Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Nat Commun ; 15(1): 4163, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38755145

RESUMEN

TAR DNA-binding protein 43 (TDP-43) proteinopathy in brain cells is the hallmark of amyotrophic lateral sclerosis (ALS) but its cause remains elusive. Asparaginase-like-1 protein (ASRGL1) cleaves isoaspartates, which alter protein folding and susceptibility to proteolysis. ASRGL1 gene harbors a copy of the human endogenous retrovirus HML-2, whose overexpression contributes to ALS pathogenesis. Here we show that ASRGL1 expression was diminished in ALS brain samples by RNA sequencing, immunohistochemistry, and western blotting. TDP-43 and ASRGL1 colocalized in neurons but, in the absence of ASRGL1, TDP-43 aggregated in the cytoplasm. TDP-43 was found to be prone to isoaspartate formation and a substrate for ASRGL1. ASRGL1 silencing triggered accumulation of misfolded, fragmented, phosphorylated and mislocalized TDP-43 in cultured neurons and motor cortex of female mice. Overexpression of ASRGL1 restored neuronal viability. Overexpression of HML-2 led to ASRGL1 silencing. Loss of ASRGL1 leading to TDP-43 aggregation may be a critical mechanism in ALS pathophysiology.


Asunto(s)
Esclerosis Amiotrófica Lateral , Proteínas de Unión al ADN , Neuronas , Proteinopatías TDP-43 , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Humanos , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Ratones , Femenino , Proteinopatías TDP-43/metabolismo , Proteinopatías TDP-43/patología , Proteinopatías TDP-43/genética , Neuronas/metabolismo , Neuronas/patología , Encéfalo/metabolismo , Encéfalo/patología , Masculino , Corteza Motora/metabolismo , Corteza Motora/patología
2.
Nat Commun ; 15(1): 907, 2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38383456

RESUMEN

Post-infectious myalgic encephalomyelitis/chronic fatigue syndrome (PI-ME/CFS) is a disabling disorder, yet the clinical phenotype is poorly defined, the pathophysiology is unknown, and no disease-modifying treatments are available. We used rigorous criteria to recruit PI-ME/CFS participants with matched controls to conduct deep phenotyping. Among the many physical and cognitive complaints, one defining feature of PI-ME/CFS was an alteration of effort preference, rather than physical or central fatigue, due to dysfunction of integrative brain regions potentially associated with central catechol pathway dysregulation, with consequences on autonomic functioning and physical conditioning. Immune profiling suggested chronic antigenic stimulation with increase in naïve and decrease in switched memory B-cells. Alterations in gene expression profiles of peripheral blood mononuclear cells and metabolic pathways were consistent with cellular phenotypic studies and demonstrated differences according to sex. Together these clinical abnormalities and biomarker differences provide unique insight into the underlying pathophysiology of PI-ME/CFS, which may guide future intervention.


Asunto(s)
Enfermedades Transmisibles , Síndrome de Fatiga Crónica , Humanos , Síndrome de Fatiga Crónica/metabolismo , Leucocitos Mononucleares/metabolismo , Enfermedades Transmisibles/metabolismo , Biomarcadores/metabolismo , Fenotipo
3.
Transl Psychiatry ; 13(1): 397, 2023 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-38104115

RESUMEN

Genome-wide (GWAS) and copy number variant (CNV) association studies have reproducibly identified numerous risk alleles associated with bipolar disorder (BD), major depressive disorder (MDD), and schizophrenia (SCZ), but biological characterization of these alleles lags gene discovery, owing to the inaccessibility of live human brain cells and inadequate animal models for human psychiatric conditions. Human-derived induced pluripotent stem cells (iPSCs) provide a renewable cellular reagent that can be differentiated into living, disease-relevant cells and 3D brain organoids carrying the full complement of genetic variants present in the donor germline. Experimental studies of iPSC-derived cells allow functional characterization of risk alleles, establishment of causal relationships between genes and neurobiology, and screening for novel therapeutics. Here we report the creation and availability of an iPSC resource comprising clinical, genomic, and cellular data obtained from genetically isolated families with BD and related conditions. Results from the first 324 study participants, 61 of whom have validated pluripotent clones, show enrichment of rare single nucleotide variants and CNVs overlapping many known risk genes and pathogenic CNVs. This growing iPSC resource is available to scientists pursuing functional genomic studies of BD and related conditions.


Asunto(s)
Trastorno Depresivo Mayor , Células Madre Pluripotentes Inducidas , Trastornos Psicóticos , Esquizofrenia , Animales , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Trastorno Depresivo Mayor/genética , Trastorno Depresivo Mayor/metabolismo , Trastornos Psicóticos/metabolismo , Esquizofrenia/genética , Esquizofrenia/metabolismo , Genómica , Estudio de Asociación del Genoma Completo
4.
Exp Neurol ; 367: 114469, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37327963

RESUMEN

Prenatal Zika virus (ZIKV) infection is a serious global concern as it can lead to brain injury and many serious birth defects, collectively known as congenital Zika syndrome. Brain injury likely results from viral mediated toxicity in neural progenitor cells. Additionally, postnatal ZIKV infections have been linked to neurological complications, yet the mechanisms driving these manifestations are not well understood. Existing data suggest that the ZIKV envelope protein can persist in the central nervous system for extended periods of time, but it is unknown if this protein can independently contribute to neuronal toxicity. Here we find that the ZIKV envelope protein is neurotoxic, leading to overexpression of poly adenosine diphosphate -ribose polymerase 1, which can induce parthanatos. Together, these data suggest that neuronal toxicity resulting from the envelope protein may contribute to the pathogenesis of post-natal ZIKV-related neurologic complications.


Asunto(s)
Lesiones Encefálicas , Enfermedades del Sistema Nervioso , Síndromes de Neurotoxicidad , Infección por el Virus Zika , Virus Zika , Embarazo , Femenino , Humanos , Virus Zika/metabolismo , Infección por el Virus Zika/complicaciones , Infección por el Virus Zika/patología , Proteínas del Envoltorio Viral/metabolismo , Neuronas/patología
5.
EMBO Rep ; 24(1): e55197, 2023 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-36367221

RESUMEN

Transposable elements (TEs) are active in neuronal cells raising the question whether TE insertions contribute to risk of neuropsychiatric disease. While genome-wide association studies (GWAS) serve as a tool to discover genetic loci associated with neuropsychiatric diseases, unfortunately GWAS do not directly detect structural variants such as TEs. To examine the role of TEs in psychiatric and neurologic disease, we evaluated 17,000 polymorphic TEs and find 76 are in linkage disequilibrium with disease haplotypes (P < 10-6 ) defined by GWAS. From these 76 polymorphic TEs, we identify potentially causal candidates based on having insertions in genomic regions of regulatory chromatin and on having associations with altered gene expression in brain tissues. We show that lead candidate insertions have regulatory effects on gene expression in human neural stem cells altering the activity of a minimal promoter. Taken together, we identify 10 polymorphic TE insertions that are potential candidates on par with other variants for having a causal role in neurologic and psychiatric disorders.


Asunto(s)
Trastornos Mentales , Retroelementos , Humanos , Retroelementos/genética , Estudio de Asociación del Genoma Completo , Genoma , Sitios Genéticos , Trastornos Mentales/genética , Elementos Transponibles de ADN/genética , Evolución Molecular
6.
Ann Neurol ; 92(4): 545-561, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35801347

RESUMEN

OBJECTIVE: Human endogenous retroviruses have been implicated in neurodegenerative diseases including amyotrophic lateral sclerosis (ALS). Expression of human endogenous retrovirus K (HERV-K) subtype HML-2 envelope (Env) in human neuronal cultures and in transgenic mice results in neurotoxicity and neurodegeneration, and mice expressing HML-2 Env display behavioral and neuromuscular characteristics resembling ALS. This study aims to characterize the neurotoxic properties of HML-2 Env. METHODS: Env neurotoxicity was detected in ALS cerebrospinal fluid and confirmed using recombinant Env protein in a cell-based assay and a mouse model. The mechanism of neurotoxicity was assessed with immunoprecipitation followed by mass spectrometry and Western blot, and by screening a panel of inhibitors. RESULTS: We observed that recombinant HML-2 Env protein caused neurotoxicity resulting in neuronal cell death, retraction of neurites, and decreased neuronal electrical activity. Injection of the Env protein into the brains of mice also resulted in neuronal cell death. HML-2 Env protein was also found in the cerebrospinal fluid of patients with sporadic ALS. The neurotoxic properties of the Env and the cerebrospinal fluid could be rescued with the anti-Env antibody. The Env was found to bind to CD98HC complexed to ß1 integrin on the neuronal cell surface. Using a panel of compounds to screen for their ability to block Env-induced neurotoxicity, we found that several compounds were protective and are linked to the ß1 integrin pathway. INTERPRETATION: HERV-K Env is released extracellularly in ALS and causes neurotoxicity via a novel mechanism. Present results pave the way for new treatment strategies in sporadic ALS. ANN NEUROL 2022;92:545-561.


Asunto(s)
Esclerosis Amiotrófica Lateral , Retrovirus Endógenos , Esclerosis Amiotrófica Lateral/genética , Animales , Productos del Gen env , Humanos , Integrina beta1 , Ratones , Ratones Transgénicos
7.
Neurotherapeutics ; 19(4): 1313-1328, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35831747

RESUMEN

There is a continuing unmet medical need to develop neuroprotective strategies to treat neurodegenerative disorders. To address this need, we screened over 2000 compounds for potential neuroprotective activity in a model of oxidative stress and found that numerous antifungal agents were neuroprotective. Of the identified compounds, fluconazole was further characterized. Fluconazole was able to prevent neurite retraction and cell death in in vitro and in vivo models of toxicity. Fluconazole protected neurons in a concentration-dependent manner and exhibited efficacy against several toxic agents, including 3-nitropropionic acid, N-methyl D-aspartate, 6-hydroxydopamine, and the HIV proteins Tat and gp120. In vivo studies indicated that systemically administered fluconazole was neuroprotective in animals treated with 3-nitropropionic acid and prevented gp120-mediated neuronal loss. In addition to neuroprotection, fluconazole also induced proliferation of neural progenitor cells in vitro and in vivo. Fluconazole mediates these effects through upregulation and signaling via the insulin growth factor-1 receptor which results in decreased cAMP production and increased phosphorylation of Akt. Blockade of the insulin growth factor-1 receptor signaling with the selective inhibitor AG1024 abrogated the effects of fluconazole. Our studies suggest that fluconazole may be an attractive candidate for treatment of neurodegenerative diseases due to its protective properties against several categories of neuronal insults and its ability to spur neural progenitor cell proliferation.


Asunto(s)
Insulinas , Enfermedades Neurodegenerativas , Fármacos Neuroprotectores , Animales , Receptor IGF Tipo 1/metabolismo , Neuroprotección , Fluconazol/farmacología , Fármacos Neuroprotectores/farmacología , Proteínas Proto-Oncogénicas c-akt , Oxidopamina , Antifúngicos , Ácido D-Aspártico
8.
Proc Natl Acad Sci U S A ; 117(49): 31365-31375, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33229545

RESUMEN

When Zika virus emerged as a public health emergency there were no drugs or vaccines approved for its prevention or treatment. We used a high-throughput screen for Zika virus protease inhibitors to identify several inhibitors of Zika virus infection. We expressed the NS2B-NS3 Zika virus protease and conducted a biochemical screen for small-molecule inhibitors. A quantitative structure-activity relationship model was employed to virtually screen ∼138,000 compounds, which increased the identification of active compounds, while decreasing screening time and resources. Candidate inhibitors were validated in several viral infection assays. Small molecules with favorable clinical profiles, especially the five-lipoxygenase-activating protein inhibitor, MK-591, inhibited the Zika virus protease and infection in neural stem cells. Members of the tetracycline family of antibiotics were more potent inhibitors of Zika virus infection than the protease, suggesting they may have multiple mechanisms of action. The most potent tetracycline, methacycline, reduced the amount of Zika virus present in the brain and the severity of Zika virus-induced motor deficits in an immunocompetent mouse model. As Food and Drug Administration-approved drugs, the tetracyclines could be quickly translated to the clinic. The compounds identified through our screening paradigm have the potential to be used as prophylactics for patients traveling to endemic regions or for the treatment of the neurological complications of Zika virus infection.


Asunto(s)
Antivirales/análisis , Antivirales/farmacología , Evaluación Preclínica de Medicamentos , Ensayos Analíticos de Alto Rendimiento , Inhibidores de Proteasas/análisis , Inhibidores de Proteasas/farmacología , Virus Zika/efectos de los fármacos , Animales , Antivirales/uso terapéutico , Inteligencia Artificial , Chlorocebus aethiops , Modelos Animales de Enfermedad , Inmunocompetencia , Concentración 50 Inhibidora , Metaciclina/farmacología , Ratones Endogámicos C57BL , Inhibidores de Proteasas/uso terapéutico , Relación Estructura-Actividad Cuantitativa , Bibliotecas de Moléculas Pequeñas , Células Vero , Infección por el Virus Zika/tratamiento farmacológico , Infección por el Virus Zika/virología
9.
Proc Natl Acad Sci U S A ; 117(30): 17842-17853, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32669437

RESUMEN

Stem cells are capable of unlimited proliferation but can be induced to form brain cells. Factors that specifically regulate human development are poorly understood. We found that human stem cells expressed high levels of the envelope protein of an endogenized human-specific retrovirus (HERV-K, HML-2) from loci in chromosomes 12 and 19. The envelope protein was expressed on the cell membrane of the stem cells and was critical in maintaining the stemness via interactions with CD98HC, leading to triggering of human-specific signaling pathways involving mammalian target of rapamycin (mTOR) and lysophosphatidylcholine acyltransferase (LPCAT1)-mediated epigenetic changes. Down-regulation or epigenetic silencing of HML-2 env resulted in dissociation of the stem cell colonies and enhanced differentiation along neuronal pathways. Thus HML-2 regulation is critical for human embryonic and neurodevelopment, while it's dysregulation may play a role in tumorigenesis and neurodegeneration.


Asunto(s)
Diferenciación Celular , Retrovirus Endógenos/fisiología , Neuronas/metabolismo , Transducción de Señal , Células Madre/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Biomarcadores , Diferenciación Celular/genética , Autorrenovación de las Células/genética , Cadena Pesada de la Proteína-1 Reguladora de Fusión/metabolismo , Regulación Viral de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuronas/citología , Unión Proteica , Células Madre/citología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas del Envoltorio Viral/genética
10.
J Vis Exp ; (151)2019 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-31589204

RESUMEN

Cytotoxicity is a critical parameter that needs to be quantified when studying drugs that may have therapeutic benefits. Because of this, many drug screening assays utilize cytotoxicity as one of the critical characteristics to be profiled for individual compounds. Cells in culture are a useful model to assess cytotoxicity before proceeding to follow up on promising lead compounds in more costly and labor-intensive animal models. We describe a strategy to identify compounds that affect cell growth in a tdTomato expressing human neural stem cells (NSC) line. The strategy uses two complementary assays to assess cell number. One assay works via the reduction of 3-(4,5-dimethylthizol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) to formazan as a proxy for cell number and the other directly counts the tdTomato expressing NSCs. The two assays can be performed simultaneously in a single experiment and are not labor intensive, rapid, and inexpensive. The strategy described in this demonstration tested 57 compounds in an exploratory primary screen for toxicity in a 96-well plate format. Three of the hits were characterized further in a six-point dose response using the same assay set-up as the primary screen. In addition to providing excellent corroboration for toxicity, comparison of results from the two assays may be effective in identifying compounds affecting other aspects of cell growth.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Medios de Cultivo/química , Animales , Recuento de Células , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Dimetilsulfóxido/toxicidad , Humanos , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos
11.
J Thorac Dis ; 10(Suppl 33): S4057-S4060, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30631554
12.
Front Microbiol ; 8: 113, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28210244

RESUMEN

Olive leaf extract (OLE) has been used traditionally as a herbal supplement since it contains polyphenolic compounds with beneficial properties ranging from increasing energy levels, lowering blood pressure, and supporting the cardiovascular and immune systems. In addition to the beneficial effects on human health, OLE also has antimicrobial properties. The aim of this work was to investigate the antimicrobial effect of OLE against major foodborne pathogens, including Listeria monocytogenes, Escherichia coli O157:H7, and Salmonella Enteritidis. Our results demonstrated that at a concentration of 62.5 mg/ml, OLE almost completely inhibited the growth of these three pathogens. In addition, OLE also reduced cell motility in L. monocytogenes, which correlated with the absence of flagella as shown by scanning electron microscopy. Moreover, OLE inhibited biofilm formation in L. monocytogenes and S. Enteritidis. Taken together, OLE, as a natural product, has the potential to be used as an antimicrobial to control foodborne pathogens.

13.
Stem Cells Transl Med ; 5(5): 613-27, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27034412

RESUMEN

UNLABELLED: Astrocytes are the predominant cell type in the nervous system and play a significant role in maintaining neuronal health and homeostasis. Recently, astrocyte dysfunction has been implicated in the pathogenesis of many neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. Astrocytes are thus an attractive new target for drug discovery for neurological disorders. Using astrocytes differentiated from human embryonic stem cells, we have developed an assay to identify compounds that protect against oxidative stress, a condition associated with many neurodegenerative diseases. This phenotypic oxidative stress assay has been optimized for high-throughput screening in a 1,536-well plate format. From a screen of approximately 4,100 bioactive tool compounds and approved drugs, we identified a set of 22 that acutely protect human astrocytes from the consequences of hydrogen peroxide-induced oxidative stress. Nine of these compounds were also found to be protective of induced pluripotent stem cell-differentiated astrocytes in a related assay. These compounds are thought to confer protection through hormesis, activating stress-response pathways and preconditioning astrocytes to handle subsequent exposure to hydrogen peroxide. In fact, four of these compounds were found to activate the antioxidant response element/nuclear factor-E2-related factor 2 pathway, a protective pathway induced by toxic insults. Our results demonstrate the relevancy and utility of using astrocytes differentiated from human stem cells as a disease model for drug discovery and development. SIGNIFICANCE: Astrocytes play a key role in neurological diseases. Drug discovery efforts that target astrocytes can identify novel therapeutics. Human astrocytes are difficult to obtain and thus are challenging to use for high-throughput screening, which requires large numbers of cells. Using human embryonic stem cell-derived astrocytes and an optimized astrocyte differentiation protocol, it was possible to screen approximately 4,100 compounds in titration to identify 22 that are cytoprotective of astrocytes. This study is the largest-scale high-throughput screen conducted using human astrocytes, with a total of 17,536 data points collected in the primary screen. The results demonstrate the relevancy and utility of using astrocytes differentiated from human stem cells as a disease model for drug discovery and development.


Asunto(s)
Antioxidantes/farmacología , Astrocitos/efectos de los fármacos , Descubrimiento de Drogas/métodos , Células Madre Embrionarias/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Neurogénesis , Estrés Oxidativo/efectos de los fármacos , Elementos de Respuesta Antioxidante/efectos de los fármacos , Astrocitos/metabolismo , Citoprotección , Relación Dosis-Respuesta a Droga , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Hep G2 , Humanos , Peróxido de Hidrógeno/toxicidad , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Oxidantes/farmacología , Fenotipo , Reproducibilidad de los Resultados , Bibliotecas de Moléculas Pequeñas
14.
Nat Med ; 22(4): 388-96, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26998835

RESUMEN

Smith-Lemli-Opitz syndrome (SLOS) is a malformation disorder caused by mutations in DHCR7, which impair the reduction of 7-dehydrocholesterol (7DHC) to cholesterol. SLOS results in cognitive impairment, behavioral abnormalities and nervous system defects, though neither affected cell types nor impaired signaling pathways are fully understood. Whether 7DHC accumulation or cholesterol loss is primarily responsible for disease pathogenesis is also unclear. Using induced pluripotent stem cells (iPSCs) from subjects with SLOS, we identified cellular defects that lead to precocious neuronal specification within SLOS derived neural progenitors. We also demonstrated that 7DHC accumulation, not cholesterol deficiency, is critical for SLOS-associated defects. We further identified downregulation of Wnt/ß-catenin signaling as a key initiator of aberrant SLOS iPSC differentiation through the direct inhibitory effects of 7DHC on the formation of an active Wnt receptor complex. Activation of canonical Wnt signaling prevented the neural phenotypes observed in SLOS iPSCs, suggesting that Wnt signaling may be a promising therapeutic target for SLOS.


Asunto(s)
Diferenciación Celular/genética , Células Madre Pluripotentes Inducidas/metabolismo , Síndrome de Smith-Lemli-Opitz/genética , Vía de Señalización Wnt/genética , Animales , Colesterol/biosíntesis , Colesterol/metabolismo , Deshidrocolesteroles/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/trasplante , Ratones , Mutación , Neuronas/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Síndrome de Smith-Lemli-Opitz/metabolismo , Síndrome de Smith-Lemli-Opitz/patología
15.
Stem Cells Dev ; 24(16): 1852-64, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25794298

RESUMEN

There is great need to develop more predictive drug discovery tools to identify new therapies to treat diseases of the central nervous system (CNS). Current nonpluripotent stem cell-based models often utilize non-CNS immortalized cell lines and do not enable the development of personalized models of disease. In this review, we discuss why in vitro models are necessary for translational research and outline the unique advantages of induced pluripotent stem cell (iPSC)-based models over those of current systems. We suggest that iPSC-based models can be patient specific and isogenic lines can be differentiated into many neural cell types for detailed comparisons. iPSC-derived cells can be combined to form small organoids, or large panels of lines can be developed that enable new forms of analysis. iPSC and embryonic stem cell-derived cells can be readily engineered to develop reporters for lineage studies or mechanism of action experiments further extending the utility of iPSC-based systems. We conclude by describing novel technologies that include strategies for the development of diversity panels, novel genomic engineering tools, new three-dimensional organoid systems, and modified high-content screens that may bring toxicology into the 21st century. The strategic integration of these technologies with the advantages of iPSC-derived cell technology, we believe, will be a paradigm shift for toxicology and drug discovery efforts.


Asunto(s)
Fármacos del Sistema Nervioso Central/farmacología , Evaluación Preclínica de Medicamentos/métodos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Neuronas/efectos de los fármacos , Animales , Técnicas de Cultivo de Célula/métodos , Humanos , Células Madre Pluripotentes Inducidas/citología , Neuronas/citología
16.
Stem Cells Transl Med ; 4(3): 230-8, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25637190

RESUMEN

Niemann-Pick disease, type C1 (NPC1) is a familial disorder that has devastating consequences on postnatal development with multisystem effects, including neurodegeneration. There is no Food and Drug Administration-approved treatment option for NPC1; however, several potentially therapeutic compounds have been identified in assays using yeast, rodent models, and NPC1 human fibroblasts. Although these discoveries were made in fibroblasts from NPC1 subjects and were in some instances validated in animal models of the disease, testing these drugs on a cell type more relevant for NPC1 neurological disease would greatly facilitate both study of the disease and identification of more relevant therapeutic compounds. Toward this goal, we have generated an induced pluripotent stem cell line from a subject homozygous for the most frequent NPC1 mutation (p.I1061T) and subsequently created a stable line of neural stem cells (NSCs). These NSCs were then used to create neurons as an appropriate disease model. NPC1 neurons display a premature cell death phenotype, and gene expression analysis of these cells suggests dysfunction of important signaling pathways, including calcium and WNT. The clear readout from these cells makes them ideal candidates for high-throughput screening and will be a valuable tool to better understand the development of NPC1 in neural cells, as well as to develop better therapeutic options for NPC1.


Asunto(s)
Señalización del Calcio , Fibroblastos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas/metabolismo , Enfermedad de Niemann-Pick Tipo C/metabolismo , Enfermedad de Niemann-Pick Tipo C/patología , Vía de Señalización Wnt , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular , Fibroblastos/patología , Humanos , Células Madre Pluripotentes Inducidas/patología , Péptidos y Proteínas de Señalización Intracelular , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Mutación , Neuronas/patología , Proteína Niemann-Pick C1 , Enfermedad de Niemann-Pick Tipo C/genética
17.
PLoS One ; 10(1): e0116032, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25587899

RESUMEN

Targeted genome engineering to robustly express transgenes is an essential methodology for stem cell-based research and therapy. Although designer nucleases have been used to drastically enhance gene editing efficiency, targeted addition and stable expression of transgenes to date is limited at single gene/locus and mostly PPP1R12C/AAVS1 in human stem cells. Here we constructed transcription activator-like effector nucleases (TALENs) targeting the safe-harbor like gene CLYBL to mediate reporter gene integration at 38%-58% efficiency, and used both AAVS1-TALENs and CLYBL-TALENs to simultaneously knock-in multiple reporter genes at dual safe-harbor loci in human induced pluripotent stem cells (iPSCs) and neural stem cells (NSCs). The CLYBL-TALEN engineered cell lines maintained robust reporter expression during self-renewal and differentiation, and revealed that CLYBL targeting resulted in stronger transgene expression and less perturbation on local gene expression than PPP1R12C/AAVS1. TALEN-mediated CLYBL engineering provides improved transgene expression and options for multiple genetic modification in human stem cells.


Asunto(s)
Endonucleasas/genética , Marcación de Gen/métodos , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/citología , Línea Celular , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células-Madre Neurales/metabolismo , Transgenes/genética
18.
Stem Cell Rev Rep ; 11(1): 194-204, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25091426

RESUMEN

We have used a four stage protocol to generate spinal motor neurons (MNs) from human embryonic stem cells (ESCs) and human induced pluripotent stem cells (iPSCs). These stages include the pluripotent stem cell (PSC) stage, neural stem cell (NSC) stage, OLIG2 expressing motor neuron precursor (MNP) stage, and HB9 expressing mature-MN stage. To optimize the differentiation protocol reporter lines marking the NSC and MNP stages were used. The NSC stage is a pro-proliferative precursor stage at which cells can be directed to differentiate to other neural types like cortical neurons also, in addition to MNs; thus, NSCs can be expanded and stored for future differentiation to different neural types thereby, shortening the differentiation interval as compared to the complete process of differentiation from ESCs or iPSCs. Additionally, we find that OLIG2 positive cells at the MNP stage can be cryopreserved and then recovered to continue the process of MN differentiation, thereby providing a highly stable and reproducible technique for bulk differentiation. MNPs were differentiated to MNs expressing the marker HB9 demonstrating that mature-MNs can be generated with this protocol.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Neuronas Motoras/citología , Células-Madre Neurales/citología , Células Madre Pluripotentes/citología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Inmunohistoquímica , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas Motoras/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Nestina/genética , Nestina/metabolismo , Células-Madre Neurales/metabolismo , Factor de Transcripción 2 de los Oligodendrocitos , Células Madre Pluripotentes/metabolismo , Reproducibilidad de los Resultados , Médula Espinal/citología , Médula Espinal/metabolismo , Factores de Tiempo , Ingeniería de Tejidos/instrumentación , Ingeniería de Tejidos/métodos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
Neurotoxicology ; 45: 192-200, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25454721

RESUMEN

Human primary neural tissue is a vital component for the quick and simple determination of chemical compound neurotoxicity in vitro. In particular, such tissue would be ideal for high-throughput screens that can be used to identify novel neurotoxic or neurotherapeutic compounds. We have previously established a high-throughput screening platform using human induced pluripotent stem cell (iPSC)-derived neural stem cells (NSCs) and neurons. In this study, we conducted a 2000 compound screen with human NSCs and rat cortical cells to identify compounds that are selectively toxic to each group. Approximately 100 of the tested compounds showed specific toxicity to human NSCs. A secondary screen of a small subset of compounds from the primary screen on human iPSCs, NSC-derived neurons, and fetal astrocytes validated the results from >80% of these compounds with some showing cell specific toxicity. Amongst those compounds were several cardiac glycosides, all of which were selectively toxic to the human cells. As the screen was able to reliably identify neurotoxicants, many with species and cell-type specificity, this study demonstrates the feasibility of this NSC-driven platform for higher-throughput neurotoxicity screens.


Asunto(s)
Astrocitos/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Células-Madre Neurales/efectos de los fármacos , Pruebas de Toxicidad/métodos , Animales , Muerte Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Ratas
20.
Stem Cells Transl Med ; 3(12): 1418-28, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25368377

RESUMEN

Induced pluripotent stem cells (iPSCs) offer an opportunity to delve into the mechanisms underlying development while also affording the potential to take advantage of a number of naturally occurring mutations that contribute to either disease susceptibility or resistance. Just as with any new field, several models of screening are being explored, and innovators are working on the most efficient methods to overcome the inherent limitations of primary cell screens using iPSCs. In the present review, we provide a background regarding why iPSCs represent a paradigm shift for central nervous system (CNS) disease modeling. We describe the efforts in the field to develop more biologically relevant CNS disease models, which should provide screening assays useful for the pharmaceutical industry. We also provide some examples of successful uses for iPSC-based screens and suggest that additional development could revolutionize the field of drug discovery. The development and implementation of these advanced iPSC-based screens will create a more efficient disease-specific process underpinned by the biological mechanism in a patient- and disease-specific manner rather than by trial-and-error. Moreover, with careful and strategic planning, shared resources can be developed that will enable exponential advances in the field. This will undoubtedly lead to more sensitive and accurate screens for early diagnosis and allow the identification of patient-specific therapies, thus, paving the way to personalized medicine.


Asunto(s)
Enfermedades del Sistema Nervioso Central/tratamiento farmacológico , Descubrimiento de Drogas/métodos , Células Madre Pluripotentes Inducidas , Modelos Neurológicos , Humanos , Medicina de Precisión/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...