Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
1.
J Dermatol Sci ; 115(3): 101-110, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39127592

RESUMEN

BACKGROUND: Local gene therapies, including in vivo genome editing, are highly anticipated for the treatment of genetic diseases in skin, especially the epidermis. While the adeno-associated virus (AAV) is a potent vector for in vivo gene delivery, the lack of efficient gene delivery methods has limited its clinical applications. OBJECTIVE: To optimize the AAV gene delivery system with higher gene delivery efficiency and specificity for epidermis and keratinocytes (KCs), using AAV capsid and promoter engineering technologies. METHODS: AAV variants with mutations in residues reported to be critical to determine the tropism of AAV2 for KCs were generated by site-directed mutagenesis of AAVDJ. The infection efficiency and specificity for KCs of these variants were compared with those of previously reported AAVs considered to be suitable for gene delivery to KCs in vitro and in vivo. Additionally, we generated an epidermis-specific promoter using the most recent short-core promoter and compared its specificity with existing promoters. RESULTS: A novel AAVDJ variant capsid termed AAVDJK2 was superior to the existing AAVs in terms of gene transduction efficiency and specificity for epidermis and KCs in vitro and in vivo. A novel tissue-specific promoter, termed the K14 SCP3 promoter, was superior to the existing promoters in terms of gene transduction efficiency and specificity for KCs. CONCLUSION: The combination of the AAVDJK2 capsid and K14 SCP3 promoter improves gene delivery to epidermis in vivo and KCs in vitro. The novel AAV system may benefit experimental research and development of new epidermis-targeted gene therapies.


Asunto(s)
Dependovirus , Epidermis , Terapia Genética , Vectores Genéticos , Queratinocitos , Regiones Promotoras Genéticas , Transducción Genética , Dependovirus/genética , Queratinocitos/metabolismo , Queratinocitos/virología , Vectores Genéticos/genética , Vectores Genéticos/administración & dosificación , Humanos , Animales , Regiones Promotoras Genéticas/genética , Terapia Genética/métodos , Epidermis/metabolismo , Ratones , Mutagénesis Sitio-Dirigida , Técnicas de Transferencia de Gen , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo
2.
EMBO J ; 43(17): 3587-3603, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38951609

RESUMEN

Transposable elements (TEs) are mobile genetic modules of viral derivation that have been co-opted to become modulators of mammalian gene expression. TEs are a major source of endogenous dsRNAs, signaling molecules able to coordinate inflammatory responses in various physiological processes. Here, we provide evidence for a positive involvement of TEs in inflammation-driven bone repair and mineralization. In newly fractured mice bone, we observed an early transient upregulation of repeats occurring concurrently with the initiation of the inflammatory stage. In human bone biopsies, analysis revealed a significant correlation between repeats expression, mechanical stress and bone mineral density. We investigated a potential link between LINE-1 (L1) expression and bone mineralization by delivering a synthetic L1 RNA to osteoporotic patient-derived mesenchymal stem cells and observed a dsRNA-triggered protein kinase (PKR)-mediated stress response that led to strongly increased mineralization. This response was associated with a strong and transient inflammation, accompanied by a global translation attenuation induced by eIF2α phosphorylation. We demonstrated that L1 transfection reshaped the secretory profile of osteoblasts, triggering a paracrine activity that stimulated the mineralization of recipient cells.


Asunto(s)
Inflamación , Elementos de Nucleótido Esparcido Largo , Células Madre Mesenquimatosas , eIF-2 Quinasa , Animales , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , Ratones , Humanos , Inflamación/metabolismo , Inflamación/genética , Inflamación/patología , Células Madre Mesenquimatosas/metabolismo , Elementos de Nucleótido Esparcido Largo/genética , Osteoblastos/metabolismo , Calcificación Fisiológica/genética
3.
Genome Biol ; 25(1): 135, 2024 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-38783323

RESUMEN

BACKGROUND: Dilated cardiomyopathy (DCM) is one of the most common causes of heart failure. Multiple identified mutations in nexilin (NEXN) have been suggested to be linked with severe DCM. However, the exact association between multiple mutations of Nexn and DCM remains unclear. Moreover, it is critical for the development of precise and effective therapeutics in treatments of DCM. RESULTS: In our study, Nexn global knockout mice and mice carrying human equivalent G645del mutation are studied using functional gene rescue assays. AAV-mediated gene delivery is conducted through systemic intravenous injections at the neonatal stage. Heart tissues are analyzed by immunoblots, and functions are assessed by echocardiography. Here, we identify functional components of Nexilin and demonstrate that exogenous introduction could rescue the cardiac function and extend the lifespan of Nexn knockout mouse models. Similar therapeutic effects are also obtained in G645del mice, providing a promising intervention for future clinical therapeutics. CONCLUSIONS: In summary, we demonstrated that a single injection of AAV-Nexn was capable to restore the functions of cardiomyocytes and extended the lifespan of Nexn knockout and G645del mice. Our study represented a long-term gene replacement therapy for DCM that potentially covers all forms of loss-of-function mutations in NEXN.


Asunto(s)
Cardiomiopatía Dilatada , Terapia Genética , Ratones Noqueados , Animales , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/terapia , Ratones , Humanos , Dependovirus/genética , Miocitos Cardíacos/metabolismo , Modelos Animales de Enfermedad , Mutación , Vectores Genéticos/administración & dosificación , Técnicas de Transferencia de Gen
4.
BMC Biol ; 22(1): 101, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38685010

RESUMEN

BACKGROUND: CRISPR-Cas9 genome editing often induces unintended, large genomic rearrangements, posing potential safety risks. However, there are no methods for mitigating these risks. RESULTS: Using long-read individual-molecule sequencing (IDMseq), we found the microhomology-mediated end joining (MMEJ) DNA repair pathway plays a predominant role in Cas9-induced large deletions (LDs). We targeted MMEJ-associated genes genetically and/or pharmacologically and analyzed Cas9-induced LDs at multiple gene loci using flow cytometry and long-read sequencing. Reducing POLQ levels or activity significantly decreases LDs, while depleting or overexpressing RPA increases or reduces LD frequency, respectively. Interestingly, small-molecule inhibition of POLQ and delivery of recombinant RPA proteins also dramatically promote homology-directed repair (HDR) at multiple disease-relevant gene loci in human pluripotent stem cells and hematopoietic progenitor cells. CONCLUSIONS: Our findings reveal the contrasting roles of RPA and POLQ in Cas9-induced LD and HDR, suggesting new strategies for safer and more precise genome editing.


Asunto(s)
Sistemas CRISPR-Cas , Reparación del ADN por Unión de Extremidades , Edición Génica , Humanos , Edición Génica/métodos , Roturas del ADN , Reparación del ADN por Recombinación , Eliminación de Secuencia , ADN Polimerasa theta , Proteína de Replicación A/metabolismo , Proteína de Replicación A/genética
5.
Cell Stem Cell ; 31(1): 52-70.e8, 2024 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-38181751

RESUMEN

Human pluripotent stem cell-derived kidney organoids offer unprecedented opportunities for studying polycystic kidney disease (PKD), which still has no effective cure. Here, we developed both in vitro and in vivo organoid models of PKD that manifested tubular injury and aberrant upregulation of renin-angiotensin aldosterone system. Single-cell analysis revealed that a myriad of metabolic changes occurred during cystogenesis, including defective autophagy. Experimental activation of autophagy via ATG5 overexpression or primary cilia ablation significantly inhibited cystogenesis in PKD kidney organoids. Employing the organoid xenograft model of PKD, which spontaneously developed tubular cysts, we demonstrate that minoxidil, a potent autophagy activator and an FDA-approved drug, effectively attenuated cyst formation in vivo. This in vivo organoid model of PKD will enhance our capability to discover novel disease mechanisms and validate candidate drugs for clinical translation.


Asunto(s)
Cilios , Enfermedades Renales Poliquísticas , Humanos , Riñón , Enfermedades Renales Poliquísticas/tratamiento farmacológico , Autofagia , Organoides
6.
Nucleic Acids Res ; 51(8): 3793-3805, 2023 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-37014011

RESUMEN

Maternal mitochondria are the sole source of mtDNA for every cell of the offspring. Heteroplasmic mtDNA mutations inherited from the oocyte are a common cause of metabolic diseases and associated with late-onset diseases. However, the origin and dynamics of mtDNA heteroplasmy remain unclear. We used our individual Mitochondrial Genome sequencing (iMiGseq) technology to study mtDNA heterogeneity, quantitate single nucleotide variants (SNVs) and large structural variants (SVs), track heteroplasmy dynamics, and analyze genetic linkage between variants at the individual mtDNA molecule level in single oocytes and human blastoids. Our study presented the first single-mtDNA analysis of the comprehensive heteroplasmy landscape in single human oocytes. Unappreciated levels of rare heteroplasmic variants well below the detection limit of conventional methods were identified in healthy human oocytes, of which many are reported to be deleterious and associated with mitochondrial disease and cancer. Quantitative genetic linkage analysis revealed dramatic shifts of variant frequency and clonal expansions of large SVs during oogenesis in single-donor oocytes. iMiGseq of a single human blastoid suggested stable heteroplasmy levels during early lineage differentiation of naïve pluripotent stem cells. Therefore, our data provided new insights of mtDNA genetics and laid a foundation for understanding mtDNA heteroplasmy at early stages of life.


Asunto(s)
ADN Mitocondrial , Células Madre Pluripotentes , Humanos , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Haplotipos , Heteroplasmia , Mitocondrias/genética , Mitocondrias/metabolismo , Oocitos/metabolismo , Células Madre Pluripotentes/metabolismo
7.
Sci Transl Med ; 14(657): eabl6057, 2022 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-35947677

RESUMEN

Constitutive heterochromatin is responsible for genome repression of DNA enriched in repetitive sequences, telomeres, and centromeres. During physiological and pathological premature aging, heterochromatin homeostasis is profoundly compromised. Here, we showed that LINE-1 (Long Interspersed Nuclear Element-1; L1) RNA accumulation was an early event in both typical and atypical human progeroid syndromes. L1 RNA negatively regulated the enzymatic activity of the histone-lysine N-methyltransferase SUV39H1 (suppression of variegation 3-9 homolog 1), resulting in heterochromatin loss and onset of senescent phenotypes in vitro. Depletion of L1 RNA in dermal fibroblast cells from patients with different progeroid syndromes using specific antisense oligonucleotides (ASOs) restored heterochromatin histone 3 lysine 9 and histone 3 lysine 27 trimethylation marks, reversed DNA methylation age, and counteracted the expression of senescence-associated secretory phenotype genes such as p16, p21, activating transcription factor 3 (ATF3), matrix metallopeptidase 13 (MMP13), interleukin 1a (IL1a), BTG anti-proliferation factor 2 (BTG2), and growth arrest and DNA damage inducible beta (GADD45b). Moreover, systemic delivery of ASOs rescued the histophysiology of tissues and increased the life span of a Hutchinson-Gilford progeria syndrome mouse model. Transcriptional profiling of human and mouse samples after L1 RNA depletion demonstrated that pathways associated with nuclear chromatin organization, cell proliferation, and transcription regulation were enriched. Similarly, pathways associated with aging, inflammatory response, innate immune response, and DNA damage were down-regulated. Our results highlight the role of L1 RNA in heterochromatin homeostasis in progeroid syndromes and identify a possible therapeutic approach to treat premature aging and related syndromes.


Asunto(s)
Envejecimiento Prematuro , Síndrome de Cockayne , Proteínas Inmediatas-Precoces , Progeria , Envejecimiento Prematuro/genética , Animales , Antígenos de Diferenciación , Heterocromatina , Histonas/metabolismo , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Elementos de Nucleótido Esparcido Largo , Lisina/metabolismo , Ratones , Fenotipo , Progeria/genética , ARN , Telómero/genética , Proteínas Supresoras de Tumor/genética
8.
Front Cell Dev Biol ; 10: 786031, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35309931

RESUMEN

It is widely believed that cellular senescence plays a critical role in both aging and cancer, and that senescence is a fundamental, permanent growth arrest that somatic cells cannot avoid. Here we show that Myc plays an important role in self-renewal of esophageal epithelial cells, contributing to their resistance to cellular senescence. Myc is homogeneously expressed in basal cells of the esophageal epithelium and Myc positively regulates their self-renewal by maintaining their undifferentiated state. Indeed, Myc knockout induced a loss of the undifferentiated state of esophageal epithelial cells resulting in cellular senescence while forced MYC expression promoted oncogenic cell proliferation. A superoxide scavenger counteracted Myc knockout-induced senescence, therefore suggesting that a mitochondrial superoxide takes part in inducing senescence. Taken together, these analyses reveal extremely low levels of cellular senescence and senescence-associated phenotypes in the esophageal epithelium, as well as a critical role for Myc in self-renewal of basal cells in this organ. This provides new avenues for studying and understanding the links between stemness and resistance to cellular senescence.

9.
Nat Commun ; 12(1): 3094, 2021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-34035273

RESUMEN

Short-term, systemic expression of the Yamanaka reprogramming factors (Oct-3/4, Sox2, Klf4 and c-Myc [OSKM]) has been shown to rejuvenate aging cells and promote tissue regeneration in vivo. However, the mechanisms by which OSKM promotes tissue regeneration are unknown. In this work, we focus on a specific tissue and demonstrate that local expression of OSKM, specifically in myofibers, induces the activation of muscle stem cells or satellite cells (SCs), which accelerates muscle regeneration in young mice. In contrast, expressing OSKM directly in SCs does not improve muscle regeneration. Mechanistically, expressing OSKM in myofibers regulates the expression of genes important for the SC microenvironment, including upregulation of p21, which in turn downregulates Wnt4. This is critical because Wnt4 is secreted by myofibers to maintain SC quiescence. Thus, short-term induction of the Yamanaka factors in myofibers may promote tissue regeneration by modifying the stem cell niche.


Asunto(s)
Diferenciación Celular/genética , Reprogramación Celular/genética , Miofibrillas/metabolismo , Regeneración/genética , Células Satélite del Músculo Esquelético/metabolismo , Nicho de Células Madre , Animales , Células Cultivadas , Femenino , Expresión Génica , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Ratones Transgénicos , Miofibrillas/fisiología , Factor 3 de Transcripción de Unión a Octámeros/genética , Proteínas Proto-Oncogénicas c-myc/genética , Factores de Transcripción SOXB1/genética , Células Satélite del Músculo Esquelético/citología , Proteína Wnt4/genética
10.
Cell ; 184(8): 2020-2032.e14, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33861963

RESUMEN

Interspecies chimera formation with human pluripotent stem cells (hPSCs) represents a necessary alternative to evaluate hPSC pluripotency in vivo and might constitute a promising strategy for various regenerative medicine applications, including the generation of organs and tissues for transplantation. Studies using mouse and pig embryos suggest that hPSCs do not robustly contribute to chimera formation in species evolutionarily distant to humans. We studied the chimeric competency of human extended pluripotent stem cells (hEPSCs) in cynomolgus monkey (Macaca fascicularis) embryos cultured ex vivo. We demonstrate that hEPSCs survived, proliferated, and generated several peri- and early post-implantation cell lineages inside monkey embryos. We also uncovered signaling events underlying interspecific crosstalk that may help shape the unique developmental trajectories of human and monkey cells within chimeric embryos. These results may help to better understand early human development and primate evolution and develop strategies to improve human chimerism in evolutionarily distant species.


Asunto(s)
Quimerismo , Embrión de Mamíferos/citología , Células Madre Pluripotentes/citología , Animales , Blastocisto/citología , Blastocisto/metabolismo , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Embrión de Mamíferos/metabolismo , Femenino , Humanos , Macaca fascicularis , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/trasplante , RNA-Seq , Análisis de la Célula Individual , Transcriptoma
11.
Med ; 2(6): 689-700.e4, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-33821249

RESUMEN

BACKGROUND: Strategies for monitoring the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are crucial for combating the pandemic. Detection and mutation surveillance of SARS-CoV-2 and other respiratory viruses require separate and complex workflows that rely on highly specialized facilities, personnel, and reagents. To date, no method can rapidly diagnose multiple viral infections and determine variants in a high-throughput manner. METHODS: We describe a method for multiplex isothermal amplification-based sequencing and real-time analysis of multiple viral genomes, termed nanopore sequencing of isothermal rapid viral amplification for near real-time analysis (NIRVANA). It can simultaneously detect SARS-CoV-2, influenza A, human adenovirus, and human coronavirus and monitor mutations for up to 96 samples in real time. FINDINGS: NIRVANA showed high sensitivity and specificity for SARS-CoV-2 in 70 clinical samples with a detection limit of 20 viral RNA copies per µL of extracted nucleic acid. It also detected the influenza A co-infection in two samples. The variant analysis results of SARS-CoV-2-positive samples mirror the epidemiology of coronavirus disease 2019 (COVID-19). Additionally, NIRVANA could simultaneously detect SARS-CoV-2 and pepper mild mottle virus (PMMoV) (an omnipresent virus and water-quality indicator) in municipal wastewater samples. CONCLUSIONS: NIRVANA provides high-confidence detection of both SARS-CoV-2 and other respiratory viruses and mutation surveillance of SARS-CoV-2 on the fly. We expect it to offer a promising solution for rapid field-deployable detection and mutational surveillance of pandemic viruses. FUNDING: M.L. is supported by KAUST Office of Sponsored Research (BAS/1/1080-01). This work is supported by KAUST Competitive Research Grant (URF/1/3412-01-01; M.L. and J.C.I.B.) and Universidad Catolica San Antonio de Murcia (J.C.I.B.). A.M.H. is supported by Saudi Ministry of Education (project 436).


Asunto(s)
COVID-19 , Gripe Humana , COVID-19/diagnóstico , Humanos , Gripe Humana/epidemiología , Mutación/genética , Pandemias , SARS-CoV-2/genética
12.
Cell ; 180(3): 585-600.e19, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-32004457

RESUMEN

Molecular mechanisms of ovarian aging and female age-related fertility decline remain unclear. We surveyed the single-cell transcriptomic landscape of ovaries from young and aged non-human primates (NHPs) and identified seven ovarian cell types with distinct gene-expression signatures, including oocyte and six types of ovarian somatic cells. In-depth dissection of gene-expression dynamics of oocytes revealed four subtypes at sequential and stepwise developmental stages. Further analysis of cell-type-specific aging-associated transcriptional changes uncovered the disturbance of antioxidant signaling specific to early-stage oocytes and granulosa cells, indicative of oxidative damage as a crucial factor in ovarian functional decline with age. Additionally, inactivated antioxidative pathways, increased reactive oxygen species, and apoptosis were observed in granulosa cells from aged women. This study provides a comprehensive understanding of the cell-type-specific mechanisms underlying primate ovarian aging at single-cell resolution, revealing new diagnostic biomarkers and potential therapeutic targets for age-related human ovarian disorders.


Asunto(s)
Envejecimiento/genética , Ovario/fisiología , Análisis de la Célula Individual/métodos , Transcriptoma , Anciano , Animales , Antioxidantes/metabolismo , Apoptosis/fisiología , Atlas como Asunto , Biomarcadores , Línea Celular Tumoral , Femenino , Células de la Granulosa/metabolismo , Humanos , Macaca fascicularis , Oocitos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología
13.
Semin Cell Dev Biol ; 97: 3-15, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31028854

RESUMEN

Epigenetic regulation of gene expression is fundamental in the maintenance of cellular identity and the regulation of cellular plasticity during tissue repair. In fact, epigenetic modulation is associated with the processes of cellular de-differentiation, proliferation, and re-differentiation that takes place during tissue regeneration. In here we explore the epigenetic events that coordinate tissue repair in lower vertebrates with high regenerative capacity, and in mammalian adult stem cells, which are responsible for the homeostasis maintenance of most of our tissues. Finally we summarize promising CRISPR-based editing technologies developed during the last years, which look as promising tools to not only study but also promote specific events during tissue regeneration.


Asunto(s)
Cromatina/metabolismo , Epigénesis Genética/genética , Regeneración/efectos de los fármacos , Humanos
14.
Cell Res ; 29(10): 804-819, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31444470

RESUMEN

In vivo genome editing represents a powerful strategy for both understanding basic biology and treating inherited diseases. However, it remains a challenge to develop universal and efficient in vivo genome-editing tools for tissues that comprise diverse cell types in either a dividing or non-dividing state. Here, we describe a versatile in vivo gene knock-in methodology that enables the targeting of a broad range of mutations and cell types through the insertion of a minigene at an intron of the target gene locus using an intracellularly linearized single homology arm donor. As a proof-of-concept, we focused on a mouse model of premature-aging caused by a dominant point mutation, which is difficult to repair using existing in vivo genome-editing tools. Systemic treatment using our new method ameliorated aging-associated phenotypes and extended animal lifespan, thus highlighting the potential of this methodology for a broad range of in vivo genome-editing applications.


Asunto(s)
Edición Génica/métodos , Animales , Sistemas CRISPR-Cas/genética , Reparación del ADN , Dependovirus/genética , Factor de Transcripción GATA3/genética , Técnicas de Sustitución del Gen , Terapia Genética/métodos , Vectores Genéticos/metabolismo , Células Madre Embrionarias Humanas , Humanos , Intrones , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Neuronas/citología , Neuronas/metabolismo , ARN Guía de Kinetoplastida/metabolismo , Ratas , Tubulina (Proteína)/genética
15.
Cell Stem Cell ; 25(3): 373-387.e9, 2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31303547

RESUMEN

Human pluripotent stem cell-derived kidney organoids recapitulate developmental processes and tissue architecture, but intrinsic limitations, such as lack of vasculature and functionality, have greatly hampered their application. Here we establish a versatile protocol for generating vascularized three-dimensional (3D) kidney organoids. We employ dynamic modulation of WNT signaling to control the relative proportion of proximal versus distal nephron segments, producing a correlative level of vascular endothelial growth factor A (VEGFA) to define a resident vascular network. Single-cell RNA sequencing identifies a subset of nephron progenitor cells as a potential source of renal vasculature. These kidney organoids undergo further structural and functional maturation upon implantation. Using this kidney organoid platform, we establish an in vitro model of autosomal recessive polycystic kidney disease (ARPKD), the cystic phenotype of which can be effectively prevented by gene correction or drug treatment. Our studies provide new avenues for studying human kidney development, modeling disease pathogenesis, and performing patient-specific drug validation.


Asunto(s)
Riñón/citología , Organoides/citología , Células Madre Pluripotentes/citología , Riñón Poliquístico Autosómico Recesivo/patología , Diferenciación Celular , Células Cultivadas , Descubrimiento de Drogas , Terapia Genética , Humanos , Riñón/irrigación sanguínea , Neovascularización Fisiológica , Técnicas de Cultivo de Órganos , Organogénesis , Organoides/irrigación sanguínea , Riñón Poliquístico Autosómico Recesivo/metabolismo , Riñón Poliquístico Autosómico Recesivo/terapia , Medicina de Precisión , Factor A de Crecimiento Endotelial Vascular/metabolismo , Vía de Señalización Wnt
16.
Nat Commun ; 10(1): 3329, 2019 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-31350386

RESUMEN

DiGeorge syndrome critical region 8 (DGCR8) is a critical component of the canonical microprocessor complex for microRNA biogenesis. However, the non-canonical functions of DGCR8 have not been studied. Here, we demonstrate that DGCR8 plays an important role in maintaining heterochromatin organization and attenuating aging. An N-terminal-truncated version of DGCR8 (DR8dex2) accelerated senescence in human mesenchymal stem cells (hMSCs) independent of its microRNA-processing activity. Further studies revealed that DGCR8 maintained heterochromatin organization by interacting with the nuclear envelope protein Lamin B1, and heterochromatin-associated proteins, KAP1 and HP1γ. Overexpression of any of these proteins, including DGCR8, reversed premature senescent phenotypes in DR8dex2 hMSCs. Finally, DGCR8 was downregulated in pathologically and naturally aged hMSCs, whereas DGCR8 overexpression alleviated hMSC aging and mouse osteoarthritis. Taken together, these analyses uncovered a novel, microRNA processing-independent role in maintaining heterochromatin organization and attenuating senescence by DGCR8, thus representing a new therapeutic target for alleviating human aging-related disorders.


Asunto(s)
Heterocromatina/metabolismo , Osteoartritis/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Senescencia Celular , Heterocromatina/genética , Humanos , Lamina Tipo B/genética , Lamina Tipo B/metabolismo , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , MicroARNs/genética , Osteoartritis/genética , Osteoartritis/fisiopatología , Estabilidad Proteica , Proteínas de Unión al ARN/genética , Proteína 28 que Contiene Motivos Tripartito/genética , Proteína 28 que Contiene Motivos Tripartito/metabolismo
17.
J Dermatol Sci ; 95(1): 8-12, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31255469

RESUMEN

Cutaneous ulcers are a common cause of morbidity. We have developed a de novo epithelialization method for treating cutaneous ulcers by means of reprogramming wound-resident mesenchymal cells in vivo into cells able to form a stratified epithelium: induced stratified epithelial progenitors (iSEPs). Administration of 4 transcription factors (DNP63A, GRHL2, TFAP2A, and cMYC) expressed via adeno-associated viral vectors enabled generation of epithelial cells and tissues, thereby acheiving de novo epithelialization from the surfaces of cutaneous ulcers in a mouse model. Generated epithelia, having barrier functions equivalent to the original epidermis, were maintained for more than 6 months. Our findings constitute a proof of concept for future development towards innovative therapies for cutaneous ulcers via de novo epithelialization.


Asunto(s)
Terapia Genética/métodos , Repitelización/genética , Úlcera Cutánea/terapia , Factores de Transcripción/genética , Animales , Diferenciación Celular/genética , Dependovirus/genética , Modelos Animales de Enfermedad , Fibroblastos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Células Madre Pluripotentes Inducidas , Queratinocitos , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Transgénicos , Piel/patología , Úlcera Cutánea/patología
18.
Methods Mol Biol ; 2005: 101-124, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31175649

RESUMEN

Interspecies chimera formation provides a unique platform for studying donor cell developmental potential, modeling disease in vivo, as well as in vivo production of tissues and organs. The derivation of human pluripotent stem cells (hPSC) from either human embryos or somatic cell reprogramming facilitates our understanding of human development, as well as accelerates our exploration of regenerative medicine for human health. Due to similar organ size, close anatomy, and physiology between pig and human, human-Pig interspecies chimeric model in which pig serves as the host species may open new avenues for studying human embryogenesis, disease pathogenesis, and generation of human organ for transplantation to solve the worldwide donor organ shortage. Our previous study demonstrated chimeric competency of different types of human PSCs in pig host. In this chapter, we introduce our workflow for the generation of human PSCs and analysis of its chimeric contribution to pre- and postimplantation pig embryos.


Asunto(s)
Blastocisto/metabolismo , Modelos Biológicos , Células Madre Pluripotentes/metabolismo , Trasplante de Células Madre , Quimera por Trasplante/embriología , Animales , Blastocisto/citología , Xenoinjertos , Humanos , Células Madre Pluripotentes/citología , Porcinos
19.
Protein Cell ; 10(7): 485-495, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31041783

RESUMEN

Identification of the precise molecular pathways involved in oncogene-induced transformation may help us gain a better understanding of tumor initiation and promotion. Here, we demonstrate that SOX2+ foregut epithelial cells are prone to oncogenic transformation upon mutagenic insults, such as KrasG12D and p53 deletion. GFP-based lineage-tracing experiments indicate that SOX2+ cells are the cells-of-origin of esophagus and stomach hyperplasia. Our observations indicate distinct roles for oncogenic KRAS mutation and P53 deletion. p53 homozygous deletion is required for the acquisition of an invasive potential, and KrasG12D expression, but not p53 deletion, suffices for tumor formation. Global gene expression analysis reveals secreting factors upregulated in the hyperplasia induced by oncogenic KRAS and highlights a crucial role for the CXCR2 pathway in driving hyperplasia. Collectively, the array of genetic models presented here demonstrate that stratified epithelial cells are susceptible to oncogenic insults, which may lead to a better understanding of tumor initiation and aid in the design of new cancer therapeutics.


Asunto(s)
Neoplasias Esofágicas/metabolismo , Mutación , Receptores de Interleucina-8B/metabolismo , Factores de Transcripción SOXB1/metabolismo , Animales , Proliferación Celular , Neoplasias Esofágicas/patología , Femenino , Masculino , Ratones , Ratones Mutantes , Transducción de Señal , Células Tumorales Cultivadas
20.
PLoS Biol ; 17(4): e3000201, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30933975

RESUMEN

Cellular senescence is a driver of various aging-associated disorders, including osteoarthritis. Here, we identified a critical role for Yes-associated protein (YAP), a major effector of Hippo signaling, in maintaining a younger state of human mesenchymal stem cells (hMSCs) and ameliorating osteoarthritis in mice. Clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR associated protein 9 nuclease (Cas9)-mediated knockout (KO) of YAP in hMSCs resulted in premature cellular senescence. Mechanistically, YAP cooperated with TEA domain transcriptional factor (TEAD) to activate the expression of forkhead box D1 (FOXD1), a geroprotective protein. YAP deficiency led to the down-regulation of FOXD1. In turn, overexpression of YAP or FOXD1 rejuvenated aged hMSCs. Moreover, intra-articular administration of lentiviral vector encoding YAP or FOXD1 attenuated the development of osteoarthritis in mice. Collectively, our findings reveal YAP-FOXD1, a novel aging-associated regulatory axis, as a potential target for gene therapy to alleviate osteoarthritis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Factores de Transcripción Forkhead/metabolismo , Osteoartritis/metabolismo , Osteoartritis/patología , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proliferación Celular/genética , Senescencia Celular/fisiología , Factores de Transcripción Forkhead/genética , Xenoinjertos , Humanos , Masculino , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Osteoartritis/genética , Transducción de Señal , Factores de Transcripción/genética , Activación Transcripcional , Regulación hacia Arriba , Proteínas Señalizadoras YAP
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA