Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 304
Filtrar
Más filtros

Base de datos
Tipo del documento
Intervalo de año de publicación
1.
Front Cell Dev Biol ; 12: 1464932, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39376628

RESUMEN

Introduction: Neurogenesis is tightly regulated in space and time, ensuring the correct development and organization of the central nervous system. Critical regulators of brain development and morphogenesis in mice include two members of the p53 family: p53 and p73. However, dissecting the in vivo functions of these factors and their various isoforms in brain development is challenging due to their pleiotropic effects. Understanding their role, particularly in neurogenesis and brain morphogenesis, requires innovative experimental approaches. Methods: To address these challenges, we developed an efficient and highly reproducible protocol to generate mouse brain organoids from pluripotent stem cells. These organoids contain neural progenitors and neurons that self-organize into rosette-like structures resembling the ventricular zone of the embryonic forebrain. Using this model, we generated organoids from p73-deficient mouse cells to investigate the roles of p73 and its isoforms (TA and DNp73) during brain development. Results and Discussion: Organoids derived from p73-deficient cells exhibited increased neuronal apoptosis and reduced neural progenitor proliferation, linked to compensatory activation of p53. This closely mirrors previous in vivo observations, confirming that p73 plays a pivotal role in brain development. Further dissection of p73 isoforms function revealed a dual role of p73 in regulating brain morphogenesis, whereby TAp73 controls transcriptional programs essential for the establishment of the neurogenic niche structure, while DNp73 is responsible for the precise and timely regulation of neural cell fate. These findings highlight the distinct roles of p73 isoforms in maintaining the balance of neural progenitor cell biology, providing a new understanding of how p73 regulates brain morphogenesis.

2.
J Am Soc Mass Spectrom ; 35(10): 2308-2314, 2024 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-39258941

RESUMEN

Single-cell proteomics has emerged as a powerful technology for unraveling the complexities of cellular heterogeneity, enabling insights into individual cell functions and pathologies. One of the primary challenges in single-cell proteomics is data generation, where low mass spectral signals often preclude the triggering of MS2 events. This challenge is addressed by Data Independent Acquisition (DIA), a data acquisition strategy that does not depend on peptide ion isotopic signatures to generate an MS2 event. In this study, we present data generated from the integration of DIA single-cell proteomics with a version of the DiagnoMass Proteomic Hub that was adapted to handle DIA data. DiagnoMass employs a hierarchical clustering methodology that enables the identification of tandem mass spectral clusters that are discriminative of biological conditions, thereby reducing the reliance on search engine biases for identifications. Nevertheless, a search engine (in this work, DIA-NN) can be integrated with DiagnoMass for spectral annotation. We used single-cell proteomic data from iPSC-derived neuroprogenitor cell cultures as a test study of this integrated approach. We were able to differentiate between control and Rett Syndrome patient cells to discern the proteomic variances potentially contributing to the disease's pathology. Our research confirms that the DiagnoMass-DIA synergy significantly enhances the identification of discriminative proteomic signatures, highlighting critical biological variations such as the presence of unique spectra that could be related to Rett Syndrome pathology.


Asunto(s)
Proteómica , Análisis de la Célula Individual , Espectrometría de Masas en Tándem , Humanos , Proteómica/métodos , Análisis de la Célula Individual/métodos , Espectrometría de Masas en Tándem/métodos , Síndrome de Rett , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/química , Proteoma/análisis , Motor de Búsqueda , Análisis por Conglomerados
3.
ACS Nano ; 2024 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-39270300

RESUMEN

Brain organoids are widely used to model brain development and diseases. However, a major challenge in their application is the insufficient supply of oxygen and nutrients to the core region, restricting the size and maturation of the organoids. In order to vascularize brain organoids and enhance the nutritional supply to their core areas, two-photon polymerization (TPP) 3D printing is employed to fabricate high-resolution meshed vessels in this study. These vessels made of photoresist with densely distributed micropores with a diameter of 20 µm on the sidewall, are cocultured with brain organoids to facilitate the diffusion of culture medium into the organoids. The vascularized organoids exhibit dimensional breaking growth and enhanced proliferation, reduced hypoxia and apoptosis, suggesting that the 3D-printed meshed vessels partially mimic vascular function to promote the culture of organoids. Furthermore, cortical, striatal and medial ganglionic eminence (MGE) organoids are respectively differentiated to generate Cortico-Striatal-MGE assembloids by 3D-printed vessels. The enhanced migration, projection and excitatory signaling transduction are observed between different brain regional organoids in the assembloids. This study presents an approach using TPP 3D printing to construct vascularized brain organoids and assembloids for enhancing the development and assembly, offering a research model and platform for neurological diseases.

4.
J Cell Mol Med ; 28(17): e18560, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39258535

RESUMEN

Neurodevelopmental disorders are mostly studied using mice as models. However, the mouse brain lacks similar cell types and structures as those of the human brain. In recent years, emergence of three-dimensional brain organoids derived from human embryonic stem cells or induced pluripotent stem cells allows for controlled monitoring and evaluation of early neurodevelopmental processes and has opened a window for studying various aspects of human brain development. However, such organoids lack original anatomical structure of the brain during maturation, and neurodevelopmental maturation processes that rely on unique cellular interactions and neural network connections are limited. Consequently, organoids are difficult to be used extensively and effectively while modelling later stages of human brain development and disease progression. To address this problem, several methods and technologies have emerged that aim to enhance the sophisticated regulation of brain organoids developmental processes through bioengineering approaches, which may alleviate some of the current limitations. This review discusses recent advances and application areas of human brain organoid culture methods, aiming to generalize optimization strategies for organoid systems, improve the ability to mimic human brain development, and enhance the application value of organoids.


Asunto(s)
Encéfalo , Trastornos del Neurodesarrollo , Organoides , Humanos , Trastornos del Neurodesarrollo/patología , Encéfalo/patología , Encéfalo/crecimiento & desarrollo , Encéfalo/citología , Animales , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Modelos Biológicos
5.
Cell Mol Life Sci ; 81(1): 410, 2024 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-39305343

RESUMEN

Rett syndrome (RTT) is a neurodevelopmental disorder caused by de novo mutations in the MECP2 gene. Although miRNAs in extracellular vesicles (EVs) have been suggested to play an essential role in several neurological conditions, no prior study has utilized brain organoids to profile EV-derived miRNAs during normal and RTT-affected neuronal development. Here we report the spatiotemporal expression pattern of EV-derived miRNAs in region-specific forebrain organoids generated from female hiPSCs with a MeCP2:R255X mutation and the corresponding isogenic control. EV miRNA and protein expression profiles were characterized at day 0, day 13, day 40, and day 75. Several members of the hsa-miR-302/367 cluster were identified as having a time-dependent expression profile with RTT-specific alterations at the latest developmental stage. Moreover, the miRNA species of the chromosome 14 miRNA cluster (C14MC) exhibited strong upregulation in RTT forebrain organoids irrespective of their spatiotemporal location. Together, our results suggest essential roles of the C14MC and hsa-miR-302/367 clusters in EVs during normal and RTT-associated neurodevelopment, displaying promising prospects as biomarkers for monitoring RTT progression.


Asunto(s)
Encéfalo , Vesículas Extracelulares , Proteína 2 de Unión a Metil-CpG , MicroARNs , Organoides , Síndrome de Rett , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Síndrome de Rett/patología , MicroARNs/genética , MicroARNs/metabolismo , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/genética , Humanos , Organoides/metabolismo , Organoides/patología , Femenino , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Mutación , Prosencéfalo/metabolismo
6.
Sci Rep ; 14(1): 22331, 2024 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-39333572

RESUMEN

Brain organoids provide a unique opportunity to model organ development in a system similar to human organogenesis in vivo. Brain organoids thus hold great promise for drug screening and disease modeling. Conventional approaches to organoid characterization predominantly rely on molecular analysis methods, which are expensive, time-consuming, labor-intensive, and involve the destruction of the valuable three-dimensional (3D) architecture of the organoids. This reliance on end-point assays makes it challenging to assess cellular and subcellular events occurring during organoid development in their 3D context. As a result, the long developmental processes are not monitored nor assessed. The ability to perform non-invasive assays is critical for longitudinally assessing features of organoid development during culture. In this paper, we demonstrate a label-free high-content imaging approach for observing changes in organoid morphology and structural changes occurring at the cellular and subcellular level. Enabled by microfluidic-based culture of 3D cell systems and a novel 3D quantitative phase imaging method, we demonstrate the ability to perform non-destructive high-resolution quantitative image analysis of the organoid. The highlighted results demonstrated in this paper provide a new approach to performing live, non-destructive monitoring of organoid systems during culture.


Asunto(s)
Encéfalo , Imagenología Tridimensional , Organoides , Organoides/citología , Encéfalo/diagnóstico por imagen , Encéfalo/citología , Imagenología Tridimensional/métodos , Humanos , Animales , Ratones , Técnicas de Cultivo Tridimensional de Células/métodos
7.
Ecotoxicol Environ Saf ; 285: 117063, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39299213

RESUMEN

Nanoplastics are ubiquitous in our daily lives, raising concerns about their potential impact on the human brain. Many studies reported that nanoplastics permeate the blood-brain barrier and influence cellular processes in mouse models. However, the neurotoxic effects of ingesting nanoplastics on human brain remain poorly understood. Here, we treated cerebral organoids with polystyrene nanoplastics to model the effects of nanoplastic exposure on human brain. Importantly, we found that mitochondria might be the significant organelles affected by polystyrene nanoplastics using immunostaing and RNA-seq analysis. Subsequently, we observed the increased cell death and decreased cell differentiation in our cerebral organoids. In conclusion, our findings shed insights on the mechanisms underlying the toxicity of nanoplastics on human brain organoids, providing an evaluation system in detection potential environmental toxicity on human brain.


Asunto(s)
Encéfalo , Diferenciación Celular , Mitocondrias , Organoides , Humanos , Organoides/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Encéfalo/efectos de los fármacos , Poliestirenos/toxicidad , Células Madre/efectos de los fármacos
8.
Neurotoxicology ; 105: 96-110, 2024 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-39276873

RESUMEN

Human brain development is a complex, multi-stage, and sensitive process, especially during the fetal stage. Animal studies over the last two decades have highlighted the potential risks of anesthetics to the developing brain, impacting its structure and function. This has raised concerns regarding the safety of anesthesia during pregnancy and its influence on fetal brain development, garnering significant attention from the anesthesiology community. Although preclinical studies predominantly indicate the neurotoxic effects of prenatal anesthesia, these findings cannot be directly extrapolated to humans due to interspecies variations. Clinical research, constrained by ethical and technical hurdles in accessing human prenatal brain tissues, often yields conflicting results compared to preclinical data. The emergence of brain organoids as a cutting-edge research tool shows promise in modeling human brain development. When integrated with single-cell sequencing, these organoids offer insights into potential neurotoxic mechanisms triggered by prenatal anesthesia. Despite several retrospective and cohort studies exploring the clinical impact of anesthesia on brain development, many findings remain inconclusive. As such, this review synthesizes preclinical and clinical evidence on the effects of prenatal anesthesia on fetal brain development and suggests areas for future research advancement.

9.
Ageing Res Rev ; : 102517, 2024 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-39321879

RESUMEN

The prevalence of neurodegenerative diseases (NDs) is increasing rapidly as the aging population accelerates, and there are still no treatments to halt or reverse the progression of these diseases. While traditional 2D cultures and animal models fail to translate into effective therapies benefit patients, 3D cultured human brain organoids (hBOs) facilitate the use of non-invasive methods to capture patient data. The purpose of this study was to review the research and application of hBO in disease models and drug screening in NDs. The pluripotent stem cells are induced in multiple stages to form cerebral organoids, brain region-specific organoids and their derived brain cells, which exhibit complex brain-like structures and perform electrophysiological activities. The brain region-specific organoids and their derived neurons or glial cells contribute to the understanding of the pathogenesis of NDs and the efficient development of drugs, including Alzheimer's disease, Parkinson's disease, Huntington's disease and Amyotrophic lateral sclerosis. Glial-rich brain organoids facilitate the study of glial function and neuroinflammation, including astrocytes, microglia, and oligodendrocytes. Further research on the maturation enhancement, vascularization and multi-organoid assembly of hBO will help to enhance the research and application of NDs cellular models.

10.
Trends Biotechnol ; 2024 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-39306492

RESUMEN

Neuroprivacy, or the privacy of neural data, has attracted considerable interest. Here, we explore the implications of neuroprivacy in human brain organoid research, detailing different interpretations of this right. Findings suggest a limited connection between neuroprivacy and brain organoid research, underscoring the importance of further examination of this critical issue.

11.
Environ Pollut ; 362: 124934, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39260546

RESUMEN

Silver nanoparticles (AgNPs) garnered significant attention and applications in the field of nanotechnology due to their unique physicochemical properties. However, with the increasing exposure of AgNPs in the environment and biological systems, concerns about their potential neurotoxicity have also risen. Recent studies on the neurotoxic effects and mechanisms of AgNPs have often relied on traditional toxicological research methods and perspectives. This reliance has limited the extrapolation of these findings to the human brain environment and hindered a deep understanding of the neurotoxicity of AgNPs. This review first outlines the molecular mechanisms of AgNPs-induced neurotoxic injury from a traditional research perspective, identifying oxidative stress, inflammatory responses, and autophagy disorders as key areas of current research. Related molecular signaling pathways, including the nuclear transcription factor-κB (NF-κB) signaling pathway, the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway, and the calcium signaling pathway, have been implicated in the neurotoxic injury process induced by AgNPs. Subsequently, we elucidated the unique advantages of the 3D brain organoids applied to the neurotoxicity study of AgNPs by drawing on relevant studies in the same field. We also emphasize that establishing a standardized 3D brain organoids construction platform is a crucial prerequisite for its widespread application. Furthermore, we suggest that future studies should explore the neurotoxicity mechanisms of AgNPs through the lenses of "adaptive homeostasis" and "structure-activity relationship analysis". In conclusion, the neurotoxicity of AgNPs should be comprehensively evaluated by integrating new research techniques and perspectives, ultimately allowing these nanoparticles to better serve human society.

12.
Ecotoxicol Environ Saf ; 284: 116876, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39146594

RESUMEN

The prevalence of environmental problems and the increasing risk of human exposure to environmental pollutants have become a global concern. The increasing environmental pollution is one of the main reasons for the rising incidence of most neurological-related diseases in recent years. However, the ethical constraints of direct human research and the racial limitations of animal models have slowed the progress of research in this area. The purpose of this study is to review the neurotoxicity of different environmental pollutants on the brain using brain organoids as a new model and to conclude that brain organoids may play a key role in assessing the mechanisms by which environmental pollutants affect neurogenesis and cause neurological pathogenesis. To accurately determine the negative effects of environmental pollutants on the nervous system, self-organizing brain organoids that are highly similar to the developing brain have become a new model system for studying the effects of environmental pollutants on human brain development and disease. This study uses brain organoids as a model to summarize the neurotoxicity of different environmental pollutants on the nervous system, including structural changes in brain organoids, inhibition of neuronal differentiation and migration, impairment of mitochondrial function, damage to cellular cilia, and influence on signaling pathways. In conclusion, exposure to environmental pollutants may cause different neurotoxicity to the nervous system. Therefore, it is crucial to understand how to use brain organoids to ameliorate neurological disorders caused by environmental pollution.


Asunto(s)
Encéfalo , Contaminantes Ambientales , Organoides , Organoides/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Humanos , Encéfalo/efectos de los fármacos , Animales , Neurogénesis/efectos de los fármacos , Síndromes de Neurotoxicidad/patología , Síndromes de Neurotoxicidad/etiología , Modelos Biológicos
13.
Cell Rep Med ; 5(8): 101680, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39121861

RESUMEN

The role of central nervous system (CNS) glia in sustaining self-autonomous inflammation and driving clinical progression in multiple sclerosis (MS) is gaining scientific interest. We applied a single transcription factor (SOX10)-based protocol to accelerate oligodendrocyte differentiation from human induced pluripotent stem cell (hiPSC)-derived neural precursor cells, generating self-organizing forebrain organoids. These organoids include neurons, astrocytes, oligodendroglia, and hiPSC-derived microglia to achieve immunocompetence. Over 8 weeks, organoids reproducibly generated mature CNS cell types, exhibiting single-cell transcriptional profiles similar to the adult human brain. Exposed to inflamed cerebrospinal fluid (CSF) from patients with MS, organoids properly mimic macroglia-microglia neurodegenerative phenotypes and intercellular communication seen in chronic active MS. Oligodendrocyte vulnerability emerged by day 6 post-MS-CSF exposure, with nearly 50% reduction. Temporally resolved organoid data support and expand on the role of soluble CSF mediators in sustaining downstream events leading to oligodendrocyte death and inflammatory neurodegeneration. Such findings support the implementation of this organoid model for drug screening to halt inflammatory neurodegeneration.


Asunto(s)
Encéfalo , Diferenciación Celular , Células Madre Pluripotentes Inducidas , Esclerosis Múltiple , Neuroglía , Organoides , Fenotipo , Humanos , Esclerosis Múltiple/patología , Esclerosis Múltiple/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Organoides/patología , Organoides/metabolismo , Neuroglía/metabolismo , Neuroglía/patología , Encéfalo/patología , Encéfalo/metabolismo , Oligodendroglía/metabolismo , Oligodendroglía/patología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Microglía/metabolismo , Microglía/patología
14.
Front Neurosci ; 18: 1434945, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39156632

RESUMEN

Age-related neurodegenerative diseases, like Alzheimer's disease (AD), are challenging diseases for those affected with no cure and limited treatment options. Functional, human derived brain tissues that represent the diverse genetic background and cellular subtypes contributing to sporadic AD (sAD) are limited. Human stem cell derived brain organoids recapitulate some features of human brain cytoarchitecture and AD-like pathology, providing a tool for illuminating the relationship between AD pathology and neural cell dysregulation leading to cognitive decline. In this review, we explore current strategies for implementing brain organoids in the study of AD as well as the challenges associated with investigating age-related brain diseases using organoid models.

15.
Biol Psychiatry Glob Open Sci ; 4(5): 100344, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39099731

RESUMEN

Background: Human brain organoids are 3-dimensional cellular models that mimic architectural features of a developing brain. Generated from human induced pluripotent stem cells, these organoids offer an unparalleled physiologically relevant in vitro system for disease modeling and drug screening. In the current study, we sought to establish a foundation for a magnetic resonance imaging (MRI)-based, label-free imaging system that offers high-resolution capabilities for deep tissue imaging of whole organoids. Methods: An 11.7T Bruker/89 mm microimaging system was used to collect high-resolution multishell 3-dimensional diffusion images of 2 induced pluripotent stem cell-derived human hippocampal brain organoids. The MRI features identified in the study were interpreted on the basis of similarities with immunofluorescence microscopy. Results: MRI microscopy at ≤40 µm isotropic resolution provided a 3-dimensional view of organoid microstructure. T2-weighted contrast showed a rosette-like internal structure and a protruding spherical structure that correlated with immunofluorescence staining for the choroid plexus. Diffusion tractography methods can be used to model tissue microstructural features and possibly map neuronal organization. This approach complements traditional immunohistochemistry imaging methods without the need for tissue clearing. Conclusions: This proof-of-concept study shows, for the first time, the application of high-resolution diffusion MRI microscopy to image 2-mm diameter spherical human brain organoids. Application of ultrahigh-field MRI and diffusion tractography is a powerful modality for whole organoid imaging and has the potential to make a significant impact for probing microstructural changes in brain organoids used to model psychiatric disorders, neurodegenerative diseases, and viral infections of the human brain, as well as for assessing neurotoxicity in drug screening.


Versace et al. present a groundbreaking approach using ultrahigh-resolution MRI (11.7T) for deep tissue imaging of whole human brain organoids. These 3D miniature brains mimic the developing brain's architecture and hold promise for disease modeling and drug discovery. This label-free MRI approach offers the potential to characterize microstructural features in human brain organoids modeling psychiatric disorders, neurodegenerative diseases, viral infections, and/or drug-induced neurotoxicity.

16.
Drug Metab Pharmacokinet ; 58: 101031, 2024 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-39146603

RESUMEN

Substance use disorders (SUDs) are complex mental health conditions involving a problematic pattern of substance use. Challenges remain in understanding their neural mechanisms, which are likely to lead to improved SUD treatments. Human brain organoids, brain-like 3D in vitro cultures derived from human stem cells, show unique potential in recapitulating the response of a developing human brain to substances. Here, we review the recent progress in understanding SUDs using human brain organoid models focusing on neurodevelopmental perspectives. We first summarize the background of SUDs in humans. Moreover, we introduce the development of various human brain organoid models and then discuss current progress and findings underlying the abuse of substances like nicotine, alcohol, and other addictive drugs using organoid models. Furthermore, we review efforts to develop organ chips and microphysiological systems to engineer better human brain organoids for advancing SUD studies. Lastly, we conclude by elaborating on the current challenges and future directions of SUD studies using human brain organoids.

17.
Mol Brain ; 17(1): 53, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-39107846

RESUMEN

Human embryonic stem cells and human induced pluripotent stem cells may be used to create 3D tissues called brain organoids. They duplicate the physiological and pathological characteristics of human brain tissue more faithfully in terms of both structure and function, and they more precisely resemble the morphology and cellular structure of the human embryonic brain. This makes them valuable models for both drug screening and in vitro studies on the development of the human brain and associated disorders. The technical breakthroughs enabled by brain organoids have a significant impact on the research of different brain regions, brain development and sickness, the connections between the brain and other tissues and organs, and brain evolution. This article discusses the development of brain organoids, their use in diabetes research, and their progress.


Asunto(s)
Encéfalo , Diabetes Mellitus , Organoides , Humanos , Organoides/patología , Encéfalo/patología , Diabetes Mellitus/patología , Animales , Células Madre Pluripotentes Inducidas/citología , Investigación Biomédica
18.
bioRxiv ; 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39091876

RESUMEN

Cryopreservation in cryovials extends cell storage at low temperatures, and advances in organoid cryopreservation improve reproducibility and reduce generation time. However, cryopreserving human organoids presents challenges due to the limited diffusion of cryoprotective agents (CPAs) into the organoid core and the potential toxicity of these agents. To overcome these obstacles, we developed a cryopreservation technique using a pillar plate platform. To illustrate cryopreservation application to human brain organoids (HBOs), early-stage HBOs were produced by differentiating induced pluripotent stem cells (iPSCs) into neuroectoderm (NEs) in an ultralow atachement (ULA) 384-well plate. These NEs were transferred and encapsulated in Matrigel on the pillar plate. The early-stage HBOs on the pillar plate were exposed to four commercially available CPAs, including PSC cryopreservation kit, CryoStor CS10, 3dGRO, and 10% DMSO, before being frozen overnight at -80°C and subsequently stored in a liquid nitrogen dewar. We examined the impact of CPA type, organoid size, and CPA exposure duration on cell viability post-thaw. Additionally, the differentiation of early-stage HBOs on the pillar plate was assessed using RT-qPCR and immunofluorescence staining. The PSC cryopreservation kit proved to be the least toxic for preserving these HBOs on the pillar plate. Notably, smaller HBOs showed higher cell viability post-cryopreservation than larger ones. An incubation period of 80 minutes with the PSC kit was essential to ensure optimal CPA diffusion into HBOs with a diameter of 400 - 600 µm. These cryopreserved early-stage HBOs successfully matured over 30 days, exhibiting gene expression patterns akin to non-cryopreserved HBOs. The cryopreserved early-stage HBOs on the pillar plate maintained high viability after thawing and successfully differentiated into mature HBOs. This on-chip cryopreservation method could extend to other small organoids, by integrating cryopreservation, thawing, culturing, staining, rinsing, and imaging processes within a single system, thereby preserving the 3D structure of the organoids.

19.
Front Neural Circuits ; 18: 1453958, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39161368

RESUMEN

Recent advances in human pluripotent stem cell (hPSC) technologies have prompted the emergence of new research fields and applications for human neurons and brain organoids. Brain organoids have gained attention as an in vitro model system that recapitulates the higher structure, cellular diversity and function of the brain to explore brain development, disease modeling, drug screening, and regenerative medicine. This progress has been accelerated by abundant interactions of brain organoid technology with various research fields. A cross-disciplinary approach with human brain organoid technology offers a higher-ordered advance for more accurately understanding the human brain. In this review, we summarize the status of neural induction in two- and three-dimensional culture systems from hPSCs and the modeling of neurodegenerative diseases using brain organoids. We also highlight the latest bioengineered technologies for the assembly of spatially higher-ordered neural tissues and prospects of brain organoid technology toward the understanding of the potential and abilities of the human brain.


Asunto(s)
Encéfalo , Organoides , Humanos , Encéfalo/fisiología , Encéfalo/citología , Organoides/fisiología , Células Madre Pluripotentes/fisiología , Animales
20.
bioRxiv ; 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39091761

RESUMEN

Human brain organoids produce anatomically relevant cellular structures and recapitulate key aspects of in vivo brain function, which holds great potential to model neurological diseases and screen therapeutics. However, the long growth time of 3D systems complicates the culturing of brain organoids and results in heterogeneity across samples hampering their applications. We developed an integrated platform to enable robust and long-term culturing of 3D brain organoids. We designed a mesofluidic bioreactor device based on a reaction-diffusion scaling theory, which achieves robust media exchange for sufficient nutrient delivery in long-term culture. We integrated this device with longitudinal tracking and machine learning-based classification tools to enable non-invasive quality control of live organoids. This integrated platform allows for sample pre-selection for downstream molecular analysis. Transcriptome analyses of organoids revealed that our mesofluidic bioreactor promoted organoid development while reducing cell death. Our platform thus offers a generalizable tool to establish reproducible culture standards for 3D cellular systems for a variety of applications beyond brain organoids.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA