Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.694
Filter
1.
Methods Mol Biol ; 2855: 505-519, 2025.
Article in English | MEDLINE | ID: mdl-39354324

ABSTRACT

Cell cultures are widely used in studies to gain mechanistic insights of metabolic processes. The foundation of these studies lies on the quantification of intracellular and extracellular metabolites, and nuclear magnetic resonance (NMR) is one of the key analytical platforms used to this aim. Among the factors influencing the quality of the produced data are the sampling procedures as well as the acquisition and processing of spectroscopic data. Here we provide our workflow for obtaining quantitative metabolic data from adherent mammalian cells using NMR spectroscopy. The described protocol is compatible with other analytical methods like LC- or GC-MS-based lipidomics and untargeted metabolomics from the same sample. We also show how the collected extracellular data can be used to extract exchange flux rates, particularly useful for flux analysis studies and metabolic engineering of human-induced pluripotent stem cells.


Subject(s)
Energy Metabolism , Magnetic Resonance Spectroscopy , Metabolomics , Humans , Metabolomics/methods , Magnetic Resonance Spectroscopy/methods , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Metabolome , Animals , Lipidomics/methods
2.
Methods Mol Biol ; 2848: 25-36, 2025.
Article in English | MEDLINE | ID: mdl-39240514

ABSTRACT

The pathological mechanisms of cataract remain largely unknown due to the lack of appropriate in vitro cellular models. We developed a stable in vitro system, namely, a "fried egg" differentiation method to generate functional lentoid bodies (LBs) from induced pluripotent stem cells (iPSCs). The iPSCs-derived LBs exhibited crystalline lens-like morphology and a transparent structure, and expressed lens-specific markers. TEM examination and optical analysis further demonstrated that it has the same cell arrangement structure and magnifying ability as lens.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells , Lens, Crystalline , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Humans , Lens, Crystalline/cytology , Lens, Crystalline/metabolism , Cell Culture Techniques/methods , Cells, Cultured , Cataract/pathology
3.
Methods Mol Biol ; 2848: 197-214, 2025.
Article in English | MEDLINE | ID: mdl-39240525

ABSTRACT

Retinal pigment epithelium (RPE) cells derived from induced pluripotent stem cells (iPSCs) serve multiple roles, including among others, modeling RPE development in normal and pathological conditions, investigating mechanisms of RPE physiology, modeling retinal diseases involving the RPE, and developing strategies for regenerative therapies. We have developed a simple and efficient protocol to generate RPE tissue from human iPSCs-derived retinal organoids. The RPE tissue present in the retinal organoids is analogous to the native human RPE in differentiation timeline, histological organization, and key features of functional maturation. Building upon this system, we established a method to generate functionally mature, polarized RPE monolayers comparable to human primary RPE. This comprehensive protocol outlines the steps for isolating and culturing RPE tissue using retinal organoids. The outcome is a pure population of cells expressing mature RPE signatures and organized in a characteristic cobblestone monolayer featuring robust ultrastructural polarization. These RPE monolayers also exhibit the functional hallmarks of bona fide mature RPE cells, providing a suitable system to mimic the biology and function of the native human RPE.


Subject(s)
Cell Culture Techniques , Cell Differentiation , Induced Pluripotent Stem Cells , Organoids , Retinal Pigment Epithelium , Humans , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Organoids/cytology , Organoids/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Cell Culture Techniques/methods , Cells, Cultured
4.
Biomaterials ; 313: 122770, 2025 Feb.
Article in English | MEDLINE | ID: mdl-39226653

ABSTRACT

Major advances have been made in utilizing human-induced pluripotent stem cells (hiPSCs) for regenerative medicine. Nevertheless, the delivery and integration of hiPSCs into target tissues remain significant challenges, particularly in the context of retinal ganglion cell (RGC) restoration. In this study, we introduce a promising avenue for providing directional guidance to regenerated cells in the retina. First, we developed a technique for construction of gradient interfaces based on functionalized conductive polymers, which could be applied with various functionalized ehthylenedioxythiophene (EDOT) monomers. Using a tree-shaped channel encapsulated with a thin PDMS and a specially designed electrochemical chamber, gradient flow generation could be converted into a functionalized-PEDOT gradient film by cyclic voltammetry. The characteristics of the successfully fabricated gradient flow and surface were analyzed using fluorescent labels, time of flight secondary ion mass spectrometry (TOF-SIMS), and X-ray photoelectron spectroscopy (XPS). Remarkably, hiPSC-RGCs seeded on PEDOT exhibited improvements in neurite outgrowth, axon guidance and neuronal electrophysiology measurements. These results suggest that our novel gradient PEDOT may be used with hiPSC-based technologies as a potential biomedical engineering scaffold for functional restoration of RGCs in retinal degenerative diseases and optic neuropathies.


