Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
1.
Int J Mol Sci ; 25(2)2024 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-38255817

RESUMEN

Demyelination in the central nervous system (CNS) resulting from injury or disease can cause loss of nerve function and paralysis. Cell therapies intended to promote remyelination of axons are a promising avenue of treatment, with mesenchymal stromal cells (MSCs) a prominent candidate. We have previously demonstrated that MSCs derived from human olfactory mucosa (hOM-MSCs) promote myelination to a greater extent than bone marrow-derived MSCs (hBM-MSCs). However, hOM-MSCs were developed using methods and materials that were not good manufacturing practice (GMP)-compliant. Before considering these cells for clinical use, it is necessary to develop a method for their isolation and expansion that is readily adaptable to a GMP-compliant environment. We demonstrate here that hOM-MSCs can be derived without enzymatic tissue digestion or cell sorting and without culture antibiotics. They grow readily in GMP-compliant media and express typical MSC surface markers. They robustly produce CXCL12 (a key secretory factor in promoting myelination) and are pro-myelinating in in vitro rodent CNS cultures. GMP-compliant hOM-MSCs are comparable in this respect to those grown in non-GMP conditions. However, when assessed in an in vivo model of demyelinating disease (experimental autoimmune encephalitis, EAE), they do not significantly improve disease scores compared with controls, indicating further pre-clinical evaluation is necessary before their advancement to clinical trials.


Asunto(s)
Antibacterianos , Células Madre Mesenquimatosas , Humanos , Técnicas de Cultivo , Axones , Transporte Biológico
2.
Glia ; 71(7): 1683-1698, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36945189

RESUMEN

There is an urgent need for therapies that target the multicellular pathology of central nervous system (CNS) disease. Modified, nonanticoagulant heparins mimic the heparan sulfate glycan family and are known regulators of multiple cellular processes. In vitro studies have demonstrated that low sulfated modified heparin mimetics (LS-mHeps) drive repair after CNS demyelination. Herein, we test LS-mHep7 (an in vitro lead compound) in experimental autoimmune encephalomyelitis (EAE) and cuprizone-induced demyelination. In EAE, LS-mHep7 treatment resulted in faster recovery and rapidly reduced inflammation which was accompanied by restoration of animal weight. LS-mHep7 treatment had no effect on remyelination or on OLIG2 positive oligodendrocyte numbers within the corpus callosum in the cuprizone model. Further in vitro investigation confirmed that LS-mHep7 likely mediates its pro-repair effect in the EAE model by sequestering inflammatory cytokines, such as CCL5 which are upregulated during immune-mediated inflammatory attacks. These data support the future clinical translation of this next generation modified heparin as a treatment for CNS diseases with active immune system involvement.


Asunto(s)
Enfermedades del Sistema Nervioso Central , Encefalomielitis Autoinmune Experimental , Animales , Ratones , Cuprizona/toxicidad , Sulfatos/efectos adversos , Oligodendroglía/patología , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/patología , Cuerpo Calloso/patología , Enfermedades del Sistema Nervioso Central/patología , Heparitina Sulfato/uso terapéutico , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Vaina de Mielina/patología
3.
Biology (Basel) ; 12(3)2023 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-36979099

RESUMEN

Therapies that target the multicellular pathology of central nervous system (CNS) disease/injury are urgently required. Modified non-anticoagulant heparins mimic the heparan sulphate (HS) glycan family and have been proposed as therapeutics for CNS repair since they are effective regulators of numerous cellular processes. Our in vitro studies have demonstrated that low-sulphated modified heparan sulphate mimetics (LS-mHeps) drive CNS repair. However, LS-mHeps are derived from pharmaceutical heparin purified from pig intestines, in a supply chain at risk of shortages and contamination. Alternatively, cellular synthesis of heparin and HS can be achieved using mammalian cell multiplex genome engineering, providing an alternative source of recombinant HS mimetics (rHS). TEGA Therapeutics (San Diego) have manufactured rHS reagents with varying degrees of sulphation and we have validated their ability to promote repair in vitro using models that mimic CNS injury, making comparisons to LS-mHep7, a previous lead compound. We have shown that like LS-mHep7, low-sulphated rHS compounds promote remyelination and reduce features of astrocytosis, and in contrast, highly sulphated rHS drive neurite outgrowth. Cellular production of heparin mimetics may, therefore, offer potential clinical benefits for CNS repair.

4.
Methods Mol Biol ; 2429: 333-344, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35507171

RESUMEN

Striatum-derived neural stem cells have been used to generate a variety of neural cell populations. They are composed of free-floating clusters of clonal neural stem cells, termed neurospheres, and can be expanded under growth factor stimulation in vitro. The multipotent nature of neurospheres means that under certain growth conditions they can differentiate into neuronal and glial progenitors of the central nervous system (CNS).Here, we describe a method for creating a population of astrocytes derived from rat striatum neurospheres, which in turn can be used to generate astrocytes with different reactivity phenotypes. Several methods and techniques are already available for the generation of neurospheres, but the method detailed herein provides an accessible, reproducible protocol for large numbers of astrocyte cultures, that can then be manipulated in an experimental format for further investigation.


Asunto(s)
Astrocitos , Células-Madre Neurales , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Inmunohistoquímica , Fenotipo , Ratas
5.
Exp Neurol ; 354: 114113, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35569511

RESUMEN

Iron released from oligodendrocytes during demyelination or derived from haemoglobin breakdown products is believed to amplify oxidative tissue injury in multiple sclerosis (MS). However, the pathophysiological significance of iron-containing haemoglobin breakdown products themselves is rarely considered in the context of MS and their cellular specificity and mode of action remain unclear. Using myelinating cell cultures, we now report the cytotoxic potential of hemin (ferriprotoporphyrin IX chloride), a major degradation product of haemoglobin, is 25-fold greater than equimolar concentrations of free iron in myelinating cultures; a model that reproduces the complex multicellular environment of the CNS. At low micro molar concentrations (3.3 - 10 µM) we observed hemin preferentially binds to myelin and axons to initiate a complex detrimental response that results in targeted demyelination and axonal loss but spares neuronal cell bodies, astrocytes and the majority of oligodendroglia. Demyelination and axonal loss in this context are executed by a combination of mechanisms that include iron-dependent peroxidation by reactive oxygen species (ROS) and ferroptosis. These effects are microglial-independent, do not require any initiating inflammatory insult and represent a direct effect that compromises the structural integrity of myelinated axons in the CNS. Our data identify hemin-mediated demyelination and axonal loss as a novel mechanism by which intracerebral degradation of haemoglobin may contribute to lesion development in MS.


Asunto(s)
Hemina , Esclerosis Múltiple , Axones/patología , Sistema Nervioso Central/patología , Hemina/metabolismo , Hemina/farmacología , Humanos , Hierro/metabolismo , Esclerosis Múltiple/patología , Vaina de Mielina/patología , Oligodendroglía/metabolismo , Estrés Oxidativo
6.
Acta Neuropathol Commun ; 10(1): 12, 2022 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-35093166

RESUMEN

One of the therapeutic approaches for the treatment of the autoimmune demyelinating disease, multiple sclerosis (MS) is bone marrow mesenchymal stromal cell (hBM-MSCs) transplantation. However, given their capacity to enhance myelination in vitro, we hypothesised that human olfactory mucosa-derived MSCs (hOM-MSCs) may possess additional properties suitable for CNS repair. Herein, we have examined the efficacy of hOM-MSCs versus hBM-MSCs using the experimental autoimmune encephalomyelitis (EAE) model. Both MSC types ameliorated disease, if delivered during the initial onset of symptomatic disease. Yet, only hOM-MSCs improved disease outcome if administered during established disease when animals had severe neurological deficits. Histological analysis of spinal cord lesions revealed hOM-MSC transplantation reduced blood-brain barrier disruption and inflammatory cell recruitment and enhanced axonal survival. At early time points post-hOM-MSC treatment, animals had reduced levels of circulating IL-16, which was reflected in both the ability of immune cells to secrete IL-16 and the level of IL-16 in spinal cord inflammatory lesions. Further in vitro investigation revealed an inhibitory role for IL-16 on oligodendrocyte differentiation and myelination. Moreover, the availability of bioactive IL-16 after demyelination was reduced in the presence of hOM-MSCs. Combined, our data suggests that human hOM-MSCs may have therapeutic benefit in the treatment of MS via an IL-16-mediated pathway, especially if administered during active demyelination and inflammation.


Asunto(s)
Encefalomielitis Autoinmune Experimental/terapia , Interleucina-16/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Vaina de Mielina/metabolismo , Mucosa Olfatoria/citología , Animales , Encefalomielitis Autoinmune Experimental/metabolismo , Humanos , Ratones , Neurogénesis/fisiología
7.
J Med Genet ; 59(4): 358-365, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-33820834

RESUMEN

BACKGROUND: Holoprosencephaly is a spectrum of developmental disorder of the embryonic forebrain in which there is failed or incomplete separation of the prosencephalon into two cerebral hemispheres. To date, dominant mutations in sonic hedgehog (SHH) pathway genes are the predominant Mendelian causes, and have marked interfamilial and intrafamilial phenotypical variabilities. METHODS: We describe two families in which offspring had holoprosencephaly spectrum and homozygous predicted-deleterious variants in phospholipase C eta-1 (PLCH1). Immunocytochemistry was used to examine the expression pattern of PLCH1 in human embryos. We used SHH as a marker of developmental stage and of early embryonic anatomy. RESULTS: In the first family, two siblings had congenital hydrocephalus, significant developmental delay and a monoventricle or fused thalami with a homozygous PLCH1 c.2065C>T, p.(Arg689*) variant. In the second family, two siblings had alobar holoprosencephaly and cyclopia with a homozygous PLCH1 c.4235delA, p.(Cys1079ValfsTer16) variant. All parents were healthy carriers, with no holoprosencephaly spectrum features. We found that the subcellular localisation of PLCH1 is cytoplasmic, but the p.(Cys1079ValfsTer16) variant was predominantly nuclear. Human embryo immunohistochemistry showed PLCH1 to be expressed in the notorcord, developing spinal cord (in a ventral to dorsal gradient), dorsal root ganglia, cerebellum and dermatomyosome, all tissues producing or responding to SHH. Furthermore, the embryonic subcellular localisation of PLCH1 was exclusively cytoplasmic, supporting protein mislocalisation contributing to the pathogenicity of the p.(Cys1079ValfsTer16) variant. CONCLUSION: Our data support the contention that PLCH1 has a role in prenatal mammalian neurodevelopment, and deleterious variants cause a clinically variable holoprosencephaly spectrum phenotype.


Asunto(s)
Holoprosencefalia , Fosfolipasas de Tipo C , Animales , Proteínas Hedgehog/genética , Holoprosencefalia/genética , Holoprosencefalia/metabolismo , Humanos , Mamíferos/metabolismo , Mutación , Fenotipo , Fosfolipasas de Tipo C/genética
8.
Cells ; 10(4)2021 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-33919910

RESUMEN

The use of mesenchymal stem/stromal cells (MSCs) for transplant-mediated repair represents an important and promising therapeutic strategy after spinal cord injury (SCI). The appeal of MSCs has been fuelled by their ease of isolation, immunosuppressive properties, and low immunogenicity, alongside the large variety of available tissue sources. However, despite reported similarities in vitro, MSCs sourced from distinct tissues may not have comparable biological properties in vivo. There is accumulating evidence that stemness, plasticity, immunogenicity, and adaptability of stem cells is largely controlled by tissue niche. The extrinsic impact of cellular niche for MSC repair potential is therefore important, not least because of its impact on ex vivo expansion for therapeutic purposes. It is likely certain niche-targeted MSCs are more suited for SCI transplant-mediated repair due to their intrinsic capabilities, such as inherent neurogenic properties. In addition, the various MSC anatomical locations means that differences in harvest and culture procedures can make cross-comparison of pre-clinical data difficult. Since a clinical grade MSC product is inextricably linked with its manufacture, it is imperative that cells can be made relatively easily using appropriate materials. We discuss these issues and highlight the importance of identifying the appropriate niche-specific MSC type for SCI repair.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Regeneración Nerviosa/fisiología , Traumatismos de la Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/terapia , Nicho de Células Madre , Humanos
9.
Biomater Sci ; 8(13): 3611-3627, 2020 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-32515439

