Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Cells ; 13(12)2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38920630

RESUMEN

Circular RNAs (circRNAs) have emerged as pivotal regulators of gene expression with diverse roles in various biological processes. In recent years, research into circRNAs' involvement in bone biology has gained significant attention, unveiling their potential as novel regulators and biomarkers in bone-related disorders and diseases. CircRNAs, characterized by their closed-loop structure, exhibit stability and resistance to degradation, underscoring their functional significance. In bone tissue, circRNAs are involved in critical processes such as osteogenic differentiation, osteoclastogenesis, and bone remodeling through intricate molecular mechanisms including microRNA regulation. Dysregulated circRNAs are associated with various bone disorders, suggesting their potential as diagnostic and prognostic biomarkers. The therapeutic targeting of these circRNAs holds promise for addressing bone-related conditions, offering new perspectives for precision medicine. Thus, circRNAs constitute integral components of bone regulatory networks, impacting both physiological bone homeostasis and pathological conditions. This review provides a comprehensive overview of circRNAs in bone biology, emphasizing their regulatory mechanisms, functional implications, and therapeutic potential.


Asunto(s)
Huesos , ARN Circular , Humanos , ARN Circular/genética , ARN Circular/metabolismo , Huesos/metabolismo , Animales , Enfermedades Óseas/genética , Enfermedades Óseas/metabolismo , Osteogénesis/genética , Biomarcadores/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Regulación de la Expresión Génica
2.
Mol Cell Neurosci ; 128: 103919, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38307302

RESUMEN

Parkinson's disease (PD) is a complex, progressive neurodegenerative disease characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta in the midbrain. Despite extensive research efforts, the molecular and cellular changes that precede neurodegeneration in PD are poorly understood. To address this, here we describe the use of patient specific human midbrain organoids harboring the SNCA triplication to investigate mechanisms underlying dopaminergic degeneration. Our midbrain organoid model recapitulates key pathological hallmarks of PD, including the aggregation of α-synuclein and the progressive loss of dopaminergic neurons. We found that these pathological hallmarks are associated with an increase in senescence associated cellular phenotypes in astrocytes including nuclear lamina defects, the presence of senescence associated heterochromatin foci, and the upregulation of cell cycle arrest genes. These results suggest a role of pathological α-synuclein in inducing astrosenescence which may, in turn, increase the vulnerability of dopaminergic neurons to degeneration.


Asunto(s)
Enfermedades Neurodegenerativas , Enfermedad de Parkinson , Humanos , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Astrocitos/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Mesencéfalo/metabolismo , Mesencéfalo/patología , Neuronas Dopaminérgicas/metabolismo , Organoides/metabolismo , Organoides/patología , Sustancia Negra/metabolismo
3.
EMBO Rep ; 25(1): 254-285, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38177910

RESUMEN

Midbrain dopaminergic neurons (mDANs) control voluntary movement, cognition, and reward behavior under physiological conditions and are implicated in human diseases such as Parkinson's disease (PD). Many transcription factors (TFs) controlling human mDAN differentiation during development have been described, but much of the regulatory landscape remains undefined. Using a tyrosine hydroxylase (TH) human iPSC reporter line, we here generate time series transcriptomic and epigenomic profiles of purified mDANs during differentiation. Integrative analysis predicts novel regulators of mDAN differentiation and super-enhancers are used to identify key TFs. We find LBX1, NHLH1 and NR2F1/2 to promote mDAN differentiation and show that overexpression of either LBX1 or NHLH1 can also improve mDAN specification. A more detailed investigation of TF targets reveals that NHLH1 promotes the induction of neuronal miR-124, LBX1 regulates cholesterol biosynthesis, and NR2F1/2 controls neuronal activity.


Asunto(s)
Neuronas Dopaminérgicas , Células Madre Pluripotentes Inducidas , Humanos , Neuronas Dopaminérgicas/metabolismo , Multiómica , Mesencéfalo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Diferenciación Celular/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética
4.
NPJ Parkinsons Dis ; 9(1): 166, 2023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-38110400

RESUMEN

The mechanisms underlying Parkinson's disease (PD) etiology are only partially understood despite intensive research conducted in the field. Recent evidence suggests that early neurodevelopmental defects might play a role in cellular susceptibility to neurodegeneration. To study the early developmental contribution of GBA mutations in PD we used patient-derived iPSCs carrying a heterozygous N370S mutation in the GBA gene. Patient-specific midbrain organoids displayed GBA-PD relevant phenotypes such as reduction of GCase activity, autophagy impairment, and mitochondrial dysfunction. Genome-scale metabolic (GEM) modeling predicted changes in lipid metabolism which were validated with lipidomics analysis, showing significant differences in the lipidome of GBA-PD. In addition, patient-specific midbrain organoids exhibited a decrease in the number and complexity of dopaminergic neurons. This was accompanied by an increase in the neural progenitor population showing signs of oxidative stress-induced damage and premature cellular senescence. These results provide insights into how GBA mutations may lead to neurodevelopmental defects thereby predisposing to PD pathology.

5.
Cells ; 12(3)2023 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-36766845

RESUMEN

Nanoparticles (NPs) are the focus of research efforts that aim to develop successful drug delivery systems for the brain. Polypeptide nanocarriers are versatile platforms and combine high functionality with good biocompatibility and biodegradability. The key to the efficient brain delivery of NPs is the specific targeting of cerebral endothelial cells that form the blood-brain barrier (BBB). We have previously discovered that the combination of two different ligands of BBB nutrient transporters, alanine and glutathione, increases the permeability of vesicular NPs across the BBB. Our aim here was to investigate whether the combination of these molecules can also promote the efficient transfer of 3-armed poly(l-glutamic acid) NPs across a human endothelial cell and brain pericyte BBB co-culture model. Alanine and glutathione dual-targeted polypeptide NPs showed good cytocompatibility and elevated cellular uptake in a time-dependent and active manner. Targeted NPs had a higher permeability across the BBB model and could subsequently enter midbrain-like organoids derived from healthy and Parkinson's disease patient-specific stem cells. These results indicate that poly(l-glutamic acid) NPs can be used as nanocarriers for nervous system application and that the right combination of molecules that target cerebral endothelial cells, in this case alanine and glutathione, can facilitate drug delivery to the brain.


Asunto(s)
Barrera Hematoencefálica , Células Endoteliales , Humanos , Alanina , Ácido Glutámico , Encéfalo , Péptidos/farmacología , Péptidos/química , Glutatión , Organoides
6.
Mol Neurobiol ; 59(4): 2129-2149, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35044626

RESUMEN

We have previously shown that the heteromer composed by the dopamine D3 receptor (D3R) and the nicotinic acetylcholine receptor (nAChR) (D3R-nAChR heteromer) is expressed in dopaminergic neurons, activated by nicotine and represents the molecular unit that, in these neurons, contributes to the modulation of critical events such as structural plasticity and neuroprotection. We now extended this study by investigating the D3R-nAChR heteromer properties using various cell models such as transfected HEK293 cells, primary cultures of mouse dopaminergic neurons and human dopaminergic neurons derived from induced pluripotent stem cells.We found that the D3R-nAChR heteromer is the molecular effector that transduces the remodeling properties not only associated with nicotine but also with D3R agonist stimulation: neither nAChR nor D3R, in fact, when express as monomers, are able to elicit these effects. Moreover, strong and sustained activation of the PI3K-ERK1/2/Akt pathways is coupled with D3R-nAChR heteromer stimulation, leading to the expression of the immediate-early gene c-Fos and to sustained phosphorylation of cytosolic p70 ribosomal S6 kinase (p70S6K), critical for dendritic remodeling. By contrast, while D3R stimulation results in rapid and transient activation of both Erk1/2 and Akt, that is PI3K-dependent, stimulation of nAChR is associated with persistent activation of Erk1/2 and Akt, in a PI3K-independent way. Thus, the D3R-nAChR heteromer and its ability to trigger the PI3K-ERK1/2/Akt signaling pathways may represent a novel target for preserving dopaminergic neurons healthy and for conferring neuronal protection against injuries.


Asunto(s)
Receptores de Dopamina D3 , Receptores Nicotínicos , Animales , Neuronas Dopaminérgicas/metabolismo , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Nicotina/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores de Dopamina D3/metabolismo , Receptores Nicotínicos/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal
7.
Mov Disord ; 37(1): 80-94, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34637165

RESUMEN

BACKGROUND: The etiology of Parkinson's disease (PD) is only partially understood despite the fact that environmental causes, risk factors, and specific gene mutations are contributors to the disease. Biallelic mutations in the phosphatase and tensin homolog (PTEN)-induced putative kinase 1 (PINK1) gene involved in mitochondrial homeostasis, vesicle trafficking, and autophagy are sufficient to cause PD. OBJECTIVES: We sought to evaluate the difference between controls' and PINK1 patients' derived neurons in their transition from neuroepithelial stem cells to neurons, allowing us to identify potential pathways to target with repurposed compounds. METHODS: Using two-dimensional and three-dimensional models of patients' derived neurons we recapitulated PD-related phenotypes. We introduced the usage of midbrain organoids for testing compounds. Using Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated protein 9 (Cas9), we corrected the point mutations of three patients' derived cells. We evaluated the effect of the selected compound in a mouse model. RESULTS: PD patient-derived cells presented differences in their energetic profile, imbalanced proliferation, apoptosis, mitophagy, and a reduced differentiation efficiency to tyrosine hydroxylase positive (TH+) neurons compared to controls' cells. Correction of a patient's point mutation ameliorated the metabolic properties and neuronal firing rates as well as reversing the differentiation phenotype, and reducing the increased astrocytic levels. Treatment with 2-hydroxypropyl-ß-cyclodextrin increased the autophagy and mitophagy capacity of neurons concomitant with an improved dopaminergic differentiation of patient-specific neurons in midbrain organoids and ameliorated neurotoxicity in a mouse model. CONCLUSION: We show that treatment with a repurposed compound is sufficient for restoring the impaired dopaminergic differentiation of PD patient-derived cells. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Asunto(s)
Enfermedad de Parkinson , 2-Hidroxipropil-beta-Ciclodextrina/metabolismo , Animales , Encéfalo/metabolismo , Neuronas Dopaminérgicas/metabolismo , Humanos , Ratones , Neuronas/metabolismo , Organoides/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Fenotipo
8.
Cells ; 12(1)2022 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-36611845

RESUMEN

Degenerative diseases affecting bone tissues and the brain represent important problems with high socio-economic impact. Certain bone diseases, such as osteoporosis, are considered risk factors for the progression of neurological disorders. Often, patients with neurodegenerative diseases have bone fractures or reduced mobility linked to osteoarthritis. The bone is a dynamic tissue involved not only in movement but also in the maintenance of mineral metabolism. Bone is also associated with the generation of both hematopoietic stem cells (HSCs), and thus the generation of the immune system, and mesenchymal stem cells (MSCs). Bone marrow is a lymphoid organ and contains MSCs and HSCs, both of which are involved in brain health via the production of cytokines with endocrine functions. Hence, it seems clear that bone is involved in the regulation of the neuronal system and vice versa. This review summarizes the recent knowledge on the interactions between the nervous system and bone and highlights the importance of the interaction between nerve and bone cells. In addition, experimental models that study the interaction between nerve and skeletal cells are discussed, and innovative models are suggested to better evaluate the molecular interactions between these two cell types.


Asunto(s)
Células de la Médula Ósea , Enfermedades Neurodegenerativas , Humanos , Diferenciación Celular/fisiología , Huesos
9.
Cell Rep ; 37(3): 109864, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34686322

RESUMEN

Increasing evidence suggests that neurodevelopmental alterations might contribute to increase the susceptibility to develop neurodegenerative diseases. We investigate the occurrence of developmental abnormalities in dopaminergic neurons in a model of Parkinson's disease (PD). We monitor the differentiation of human patient-specific neuroepithelial stem cells (NESCs) into dopaminergic neurons. Using high-throughput image analyses and single-cell RNA sequencing, we observe that the PD-associated LRRK2-G2019S mutation alters the initial phase of neuronal differentiation by accelerating cell-cycle exit with a concomitant increase in cell death. We identify the NESC-specific core regulatory circuit and a molecular mechanism underlying the observed phenotypes. The expression of NR2F1, a key transcription factor involved in neurogenesis, decreases in LRRK2-G2019S NESCs, neurons, and midbrain organoids compared to controls. We also observe accelerated dopaminergic differentiation in vivo in NR2F1-deficient mouse embryos. This suggests a pathogenic mechanism involving the LRRK2-G2019S mutation, where the dynamics of dopaminergic differentiation are modified via NR2F1.


Asunto(s)
Encéfalo/enzimología , Factor de Transcripción COUP I/metabolismo , Neuronas Dopaminérgicas/enzimología , Células Madre Pluripotentes Inducidas/enzimología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Células-Madre Neurales/enzimología , Neurogénesis , Enfermedad de Parkinson/enzimología , Animales , Encéfalo/patología , Factor de Transcripción COUP I/genética , Ciclo Celular , Línea Celular , Proliferación Celular , Supervivencia Celular , Neuronas Dopaminérgicas/patología , Femenino , Humanos , Células Madre Pluripotentes Inducidas/patología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Masculino , Ratones de la Cepa 129 , Ratones Noqueados , Mutación , Células-Madre Neurales/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Fenotipo , RNA-Seq , Transducción de Señal , Análisis de la Célula Individual , Factores de Tiempo
10.
Mikrochim Acta ; 188(6): 203, 2021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-34043106

RESUMEN

Molecularly imprinted polymer (MIP)-based electrochemical sensors for the protein α-synuclein (a marker for Parkinson's disease) were developed using a peptide epitope from the protein. MIPs doped with various concentrations and species of transition metal dichalcogenides (TMDs) to enhance conductivity were electropolymerized with and without template molecules. The current during the electropolymerization was compared with that associated with the electrochemical response (at 0.24~0.29 V vs. ref. electrode) to target peptide molecules in the finished sensor. We found that this relationship can aid in the rational design of conductive MIPs for the recognition of biomarkers in biological fluids. The sensing range and limit of detection of TMD-doped imprinted poly(AN-co-MSAN)-coated electrodes were 0.001-100 pg/mL and 0.5 fg/mL (SNR = 3), respectively. To show the potential applicability of the MIP electrochemical sensor, cell culture medium from PD patient-specific midbrain organoids generated from induced pluripotent stem cells was analyzed. α-Synuclein levels were found to be significantly reduced in the organoids from PD patients, compared to those generated from age-matched controls. The relative standard deviation and recovery are less than 5% and 95-115%, respectively. Preparation of TMD-doped α-synuclein (SNCA) peptide-imprinted poly(AN-co-MSAN)-coated electrodes.


Asunto(s)
Disulfuros/química , Polímeros Impresos Molecularmente/química , Molibdeno/química , Sulfuros/química , Compuestos de Tungsteno/química , alfa-Sinucleína/análisis , Técnicas Electroquímicas/métodos , Humanos , Límite de Detección , Mesencéfalo/química , Organoides/química , Enfermedad de Parkinson/diagnóstico , Fragmentos de Péptidos/química , alfa-Sinucleína/química
11.
Analyst ; 146(7): 2358-2367, 2021 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-33625407

RESUMEN

In this study, we have aimed at developing a novel electrochemical sensing approach capable of detecting dopamine, the main biomarker in Parkinson's disease, within the highly complex cell culture matrix of human midbrain organoids in a non-invasive and label-free manner. With its ability to generate organotypic structures in vitro, induced pluripotent stem cell technology has provided the basis for the development of advanced patient-derived disease models. These include models of the human midbrain, the affected region in the neurodegenerative disorder Parkinson's disease. Up to now, however, the analysis of so-called human midbrain organoids has relied on time-consuming and invasive strategies, incapable of monitoring organoid development. Using a redox-cycling approach in combination with a 3-mercaptopropionic acid self-assembled monolayer modification enabled the increase of sensor selectivity and sensitivity towards dopamine, while simultaneously reducing matrix-mediated interferences. In this work, we demonstrate the ability to detect and monitor even small differences in dopamine release between healthy and Parkinson`s disease-specific midbrain organoids over prolonged cultivation periods, which was additionally verified using liquid chromatography-multiple reaction monitoring mass spectrometry. Furthermore, the detection of a phenotypic rescue in midbrain organoids carrying a pathogenic mutation in leucine-rich repeat kinase 2, upon treatment with the leucine-rich repeat kinase 2 inhibitor II underlines the practical implementability of our sensing approach for drug screening applications as well as personalized disease modelling.


Asunto(s)
Organoides , Enfermedad de Parkinson , Evaluación Preclínica de Medicamentos , Humanos , Mesencéfalo , Neurotransmisores , Organoides/metabolismo , Oxidación-Reducción , Enfermedad de Parkinson/metabolismo
12.
Sci Rep ; 11(1): 1439, 2021 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-33446877

RESUMEN

Patient-derived cellular models become an increasingly powerful tool to model human diseases for precision medicine approaches. The identification of robust cellular disease phenotypes in these models paved the way towards high throughput screenings (HTS) including the implementation of laboratory advanced automation. However, maintenance and expansion of cells for HTS remains largely manual work. Here, we describe an integrated, complex automated platform for HTS in a translational research setting also designed for maintenance and expansion of different cell types. The comprehensive design allows automation of all cultivation steps and is flexible for development of methods for variable cell types. We demonstrate protocols for controlled cell seeding, splitting and expansion of human fibroblasts, induced pluripotent stem cells (iPSC), and neural progenitor cells (NPC) that allow for subsequent differentiation into different cell types and image-based multiparametric screening. Furthermore, we provide automated protocols for neuronal differentiation of NPC in 2D culture and 3D midbrain organoids for HTS. The flexibility of this multitask platform makes it an ideal solution for translational research settings involving experiments on different patient-derived cellular models for precision medicine.


Asunto(s)
Automatización de Laboratorios , Técnicas de Cultivo de Célula , Modelos Biológicos , Organoides/citología , Medicina de Precisión , Evaluación Preclínica de Medicamentos , Humanos
13.
Neurobiol Aging ; 99: 65-78, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33422895

RESUMEN

Mutations in the leucine-rich repeat kinase 2 (LRRK2) are the most common genetic determinants of Parkinson's disease (PD), with the G2019S accounting for about 3% of PD cases. LRRK2 regulates various cellular processes, including vesicle trafficking that is crucial for receptor localization at the plasma membrane. In this study, induced pluripotent stem cells derived from 2 PD patients bearing the G2019S LRRK2 kinase activating mutation were used to generate neuronal cultures enriched in dopaminergic neurons. The results show that mutant LRRK2 prevents the membrane localization of both the dopamine D3 receptors (D3R) and the nicotinic acetylcholine receptors (nAChR) and the formation of the D3R-nAChR heteromer, a molecular unit crucial for promoting neuronal homeostasis and preserving dopaminergic neuron health. Interestingly, D3R and nAChR as well as the corresponding heteromer membrane localization were rescued by inhibiting the abnormally increased kinase activity. Thus, the altered membrane localization of the D3R-nAChR heteromer associated with mutation in LRRK2 might represent a pre-degenerative feature of dopaminergic neurons contributing to the special vulnerability of this neuronal population.


Asunto(s)
Membrana Celular/metabolismo , Neuronas Dopaminérgicas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Mutación , Enfermedad de Parkinson/genética , Receptores de Dopamina D3/metabolismo , Receptores Nicotínicos/metabolismo , Humanos
14.
Pharmaceutics ; 14(1)2021 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-35056983

RESUMEN

Nanosized drug delivery systems targeting transporters of the blood-brain barrier (BBB) are promising carriers to enhance the penetration of therapeutics into the brain. The expression of solute carriers (SLC) is high and shows a specific pattern at the BBB. Here we show that targeting ligands ascorbic acid, leucine and glutathione on nanoparticles elevated the uptake of albumin cargo in cultured primary rat brain endothelial cells. Moreover, we demonstrated the ability of the triple-targeted nanovesicles to deliver their cargo into midbrain organoids after crossing the BBB model. The cellular uptake was temperature- and energy-dependent based on metabolic inhibition. The process was decreased by filipin and cytochalasin D, indicating that the cellular uptake of nanoparticles was partially mediated by endocytosis. The uptake of the cargo encapsulated in triple-targeted nanoparticles increased after modification of the negative zeta potential of endothelial cells by treatment with a cationic lipid or after cleaving the glycocalyx with an enzyme. We revealed that targeted nanoparticles elevated plasma membrane fluidity, indicating the fusion of nanovesicles with endothelial cell membranes. Our data indicate that labeling nanoparticles with three different ligands of multiple transporters of brain endothelial cells can promote the transfer and delivery of molecules across the BBB.

15.
Biosens Bioelectron ; 175: 112852, 2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33288425

RESUMEN

Parkinson's disease (PD) is a progressive nervous system disorder that affects movement, whose early signs may be mild and unnoticed. α-Synuclein has been identified as the major component of Lewy bodies and Lewy neurites, which are the characteristic proteinaceous deposits that are the hallmarks of PD. In this work, three alpha-synuclein peptides were synthesized as templates for the molecular imprinting of conductive polymers to enable recognition of alpha-synuclein via ultrasensitive electrochemical measurements. The peptide sequences encompassed specific residues where mutations are known to accelerate PD (though the target sequences, in this study, were wild-type.) The different peptide targets were all successfully imprinted, but with differing imprinting effectiveness, probably owing to differences in target carboxylic acids (which can bind to the aniline (AN) m-aminobenzenesulfonic acid (MSAN) MIP polymers.) Composition of the imprinted polymer, (the mole proportions of AN and MSAN), and the concentrations and sequences of imprinted peptide templates were optimized by measuring the electrochemical responses to target peptides. The imprinted electrode can detect alpha-synuclein at fg/mL levels, and was therefore used to measure alpha-synuclein in the culture medium of human brain organoids generated from normal and idiopathic PD patients.


Asunto(s)
Técnicas Biosensibles , Enfermedad de Parkinson , Encéfalo/metabolismo , Epítopos , Humanos , Organoides/metabolismo , alfa-Sinucleína
16.
NPJ Parkinsons Dis ; 6(1): 33, 2020 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-33298969

RESUMEN

Parkin (encoded by PRKN) is a ubiquitin ligase that plays an important role in cellular mitochondrial quality control. Mutations in PRKN cause selective dopaminergic cell loss in the substantia nigra and are presumed to induce a decrease in mitochondrial function caused by the defective clearance of mitochondria. Several studies have demonstrated that parkin dysfunction causes mitochondrial injury and astrocytic dysfunction. Using immunohistochemical methods, we analyzed astrocytic changes in human brains from individuals with PRKN mutations. Few glial fibrillary acidic protein- and vimentin-positive astrocytes were observed in the substantia nigra in PRKN-mutated subjects compared with subjects with idiopathic Parkinson's disease. We also differentiated patient-specific induced pluripotent stem cells into midbrain organoids and confirmed decreased numbers of glial fibrillary acidic protein-positive astrocytes in PRKN-mutated organoids compared with age- and sex-matched controls. Our study reveals PRKN-mutation-induced astrocytic alteration and suggests the possibility of an astrocyte-related non-autonomous cell death mechanism for dopaminergic neurons in brains of PRKN-mutated patients.

17.
Sci Transl Med ; 12(560)2020 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-32908004

RESUMEN

Parkinson's disease (PD) is a heterogeneous neurodegenerative disorder with monogenic forms representing prototypes of the underlying molecular pathology and reproducing to variable degrees the sporadic forms of the disease. Using a patient-based in vitro model of PARK7-linked PD, we identified a U1-dependent splicing defect causing a drastic reduction in DJ-1 protein and, consequently, mitochondrial dysfunction. Targeting defective exon skipping with genetically engineered U1-snRNA recovered DJ-1 protein expression in neuronal precursor cells and differentiated neurons. After prioritization of candidate drugs, we identified and validated a combinatorial treatment with the small-molecule compounds rectifier of aberrant splicing (RECTAS) and phenylbutyric acid, which restored DJ-1 protein and mitochondrial dysfunction in patient-derived fibroblasts as well as dopaminergic neuronal cell loss in mutant midbrain organoids. Our analysis of a large number of exomes revealed that U1 splice-site mutations were enriched in sporadic PD patients. Therefore, our study suggests an alternative strategy to restore cellular abnormalities in in vitro models of PD and provides a proof of concept for neuroprotection based on precision medicine strategies in PD.


Asunto(s)
Enfermedad de Parkinson , Neuronas Dopaminérgicas , Exones/genética , Humanos , Mutación/genética , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/genética , Empalme del ARN
18.
Cell Tissue Res ; 382(3): 463-476, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32737576

RESUMEN

Human stem cell-derived organoids have great potential for modelling physiological and pathological processes. They recapitulate in vitro the organization and function of a respective organ or part of an organ. Human midbrain organoids (hMOs) have been described to contain midbrain-specific dopaminergic neurons that release the neurotransmitter dopamine. However, the human midbrain contains also additional neuronal cell types, which are functionally interacting with each other. Here, we analysed hMOs at high-resolution by means of single-cell RNA sequencing (scRNA-seq), imaging and electrophysiology to unravel cell heterogeneity. Our findings demonstrate that hMOs show essential neuronal functional properties as spontaneous electrophysiological activity of different neuronal subtypes, including dopaminergic, GABAergic, glutamatergic and serotonergic neurons. Recapitulating these in vivo features makes hMOs an excellent tool for in vitro disease phenotyping and drug discovery.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Organoides/metabolismo , Análisis de Secuencia de ARN/métodos , Transcriptoma/fisiología , Diferenciación Celular , Humanos
19.
Parkinsonism Relat Disord ; 75: 105-109, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32534431

RESUMEN

INTRODUCTION: Brain organoids are highly complex multi-cellular tissue proxies, which have recently risen as novel tools to study neurodegenerative diseases such as Parkinson's disease (PD). However, with increasing complexity of the system, usage of quantitative tools becomes challenging. OBJECTIVES: The primary objective of this study was to develop a neurotoxin-induced PD organoid model and to assess the neurotoxic effect on dopaminergic neurons using microscopy-based phenotyping in a high-content fashion. METHODS: We describe a pipeline for a machine learning-based analytical method, allowing for detailed image-based cell profiling and toxicity prediction in brain organoids treated with the neurotoxic compound 6-hydroxydopamine (6-OHDA). RESULTS: We quantified features such as dopaminergic neuron count and neuronal complexity and built a machine learning classifier with the data to optimize data processing strategies and to discriminate between different treatment conditions. We validated the approach with high content imaging data from PD patient derived midbrain organoids. CONCLUSIONS: The here described model is a valuable tool for advanced in vitro PD modeling and to test putative neurotoxic compounds.


Asunto(s)
Neuronas Dopaminérgicas , Aprendizaje Automático , Mesencéfalo , Síndromes de Neurotoxicidad , Organoides , Oxidopamina/toxicidad , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/patología , Citometría de Flujo , Humanos , Células Madre Pluripotentes Inducidas , Mesencéfalo/diagnóstico por imagen , Mesencéfalo/efectos de los fármacos , Mesencéfalo/patología , Microscopía Confocal , Síndromes de Neurotoxicidad/diagnóstico por imagen , Síndromes de Neurotoxicidad/patología , Organoides/diagnóstico por imagen , Organoides/efectos de los fármacos , Organoides/patología , Prueba de Estudio Conceptual
20.
Parkinsonism Relat Disord ; 67: 48-55, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31621607

RESUMEN

Parkinson's disease (PD) is a multifactorial disorder with complex etiology. The most prevalent PD associated mutation, LRRK2-G2019S is linked to familial and sporadic cases. Based on the multitude of genetic predispositions in PD and the incomplete penetrance of LRRK2-G2019S, we hypothesize that modifiers in the patients' genetic background act as susceptibility factors for developing PD. To assess LRRK2-G2019S modifiers, we used human induced pluripotent stem cell-derived neuroepithelial stem cells (NESCs). Isogenic controls distinguish between LRRK2-G2019S dependent and independent cellular phenotypes. LRRK2-G2019S patient and healthy mutagenized lines showed altered NESC self-renewal and viability, as well as impaired serine metabolism. In patient cells, phenotypes were only partly LRRK2-G2019S dependent, suggesting a significant contribution of the genetic background. In this context we identified the gene serine racemase (SRR) as a novel patient-specific, developmental, genetic modifier contributing to the aberrant phenotypes. Its enzymatic product, d-serine, rescued altered cellular phenotypes. Susceptibility factors in the genetic background, such as SRR, could be new targets for early PD diagnosis and treatment.


Asunto(s)
Autorrenovación de las Células/genética , Enfermedad de Parkinson/genética , Racemasas y Epimerasas/genética , Serina/metabolismo , Estudios de Casos y Controles , Línea Celular , Supervivencia Celular/genética , Predisposición Genética a la Enfermedad , Humanos , Células Madre Pluripotentes Inducidas , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Células-Madre Neurales , Enfermedad de Parkinson/metabolismo , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...