Subject(s)
Induced Pluripotent Stem Cells , Polymers , Retinal Ganglion Cells , Humans , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/cytology , Induced Pluripotent Stem Cells/cytology , Polymers/chemistry , Axon Guidance , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Surface Properties , Electric Conductivity , Nerve Growth Factors/metabolism , Axons/metabolism , Axons/physiology
5.
Rinsho Ketsueki ; 65(9): 1075-1086, 2024.
Article in Japanese | MEDLINE | ID: mdl-39358263

ABSTRACT

Methods in which patient-derived T cells are genetically modified in vitro and administered to patients have been demonstrated effective in the area of cancer immunotherapy. However, these methods have some unresolved issues such as cost, time, and unstable quality. Several groups have developed strategies to overcome these barriers by regenerating T cells from iPSCs. We have been developing a method in which specific TCR genes are introduced into iPSCs and T cells are regenerated from these iPSCs (TCR-iPSC method). We are now using starting iPSCs from the iPSC stock lines provided by CiRA-F, as the iPSC stock cells are less likely to be rejected. A study aimed at application to solid tumors demonstrated the therapeutic effect of regenerated T cells in a patient tissue xenograft model of WT1 antigen-positive renal cell carcinoma. This article will also discuss strategies by other groups to regenerate various types of T cells from iPSCs.


Subject(s)
Induced Pluripotent Stem Cells , Neoplasms , T-Lymphocytes , Humans , Induced Pluripotent Stem Cells/cytology , Animals , Neoplasms/therapy , Neoplasms/immunology , T-Lymphocytes/immunology , Embryonic Stem Cells/cytology , Virus Diseases/therapy , Virus Diseases/immunology
6.
Mol Med ; 30(1): 168, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39354344

ABSTRACT

BACKGROUND: A spinal cord injury (SCI) can result in severe impairment and fatality as well as significant motor and sensory abnormalities. Exosomes produced from IPSCs have demonstrated therapeutic promise for accelerating spinal cord injury recovery, according to a recent study. OBJECTIVE: This study aims to develop engineered IPSCs-derived exosomes (iPSCs-Exo) capable of targeting and supporting neurons, and to assess their therapeutic potential in accelerating recovery from spinal cord injury (SCI). METHODS: iPSCs-Exo were characterized using Transmission Electron Microscopy (TEM), Nanoparticle Tracking Analysis (NTA), and western blot. To enhance neuronal targeting, iPSCs-Exo were bioengineered, and their uptake by neurons was visualized using PKH26 labeling and fluorescence microscopy. In vitro, the anti-inflammatory effects of miRNA-loaded engineered iPSCs-Exo were evaluated by exposing neurons to LPS and IFN-γ. In vivo, biodistribution of engineered iPSC-Exo was monitored using a vivo imaging system. The therapeutic efficacy of miRNA-loaded engineered iPSC-Exo in a SCI mouse model was assessed by Basso Mouse Scale (BMS) scores, H&E, and Luxol Fast Blue (LFB) staining. RESULTS: The results showed that engineered iPSC-Exo loaded with miRNA promoted the spinal cord injure recovery. Thorough safety assessments using H&E staining on major organs revealed no evidence of systemic toxicity, with normal organ histology and biochemistry profiles following engineered iPSC-Exo administration. CONCLUSION: These results suggest that modified iPSC-derived exosomes loaded with miRNA have great potential as a cutting-edge therapeutic approach to improve spinal cord injury recovery. The observed negligible systemic toxicity further underscores their potential safety and efficacy in clinical applications.


Subject(s)
Exosomes , Induced Pluripotent Stem Cells , MicroRNAs , Spinal Cord Injuries , Exosomes/metabolism , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Animals , MicroRNAs/genetics , Spinal Cord Injuries/therapy , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/genetics , Mice , Disease Models, Animal , Neurons/metabolism , Bioengineering/methods , Female , Spinal Cord Regeneration , Humans
7.
Mol Autism ; 15(1): 42, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39350244

ABSTRACT

BACKGROUND: SETBP1 Haploinsufficiency Disorder (SETBP1-HD) is characterised by mild to moderate intellectual disability, speech and language impairment, mild motor developmental delay, behavioural issues, hypotonia, mild facial dysmorphisms, and vision impairment. Despite a clear link between SETBP1 mutations and neurodevelopmental disorders the precise role of SETBP1 in neural development remains elusive. We investigate the functional effects of three SETBP1 genetic variants including two pathogenic mutations p.Glu545Ter and SETBP1 p.Tyr1066Ter, resulting in removal of SKI and/or SET domains, and a point mutation p.Thr1387Met in the SET domain. METHODS: Genetic variants were introduced into induced pluripotent stem cells (iPSCs) and subsequently differentiated into neurons to model the disease. We measured changes in cellular differentiation, SETBP1 protein localisation, and gene expression changes. RESULTS: The data indicated a change in the WNT pathway, RNA polymerase II pathway and identified GATA2 as a central transcription factor in disease perturbation. In addition, the genetic variants altered the expression of gene sets related to neural forebrain development matching characteristics typical of the SETBP1-HD phenotype. LIMITATIONS: The study investigates changes in cellular function in differentiation of iPSC to neural progenitor cells as a human model of SETBP1 HD disorder. Future studies may provide additional information relevant to disease on further neural cell specification, to derive mature neurons, neural forebrain cells, or brain organoids. CONCLUSIONS: We developed a human SETBP1-HD model and identified perturbations to the WNT and POL2RA pathway, genes regulated by GATA2. Strikingly neural cells for both the SETBP1 truncation mutations and the single nucleotide variant displayed a SETBP1-HD-like phenotype.


Subject(s)
Carrier Proteins , Cell Differentiation , Haploinsufficiency , Induced Pluripotent Stem Cells , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Humans , Carrier Proteins/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Mutation , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Neurons/metabolism , Neural Stem Cells/metabolism , Wnt Signaling Pathway/genetics , Intellectual Disability/genetics , Phenotype
9.
Nat Commun ; 15(1): 8547, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39358374

ABSTRACT

Human induced pluripotent stem cells (iPSCs) have great potential in research, but pluripotency testing faces challenges due to non-standardized methods and ambiguous markers. Here, we use long-read nanopore transcriptome sequencing to discover 172 genes linked to cell states not covered by current guidelines. We validate 12 genes by qPCR as unique markers for specific cell fates: pluripotency (CNMD, NANOG, SPP1), endoderm (CER1, EOMES, GATA6), mesoderm (APLNR, HAND1, HOXB7), and ectoderm (HES5, PAMR1, PAX6). Using these genes, we develop a machine learning-based scoring system, "hiPSCore", trained on 15 iPSC lines and validated on 10 more. hiPSCore accurately classifies pluripotent and differentiated cells and predicts their potential to become specialized 2D cells and 3D organoids. Our re-evaluation of cell fate marker genes identifies key targets for future studies on cell fate assessment. hiPSCore improves iPSC testing by reducing time, subjectivity, and resource use, thus enhancing iPSC quality for scientific and medical applications.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells , Quality Control , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Humans , Cell Differentiation/genetics , Biomarkers/metabolism , Machine Learning , Endoderm/cytology , Endoderm/metabolism , Transcriptome , Mesoderm/metabolism , Mesoderm/cytology , Cell Line , Ectoderm/metabolism , Ectoderm/cytology , Organoids/metabolism , Gene Expression Profiling/methods , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , T-Box Domain Proteins
10.
Nat Commun ; 15(1): 8209, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39294132

ABSTRACT

CRISPR-based gene activation (CRISPRa) is a strategy for upregulating gene expression by targeting promoters or enhancers in a tissue/cell-type specific manner. Here, we describe an experimental framework that combines highly multiplexed perturbations with single-cell RNA sequencing (sc-RNA-seq) to identify cell-type-specific, CRISPRa-responsive cis-regulatory elements and the gene(s) they regulate. Random combinations of many gRNAs are introduced to each of many cells, which are then profiled and partitioned into test and control groups to test for effect(s) of CRISPRa perturbations of both enhancers and promoters on the expression of neighboring genes. Applying this method to a library of 493 gRNAs targeting candidate cis-regulatory elements in both K562 cells and iPSC-derived excitatory neurons, we identify gRNAs capable of specifically upregulating intended target genes and no other neighboring genes within 1 Mb, including gRNAs yielding upregulation of six autism spectrum disorder (ASD) and neurodevelopmental disorder (NDD) risk genes in neurons. A consistent pattern is that the responsiveness of individual enhancers to CRISPRa is restricted by cell type, implying a dependency on either chromatin landscape and/or additional trans-acting factors for successful gene activation. The approach outlined here may facilitate large-scale screens for gRNAs that activate genes in a cell type-specific manner.


Subject(s)
CRISPR-Cas Systems , Enhancer Elements, Genetic , Single-Cell Analysis , Humans , Single-Cell Analysis/methods , K562 Cells , Enhancer Elements, Genetic/genetics , Promoter Regions, Genetic/genetics , RNA, Guide, CRISPR-Cas Systems/genetics , Autism Spectrum Disorder/genetics , Neurons/metabolism , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Clustered Regularly Interspaced Short Palindromic Repeats/genetics
11.
Cell Mol Life Sci ; 81(1): 410, 2024 Sep 21.
Article in English | MEDLINE | ID: mdl-39305343

ABSTRACT

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.


Subject(s)
Brain , Extracellular Vesicles , Methyl-CpG-Binding Protein 2 , MicroRNAs , Organoids , Rett Syndrome , Rett Syndrome/genetics , Rett Syndrome/metabolism , Rett Syndrome/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Humans , Organoids/metabolism , Organoids/pathology , Female , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Brain/metabolism , Brain/pathology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Mutation , Prosencephalon/metabolism
12.
Invest Ophthalmol Vis Sci ; 65(11): 5, 2024 09 03.
Article in English | MEDLINE | ID: mdl-39230994

ABSTRACT

Purpose: Metabolic defects in the retinal pigment epithelium (RPE) underlie many retinal degenerative diseases. This study aims to identify the nutrient requirements of healthy and diseased human RPE cells. Methods: We profiled nutrient use of various human RPE cells, including differentiated and dedifferentiated fetal RPE (fRPE), induced pluripotent stem cell-derived RPE (iPSC RPE), Sorsby fundus dystrophy (SFD) patient-derived iPSC RPE, CRISPR-corrected isogenic SFD (cSFD) iPSC RPE, and ARPE-19 cell lines using Biolog Phenotype MicroArray Assays. Results: Differentiated fRPE cells and healthy iPSC RPE cells can use 51 and 48 nutrients respectively, including sugars, intermediates from glycolysis and tricarboxylic acid (TCA) cycle, fatty acids, ketone bodies, amino acids, and dipeptides. However, when fRPE cells lose their epithelial phenotype through dedifferentiation, nutrient use becomes restricted to 17 nutrients, primarily sugar and glutamine-related amino acids. SFD RPE cells can use 37 nutrients; however, compared to cSFD RPE and healthy iPSC RPE, they are unable to use lactate, some TCA cycle intermediates, and short-chain fatty acids. Nonetheless, they show increased use of branch-chain amino acids (BCAAs) and BCAA-containing dipeptides. Dedifferentiated ARPE-19 cells grown in traditional culture media cannot use lactate and ketone bodies. In contrast, nicotinamide supplementation promotes differentiation toward an epithelial phenotype, restoring the ability to use these nutrients. Conclusions: Epithelial phenotype confers metabolic flexibility to healthy RPE for using various nutrients. SFD RPE cells have reduced metabolic flexibility, relying on the oxidation of BCAAs. Our findings highlight the potentially important roles of nutrient availability and use in RPE differentiation and diseases.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells , Phenotype , Retinal Pigment Epithelium , Humans , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/cytology , Cell Differentiation/physiology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Cells, Cultured , Cell Line
13.
Biomed Phys Eng Express ; 10(6)2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39260389

ABSTRACT

The rapid advancements in 3D printing technology have revolutionized the field of tissue engineering, particularly in the development of neural tissues for the treatment of nervous system diseases. Brain neural tissue, composed of neurons and glial cells, plays a crucial role in the functioning of the brain, spinal cord, and peripheral nervous system by transmitting nerve impulses and processing information. By leveraging 3D bioprinting and bioinks, researchers can create intricate neural scaffolds that facilitate the proliferation and differentiation of nerve cells, thereby promoting the repair and regeneration of damaged neural tissues. This technology allows for the precise spatial arrangement of various cell types and scaffold materials, enabling the construction of complex neural tissue models that closely mimic the natural architecture of the brain. Human-induced pluripotent stem cells (hiPSCs) have emerged as a groundbreaking tool in neuroscience research and the potential treatment of neurological diseases. These cells can differentiate into diverse cell types within the nervous system, including neurons, astrocytes, microglia, oligodendrocytes, and Schwann cells, providing a versatile platform for studying neural networks, neurodevelopment, and neurodegenerative disorders. The use of hiPSCs also opens new avenues for personalized medicine, allowing researchers to model diseases and develop targeted therapies based on individual patient profiles. Despite the promise of direct hiPSC injections for therapeutic purposes, challenges such as poor localization and limited integration have led to the exploration of biomaterial scaffolds as supportive platforms for cell delivery and tissue regeneration. This paper reviews the integration of 3D bioprinting technologies and bioink materials in neuroscience applications, offering a unique platform to create complex brain and tissue architectures that mimic the mechanical, architectural, and biochemical properties of native tissues. These advancements provide robust tools for modelling, repair, and drug screening applications. The review highlights current research, identifies research gaps, and offers recommendations for future studies on 3D bioprinting in neuroscience. The investigation demonstrates the significant potential of 3D bioprinting to fabricate brain-like tissue constructs, which holds great promise for regenerative medicine and drug testing models. This approach offers new avenues for studying brain diseases and potential treatments.


Subject(s)
Bioprinting , Brain , Induced Pluripotent Stem Cells , Printing, Three-Dimensional , Tissue Engineering , Tissue Scaffolds , Humans , Bioprinting/methods , Tissue Engineering/methods , Induced Pluripotent Stem Cells/cytology , Neurons , Animals , Cell Differentiation
14.
Sci Data ; 11(1): 987, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39256463

ABSTRACT

Astrocytes, the predominant glial cells in the central nervous system, play essential roles in maintaining brain function. Reprogramming induced pluripotent stem cells (iPSCs) to become astrocytes through overexpression of the transcription factors, NFIB and SOX9, is a rapid and efficient approach for studying human neurological diseases and identifying therapeutic targets. However, the precise differentiation path and molecular signatures of induced astrocytes remain incompletely understood. Accordingly, we performed single-cell RNA sequencing analysis on 64,736 cells to establish a comprehensive atlas of NFIB/SOX9-directed astrocyte differentiation from human iPSCs. Our dataset provides detailed information about the path of astrocyte differentiation, highlighting the stepwise molecular changes that occur throughout the differentiation process. This dataset serves as a valuable reference for dissecting uncharacterized transcriptomic features of NFIB/SOX9-induced astrocytes and investigating lineage progression during astrocyte differentiation. Moreover, these findings pave the way for future studies on neurological diseases using the NFIB/SOX9-induced astrocyte model.


Subject(s)
Astrocytes , Cell Differentiation , Induced Pluripotent Stem Cells , NFI Transcription Factors , SOX9 Transcription Factor , Transcriptome , SOX9 Transcription Factor/genetics , Astrocytes/metabolism , Astrocytes/cytology , Humans , NFI Transcription Factors/genetics , NFI Transcription Factors/metabolism , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Single-Cell Analysis
15.
Stem Cell Res Ther ; 15(1): 291, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39256865

ABSTRACT

BACKGROUND: Stem-cell-derived therapy is a promising option for tissue regeneration. Human iPSC-derived progenitors of smooth muscle cells (pSMCs) exhibit limited proliferation and differentiation, which minimizes the risk of tumor formation while restoring smooth muscle cells (SMCs). Up to 29% of women suffer from recurrence of vaginal prolapse after prolapse surgery. Therefore, there is a need for therapies that can restore vaginal function. SMCs contribute to vaginal tone and contractility. We sought to examine whether human pSMCs can restore vaginal function in a rat model. METHODS: Female immunocompromised RNU rats were divided into 5 groups: intact controls (n = 12), VSHAM (surgery + saline injection, n = 35), and three cell-injection groups (surgery + cell injection using pSMCs from three patients, n = 14/cell line). The surgery to induce vaginal injury was analogous to prolapse surgery. Menopause was induced by surgical ovariectomy. The vagina, urethra, bladder were harvested 10 weeks after surgery (5 weeks after cell injection). Organ bath myography was performed to evaluate the contractile function of the vagina, and smooth muscle thickness was examined by tissue immunohistochemistry. Collagen I, collagen III, and elastin mRNA and protein expressions in tissues were assessed. RESULTS: Vaginal smooth muscle contractions induced by carbachol and KCl in the cell-injection groups were significantly greater than those in the VSHAM group. Collagen I protein expression in the vagina of the cell-injections groups was significantly higher than in the VSHAM group. Vaginal elastin protein expression was similar between the cell-injection and VSHAM groups. In the urethra, gene expression levels of collagen I, III, and elastin were all significantly greater in the cell-injection groups than in the VSHAM group. Collagen I, III, and elastin protein expression of the urethra did not show a consistent trend between cell-injection groups and the VSHAM group. CONCLUSIONS: Human iPSC-derived pSMCs transplantation appears to be associated with improved contractile function of the surgically injured vagina in a rat model. This is accompanied by changes in extracellular protein expression the vagina and urethra. These observations support further efforts in the translation of pSMCs into a treatment for regenerating the surgically injured vagina in women who suffer recurrent prolapse after surgery.


Subject(s)
Disease Models, Animal , Myocytes, Smooth Muscle , Vagina , Animals , Female , Rats , Humans , Myocytes, Smooth Muscle/metabolism , Stem Cell Transplantation/methods , Elastin/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Muscle Contraction , Cell Differentiation
16.
J Cell Mol Med ; 28(17): e18560, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39258535

ABSTRACT

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.


Subject(s)
Brain , Neurodevelopmental Disorders , Organoids , Humans , Neurodevelopmental Disorders/pathology , Brain/pathology , Brain/growth & development , Brain/cytology , Animals , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Models, Biological
17.
Sci Rep ; 14(1): 22572, 2024 09 29.
Article in English | MEDLINE | ID: mdl-39343771

ABSTRACT

In the present study, we aimed to establish and characterize a mature cortical spheroid model system for Kleefstra syndrome (KS) using patient-derived iPSC. We identified key differences in the growth behavior of KS spheroids determined by reduced proliferation marked by low Ki67 and high E-cadherin expression. Conversely, in the spheroid-based neurite outgrowth assay KS outperformed the control neurite outgrowth due to higher BDNF expression. KS spheroids were highly enriched in VGLUT1/2-expressing glutamatergic and ChAT-expressing cholinergic neurons, while TH-positive catecholamine neurons were significantly underrepresented. Furthermore, high NMDAR1 expression was also detected in the KS spheroid, similarly to other patients-derived neuronal cultures, denoting high NMDAR1 expression as a general, KS-specific marker. Control and KS neuronal progenitors and neurospheres were exposed to different toxicants (paraquat, rotenone, bardoxolone, and doxorubicin), and dose-response curves were assessed after acute exposure. Differentiation stage and compound-specific differences were detected with KS neurospheres being the most sensitive to paraquat. Altogether this study describes a robust 3D model system expressing the disease-specific markers and recapitulating the characteristic pathophysiological traits. This platform is suitable for testing developing brain-adverse environmental effects interactions, drug development, and screening towards individual therapeutic strategies.


Subject(s)
Cell Differentiation , Chromosome Deletion , Chromosomes, Human, Pair 9 , Induced Pluripotent Stem Cells , Spheroids, Cellular , Humans , Chromosomes, Human, Pair 9/genetics , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/cytology , Cell Differentiation/drug effects , Craniofacial Abnormalities/pathology , Craniofacial Abnormalities/metabolism , Intellectual Disability/metabolism , Cell Proliferation/drug effects , Neurons/metabolism , Neurons/drug effects , Neurons/pathology , Receptors, N-Methyl-D-Aspartate/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebral Cortex/drug effects , Cells, Cultured , Brain-Derived Neurotrophic Factor/metabolism , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/drug effects , Rotenone/toxicity , Heart Defects, Congenital , Nerve Tissue Proteins
18.
Nat Commun ; 15(1): 8392, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39349465

ABSTRACT

Mural cells are central to vascular integrity and function. In this study, we demonstrate the innovative use of the transcription factor NKX3.1 to guide the differentiation of human induced pluripotent stem cells into mural progenitor cells (iMPCs). By transiently activating NKX3.1 in mesodermal intermediates, we developed a method that diverges from traditional growth factor-based differentiation techniques. This approach efficiently generates a robust iMPC population capable of maturing into diverse functional mural cell subtypes, including smooth muscle cells and pericytes. These iMPCs exhibit key mural cell functionalities such as contractility, deposition of extracellular matrix, and the ability to support endothelial cell-mediated vascular network formation in vivo. Our study not only underscores the fate-determining significance of NKX3.1 in mural cell differentiation but also highlights the therapeutic potential of these iMPCs. We envision these insights could pave the way for a broader use of iMPCs in vascular biology and regenerative medicine.


Subject(s)
Cell Differentiation , Homeodomain Proteins , Induced Pluripotent Stem Cells , Myocytes, Smooth Muscle , Pericytes , Transcription Factors , Humans , Transcription Factors/metabolism , Transcription Factors/genetics , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Pericytes/cytology , Pericytes/metabolism , Animals , Mice , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Extracellular Matrix/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism
19.
Development ; 151(19)2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39348466

ABSTRACT

Cellular plasticity progressively declines with development and differentiation, yet these processes can be experimentally reversed by reprogramming somatic cells to induced pluripotent stem cells (iPSCs) using defined transcription factors. Advances in reprogramming technology over the past 15 years have enabled researchers to study diseases with patient-specific iPSCs, gain fundamental insights into how cell identity is maintained, recapitulate early stages of embryogenesis using various embryo models, and reverse aspects of aging in cultured cells and animals. Here, we review and compare currently available reprogramming approaches, including transcription factor-based methods and small molecule-based approaches, to derive pluripotent cells characteristic of early embryos. Additionally, we discuss our current understanding of mechanisms that resist reprogramming and their role in cell identity maintenance. Finally, we review recent efforts to rejuvenate cells and tissues with reprogramming factors, as well as the application of iPSCs in deriving novel embryo models to study pre-implantation development.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells , Animals , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Cell Differentiation , Transcription Factors/metabolism , Transcription Factors/genetics , Cell Lineage , Embryonic Development
20.
Cell Rep Methods ; 4(9): 100858, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39255791

ABSTRACT

NGN2-driven induced pluripotent stem cell (iPSC)-to-neuron conversion is a popular method for human neurological disease modeling. In this study, we present a standardized approach for generating neurons utilizing clonal, targeted-engineered iPSC lines with defined reagents. We demonstrate consistent production of excitatory neurons at scale and long-term maintenance for at least 150 days. Temporal omics, electrophysiological, and morphological profiling indicate continued maturation to postnatal-like neurons. Quantitative characterizations through transcriptomic, imaging, and functional assays reveal coordinated actions of multiple pathways that drive neuronal maturation. We also show the expression of disease-related genes in these neurons to demonstrate the relevance of our protocol for modeling neurological disorders. Finally, we demonstrate efficient generation of NGN2-integrated iPSC lines. These workflows, profiling data, and functional characterizations enable the development of reproducible human in vitro models of neurological disorders.


Subject(s)
Induced Pluripotent Stem Cells , Nerve Tissue Proteins , Neurons , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Neurons/physiology , Neurons/cytology , Neurons/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Cell Differentiation , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Neurogenesis/physiology , Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL