Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 3873, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38719882

ABSTRACT

Human glial progenitor cells (hGPCs) exhibit diminished expansion competence with age, as well as after recurrent demyelination. Using RNA-sequencing to compare the gene expression of fetal and adult hGPCs, we identify age-related changes in transcription consistent with the repression of genes enabling mitotic expansion, concurrent with the onset of aging-associated transcriptional programs. Adult hGPCs develop a repressive transcription factor network centered on MYC, and regulated by ZNF274, MAX, IKZF3, and E2F6. Individual over-expression of these factors in iPSC-derived hGPCs lead to a loss of proliferative gene expression and an induction of mitotic senescence, replicating the transcriptional changes incurred during glial aging. miRNA profiling identifies the appearance of an adult-selective miRNA signature, imposing further constraints on the expansion competence of aged GPCs. hGPC aging is thus associated with acquisition of a MYC-repressive environment, suggesting that suppression of these repressors of glial expansion may permit the rejuvenation of aged hGPCs.


Subject(s)
Aging , MicroRNAs , Neuroglia , Transcription Factors , Humans , Neuroglia/metabolism , Neuroglia/cytology , Aging/genetics , Aging/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Cellular Senescence/genetics , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Stem Cells/metabolism , Stem Cells/cytology , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Adult , Gene Regulatory Networks , Cell Proliferation/genetics , Gene Expression Regulation, Developmental , Gene Expression Profiling
2.
Nat Biotechnol ; 2023 Jul 17.
Article in English | MEDLINE | ID: mdl-37460676

ABSTRACT

Competition among adult brain cells has not been extensively researched. To investigate whether healthy glia can outcompete diseased human glia in the adult forebrain, we engrafted wild-type (WT) human glial progenitor cells (hGPCs) produced from human embryonic stem cells into the striata of adult mice that had been neonatally chimerized with mutant Huntingtin (mHTT)-expressing hGPCs. The WT hGPCs outcompeted and ultimately eliminated their human Huntington's disease (HD) counterparts, repopulating the host striata with healthy glia. Single-cell RNA sequencing revealed that WT hGPCs acquired a YAP1/MYC/E2F-defined dominant competitor phenotype upon interaction with the host HD glia. WT hGPCs also outcompeted older resident isogenic WT cells that had been transplanted neonatally, suggesting that competitive success depended primarily on the relative ages of competing populations, rather than on the presence of mHTT. These data indicate that aged and diseased human glia may be broadly replaced in adult brain by younger healthy hGPCs, suggesting a therapeutic strategy for the replacement of aged and diseased human glia.

3.
Biochem Biophys Res Commun ; 530(1): 252-258, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32828295

ABSTRACT

Human adipose-derived stem cells (ASCs) are a commonly used cell type for cartilage tissue engineering. However, donor-to-donor variability, cell heterogeneity, inconsistent chondrogenic potential, and limited expansion potential can hinder the use of these cells for modeling chondrogenesis, in vitro screening of drugs and treatments for joint diseases, or translational applications for tissue engineered cartilage repair. The goal of this study was to create an immortalized ASC line that showed enhanced and consistent chondrogenic potential for applications in cartilage tissue engineering as well as to provide a platform for investigation of biological and mechanobiological pathways involved in cartilage homeostasis and disease. Starting with the ASC52telo cell line, a hTERT-immortalized ASC line, we used lentivirus to overexpress SOX9, a master regulator of chondrogenesis, and screened several clonal populations of SOX9 overexpressing cells to form a new stable cell line with high chondrogenic potential. One clonal line, named ASC52telo-SOX9, displayed increased GAG and type II collagen synthesis and was found to be responsive to both mechanical and inflammatory stimuli in a manner similar to native chondrocytes. The development of a clonal line such as ASC52telo-SOX9 has the potential to be a powerful tool for studying cartilage homeostasis and disease mechanisms in vitro, and potentially as a platform for in vitro drug screening for diseases that affect articular cartilage. Our findings provide an approach for the development of other immortalized cell lines with improved chondrogenic capabilities in ASCs or other adult stem cells.


Subject(s)
Chondrocytes/cytology , Chondrogenesis , Mesenchymal Stem Cells/cytology , Cell Line , Chondrocytes/metabolism , Humans , Mesenchymal Stem Cells/metabolism , NF-kappa B/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , TRPV Cation Channels/metabolism , Up-Regulation
4.
Matrix Biol ; 89: 1-10, 2020 07.
Article in English | MEDLINE | ID: mdl-31874220

ABSTRACT

The coordinated spatial and temporal regulation of gene expression in the murine hindlimb determines the identity of mesenchymal progenitors and the development of diversity of musculoskeletal tissues they form. Hindlimb development has historically been studied with lineage tracing of individual genes selected a priori, or at the bulk tissue level, which does not allow for the determination of single cell transcriptional programs yielding mature cell types and tissues. To identify the cellular trajectories of lineage specification during limb bud development, we used single cell mRNA sequencing (scRNA-seq) to profile the developing murine hindlimb between embryonic days (E)11.5-E18.5. We found cell type heterogeneity at all time points, and the expected cell types that form the mouse hindlimb. In addition, we used RNA fluorescence in situ hybridization (FISH) to examine the spatial locations of cell types and cell trajectories to understand the ancestral continuum of cell maturation. This data provides a resource for the transcriptional program of hindlimb development that will support future studies of musculoskeletal development and generate hypotheses for tissue regeneration.


Subject(s)
Gene Expression Profiling/methods , Hindlimb/growth & development , Single-Cell Analysis/methods , Animals , Cell Differentiation , Cell Lineage , Embryonic Development , Female , Gene Expression Regulation, Developmental , Hindlimb/embryology , Hindlimb/metabolism , In Situ Hybridization, Fluorescence , Male , Mice , Sequence Analysis, RNA
5.
Cell Rep ; 27(13): 3832-3843.e6, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31242417

ABSTRACT

Astrocytic differentiation is developmentally impaired in patients with childhood-onset schizophrenia (SCZ). To determine why, we used genetic gain- and loss-of-function studies to establish the contributions of differentially expressed transcriptional regulators to the defective differentiation of glial progenitor cells (GPCs) produced from SCZ patient-derived induced pluripotent cells (iPSCs). Negative regulators of the bone morphogenetic protein (BMP) pathway were upregulated in SCZ GPCs, including BAMBI, FST, and GREM1, whose overexpression retained SCZ GPCs at the progenitor stage. SMAD4 knockdown (KD) suppressed the production of these BMP inhibitors by SCZ GPCs and rescued normal astrocytic differentiation. In addition, the BMP-regulated transcriptional repressor REST was upregulated in SCZ GPCs, and its KD similarly restored normal glial differentiation. REST KD also rescued potassium-transport-associated gene expression and K+ uptake, which were otherwise deficient in SCZ glia. These data suggest that the glial differentiation defect in childhood-onset SCZ, and its attendant disruption in K+ homeostasis, may be rescued by targeting BMP/SMAD4- and REST-dependent transcription.


Subject(s)
Cell Differentiation , Neuroglia/metabolism , Repressor Proteins/metabolism , Schizophrenia/metabolism , Signal Transduction , Smad4 Protein/metabolism , Adolescent , Adult , Cell Line , Child , Female , Humans , Male , Neuroglia/pathology , Repressor Proteins/genetics , Schizophrenia/genetics , Schizophrenia/pathology , Smad4 Protein/genetics
6.
FASEB J ; 33(1): 358-372, 2019 01.
Article in English | MEDLINE | ID: mdl-29985644

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) provide an attractive cell source for cartilage repair and cell therapy; however, the underlying molecular pathways that drive chondrogenesis of these populations of adult stem cells remain poorly understood. We generated a rich data set of high-throughput RNA sequencing of human MSCs throughout chondrogenesis at 6 different time points. Our data consisted of 18 libraries with 3 individual donors as biologic replicates, with each library possessing a sequencing depth of 100 million reads. Computational analyses with differential gene expression, gene ontology, and weighted gene correlation network analysis identified dynamic changes in multiple biologic pathways and, most importantly, a chondrogenic gene subset, whose functional characterization promises to further harness the potential of MSCs for cartilage tissue engineering. Furthermore, we created a graphic user interface encyclopedia built with the goal of producing an open resource of transcriptomic regulation for additional data mining and pathway analysis of the process of MSC chondrogenesis.-Huynh, N. P. T., Zhang, B., Guilak, F. High-depth transcriptomic profiling reveals the temporal gene signature of human mesenchymal stem cells during chondrogenesis.


Subject(s)
Adult Stem Cells/metabolism , Cartilage/metabolism , Cell Differentiation , Chondrocytes/metabolism , Gene Expression Profiling , Gene Regulatory Networks , Mesenchymal Stem Cells/metabolism , Adult Stem Cells/cytology , Cartilage/cytology , Cells, Cultured , Chondrocytes/cytology , High-Throughput Nucleotide Sequencing , Humans , Mesenchymal Stem Cells/cytology , Signal Transduction , Tissue Engineering
7.
Biomaterials ; 177: 161-175, 2018 09.
Article in English | MEDLINE | ID: mdl-29894913

ABSTRACT

Cartilage-derived matrix (CDM) has emerged as a promising scaffold material for tissue engineering of cartilage and bone due to its native chondroinductive capacity and its ability to support endochondral ossification. Because it consists of native tissue, CDM can undergo cellular remodeling, which can promote integration with host tissue and enables it to be degraded and replaced by neotissue over time. However, enzymatic degradation of decellularized tissues can occur unpredictably and may not allow sufficient time for mechanically competent tissue to form, especially in the harsh inflammatory environment of a diseased joint. The goal of the current study was to engineer cartilage and bone constructs with the ability to inhibit aberrant inflammatory processes caused by the cytokine interleukin-1 (IL-1), through scaffold-mediated delivery of lentiviral particles containing a doxycycline-inducible IL-1 receptor antagonist (IL-1Ra) transgene on anatomically-shaped CDM constructs. Additionally, scaffold-mediated lentiviral gene delivery was used to facilitate spatial organization of simultaneous chondrogenic and osteogenic differentiation via site-specific transduction of a single mesenchymal stem cell (MSC) population to overexpress either chondrogenic, transforming growth factor-beta 3 (TGF-ß3), or osteogenic, bone morphogenetic protein-2 (BMP-2), transgenes. Controlled induction of IL-1Ra expression protected CDM hemispheres from inflammation-mediated degradation, and supported robust bone and cartilage tissue formation even in the presence of IL-1. In the absence of inflammatory stimuli, controlled cellular remodeling was exploited as a mechanism for fusing concentric CDM hemispheres overexpressing BMP-2 and TGF-ß3 into a single bi-layered osteochondral construct. Our findings demonstrate that site-specific delivery of inducible and tunable transgenes confers spatial and temporal control over both CDM scaffold remodeling and neotissue composition. Furthermore, these constructs provide a microphysiological in vitro joint organoid model with site-specific, tunable, and inducible protein delivery systems for examining the spatiotemporal response to pro-anabolic and/or inflammatory signaling across the osteochondral interface.


Subject(s)
Cartilage, Articular/chemistry , Gene Transfer Techniques , Lentivirus/genetics , Mesenchymal Stem Cells/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Bone Morphogenetic Protein 2/genetics , Cells, Cultured , Chondrogenesis , Humans , Osteogenesis , Swine , Transduction, Genetic , Transforming Growth Factor beta3/genetics , Transgenes
8.
Tissue Eng Part A ; 24(19-20): 1531-1544, 2018 10.
Article in English | MEDLINE | ID: mdl-29756533

ABSTRACT

Tissue engineering approaches for the repair of osteochondral defects using biomaterial scaffolds and stem cells have remained challenging due to the inherent complexities of inducing cartilage-like matrix and bone-like matrix within the same local environment. Members of the transforming growth factor ß (TGFß) family have been extensively utilized in the engineering of skeletal tissues, but have distinct effects on chondrogenic and osteogenic differentiation of progenitor cells. The goal of this study was to develop a method to direct human bone marrow-derived mesenchymal stem cells (MSCs) to deposit either mineralized matrix or a cartilaginous matrix rich in glycosaminoglycan and type II collagen within the same biochemical environment. This differential induction was performed by culturing cells on engineered three-dimensionally woven poly(ɛ-caprolactone) (PCL) scaffolds in a chondrogenic environment for cartilage-like matrix production while inhibiting TGFß3 signaling through Mothers against DPP homolog 3 (SMAD3) knockdown, in combination with overexpressing RUNX2, to achieve mineralization. The highest levels of mineral deposition and alkaline phosphatase activity were observed on scaffolds with genetically engineered MSCs and exhibited a synergistic effect in response to SMAD3 knockdown and RUNX2 expression. Meanwhile, unmodified MSCs on PCL scaffolds exhibited accumulation of an extracellular matrix rich in glycosaminoglycan and type II collagen in the same biochemical environment. This ability to derive differential matrix deposition in a single culture condition opens new avenues for developing complex tissue replacements for chondral or osteochondral defects.


Subject(s)
Extracellular Matrix/metabolism , Genetic Engineering/methods , Mesenchymal Stem Cells/metabolism , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Adult , Cells, Cultured , Chondrogenesis/drug effects , Collagen Type II/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Extracellular Matrix/drug effects , Gene Knockdown Techniques , Glycosaminoglycans/metabolism , Humans , Mesenchymal Stem Cells/drug effects , Minerals/metabolism , Osteogenesis/drug effects , Smad3 Protein/metabolism , Transforming Growth Factor beta3/pharmacology
9.
Connect Tissue Res ; 58(1): 116-141, 2017 01.
Article in English | MEDLINE | ID: mdl-27254479

ABSTRACT

Normal skeletal development requires tight coordination of transcriptional networks, signaling pathways, and biomechanical cues, and many of these pathways are dysregulated in pathological conditions affecting cartilage and bone. Recently, a significant role has been identified for long noncoding RNAs (lncRNAs) in developing and maintaining cellular phenotypes, and improvements in sequencing technologies have led to the identification of thousands of lncRNAs across diverse cell types, including the cells within cartilage and bone. It is clear that lncRNAs play critical roles in regulating gene expression. For example, they can function as epigenetic regulators in the nucleus via chromatin modulation to control gene transcription, or in the cytoplasm, where they can function as scaffolds for protein-binding partners or modulate the activity of other coding and noncoding RNAs. In this review, we discuss the growing list of lncRNAs involved in normal development and/or homeostasis of the skeletal system, the potential mechanisms by which these lncRNAs might function, and recent improvements in the methodologies available to study lncRNA functions in vitro and in vivo. Finally, we address the likely utility of lncRNAs as biomarkers and therapeutic targets for diseases of the skeletal system, including osteoarthritis, osteoporosis, and in cancers of the skeletal system.


Subject(s)
Bone Neoplasms , Gene Expression Regulation, Neoplastic , Osteoarthritis , Osteoporosis , RNA, Long Noncoding , RNA, Neoplasm , Animals , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Humans , Osteoarthritis/genetics , Osteoarthritis/metabolism , Osteoarthritis/pathology , Osteoporosis/genetics , Osteoporosis/metabolism , Osteoporosis/pathology , RNA, Long Noncoding/biosynthesis , RNA, Long Noncoding/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics
10.
Proc Natl Acad Sci U S A ; 111(9): E798-806, 2014 Mar 04.
Article in English | MEDLINE | ID: mdl-24550481

ABSTRACT

The ability to develop tissue constructs with matrix composition and biomechanical properties that promote rapid tissue repair or regeneration remains an enduring challenge in musculoskeletal engineering. Current approaches require extensive cell manipulation ex vivo, using exogenous growth factors to drive tissue-specific differentiation, matrix accumulation, and mechanical properties, thus limiting their potential clinical utility. The ability to induce and maintain differentiation of stem cells in situ could bypass these steps and enhance the success of engineering approaches for tissue regeneration. The goal of this study was to generate a self-contained bioactive scaffold capable of mediating stem cell differentiation and formation of a cartilaginous extracellular matrix (ECM) using a lentivirus-based method. We first showed that poly-L-lysine could immobilize lentivirus to poly(ε-caprolactone) films and facilitate human mesenchymal stem cell (hMSC) transduction. We then demonstrated that scaffold-mediated gene delivery of transforming growth factor ß3 (TGF-ß3), using a 3D woven poly(ε-caprolactone) scaffold, induced robust cartilaginous ECM formation by hMSCs. Chondrogenesis induced by scaffold-mediated gene delivery was as effective as traditional differentiation protocols involving medium supplementation with TGF-ß3, as assessed by gene expression, biochemical, and biomechanical analyses. Using lentiviral vectors immobilized on a biomechanically functional scaffold, we have developed a system to achieve sustained transgene expression and ECM formation by hMSCs. This method opens new avenues in the development of bioactive implants that circumvent the need for ex vivo tissue generation by enabling the long-term goal of in situ tissue engineering.


Subject(s)
Cell Differentiation/physiology , Chondrogenesis/physiology , Extracellular Matrix/physiology , Tissue Engineering/methods , Tissue Scaffolds/virology , Transduction, Genetic/methods , Analysis of Variance , Biomechanical Phenomena , DNA Primers/genetics , Flow Cytometry , Gene Transfer Techniques , Humans , Immunohistochemistry , Lentivirus , Mesenchymal Stem Cells/metabolism , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Polyesters , Polylysine , Regenerative Medicine/methods , Transforming Growth Factor beta3/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...