Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 81(1): 305, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-39012348

RESUMEN

Lentiviral vectors have markedly enhanced gene therapy efficiency in treating congenital diseases, but their long-term safety remains controversial. Most gene therapies for congenital eye diseases need to be carried out at early ages, yet the assessment of related risks to ocular development posed by lentiviral vectors is challenging. Utilizing single-cell transcriptomic profiling on human retinal organoids, this study explored the impact of lentiviral vectors on the retinal development and found that lentiviral vectors can cause retinal precursor cells to shift toward photoreceptor fate through the up-regulation of key fate-determining genes such as PRDM1. Further investigation demonstrated that the intron and intergenic region of PRDM1 was bound by PHLDA1, which was also up-regulated by lentiviral vectors exposure. Importantly, knockdown of PHLDA1 successfully suppressed the lentivirus-induced differentiation bias of photoreceptor cells. The findings also suggest that while lentiviral vectors may disrupt the fate determination of retinal precursor cells, posing risks in early-stage retinal gene therapy, these risks could potentially be reduced by inhibiting the PHLDA1-PRDM1 axis.


Asunto(s)
Diferenciación Celular , Vectores Genéticos , Lentivirus , Retina , Células Madre , Factores de Transcripción , Humanos , Retina/metabolismo , Retina/citología , Lentivirus/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Vectores Genéticos/metabolismo , Vectores Genéticos/genética , Diferenciación Celular/genética , Células Madre/metabolismo , Células Madre/citología , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Organoides/metabolismo , Organoides/citología , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Terapia Genética/métodos
2.
Proc Natl Acad Sci U S A ; 117(41): 25712-25721, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-32989127

RESUMEN

Vogt-Koyanagi-Harada (VKH) disease is a systemic autoimmune disorder affecting multiple organs, including eyes, skin, and central nervous system. It is known that monocytes significantly contribute to the development of autoimmune disease. However, the subset heterogeneity with unique functions and signatures in human circulating monocytes and the identity of disease-specific monocytic populations remain largely unknown. Here, we employed an advanced single-cell RNA sequencing technology to systematically analyze 11,259 human circulating monocytes and genetically defined their subpopulations. We constructed a precise atlas of human blood monocytes, identified six subpopulations-including S100A12, HLA, CD16, proinflammatory, megakaryocyte-like, and NK-like monocyte subsets-and uncovered two previously unidentified subsets: HLA and megakaryocyte-like monocyte subsets. Relative to healthy individuals, cellular composition, gene expression signatures, and activation states were markedly alternated in VKH patients utilizing cell type-specific programs, especially the CD16 and proinflammatory monocyte subpopulations. Notably, we discovered a disease-relevant subgroup, proinflammatory monocytes, which showed a discriminative gene expression signature indicative of inflammation, antiviral activity, and pathologic activation, and converted into a pathologic activation state implicating the active inflammation during VKH disease. Additionally, we found the cell type-specific transcriptional signature of proinflammatory monocytes, ISG15, whose production might reflect the treatment response. Taken together, in this study, we present discoveries on accurate classification, molecular markers, and signaling pathways for VKH disease-associated monocytes. Therapeutically targeting this proinflammatory monocyte subpopulation would provide an attractive approach for treating VKH, as well as other autoimmune diseases.


Asunto(s)
Monocitos/inmunología , Síndrome Uveomeningoencefálico/genética , Síndrome Uveomeningoencefálico/inmunología , Adulto , Autoinmunidad , Citocinas/genética , Citocinas/inmunología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Receptores de IgG/genética , Receptores de IgG/inmunología , Proteína S100A12/genética , Proteína S100A12/inmunología , Ubiquitinas/genética , Ubiquitinas/inmunología
3.
Dev Dyn ; 250(7): 974-985, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33449399

RESUMEN

BACKGROUND: Genetic programs underlying preimplantation development and early lineage segregation are highly conserved across mammals. It has been suggested that nonhuman primates would be better model organisms for human embryogenesis, but a limited number of studies have investigated the monkey preimplantation development. In this study, we collect single cells from cynomolgus monkey preimplantation embryos for transcriptome profiling and compare with single-cell RNA-seq data derived from human and mouse embryos. RESULTS: By weighted gene-coexpression network analysis, we found that cynomolgus gene networks have greater conservation with human embryos including a greater number of conserved hub genes than that of mouse embryos. Consistently, we found that early ICM/TE lineage-segregating genes in monkeys exhibit greater similarity with human when compared to mouse, so are the genes in signaling pathways such as LRP1 and TCF7 involving in WNT pathway. Last, we tested the role of one conserved pre-EGA hub gene, SIN3A, using a morpholino knockdown of maternal RNA transcripts in monkey embryos followed by single-cell RNA-seq. We found that SIN3A knockdown disrupts the gene-silencing program during the embryonic genome activation transition and results in developmental delay of cynomolgus embryos. CONCLUSION: Taken together, our study provided new insight into evolutionarily conserved and divergent transcriptome dynamics during mammalian preimplantation development.


Asunto(s)
Blastómeros/metabolismo , Desarrollo Embrionario/genética , Macaca fascicularis/embriología , Adulto , Animales , Blastocisto , Blastómeros/citología , Linaje de la Célula/genética , Células Cultivadas , Embrión de Mamíferos , Desarrollo Embrionario/fisiología , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes/fisiología , Humanos , Macaca fascicularis/genética , Macaca mulatta , Masculino , Ratones , Embarazo , Complejo Correpresor Histona Desacetilasa y Sin3/genética , Complejo Correpresor Histona Desacetilasa y Sin3/fisiología , Análisis de la Célula Individual/veterinaria , Transcriptoma/genética
4.
Proteomics ; 20(13): e1900271, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32223079

RESUMEN

In recent years, the emergence of single-cell omics technologies, which can profile genomics, transcriptomics, epigenomics, and proteomics, has provided unprecedented insights into characteristics of cancer, enabling higher resolution and accuracy to decipher the cellular and molecular mechanisms relating to tumorigenesis, evolution, metastasis, and immune responses. Single-cell multi-omics technologies, which are developed based on the combination of multiple single-cell mono-omics technologies, can simultaneously analyze RNA expression, single nucleotide polymorphism, epigenetic modification, or protein abundance, enabling the in-depth understanding of gene expression regulatory mechanisms. In this review, the state-of-the-art single-cell multi-omics technologies are summarized and the prospects of their application in cancer biology are discussed.


Asunto(s)
Metabolómica , Neoplasias , Epigenómica , Genómica , Humanos , Estudios Prospectivos , Proteómica
5.
Graefes Arch Clin Exp Ophthalmol ; 257(3): 455-463, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30684011

RESUMEN

PURPOSE: The aim of this study was to identify the morphological features of the retina and choroid in Macaca fascicularis of different ages using multimodal imaging. METHODS: A total of 27 Macaca fascicularis with no ocular diseases were studied (mean age, 104.2 months; range, 1.2-223.6 months). Multimodal imaging was obtained from each subject. The morphological features were compared within four subgroups according to age. RESULTS: On spectrum-domain optical coherence tomography (SD-OCT), four hyper-reflective bands could be observed in the outer retina in non-infant macaques (21/21, 100%), while the interdigitation zone could not be observed in the six infant macaques. A narrow hypo-reflective band just posterior to the retinal pigment epithelium (RPE) was noted in most eyes (25/27, 92.6%). The choroidal-scleral junction (CSJ) was visible in 83.3% of infants but only in 12.5% of adults and 14.3% of the geriatric population, and it could not be seen in juveniles. There was a significant difference in CSJ visibility between the infant group and the other three groups (P < 0.001). Tessellated fundus, in which the choroidal vessels are visible through the retina, could be observed clearly with near-infrared reflectance imaging (NIR). Some granular spots were noted in juveniles, and they accumulated dramatically with age, but were absent in infants. CONCLUSION: Notable morphological features can be observed in the Macaca fascicularis subjects using multimodal imaging, and these features vary distinctly according to their age. It is important to note that infant macaques had no interdigitation zone, while the other macaques had no visible CSJ but did have well-defined choroidal capillaries. Age and the features should be considered seriously in future animal studies.


Asunto(s)
Envejecimiento , Coroides/diagnóstico por imagen , Imagen Multimodal , Retina/diagnóstico por imagen , Animales , Angiografía con Fluoresceína/métodos , Fondo de Ojo , Macaca fascicularis , Modelos Animales , Valores de Referencia , Reproducibilidad de los Resultados , Tomografía de Coherencia Óptica/métodos , Pruebas del Campo Visual
6.
Biochem Biophys Res Commun ; 446(1): 261-6, 2014 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-24589733

RESUMEN

Although targeted gene addition could be stimulated strikingly by a DNA double strand break (DSB) created by either zinc finger nucleases (ZFNs) or TALE nucleases (TALENs), the DSBs are really mutagenic and toxic to human cells. As a compromised solution, DNA single-strand break (SSB) or nick has been reported to mediate high efficient gene addition but with marked reduction of random mutagenesis. We previously demonstrated effective targeted gene addition at the human multicopy ribosomal DNA (rDNA) locus, a genomic safe harbor for the transgene with therapeutic potential. To improve the transgene integration efficiency by using TALENs while lowering the cytotoxicity of DSBs, we created both TALENs and TALE nickases (TALENickases) targeting this multicopy locus. A targeting vector which could integrate a GFP cassette at the rDNA locus was constructed and co-transfected with TALENs or TALENickases. Although the fraction of GFP positive cells using TALENs was greater than that using TALENickases during the first few days after transfection, it reduced to a level less than that using TALENickases after continuous culture. Our findings showed that the TALENickases were more effective than their TALEN counterparts at the multi-copy rDNA locus, though earlier studies using ZFNs and ZFNickases targeting the single-copy loci showed the reverse. Besides, TALENickases mediated the targeted integration of a 5.4 kb fragment at a frequency of up to 0.62% in HT1080 cells after drug selection, suggesting their potential application in targeted gene modification not being limited at the rDNA locus.


Asunto(s)
ADN Ribosómico/genética , Desoxirribonucleasa I/metabolismo , Técnicas de Sustitución del Gen/métodos , Mutagénesis Insercional , Línea Celular , Desoxirribonucleasa I/genética , Marcación de Gen , Terapia Genética , Células HEK293 , Humanos , Regiones Promotoras Genéticas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfección , Dedos de Zinc
7.
Spectrochim Acta A Mol Biomol Spectrosc ; 310: 123932, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38266606

RESUMEN

Perfluorooctanoic acid (PFOA), a novel organic pollutant, has been shown to be toxic, persistent, bioaccumulative, long-range transportable, and globally prevalent. This article is based on surface enhanced Raman scattering (SERS) spectroscopy analysis technology. The monolayer of SiO2 was prepared by chemical reduction etching self-assembly method and silver dendrites were grown on it, thus forming the SERS substrate with silver dendrite Metasurface structure with Raman detection enhancement factor up to 2.32 × 105. The prepared silver dendrite Metasurface SERS substrate was applied to the qualitative and quantitative detection of PFOA, with a quantitative detection limit of 15.89 ppb. The results of this paper provide a new, simple, and quick method for the detection of PFOA in the environment.

8.
Nat Commun ; 14(1): 7126, 2023 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-37932255

RESUMEN

Age is closely related to human health and disease risks. However, chronologically defined age often disagrees with biological age, primarily due to genetic and environmental variables. Identifying effective indicators for biological age in clinical practice and self-monitoring is important but currently lacking. The human lens accumulates age-related changes that are amenable to rapid and objective assessment. Here, using lens photographs from 20 to 96-year-olds, we develop LensAge to reflect lens aging via deep learning. LensAge is closely correlated with chronological age of relatively healthy individuals (R2 > 0.80, mean absolute errors of 4.25 to 4.82 years). Among the general population, we calculate the LensAge index by contrasting LensAge and chronological age to reflect the aging rate relative to peers. The LensAge index effectively reveals the risks of age-related eye and systemic disease occurrence, as well as all-cause mortality. It outperforms chronological age in reflecting age-related disease risks (p < 0.001). More importantly, our models can conveniently work based on smartphone photographs, suggesting suitability for routine self-examination of aging status. Overall, our study demonstrates that the LensAge index may serve as an ideal quantitative indicator for clinically assessing and self-monitoring biological age in humans.


Asunto(s)
Aprendizaje Profundo , Cristalino , Humanos , Preescolar , Envejecimiento/genética
9.
Nanomaterials (Basel) ; 12(6)2022 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-35335783

RESUMEN

Surface-enhanced Raman scattering (SERS) enables trace-detection for biosensing and environmental monitoring. Optimized enhancement of SERS can be achieved when the energy of the localized surface plasmon resonance (LSPR) is close to the energy of the Raman excitation wavelength. The LSPR can be tuned using a plasmonic superstructure array with controlled periods. In this paper, we develop a new technique based on laser near-field reduction to fabricate a superstructure array, which provides distinct features in the formation of periodic structures with hollow nanoclusters and flexible control of the LSPR in fewer steps than current techniques. Fabrication involves irradiation of a continuous wave laser or femtosecond laser onto a monolayer of self-assembled silica microspheres to grow silver nanoparticles along the silica microsphere surfaces by laser near-field reduction. The LSPR of superstructure array can be flexibly tuned to match the Raman excitation wavelengths from the visible to the infrared regions using different diameters of silica microspheres. The unique nanostructure formed can contribute to an increase in the sensitivity of SERS sensing. The fabricated superstructure array thus offers superior characteristics for the quantitative analysis of fluorescent perfluorooctanoic acid with a wide detection range from 11 ppb to 400 ppm.

10.
Cell Prolif ; 55(7): e13254, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35633292

RESUMEN

OBJECTIVES: Stem cell-derived photoreceptor replacement therapy is a promising strategy for the treatment of retinal degenerative disease. The development of 3D retinal organoids has permitted the production of photoreceptors. However, there is no strategy to enrich a specific photoreceptor subtype due to inadequate knowledge of the molecular mechanism underlying the photoreceptor fate determination. Hence, our aim is to explore the uncharacterized function of somatostatin signalling in human pluripotent stem cell-derived photoreceptor differentiation. MATERIALS AND METHODS: 3D retinal organoids were achieved from human embryonic stem cell. The published single-cell RNA-sequencing datasets of human retinal development were utilized to further investigate the transcriptional regulation of photoreceptor differentiation. The assays of immunofluorescence staining, lentivirus transfection, real-time quantitative polymerase chain reaction and western blotting were performed. RESULTS: We identified that the somatostatin receptor 2 (SSTR2)-mediated signalling was essential for rod photoreceptor differentiation at the precursor stage. The addition of the cognate ligand somatostatin in human 3D retinal organoids promoted rod photoreceptor differentiation and inhibited cone photoreceptor production. Furthermore, we found that the genesis of rod photoreceptors was modulated by endogenous somatostatin specifically secreted by developing retinal ganglion cells. CONCLUSIONS: Our study identified SSTR2 signalling as a novel extrinsic regulator for rod photoreceptor fate determination in photoreceptor precursors, which expands the repertoire of functional signalling pathways in photoreceptor development and sheds light on the optimization of the photoreceptor enrichment strategy.


Asunto(s)
Organoides , Células Madre Pluripotentes , Diferenciación Celular/fisiología , Humanos , Retina/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Somatostatina/metabolismo
11.
Invest Ophthalmol Vis Sci ; 63(12): 9, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36331259

RESUMEN

Purpose: Cone and rod photoreceptors in the retina convert light to electrical signals which are transmitted to the visual cortex of the brain. Abnormal photoreceptor development and degeneration results in blindness. So far, the mechanism that controls photoreceptor specification and its subsequent fate bifurcation remain elusive. Methods: To trace and enrich the human photoreceptor lineage, we first engineered H9 human embryonic stem cell (hESC) reporter line by fusing EGFP to endogenous BLIMP1 using CRISPR/CAS9 gene-editing technology, and then used the cell line to generate 3D retinal organoids. Following EGFP-based cell sorting, single-cell RNA-sequencing was conducted via 10x Genomics Chromium system, and the data were analyzed using Seurat. Immunofluorescence combined with lentivirus-mediated knockdown and overexpression experiments were used as validation approaches. Results: Single-cell transcriptomic profiling revealed that retinal progenitor cells were temporally programmed to differentiate to cone and rod sequentially. We identified PHLDA1 as a novel regulator of photoreceptor specification. PHLDA1 mediated the effects of IGF1 through IGF1R, and inhibited AKT phosphorylation during photoreceptor development. Conclusions: Our data established a transcriptomic cell atlas of the human photoreceptor lineage, and identified IGF1-PHLDA1 axis to regulate human photoreceptor development.


Asunto(s)
Organoides , Células Fotorreceptoras Retinianas Bastones , Humanos , Organoides/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Transcriptoma , Diferenciación Celular/fisiología , Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Madre Embrionarias , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo
12.
Nat Med ; 28(9): 1883-1892, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36109638

RESUMEN

The storage of facial images in medical records poses privacy risks due to the sensitive nature of the personal biometric information that can be extracted from such images. To minimize these risks, we developed a new technology, called the digital mask (DM), which is based on three-dimensional reconstruction and deep-learning algorithms to irreversibly erase identifiable features, while retaining disease-relevant features needed for diagnosis. In a prospective clinical study to evaluate the technology for diagnosis of ocular conditions, we found very high diagnostic consistency between the use of original and reconstructed facial videos (κ ≥ 0.845 for strabismus, ptosis and nystagmus, and κ = 0.801 for thyroid-associated orbitopathy) and comparable diagnostic accuracy (P ≥ 0.131 for all ocular conditions tested) was observed. Identity removal validation using multiple-choice questions showed that compared to image cropping, the DM could much more effectively remove identity attributes from facial images. We further confirmed the ability of the DM to evade recognition systems using artificial intelligence-powered re-identification algorithms. Moreover, use of the DM increased the willingness of patients with ocular conditions to provide their facial images as health information during medical treatment. These results indicate the potential of the DM algorithm to protect the privacy of patients' facial images in an era of rapid adoption of digital health technologies.


Asunto(s)
Inteligencia Artificial , Privacidad , Algoritmos , Confidencialidad , Cara , Humanos , Estudios Prospectivos
13.
Front Cell Dev Biol ; 9: 680947, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34434927

RESUMEN

Diabetic retinopathy, as one of the common complications of diabetes mellitus, is the leading cause of blindness in the working-age population worldwide. The disease is characterized by damage to retinal vasculature, which is associated with the activation of retina microglial and induces chronic neurodegeneration. Previous studies have identified the effects of activated microglial on the retinal neurons, but the cellular and molecular mechanisms underlying microglial activation is largely unknown. Here, we performed scRNA-seq on the retina of non-human primates with diabetes mellitus, and identified cell-type-specific molecular changes of the six major cell types. By identifying the ligand-receptor expression patterns among different cells, we established the interactome of the whole retina. The data showed that TNF-α signal mediated the activation of microglia through an autocrine manner. And we found TGFß2, which was upregulated in cone dramatically by hyperglycemia, inhibited microglia activation at the early stage of diabetic retinopathy. In summary, our study is the first to profile cell-specific molecular changes and the cell-cell interactome of retina under diabetes mellitus, paving a way to dissect the cellular and molecular mechanisms underlying early-stage diabetic retinopathy.

14.
Invest Ophthalmol Vis Sci ; 62(3): 30, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33749721

RESUMEN

Purpose: This study aims to reveal retinal abnormities in a spontaneous diabetic nonhuman primate model and explore the mechanism of featured injuries. Methods: Twenty-eight cynomolgus monkeys were identified to suffer from spontaneous type 2 diabetes from a colony of more than eight-hundred aged monkeys, and twenty-six age-matched ones were chosen as controls. Their blood biochemistry profiles were determined and retinal changes were examined by multimodal imaging, hematoxylin and eosin staining, and immunofluorescence. Retinal pigment epithelium (RPE) cells were further investigated by RNA sequencing and computational analyses. Results: These diabetic monkeys were characterized by early retinal vascular and neural damage and dyslipidemia. The typical acellular capillaries and pericyte ghost were found in the diabetic retina, which also exhibited reduced retinal nerve fiber layer thickness compared to controls (all P < 0.05). Of note, distinct sub-RPE drusenoid lesions were extensively observed in these diabetic monkeys (46.43% vs. 7.69%), and complements including C3 and C5b-9 were deposited in these lesions. RNA-seq analysis revealed complement activation, AGE/RAGE activation and inflammatory response in diabetic RPE cells. Consistently, the plasma C3 and C4 were particularly increased in the diabetic monkeys with drusenoid lesions (P = 0.028 and 0.029). Conclusions: The spontaneous type 2 diabetic monkeys featured with early-stage retinopathy including not only typical vascular and neural damage but also a distinct sub-RPE deposition. The complement activation of RPE cells in response to hyperglycemia might contribute to the deposition, revealing an unrecognized role of RPE cells in the early-stage pathological process of diabetic retinopathy.


Asunto(s)
Complemento C3/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Retinopatía Diabética/metabolismo , Modelos Animales de Enfermedad , Epitelio Pigmentado de la Retina/metabolismo , Animales , Activación de Complemento , Diabetes Mellitus Tipo 2 , Retinopatía Diabética/diagnóstico , Dislipidemias/diagnóstico , Hiperglucemia/diagnóstico , Macaca fascicularis , Imagen Multimodal , Drusas Retinianas/diagnóstico , Epitelio Pigmentado de la Retina/patología , Vasos Retinianos/patología
15.
Nat Commun ; 11(1): 5148, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-33051455

RESUMEN

The differences in transcription start sites (TSS) and transcription end sites (TES) among gene isoforms can affect the stability, localization, and translation efficiency of mRNA. Gene isoforms allow a single gene diverse functions across different cell types, and isoform dynamics allow different functions over time. However, methods to efficiently identify and quantify RNA isoforms genome-wide in single cells are still lacking. Here, we introduce single cell RNA Cap And Tail sequencing (scRCAT-seq), a method to demarcate the boundaries of isoforms based on short-read sequencing, with higher efficiency and lower cost than existing long-read sequencing methods. In conjunction with machine learning algorithms, scRCAT-seq demarcates RNA transcripts with unprecedented accuracy. We identified hundreds of previously uncharacterized transcripts and thousands of alternative transcripts for known genes, revealed cell-type specific isoforms for various cell types across different species, and generated a cell atlas of isoform dynamics during the development of retinal cones.


Asunto(s)
Caperuzas de ARN/genética , Isoformas de ARN/genética , Análisis de Secuencia de ARN/métodos , Transcripción Genética , Humanos , Análisis de Secuencia de ARN/economía , Análisis de la Célula Individual , Especificidad de la Especie , Sitio de Iniciación de la Transcripción
16.
Sci Adv ; 6(22): eaaz5858, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32523990

RESUMEN

Neural organoids provide a powerful tool for investigating neural development, modeling neural diseases, screening drugs, and developing cell-based therapies. Somatic cells have previously been reprogrammed by transcription factors (TFs) into sensory ganglion (SG) neurons but not SG organoids. We identify a combination of triple TFs Ascl1, Brn3b/3a, and Isl1 (ABI) as an efficient means to reprogram mouse and human fibroblasts into self-organized and networked induced SG (iSG) organoids. The iSG neurons exhibit molecular features, subtype diversity, electrophysiological and calcium response properties, and innervation patterns characteristic of peripheral sensory neurons. Moreover, we have defined retinal ganglion cell (RGC)-specific identifiers to demonstrate the ability for ABI to reprogram induced RGCs (iRGCs) from fibroblasts. Unlike iSG neurons, iRGCs maintain a scattering distribution pattern characteristic of endogenous RGCs. iSG organoids may serve as a model to decipher the pathogenesis of sensorineural diseases and screen effective drugs and a source for cell replacement therapy.


Asunto(s)
Organoides , Células Ganglionares de la Retina , Animales , Fibroblastos/fisiología , Ganglios Sensoriales , Ratones , Células Ganglionares de la Retina/fisiología , Factores de Transcripción/genética
17.
Acta Biochim Biophys Sin (Shanghai) ; 41(12): 1053-60, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20011980

RESUMEN

Gene therapy has emerged as a promising approach for the lethal disorder of Duchenne muscular dystrophy (DMD). Using a novel non-viral delivery system, the human ribosomal DNA (hrDNA) targeting vector, we targeted a minidystrophin-GFP fusion gene into the hrDNA locus of HT1080 cells with a high site-specific integrated efficiency of 10(-5), in which the transgene could express efficiently and continuously. The minidystrophin-GFP fusion protein was easily found to localize on the plasma membrane of HT1080 cells, indicating its possible physiologic performance. Our findings showed that the hrDNA-targeting vector might be highly useful for DMD gene therapy study.


Asunto(s)
ADN Ribosómico/genética , Distrofina/genética , Marcación de Gen , Terapia Genética , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Western Blotting , Distrofina/metabolismo , Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Distrofia Muscular de Duchenne/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transgenes/genética , Células Tumorales Cultivadas
18.
Precis Clin Med ; 2(3): 131-139, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31598384

RESUMEN

Circulating cell-free DNAs (cfDNAs) are fragmented DNA molecules released into the blood by cells. Previous studies have suggested that mitochondria-originated cfDNA fragments (mt-cfDNAs) in cancer patients are more fragmented than those from healthy controls. However, it is still unknown where these short mt-cfDNAs originate, and whether the length of mt-cfDNAs can be correlated with tumor burden and cancer progression. In this study, we first performed whole-genome sequencing analysis (WGS) of cfDNAs from a human tumor cell line-xenotransplantation mouse model and found that mt-cfDNAs released from transplanted tumor cells were shorter than the mouse counterpart. We next analyzed blood cfDNA samples from hepatocellular carcinoma and prostate cancer patients and found that mt-cfDNA lengths were inversely related to tumor size as well as the concentration of circulating tumor DNA. Our study suggested that monitoring the size of mt-cfDNAs in cancer patients would be a useful way to estimate tumor burden and cancer progression.

19.
Methods Mol Biol ; 1979: 363-377, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31028648

RESUMEN

Single-cell transcriptome and single-cell methylome analysis have successfully revealed the heterogeneity in transcriptome and DNA methylome between single cells, and have become powerful tools to understand the dynamics of transcriptome and DNA methylome during the complicated biological processes, such as differentiation and carcinogenesis.Inspired by the success of using these single-cell -omics methods to understand the regulation of a particular "-ome," more interests have been put on elucidating the regulatory relationship among multiple-omics at single-cell resolution. The simultaneous profiling of multiple-omics from the same single cell would provide us the ultimate power to understand the relationship among different "-omes," but this idea is not materialized for decades due to difficulties to assay extremely tiny amount of DNA or RNA in a single cell.To address this technical challenge, we have recently developed a novel method named scMT-seq that can simultaneously profile both DNA methylome and RNA transcriptome from the same cell. This method enabled us to measure, from a single cell, the DNA methylation status of the most informative 0.5-1 million CpG sites and mRNA level of 10,000 genes, of which 3200 genes can be further analyzed with both promoter DNA methylation and RNA transcription. Using the scMT-seq data, we have successfully shown the regulatory relationship between DNA methylation and transcriptional level in a single dorsal root ganglion neuron (Hu et al., Genome Biol 17:88, 2016). We believe the scMT-seq would be a powerful technique to uncover the regulatory mechanism between transcription and DNA methylation, and would be of wide interest beyond the epigenetics community.


Asunto(s)
Metilación de ADN , Epigenómica/métodos , Perfilación de la Expresión Génica/métodos , ARN Mensajero/genética , Análisis de la Célula Individual/métodos , Animales , Islas de CpG , ADN/genética , Epigénesis Genética , Humanos , Reacción en Cadena de la Polimerasa/métodos , Análisis de Secuencia de ARN/métodos , Programas Informáticos , Transcriptoma
20.
Stem Cell Reports ; 13(4): 747-760, 2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31543471

RESUMEN

The development of the mammalian retina is a complicated process involving the generation of distinct types of neurons from retinal progenitor cells (RPCs) in a spatiotemporal-specific manner. The progression of RPCs during retinogenesis includes RPC proliferation, cell-fate commitment, and specific neuronal differentiation. In this study, by performing single-cell RNA sequencing of cells isolated from human embryonic stem cell (hESC)-derived 3D retinal organoids, we successfully deconstructed the temporal progression of RPCs during early human retinogenesis. We identified two distinctive subtypes of RPCs with unique molecular profiles, namely multipotent RPCs and neurogenic RPCs. We found that genes related to the Notch and Wnt signaling pathways, as well as chromatin remodeling, were dynamically regulated during RPC commitment. Interestingly, our analysis identified that CCND1, a G1-phase cell-cycle regulator, was coexpressed with ASCL1 in a cell-cycle-independent manner. Temporally controlled overexpression of CCND1 in retinal organoids demonstrated a role for CCND1 in promoting early retinal neurogenesis. Together, our results revealed critical pathways and novel genes in early retinogenesis of humans.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Organogénesis/genética , Organoides , Retina/citología , Retina/metabolismo , Biomarcadores , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inmunofenotipificación , Análisis de la Célula Individual , Técnicas de Cultivo de Tejidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA