Cancer Genome Atlas (TCGA) transcriptome (RNA-seq) data on 466 ccRCC cases (randomized into discovery and validation sets) annotated for ~21,000 lncRNAs. A previously uncharacterized lincRNA, SLINKY (Survival-predictive LINcRNA in KidneY cancer), was the top-ranked prognostic lincRNA, and validated in an independent University of Tokyo cohort (P=0.004). In multivariable analysis, SLINKY expression predicted overall survival independent of tumor stage and grade [TCGA HR=3.5 (CI, 2.2-5.7), P < 0.001; Tokyo HR=8.4 (CI, 1.8-40.2), P = 0.007], and by decision tree, ROC and decision curve analysis, added independent prognostic value. In ccRCC cell lines, SLINKY knockdown reduced cancer cell proliferation (with cell-cycle G1 arrest) and induced transcriptome changes enriched for cell proliferation and survival processes. Notably, the genes affected by SLINKY knockdown in cell lines were themselves prognostic and correlated with SLINKY expression in the ccRCC patient samples. From a screen for binding partners, we identified direct binding of SLINKY to Heterogeneous Nuclear Ribonucleoprotein K (HNRNPK), whose knockdown recapitulated SLINKY knockdown phenotypes. Thus, SLINKY is a robust prognostic biomarker in ccRCC, where it functions possibly together with HNRNPK in cancer cell proliferation.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma de Células Renales/patología , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias Renales/patología , ARN Largo no Codificante/genética , Adulto , Anciano , Área Bajo la Curva , Biomarcadores de Tumor/análisis , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/mortalidad , Proliferación Celular , Supervivencia sin Enfermedad , Femenino , Citometría de Flujo , Ribonucleoproteína Heterogénea-Nuclear Grupo K/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo K/metabolismo , Humanos , Inmunoprecipitación , Estimación de Kaplan-Meier , Neoplasias Renales/genética , Neoplasias Renales/mortalidad , Masculino , Persona de Mediana Edad , Pronóstico , Modelos de Riesgos Proporcionales , Análisis por Matrices de Proteínas , Curva ROC , Sensibilidad y Especificidad
9.
JAMA ; 316(17): 1808-1817, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27802546

RESUMEN

Importance: Recessive dystrophic epidermolysis bullosa (RDEB) is a devastating, often fatal, inherited blistering disorder caused by mutations in the COL7A1 gene encoding type VII collagen. Support and palliation are the only current therapies. Objective: To evaluate the safety and wound outcomes following genetically corrected autologous epidermal grafts in patients with RDEB. Design, Setting, and Participants: Single-center phase 1 clinical trial conducted in the United States of 4 patients with severe RDEB with a measured area of wounds suitable for grafting of at least 100 cm2. Patients with undetectable type VII collagen keratinocyte expression were excluded. Interventions: Autologous keratinocytes isolated from biopsy samples collected from 4 patients with RDEB were transduced with good manufacturing practice-grade retrovirus carrying full-length human COL7A1 and assembled into epidermal sheet grafts. Type VII collagen gene-corrected grafts (approximately 35 cm2) were transplanted onto 6 wounds in each of the patients (n = 24 grafts). Main Outcomes and Measures: The primary safety outcomes were recombination competent retrovirus, cancer, and autoimmune reaction. Molecular correction was assessed as type VII collagen expression measured by immunofluorescence and immunoelectron microscopy. Wound healing was assessed using serial photographs taken at 3, 6, and 12 months after grafting. Results: The 4 patients (mean age, 23 years [range, 18-32 years]) were all male with an estimated body surface area affected with RDEB of 4% to 30%. All 24 grafts were well tolerated without serious adverse events. Type VII collagen expression at the dermal-epidermal junction was demonstrated on the graft sites by immunofluorescence microscopy in 9 of 10 biopsy samples (90%) at 3 months, in 8 of 12 samples (66%) at 6 months, and in 5 of 12 samples (42%) at 12 months, including correct type VII collagen localization to anchoring fibrils. Wounds with recombinant type VII collagen graft sites displayed 75% or greater healing at 3 months (21 intact graft sites of 24 wound sites; 87%), 6 months (16/24; 67%), and 12 months (12/24; 50%) compared with baseline wound sites. Conclusions and Relevance: In this preliminary study of 4 patients with RDEB, there was wound healing in some type VII collagen gene-corrected grafts, but the response was variable among patients and among grafted sites and generally declined over 1 year. Long-term follow-up is necessary for these patients, and controlled trials are needed with a broader range of patients to better understand the potential long-term efficacy of genetically corrected autologous epidermal grafts. Trial Registration: clinicaltrials.gov Identifier: NCT01263379.


Asunto(s)
Colágeno Tipo VII/genética , Epidermólisis Ampollosa Distrófica/terapia , Técnicas de Transferencia de Gen , Queratinocitos/trasplante , Cicatrización de Heridas , Adolescente , Adulto , Colágeno Tipo VII/metabolismo , Colágeno Tipo VII/uso terapéutico , Epidermólisis Ampollosa Distrófica/genética , Epidermólisis Ampollosa Distrófica/metabolismo , Epidermólisis Ampollosa Distrófica/patología , Humanos , Masculino , Virus de la Leucemia Murina de Moloney/genética , Pirimidinas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapéutico , Colgajos Quirúrgicos , Factores de Tiempo , Heridas y Lesiones/metabolismo , Heridas y Lesiones/terapia , Adulto Joven
10.
Nat Genet ; 48(1): 53-8, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26595770

RESUMEN

Small nucleolar RNAs (snoRNAs) are conserved noncoding RNAs best studied as ribonucleoprotein (RNP) guides in RNA modification. To explore their role in cancer, we compared 5,473 tumor-normal genome pairs to identify snoRNAs with frequent copy number loss. The SNORD50A-SNORD50B snoRNA locus was deleted in 10-40% of 12 common cancers, where its loss was associated with reduced survival. A human protein microarray screen identified direct SNORD50A and SNORD50B RNA binding to K-Ras. Loss of SNORD50A and SNORD50B increased the amount of GTP-bound, active K-Ras and hyperactivated Ras-ERK1/ERK2 signaling. Loss of these snoRNAs also increased binding by farnesyltransferase to K-Ras and increased K-Ras prenylation, suggesting that KRAS mutation might synergize with SNORD50A and SNORD50B loss in cancer. In agreement with this hypothesis, CRISPR-mediated deletion of SNORD50A and SNORD50B in KRAS-mutant tumor cells enhanced tumorigenesis, and SNORD50A and SNORD50B deletion and oncogenic KRAS mutation co-occurred significantly in multiple human tumor types. SNORD50A and SNORD50B snoRNAs thus directly bind and inhibit K-Ras and are recurrently deleted in human cancer.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , ARN no Traducido/genética , ARN no Traducido/metabolismo , Proteínas ras/metabolismo , Animales , Línea Celular Tumoral , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Femenino , Eliminación de Gen , Guanosina Trifosfato/metabolismo , Humanos , Ratones Endogámicos NOD , Mutación , Neoplasias/mortalidad , Prenilación , ARN Nucleolar Pequeño/genética , ARN Nucleolar Pequeño/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas ras/genética
11.
Genes Dev ; 29(21): 2225-30, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26545810

RESUMEN

Outward migration of epidermal progenitors occurs with induction of hundreds of differentiation genes, but the identities of all regulators required for this process are unknown. We used laser capture microdissection followed by RNA sequencing to identify calmodulin-like 5 (CALML5) as the most enriched gene in differentiating outer epidermis. CALML5 mRNA was up-regulated by the ZNF750 transcription factor and then stabilized by the long noncoding RNA TINCR. CALML5 knockout impaired differentiation, abolished keratohyalin granules, and disrupted epidermal barrier function. Mass spectrometry identified SFN (stratifin/14-3-3σ) as a CALML5-binding protein. CALML5 interacts with SFN in suprabasal epidermis, cocontrols 13% of late differentiation genes, and modulates interaction of SFN to some of its binding partners. A ZNF750-TINCR-CALML5-SFN network is thus essential for epidermal differentiation.


Asunto(s)
Proteínas 14-3-3/metabolismo , Biomarcadores de Tumor/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Diferenciación Celular/genética , Células Epidérmicas , Exorribonucleasas/metabolismo , ARN no Traducido/metabolismo , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Fosfoproteínas/metabolismo , Unión Proteica , Transporte de Proteínas , Células Madre/citología , Proteínas Supresoras de Tumor , Proteínas Señalizadoras YAP
12.
Dev Cell ; 32(6): 693-706, 2015 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-25805135

RESUMEN

Progenitor differentiation requires remodeling of genomic expression; however, in many tissues, such as epidermis, the spectrum of remodeled genes and the transcription factors (TFs) that control them are not fully defined. We performed kinetic transcriptome analysis during regeneration of differentiated epidermis and identified gene sets enriched in progenitors (594 genes), in early (159 genes), and in late differentiation (387 genes). Module mapping of 1,046 TFs identified MAF and MAFB as necessary and sufficient for progenitor differentiation. MAF:MAFB regulated 393 genes altered in this setting. Integrative analysis identified ANCR and TINCR lncRNAs as essential upstream MAF:MAFB regulators. ChIP-seq analysis demonstrated MAF:MAFB binding to known epidermal differentiation TF genes whose expression they controlled, including GRHL3, ZNF750, KLF4, and PRDM1. Each of these TFs rescued expression of specific MAF:MAFB target gene subsets in the setting of MAF:MAFB loss, indicating they act downstream of MAF:MAFB. A lncRNA-TF network is thus essential for epidermal differentiation.


Asunto(s)
Diferenciación Celular/genética , Células Epidérmicas , Factor de Transcripción MafB/genética , Proteínas Proto-Oncogénicas c-maf/genética , ARN Largo no Codificante/genética , Animales , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Técnicas de Transferencia de Gen , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/biosíntesis , Ratones , Ratones Endogámicos NOD , Ratones SCID , Organogénesis/genética , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Interferencia de ARN , ARN Interferente Pequeño , Proteínas Represoras/biosíntesis , Factores de Transcripción/biosíntesis , Proteínas Supresoras de Tumor
13.
Science ; 346(6212): 941-5, 2014 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-25414301

RESUMEN

The ability of the skin to repair itself after injury is vital to human survival and is disrupted in a spectrum of disorders. The process of cutaneous wound healing is complex, requiring a coordinated response by immune cells, hematopoietic cells, and resident cells of the skin. We review the classic paradigms of wound healing and evaluate how recent discoveries have enriched our understanding of this process. We evaluate current and experimental approaches to treating cutaneous wounds, with an emphasis on cell-based therapies and skin transplantation.


Asunto(s)
Trasplante de Piel/tendencias , Piel/lesiones , Cicatrización de Heridas/fisiología , Heridas y Lesiones/cirugía , Ingeniería Genética , Humanos , Piel Artificial , Células Madre , Ingeniería de Tejidos
14.
Sci Transl Med ; 6(264): 264ra163, 2014 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-25429056

RESUMEN

Patients with recessive dystrophic epidermolysis bullosa (RDEB) lack functional type VII collagen owing to mutations in the gene COL7A1 and suffer severe blistering and chronic wounds that ultimately lead to infection and development of lethal squamous cell carcinoma. The discovery of induced pluripotent stem cells (iPSCs) and the ability to edit the genome bring the possibility to provide definitive genetic therapy through corrected autologous tissues. We generated patient-derived COL7A1-corrected epithelial keratinocyte sheets for autologous grafting. We demonstrate the utility of sequential reprogramming and adenovirus-associated viral genome editing to generate corrected iPSC banks. iPSC-derived keratinocytes were produced with minimal heterogeneity, and these cells secreted wild-type type VII collagen, resulting in stratified epidermis in vitro in organotypic cultures and in vivo in mice. Sequencing of corrected cell lines before tissue formation revealed heterogeneity of cancer-predisposing mutations, allowing us to select COL7A1-corrected banks with minimal mutational burden for downstream epidermis production. Our results provide a clinical platform to use iPSCs in the treatment of debilitating genodermatoses, such as RDEB.


Asunto(s)
Colágeno Tipo VII/genética , Colágeno Tipo VII/uso terapéutico , Epidermólisis Ampollosa Distrófica/terapia , Genes Recesivos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/trasplante , Animales , Secuencia de Bases , Epidermólisis Ampollosa Distrófica/genética , Predisposición Genética a la Enfermedad , Terapia Genética , Genoma Humano , Recombinación Homóloga/genética , Humanos , Células Madre Pluripotentes Inducidas/citología , Queratinocitos/patología , Ratones , Datos de Secuencia Molecular , Mutación/genética , Análisis de Secuencia de ADN
15.
J Clin Invest ; 124(12): 5275-90, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25401475

RESUMEN

Chromosomal translocation that results in fusion of the genes encoding RNA-binding protein EWS and transcription factor FLI1 (EWS-FLI1) is pathognomonic for Ewing sarcoma. EWS-FLI1 alters gene expression through mechanisms that are not completely understood. We performed RNA sequencing (RNAseq) analysis on primary pediatric human mesenchymal progenitor cells (pMPCs) expressing EWS-FLI1 in order to identify gene targets of this oncoprotein. We determined that long noncoding RNA-277 (Ewing sarcoma-associated transcript 1 [EWSAT1]) is upregulated by EWS-FLI1 in pMPCs. Inhibition of EWSAT1 expression diminished the ability of Ewing sarcoma cell lines to proliferate and form colonies in soft agar, whereas EWSAT1 inhibition had no effect on other cell types tested. Expression of EWS-FLI1 and EWSAT1 repressed gene expression, and a substantial fraction of targets that were repressed by EWS-FLI1 were also repressed by EWSAT1. Analysis of RNAseq data from primary human Ewing sarcoma further supported a role for EWSAT1 in mediating gene repression. We identified heterogeneous nuclear ribonucleoprotein (HNRNPK) as an RNA-binding protein that interacts with EWSAT1 and found a marked overlap in HNRNPK-repressed genes and those repressed by EWS-FLI1 and EWSAT1, suggesting that HNRNPK participates in EWSAT1-mediated gene repression. Together, our data reveal that EWSAT1 is a downstream target of EWS-FLI1 that facilitates the development of Ewing sarcoma via the repression of target genes.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Regulación Neoplásica de la Expresión Génica , ARN Largo no Codificante/biosíntesis , ARN Neoplásico/biosíntesis , Ribonucleoproteínas/metabolismo , Sarcoma de Ewing/metabolismo , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Regulación hacia Abajo/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo K , Humanos , Proteínas de Fusión Oncogénica/biosíntesis , Proteínas de Fusión Oncogénica/genética , Proteína Proto-Oncogénica c-fli-1/biosíntesis , Proteína Proto-Oncogénica c-fli-1/genética , ARN Largo no Codificante/genética , ARN Neoplásico/genética , Proteína EWS de Unión a ARN/biosíntesis , Proteína EWS de Unión a ARN/genética , Ribonucleoproteínas/genética , Sarcoma de Ewing/genética , Sarcoma de Ewing/patología , Análisis de Secuencia de ARN , Regulación hacia Arriba/genética
16.
Nat Genet ; 46(10): 1060-2, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25194279

RESUMEN

Here we report the discovery of recurrent mutations concentrated at an ultraviolet signature hotspot in KNSTRN, which encodes a kinetochore protein, in 19% of cutaneous squamous cell carcinomas (SCCs). Cancer-associated KNSTRN mutations, most notably those encoding p.Ser24Phe, disrupt chromatid cohesion in normal cells, occur in SCC precursors, correlate with increased aneuploidy in primary tumors and enhance tumorigenesis in vivo. These findings suggest a role for KNSTRN mutagenesis in SCC development.


Asunto(s)
Carcinoma de Células Escamosas/genética , Proteínas de Ciclo Celular/genética , Cinetocoros/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Mutación Puntual , Neoplasias Cutáneas/genética , Aneuploidia , Animales , Carcinogénesis/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Células Cultivadas , Femenino , Humanos , Queratinocitos/citología , Queratinocitos/metabolismo , Queratinocitos/trasplante , Ratones Endogámicos NOD , Ratones SCID , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Transfección , Trasplante Heterólogo
17.
BMC Genomics ; 13: 633, 2012 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-23157412

RESUMEN

BACKGROUND: The regulation and function of mammalian RNAs has been increasingly appreciated to operate via RNA-protein interactions. With the recent discovery of thousands of novel human RNA molecules by high-throughput RNA sequencing, efficient methods to uncover RNA-protein interactions are urgently required. Existing methods to study proteins associated with a given RNA are laborious and require substantial amounts of cell-derived starting material. To overcome these limitations, we have developed a rapid and large-scale approach to characterize binding of in vitro transcribed labeled RNA to ~9,400 human recombinant proteins spotted on protein microarrays. RESULTS: We have optimized methodology to probe human protein microarrays with full-length RNA molecules and have identified 137 RNA-protein interactions specific for 10 coding and non-coding RNAs. Those proteins showed strong enrichment for common human RNA binding domains such as RRM, RBD, as well as K homology and CCCH type zinc finger motifs. Previously unknown RNA-protein interactions were discovered using this technique, and these interactions were biochemically verified between TP53 mRNA and Staufen1 protein as well as between HRAS mRNA and CNBP protein. Functional characterization of the interaction between Staufen 1 protein and TP53 mRNA revealed a novel role for Staufen 1 in preserving TP53 RNA stability. CONCLUSIONS: Our approach demonstrates a scalable methodology, allowing rapid and efficient identification of novel human RNA-protein interactions using RNA hybridization to human protein microarrays. Biochemical validation of newly identified interactions between TP53-Stau1 and HRAS-CNBP using reciprocal pull-down experiments, both in vitro and in vivo, demonstrates the utility of this approach to study uncharacterized RNA-protein interactions.


Asunto(s)
Análisis por Matrices de Proteínas , Proteínas/genética , Proteínas/metabolismo , ARN no Traducido/metabolismo , Proteínas del Citoesqueleto/genética , Humanos , Unión Proteica , Proteínas Proto-Oncogénicas p21(ras)/genética , Estabilidad del ARN , ARN no Traducido/química , Proteínas de Unión al ARN/genética , Transcripción Genética , Proteína p53 Supresora de Tumor/genética
18.
Dev Cell ; 22(3): 669-77, 2012 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-22364861

RESUMEN

Disrupted epidermal differentiation characterizes numerous diseases that impact >25% of the population. In a search for dominant mediators of differentiation, we defined a requirement for ZNF750 in terminal epidermal differentiation. ZNF750 controlled genes mutated in numerous human skin diseases, including FLG, LOR, LCE3B, ALOXE3, and SPINK5. ZNF750 induced progenitor differentiation via an evolutionarily conserved C2H2 zinc finger motif. The epidermal master regulator, p63, bound the ZNF750 promoter and was necessary for its induction. ZNF750 restored differentiation to p63-deficient tissue, suggesting that it acts downstream of p63. A search for functionally important ZNF750 targets via analysis of ZNF750-regulated genes identified KLF4, a transcription factor that activates late epidermal differentiation. ZNF750 binds to KLF4 at multiple sites flanking the transcriptional start site and controls its expression. ZNF750 thus directly links a tissue-specifying factor, p63, to an effector of terminal differentiation, KLF4, and represents a potential future target for disorders of this process.


Asunto(s)
Diferenciación Celular , Células Epidérmicas , Factores de Transcripción de Tipo Kruppel/fisiología , Proteínas de la Membrana/fisiología , Factores de Transcripción/fisiología , Secuencia de Aminoácidos , Células Cultivadas , Epidermis/metabolismo , Proteínas Filagrina , Prepucio/fisiología , Regulación de la Expresión Génica/fisiología , Humanos , Queratinocitos/fisiología , Factor 4 Similar a Kruppel , Masculino , Datos de Secuencia Molecular , Proteínas Supresoras de Tumor
19.
Hum Gene Ther ; 21(10): 1299-310, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20497034

RESUMEN

In spite of advances in the molecular diagnosis of recessive dystrophic epidermolysis bullosa (RDEB), an inherited blistering disease due to a deficiency of type VII collagen at the basement membrane zone (BMZ) of stratified epithelium, current therapy is limited to supportive palliation. Gene delivery has shown promise in short-term experiments; however, its long-term sustainability through multiple turnover cycles in human tissue has awaited confirmation. To characterize approaches for long-term genetic correction, retroviral vectors were constructed containing long terminal repeat-driven full-length and epitope-tagged COL7A1 cDNA and evaluated for durability of type VII collagen expression and function in RDEB skin tissue regenerated on immune-deficient mice. Type VII collagen expression was maintained for 1 year in vivo, or over 12 epidermal turnover cycles, with no abnormalities in skin morphology or self-renewal. Type VII collagen restoration led to correction of RDEB disease features, including reestablishment of anchoring fibrils at the BMZ. This approach confirms durably corrective and noninjurious gene delivery to long-lived epidermal progenitors and provides the foundation for a human clinical trial of ex vivo gene delivery in RDEB.


Asunto(s)
Epidermólisis Ampollosa Distrófica/terapia , Terapia Genética/métodos , Vectores Genéticos , Queratinocitos/metabolismo , Transducción Genética , Animales , Transformación Celular Viral , Colágeno , ADN Complementario , Epidermólisis Ampollosa Distrófica/genética , Epidermólisis Ampollosa Distrófica/metabolismo , Expresión Génica , Humanos , Ratones , Ratones SCID , Virus de la Leucemia Murina de Moloney/genética , Piel/metabolismo , Trasplante de Piel , Transgenes
20.
Mol Ther ; 9(5): 721-8, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15120333

RESUMEN

Despite numerous attractive intracellular targets, protein therapeutics have been principally confined to the extracellular space due to the lack of a straightforward way to deliver functional polypeptides to the cell interior. Peptide sequences facilitating intracellular protein delivery have been identified; however, current strategies to apply them require problematic steps, such as generation of new in-frame fusion proteins, covalent chemical conjugation, and denaturation. We have developed a new approach to protein transfer into cells and tissues that relies on single-step decoration by cysteine-flanked, arginine-rich transporter peptides. This approach facilitated cell and tissue delivery of a variety of functional proteins, including antibodies and enzymes. Decoration with transporter peptides thus provides an attractive general means of intracellular delivery of functional proteins in vitro and in tissue.


Asunto(s)
Proteínas Portadoras/química , Transporte de Proteínas , Animales , Apoptosis , Proteínas Portadoras/metabolismo , Caspasas/metabolismo , Caspasas/uso terapéutico , Línea Celular , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Péptidos/síntesis química , Péptidos/química , Péptidos/metabolismo , Proteínas/metabolismo , Proteínas/uso terapéutico , Piel/citología , beta-Galactosidasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA