Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Adv Sci (Weinh) ; 10(8): e2205037, 2023 03.
Article in English | MEDLINE | ID: mdl-36642841

ABSTRACT

Creating a cellular model of Alzheimer's disease (AD) that accurately recapitulates disease pathology has been a longstanding challenge. Recent studies showed that human AD neural cells, integrated into three-dimensional (3D) hydrogel matrix, display key features of AD neuropathology. Like in the human brain, the extracellular matrix (ECM) plays a critical role in determining the rate of neuropathogenesis in hydrogel-based 3D cellular models. Aging, the greatest risk factor for AD, significantly alters brain ECM properties. Therefore, it is important to understand how age-associated changes in ECM affect accumulation of pathogenic molecules, neuroinflammation, and neurodegeneration in AD patients and in vitro models. In this review, mechanistic hypotheses is presented to address the impact of the ECM properties and their changes with aging on AD and AD-related dementias. Altered ECM characteristics in aged brains, including matrix stiffness, pore size, and composition, will contribute to disease pathogenesis by modulating the accumulation, propagation, and spreading of pathogenic molecules of AD. Emerging hydrogel-based disease models with differing ECM properties provide an exciting opportunity to study the impact of brain ECM aging on AD pathogenesis, providing novel mechanistic insights. Understanding the role of ECM aging in AD pathogenesis should also improve modeling AD in 3D hydrogel systems.


Subject(s)
Alzheimer Disease , Humans , Aged , Brain/pathology , Aging , Cell Culture Techniques, Three Dimensional , Hydrogels
2.
Mol Cell Neurosci ; 110: 103568, 2021 01.
Article in English | MEDLINE | ID: mdl-33068718

ABSTRACT

The incidence of Alzheimer's disease is increasing with the aging population, and it has become one of the main health concerns of modern society. The dissection of the underlying pathogenic mechanisms and the development of effective therapies remain extremely challenging, also because available animal and cell culture models do not fully recapitulate the whole spectrum of pathological changes. The advent of human pluripotent stem cells and cell reprogramming has provided new prospects for tackling these challenges in a human and even patient-specific setting. Yet, experimental modeling of non-cell autonomous and extracellular disease-related alterations has remained largely inaccessible. These limitations are about to be overcome by advances in the development of 3D cell culture systems including organoids, neurospheroids and matrix-embedded 3D cultures, which have been shown to recapitulate extracellular pathologies such as plaque formation in vitro. Recent xenograft studies have even taken human stem cell-based disease modeling to an in vivo scenario where grafted neurons are probed in a disease background in the context of a rodent brain. Here, we review the latest developments in this emerging field along with their advantages, challenges, and future prospects.


Subject(s)
Alzheimer Disease/metabolism , Precision Medicine/methods , Primary Cell Culture/methods , Alzheimer Disease/etiology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Gene Editing/methods , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Transplantation, Heterologous/methods
3.
Cell Rep ; 27(7): 2199-2211.e6, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31091456

ABSTRACT

Synaptic dysfunction is associated with many brain disorders, but robust human cell models to study synaptic transmission and plasticity are lacking. Instead, current in vitro studies on human neurons typically rely on spontaneous synaptic events as a proxy for synapse function. Here, we describe a standardized in vitro approach using human neurons cultured individually on glia microdot arrays that allow single-cell analysis of synapse formation and function. We show that single glutamatergic or GABAergic forebrain neurons differentiated from human induced pluripotent stem cells form mature synapses that exhibit robust evoked synaptic transmission. These neurons show plasticity features such as synaptic facilitation, depression, and recovery. Finally, we show that spontaneous events are a poor predictor of synaptic maturity and do not correlate with the robustness of evoked responses. This methodology can be deployed directly to evaluate disease models for synaptic dysfunction and can be leveraged for drug development and precision medicine.


Subject(s)
GABAergic Neurons/metabolism , Induced Pluripotent Stem Cells/metabolism , Neurogenesis/genetics , Neuronal Plasticity/physiology , Single-Cell Analysis/methods , Synaptic Transmission/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cells, Cultured , Excitatory Amino Acid Agents/pharmacology , GABAergic Neurons/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurogenesis/drug effects , Neurogenesis/physiology , Neuroglia/cytology , Neuroglia/physiology , Rats , Synapses/physiology
4.
Cell Rep ; 27(7): 2212-2228.e7, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31091457

ABSTRACT

iPSC-derived human neurons are expected to revolutionize studies on brain diseases, but their functional heterogeneity still poses a problem. Key sources of heterogeneity are the different cell culture systems used. We show that an optimized autaptic culture system, with single neurons on astrocyte feeder islands, is well suited to culture, and we analyze human iPSC-derived neurons in a standardized, systematic, and reproducible manner. Using classically differentiated and transcription factor-induced human glutamatergic and GABAergic neurons, we demonstrate that key features of neuronal morphology and function, including dendrite structure, synapse number, membrane properties, synaptic transmission, and short-term plasticity, can be assessed with substantial throughput and reproducibility. We propose our optimized autaptic culture system as a tool to study functional features of human neurons, particularly in the context of disease phenotypes and experimental therapy.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation/physiology , GABAergic Neurons/metabolism , Induced Pluripotent Stem Cells/metabolism , Synapses/metabolism , Synaptic Transmission/physiology , Animals , Astrocytes/cytology , Astrocytes/physiology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Membrane/metabolism , Cell Membrane/physiology , Cell Proliferation/physiology , Cell Survival/physiology , Cells, Cultured , Dendrites/physiology , Excitatory Amino Acid Agents/pharmacology , GABAergic Neurons/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Mice , Mice, Inbred C57BL , Neural Inhibition/physiology , Neuronal Plasticity/physiology , Rats, Wistar , Reproducibility of Results
5.
Eur J Neurosci ; 49(4): 561-589, 2019 02.
Article in English | MEDLINE | ID: mdl-30656775

ABSTRACT

Recent advances in cell reprogramming have enabled assessment of disease-related cellular traits in patient-derived somatic cells, thus providing a versatile platform for disease modeling and drug development. Given the limited access to vital human brain cells, this technology is especially relevant for neurodegenerative disorders such as Parkinson's disease (PD) as a tool to decipher underlying pathomechanisms. Importantly, recent progress in genome-editing technologies has provided an ability to analyze isogenic induced pluripotent stem cell (iPSC) pairs that differ only in a single genetic change, thus allowing a thorough assessment of the molecular and cellular phenotypes that result from monogenetic risk factors. In this review, we summarize the current state of iPSC-based modeling of PD with a focus on leucine-rich repeat kinase 2 (LRRK2), one of the most prominent monogenetic risk factors for PD linked to both familial and idiopathic forms. The LRRK2 protein is a primarily cytosolic multi-domain protein contributing to regulation of several pathways including autophagy, mitochondrial function, vesicle transport, nuclear architecture and cell morphology. We summarize iPSC-based studies that contributed to improving our understanding of the function of LRRK2 and its variants in the context of PD etiopathology. These data, along with results obtained in our own studies, underscore the multifaceted role of LRRK2 in regulating cellular homeostasis on several levels, including proteostasis, mitochondrial dynamics and regulation of the cytoskeleton. Finally, we expound advantages and limitations of reprogramming technologies for disease modeling and drug development and provide an outlook on future challenges and expectations offered by this exciting technology.


Subject(s)
Induced Pluripotent Stem Cells , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mitophagy , Models, Neurological , Parkinson Disease , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Parkinson Disease/genetics , Parkinson Disease/therapy
6.
Nat Commun ; 9(1): 4047, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30279449

ABSTRACT

Recent reports suggest that induced neurons (iNs), but not induced pluripotent stem cell (iPSC)-derived neurons, largely preserve age-associated traits. Here, we report on the extent of preserved epigenetic and transcriptional aging signatures in directly converted induced neural stem cells (iNSCs). Employing restricted and integration-free expression of SOX2 and c-MYC, we generated a fully functional, bona fide NSC population from adult blood cells that remains highly responsive to regional patterning cues. Upon conversion, low passage iNSCs display a profound loss of age-related DNA methylation signatures, which further erode across extended passaging, thereby approximating the DNA methylation age of isogenic iPSC-derived neural precursors. This epigenetic rejuvenation is accompanied by a lack of age-associated transcriptional signatures and absence of cellular aging hallmarks. We find iNSCs to be competent for modeling pathological protein aggregation and for neurotransplantation, depicting blood-to-NSC conversion as a rapid alternative route for both disease modeling and neuroregeneration.


Subject(s)
Aging/genetics , Induced Pluripotent Stem Cells , Neural Stem Cells , Aging/metabolism , DNA Methylation , Epigenesis, Genetic , Humans , Machado-Joseph Disease/blood , Peripheral Blood Stem Cells
7.
Nat Protoc ; 10(7): 985-1006, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26068894

ABSTRACT

Stem cell technologies have facilitated the development of human cellular disease models that can be used to study pathogenesis and test therapeutic candidates. These models hold promise for complex neurological diseases such as Alzheimer's disease (AD), because existing animal models have been unable to fully recapitulate all aspects of pathology. We recently reported the characterization of a novel 3D culture system that exhibits key events in AD pathogenesis, including extracellular aggregation of amyloid-ß (Aß) and accumulation of hyperphosphorylated tau. Here we provide instructions for the generation and analysis of 3D human neural cell cultures, including the production of genetically modified human neural progenitor cells (hNPCs) with familial AD mutations, the differentiation of the hNPCs in a 3D matrix and the analysis of AD pathogenesis. The 3D culture generation takes 1-2 d. The aggregation of Aß is observed after 6 weeks of differentiation, followed by robust tau pathology after 10-14 weeks.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Models, Neurological , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Cell Culture Techniques/methods , Cell Differentiation , Cell Line , Humans , Mutation , Protein Aggregation, Pathological/metabolism , tau Proteins/metabolism
8.
Nature ; 515(7526): 274-8, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-25307057

ABSTRACT

Alzheimer's disease is the most common form of dementia, characterized by two pathological hallmarks: amyloid-ß plaques and neurofibrillary tangles. The amyloid hypothesis of Alzheimer's disease posits that the excessive accumulation of amyloid-ß peptide leads to neurofibrillary tangles composed of aggregated hyperphosphorylated tau. However, to date, no single disease model has serially linked these two pathological events using human neuronal cells. Mouse models with familial Alzheimer's disease (FAD) mutations exhibit amyloid-ß-induced synaptic and memory deficits but they do not fully recapitulate other key pathological events of Alzheimer's disease, including distinct neurofibrillary tangle pathology. Human neurons derived from Alzheimer's disease patients have shown elevated levels of toxic amyloid-ß species and phosphorylated tau but did not demonstrate amyloid-ß plaques or neurofibrillary tangles. Here we report that FAD mutations in ß-amyloid precursor protein and presenilin 1 are able to induce robust extracellular deposition of amyloid-ß, including amyloid-ß plaques, in a human neural stem-cell-derived three-dimensional (3D) culture system. More importantly, the 3D-differentiated neuronal cells expressing FAD mutations exhibited high levels of detergent-resistant, silver-positive aggregates of phosphorylated tau in the soma and neurites, as well as filamentous tau, as detected by immunoelectron microscopy. Inhibition of amyloid-ß generation with ß- or γ-secretase inhibitors not only decreased amyloid-ß pathology, but also attenuated tauopathy. We also found that glycogen synthase kinase 3 (GSK3) regulated amyloid-ß-mediated tau phosphorylation. We have successfully recapitulated amyloid-ß and tau pathology in a single 3D human neural cell culture system. Our unique strategy for recapitulating Alzheimer's disease pathology in a 3D neural cell culture model should also serve to facilitate the development of more precise human neural cell models of other neurodegenerative disorders.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Cell Culture Techniques/methods , Models, Biological , Neural Stem Cells/metabolism , Alzheimer Disease/genetics , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Cell Differentiation , Drug Evaluation, Preclinical/methods , Extracellular Space/metabolism , Glycogen Synthase Kinase 3/metabolism , Humans , Microtubule-Associated Proteins/metabolism , Neural Stem Cells/pathology , Neurites/metabolism , Phosphorylation , Presenilin-1/metabolism , Protein Aggregation, Pathological , Reproducibility of Results , tau Proteins/chemistry , tau Proteins/metabolism
9.
Mol Neurodegener ; 9: 4, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24405708

ABSTRACT

BACKGROUND: Although BACE1 is a major therapeutic target for Alzheimer's disease (AD), potential side effects of BACE1 inhibition are not well characterized. BACE1 cleaves over 60 putative substrates, however the majority of these cleavages have not been characterized. Here we investigated BACE1-mediated cleavage of human contactin-2, a GPI-anchored cell adhesion molecule. RESULTS: Our initial protein sequence analysis showed that contactin-2 harbors a strong putative BACE1 cleavage site close to its GPI membrane linker domain. When we overexpressed BACE1 in CHO cells stably transfected with human contactin-2, we found increased release of soluble contactin-2 in the conditioned media. Conversely, pharmacological inhibition of BACE1 in CHO cells expressing human contactin-2 and mouse primary neurons decreased soluble contactin-2 secretion. The BACE1 cleavage site mutation 1008MM/AA dramatically impaired soluble contactin-2 release. We then asked whether contactin-2 release induced by BACE1 expression would concomitantly decrease cell surface levels of contactin-2. Using immunofluorescence and surface-biotinylation assays, we showed that BACE1 activity tightly regulates contactin-2 surface levels in CHO cells as well as in mouse primary neurons. Finally, contactin-2 levels were decreased in Alzheimer's disease brain samples correlating inversely with elevated BACE1 levels in the same samples. CONCLUSION: Our results clearly demonstrate that mouse and human contactin-2 are physiological substrates for BACE1. BACE1-mediated contactin-2 cleavage tightly regulates the surface expression of contactin-2 in neuronal cells. Given the role of contactin-2 in cell adhesion, neurite outgrowth and axon guidance, our data suggest that BACE1 may play an important role in these physiological processes by regulating contactin-2 surface levels.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Contactin 2/metabolism , Animals , Blotting, Western , Brain/metabolism , CHO Cells , Cell Membrane/metabolism , Cricetinae , Cricetulus , Fluorescent Antibody Technique , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...