RESUMEN

The limited regenerative capacity of the CNS poses formidable challenges to the repair of spinal cord injury (SCI). Two key barriers to repair are (i) the physical gap left by the injury, and (ii) the inhibitory milieu surrounding the injury, the glial scar. Biomaterial implantation into the injury site can fill the cavity, provide a substrate for cell migration, and potentially attenuate the glial scar. We investigated the biological viability of a biocompatible and biodegradable poly-ε-lysine based biomaterial, Proliferate®, in low and high cross-linked forms and when coated with IKVAV peptide, for SCI implantation. We demonstrate altered astrocyte morphology and nestin expression on Proliferate® compared to conventional glass cell coverslips suggesting a less reactive phenotype. Moreover Proliferate® supported myelination in vitro, with myelination observed sooner on IKVAV-coated constructs compared with uncoated Proliferate®, and delayed overall compared with maintenance on glass coverslips. For in vivo implantation, parallel-aligned channels were fabricated into Proliferate® to provide cell guidance cues. Extensive vascularisation and cellular infiltration were observed in constructs implanted in vivo, along with an astrocyte border and microglial response. Axonal ingrowth was observed at the construct border and inside implants in intact channels. We conclude that Proliferate® is a promising biomaterial for implantation following SCI.


Asunto(s)
Materiales Biocompatibles/química , Enfermedades del Sistema Nervioso Central/terapia , Polilisina/química , Prótesis e Implantes , Traumatismos de la Médula Espinal/terapia , Animales , Materiales Biocompatibles/síntesis química , Células Cultivadas , Polilisina/síntesis química , Ratas , Ratas Sprague-Dawley
10.
Nat Rev Neurol ; 16(4): 229-240, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32099190

RESUMEN

Spinal cord injury (SCI) remains one of the biggest challenges in the development of neuroregenerative therapeutics. Cell transplantation is one of numerous experimental strategies that have been identified and tested for efficacy at both preclinical and clinical levels in recent years. In this Review, we briefly discuss the state of human olfactory cell transplantation as a therapy, considering both its current clinical status and its limitations. Furthermore, we introduce a mesenchymal stromal cell derived from human olfactory tissue, which has the potential to induce multifaceted reparative effects in the environment within and surrounding the lesion. We argue that no single therapy will be sufficient to treat SCI effectively and that a combination of cell-based, rehabilitation and pharmaceutical interventions is the most promising approach to aid repair. For this reason, we also introduce a novel pharmaceutical strategy based on modifying the activity of heparan sulfate, an important regulator of a wide range of biological cell functions. The multi-target approach that is exemplified by these types of strategies will probably be necessary to optimize SCI treatment.


Asunto(s)
Heparitina Sulfato/uso terapéutico , Trasplante de Células Madre Mesenquimatosas/métodos , Mucosa Olfatoria/citología , Traumatismos de la Médula Espinal/terapia , Regeneración de la Medula Espinal , Trasplante de Células/métodos , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Proteoglicanos de Heparán Sulfato/metabolismo , Heparina/uso terapéutico , Heparitina Sulfato/análogos & derivados , Humanos , Células Madre Mesenquimatosas/citología , Regeneración Nerviosa , Neuroglía , Mucosa Olfatoria/fisiología , Neuronas Receptoras Olfatorias
11.
Nature ; 574(7778): 365-371, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31597962

RESUMEN

Definitive haematopoiesis in the fetal liver supports self-renewal and differentiation of haematopoietic stem cells and multipotent progenitors (HSC/MPPs) but remains poorly defined in humans. Here, using single-cell transcriptome profiling of approximately 140,000 liver and 74,000 skin, kidney and yolk sac cells, we identify the repertoire of human blood and immune cells during development. We infer differentiation trajectories from HSC/MPPs and evaluate the influence of the tissue microenvironment on blood and immune cell development. We reveal physiological erythropoiesis in fetal skin and the presence of mast cells, natural killer and innate lymphoid cell precursors in the yolk sac. We demonstrate a shift in the haemopoietic composition of fetal liver during gestation away from being predominantly erythroid, accompanied by a parallel change in differentiation potential of HSC/MPPs, which we functionally validate. Our integrated map of fetal liver haematopoiesis provides a blueprint for the study of paediatric blood and immune disorders, and a reference for harnessing the therapeutic potential of HSC/MPPs.


Asunto(s)
Feto/citología , Hematopoyesis , Hígado/citología , Hígado/embriología , Células Sanguíneas/citología , Microambiente Celular , Femenino , Feto/metabolismo , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Hígado/metabolismo , Tejido Linfoide/citología , Análisis de la Célula Individual , Células Madre/metabolismo
12.
Science ; 365(6460): 1461-1466, 2019 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-31604275

RESUMEN

Tissue-resident immune cells are important for organ homeostasis and defense. The epithelium may contribute to these functions directly or by cross-talk with immune cells. We used single-cell RNA sequencing to resolve the spatiotemporal immune topology of the human kidney. We reveal anatomically defined expression patterns of immune genes within the epithelial compartment, with antimicrobial peptide transcripts evident in pelvic epithelium in the mature, but not fetal, kidney. A network of tissue-resident myeloid and lymphoid immune cells was evident in both fetal and mature kidney, with postnatal acquisition of transcriptional programs that promote infection-defense capabilities. Epithelial-immune cross-talk orchestrated localization of antibacterial macrophages and neutrophils to the regions of the kidney most susceptible to infection. Overall, our study provides a global overview of how the immune landscape of the human kidney is zonated to counter the dominant immunological challenge.


Asunto(s)
Riñón/inmunología , Macrófagos/citología , Neutrófilos/citología , Adulto , Animales , Células Epiteliales/citología , Femenino , Feto , Regulación del Desarrollo de la Expresión Génica , Humanos , Riñón/anatomía & histología , Riñón/citología , Linfocitos/citología , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Mieloides/citología , RNA-Seq , Análisis de la Célula Individual , Infecciones Urinarias/inmunología
13.
Science ; 366(6464): 454-460, 2019 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-31624095

RESUMEN

We present histological and molecular analyses of the developing human cerebellum from 30 days after conception to 9 months after birth. Differences in developmental patterns between humans and mice include spatiotemporal expansion of both ventricular and rhombic lip primary progenitor zones to include subventricular zones containing basal progenitors. The human rhombic lip persists longer through cerebellar development than in the mouse and undergoes morphological changes to form a progenitor pool in the posterior lobule, which is not seen in other organisms, not even in the nonhuman primate the macaque. Disruptions in human rhombic lip development are associated with posterior cerebellar vermis hypoplasia and Dandy-Walker malformation. The presence of these species-specific neural progenitor populations refines our insight into human cerebellar developmental disorders.


Asunto(s)
Cerebelo/embriología , Cerebelo/crecimiento & desarrollo , Células Madre/citología , Animales , Síndrome de Dandy-Walker , Humanos , Ratones , Malformaciones del Sistema Nervioso , Análisis Espacio-Temporal , Especificidad de la Especie , Transcriptoma
14.
Biology (Basel) ; 8(3)2019 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-31261710

RESUMEN

In vitro cell-based assays have been fundamental in modern drug discovery and have led to the identification of novel therapeutics. We have developed complex mixed central nervous system (CNS) cultures, which recapitulate the normal process of myelination over time and allow the study of several parameters associated with CNS damage, both during development and after injury or disease. In particular, they have been used as a reliable screen to identify drug candidates that may promote (re)myelination and/or neurite outgrowth. Previously, using these cultures, we demonstrated that a panel of low sulphated heparin mimetics, with structures similar to heparan sulphates (HSs), can reduce astrogliosis, and promote myelination and neurite outgrowth. HSs reside in either the extracellular matrix or on the surface of cells and are thought to modulate cell signaling by both sequestering ligands, and acting as co-factors in the formation of ligand-receptor complexes. In this study, we have used these cultures as a screen to address the repair potential of numerous other commercially available sulphated glycomolecules, namely heparosans, ulvans, and fucoidans. These compounds are all known to have certain characteristics that mimic cellular glycosaminoglycans, similar to heparin mimetics. We show that the N-sulphated heparosans promoted myelination. However, O-sulphated heparosans did not affect myelination but promoted neurite outgrowth, indicating the importance of structure in HS function. Moreover, neither highly sulphated ulvans nor fucoidans had any effect on remyelination but CX-01, a low sulphated porcine intestinal heparin, promoted remyelination in vitro. These data illustrate the use of myelinating cultures as a screen and demonstrate the potential of heparin mimetics as CNS therapeutics.

15.
Nature ; 571(7766): 505-509, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31243369

RESUMEN

The evolution of gene expression in mammalian organ development remains largely uncharacterized. Here we report the transcriptomes of seven organs (cerebrum, cerebellum, heart, kidney, liver, ovary and testis) across developmental time points from early organogenesis to adulthood for human, rhesus macaque, mouse, rat, rabbit, opossum and chicken. Comparisons of gene expression patterns identified correspondences of developmental stages across species, and differences in the timing of key events during the development of the gonads. We found that the breadth of gene expression and the extent of purifying selection gradually decrease during development, whereas the amount of positive selection and expression of new genes increase. We identified differences in the temporal trajectories of expression of individual genes across species, with brain tissues showing the smallest percentage of trajectory changes, and the liver and testis showing the largest. Our work provides a resource of developmental transcriptomes of seven organs across seven species, and comparative analyses that characterize the development and evolution of mammalian organs.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Organogénesis/genética , Transcriptoma/genética , Animales , Evolución Biológica , Pollos/genética , Femenino , Humanos , Macaca mulatta/genética , Masculino , Ratones , Zarigüeyas/genética , Conejos , Ratas
17.
Development ; 146(2)2019 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-30696714

RESUMEN

The scarcity of embryonic/foetal material as a resource for direct study means that there is still limited understanding of human retina development. Here, we present an integrated transcriptome analysis combined with immunohistochemistry in human eye and retinal samples from 4 to 19 post-conception weeks. This analysis reveals three developmental windows with specific gene expression patterns that informed the sequential emergence of retinal cell types and enabled identification of stage-specific cellular and biological processes, and transcriptional regulators. Each stage is characterised by a specific set of alternatively spliced transcripts that code for proteins involved in the formation of the photoreceptor connecting cilium, pre-mRNA splicing and epigenetic modifiers. Importantly, our data show that the transition from foetal to adult retina is characterised by a large increase in the percentage of mutually exclusive exons that code for proteins involved in photoreceptor maintenance. The circular RNA population is also defined and shown to increase during retinal development. Collectively, these data increase our understanding of human retinal development and the pre-mRNA splicing process, and help to identify new candidate disease genes.


Asunto(s)
Perfilación de la Expresión Génica , Retina/embriología , Retina/metabolismo , Empalme Alternativo/genética , Animales , Biomarcadores/metabolismo , Cilios/metabolismo , Feto/metabolismo , Regulación del Desarrollo de la Expresión Génica , Organogénesis/genética , Células Fotorreceptoras de Vertebrados/citología , Células Fotorreceptoras de Vertebrados/metabolismo , Análisis de Componente Principal , ARN/genética , ARN/metabolismo , Precursores del ARN/genética , Precursores del ARN/metabolismo , ARN Circular , Retina/citología , Retina/ultraestructura , Transcriptoma/genética
18.
Nat Commun ; 9(1): 4234, 2018 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-30315276

RESUMEN

Mutations in pre-mRNA processing factors (PRPFs) cause autosomal-dominant retinitis pigmentosa (RP), but it is unclear why mutations in ubiquitously expressed genes cause non-syndromic retinal disease. Here, we generate transcriptome profiles from RP11 (PRPF31-mutated) patient-derived retinal organoids and retinal pigment epithelium (RPE), as well as Prpf31+/- mouse tissues, which revealed that disrupted alternative splicing occurred for specific splicing programmes. Mis-splicing of genes encoding pre-mRNA splicing proteins was limited to patient-specific retinal cells and Prpf31+/- mouse retinae and RPE. Mis-splicing of genes implicated in ciliogenesis and cellular adhesion was associated with severe RPE defects that include disrupted apical - basal polarity, reduced trans-epithelial resistance and phagocytic capacity, and decreased cilia length and incidence. Disrupted cilia morphology also occurred in patient-derived photoreceptors, associated with progressive degeneration and cellular stress. In situ gene editing of a pathogenic mutation rescued protein expression and key cellular phenotypes in RPE and photoreceptors, providing proof of concept for future therapeutic strategies.


Asunto(s)
Proteínas del Ojo/metabolismo , Retinitis Pigmentosa/etiología , Retinitis Pigmentosa/metabolismo , Empalme Alternativo/genética , Empalme Alternativo/fisiología , Animales , Adhesión Celular/genética , Adhesión Celular/fisiología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Cilios/genética , Cilios/metabolismo , Cilios/fisiología , Proteínas del Ojo/genética , Citometría de Flujo , Humanos , Inmunohistoquímica , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Mutación/genética , Organoides/citología , Organoides/metabolismo , Empalme del ARN/genética , Empalme del ARN/fisiología , Retina/citología , Retina/metabolismo , Retinitis Pigmentosa/genética
19.
Science ; 361(6402): 594-599, 2018 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-30093597

RESUMEN

Messenger RNA encodes cellular function and phenotype. In the context of human cancer, it defines the identities of malignant cells and the diversity of tumor tissue. We studied 72,501 single-cell transcriptomes of human renal tumors and normal tissue from fetal, pediatric, and adult kidneys. We matched childhood Wilms tumor with specific fetal cell types, thus providing evidence for the hypothesis that Wilms tumor cells are aberrant fetal cells. In adult renal cell carcinoma, we identified a canonical cancer transcriptome that matched a little-known subtype of proximal convoluted tubular cell. Analyses of the tumor composition defined cancer-associated normal cells and delineated a complex vascular endothelial growth factor (VEGF) signaling circuit. Our findings reveal the precise cellular identities and compositions of human kidney tumors.


Asunto(s)
Neoplasias Renales/genética , Neoplasias Renales/patología , Riñón/metabolismo , Transcriptoma , Adulto , Carcinoma de Células Renales/clasificación , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Niño , Variación Genética , Humanos , Riñón/embriología , Neoplasias Renales/clasificación , Análisis de la Célula Individual , Tumor de Wilms/clasificación , Tumor de Wilms/genética , Tumor de Wilms/patología
20.
Acta Biomater ; 74: 207-221, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29777959

RESUMEN

The extracellular matrix (ECM) plays an important role in numerous processes including cellular proliferation, differentiation, migration, maturation, adhesion guidance and axonal growth. To date, there has been no detailed analysis of the ECM distribution during retinal ontogenesis in humans and the functional importance of many ECM components is poorly understood. In this study, the expression of key ECM components in adult mouse and monkey retina, developing and adult human retina and retinal organoids derived from human pluripotent stem cells was studied. Our data indicate that basement membrane ECMs (Fibronectin and Collagen IV) were expressed in Bruch's membrane and the inner limiting membrane of the developing human retina, whilst the hyalectins (Versican and Brevican), cluster of differentiation 44 (CD44), photoreceptor-specific ECMs Interphotoreceptor Matrix Proteoglycan 1 (IMPG1) and Interphotoreceptor Matrix Proteoglycan 2 (IMPG2) were detected in the developing interphotoreceptor matrix (IPM). The expression of IMPG1, Versican and Brevican in the developing IPM was conserved between human developing retina and human pluripotent stem cell-derived retinal organoids. Blocking the action of CD44 and IMPG1 in pluripotent stem cell derived retinal organoids affected the development of photoreceptors, their inner/outer segments and connecting cilia and disrupted IPM formation, with IMPG1 having an earlier and more significant impact. Together, our data suggest an important role for IMPG1 and CD44 in the development of photoreceptors and IPM formation during human retinogenesis. STATEMENT OF SIGNIFICANCE: The expression and the role of many extracellular matrix (ECM) components during human retinal development is not fully understood. In this study, expression of key ECM components (Collagen IV, Fibronectin, Brevican, Versican, IMPG1 and IMPG2) was investigated during human retinal ontogenesis. Collagen IV and Fibronectin were expressed in Bruch's membrane; whereas Brevican, Versican, IMPG1 & IMPG2 in the developing interphotoreceptor matrix (IPM). Retinal organoids were successfully generated from pluripotent stem cells. The expression of ECM components was examined in the retinal organoids and found to recapitulate human retinal development in vivo. Using functional blocking experiments, we were able to highlight an important role for IMPG1 and CD44 in the development of photoreceptors and IPM formation.


Asunto(s)
Proteínas de la Matriz Extracelular/metabolismo , Matriz Extracelular/clasificación , Proteínas del Ojo/metabolismo , Receptores de Hialuranos/metabolismo , Organoides/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Células Madre Pluripotentes/metabolismo , Proteoglicanos/metabolismo , Animales , Humanos , Macaca , Ratones , Organoides/citología , Células Fotorreceptoras de Vertebrados/citología , Células Madre Pluripotentes/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA