Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 361
Filter
1.
Front Biosci (Landmark Ed) ; 29(3): 114, 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38538275

ABSTRACT

Amyotrophic Lateral Sclerosis (ALS) is a neurodegenerative disorder, characterized by progressive loss of both upper and lower motor neurons, resulting in clinical features such as muscle weakness, paralysis, and ultimately, respiratory failure. Nowadays, there is not effective treatment to reverse the progression of the disease, that leads to death within 3-5 years after the onset. Nevertheless, the induced pluripotent stem cells (iPS) technology could be the answer, providing disease modelling, drug testing, and cell-based therapies for this pathology. The aim of this work was to conduct a literature review of the past 5 years about the role of iPS in ALS, to better define the neurobiological mechanisms involved in the pathogenesis and the potential future therapies. The review also deals with advanced and currently available technologies used to reprogram cell lines and generate human motor neurons in vitro, which represent the source to study the pathological processes, the relationship between phenotype and genotype, the disease progression and the potential therapeutic targets of these group of disorders. Specific treatment options with stem cells involve Advance Gene Editing Technology, neuroprotective agents, and cells or exosomes transplantation, aimed to replace dead or damaged nerve cells. In summary, this review comprehensively addresses the role of human pluripotent stem cells (hPSCs) in motor neuron diseases (MND), with a focus on physiopathology, diagnostic and prognostic implications, specific and potential future treatment options. Understanding the biological mechanisms and practical implications of hPSCs in MND is crucial for advancing therapeutic strategies and improving outcomes for patients affected by these devastating diseases.


Subject(s)
Amyotrophic Lateral Sclerosis , Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Humans , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/therapy , Amyotrophic Lateral Sclerosis/metabolism , Motor Neurons/metabolism , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology
2.
Stem Cell Res ; 71: 103174, 2023 09.
Article in English | MEDLINE | ID: mdl-37531724

ABSTRACT

Hypomyelinating Leukodystrophy 22 (HLD22) is caused by a stoploss mutation in CLDN11. To study the molecular mechanisms underlying HLD22, human induced pluripotent stem cells (hiPSCs) were generated from patient fibroblasts carrying the stop-loss mutation in CLDN11.


Subject(s)
Cell Line , Hereditary Central Nervous System Demyelinating Diseases , Pluripotent Stem Cells , Humans , Male , Child , Fibroblasts/pathology , Hereditary Central Nervous System Demyelinating Diseases/genetics , Hereditary Central Nervous System Demyelinating Diseases/pathology , Pluripotent Stem Cells/pathology
3.
Stem Cell Reports ; 18(2): 585-596, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36638788

ABSTRACT

Macrophages armed with chimeric antigen receptors (CARs) provide a potent new option for treating solid tumors. However, genetic engineering and scalable production of somatic macrophages remains significant challenges. Here, we used CRISPR-Cas9 gene editing methods to integrate an anti-GD2 CAR into the AAVS1 locus of human pluripotent stem cells (hPSCs). We then established a serum- and feeder-free differentiation protocol for generating CAR macrophages (CAR-Ms) through arterial endothelial-to-hematopoietic transition (EHT). CAR-M produced by this method displayed a potent cytotoxic activity against GD2-expressing neuroblastoma and melanoma in vitro and neuroblastoma in vivo. This study provides a new platform for the efficient generation of off-the-shelf CAR-Ms for antitumor immunotherapy.


Subject(s)
Melanoma , Neuroblastoma , Pluripotent Stem Cells , Receptors, Chimeric Antigen , Humans , Receptors, Chimeric Antigen/genetics , Receptors, Antigen, T-Cell/genetics , Immunotherapy/methods , Pluripotent Stem Cells/pathology , Melanoma/therapy , Neuroblastoma/therapy , Neuroblastoma/pathology , Macrophages/pathology
4.
J Mol Cell Cardiol ; 174: 77-87, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36403760

ABSTRACT

Advances in stem cell biology have facilitated cardiac regeneration, and many animal studies and several initial clinical trials have been conducted using human pluripotent stem cell-derived cardiomyocytes (PSC-CMs). Most preclinical and clinical studies have typically transplanted PSC-CMs via the following two distinct approaches: direct intramyocardial injection or epicardial delivery of engineered heart tissue. Both approaches present common disadvantages, including a mandatory thoracotomy and poor engraftment. Furthermore, a standard transplantation approach has yet to be established. In this study, we tested the feasibility of performing intracoronary administration of PSC-CMs based on a commonly used method of transplanting somatic stem cells. Six male cynomolgus monkeys underwent intracoronary administration of dispersed human PSC-CMs or PSC-CM aggregates, which are called cardiac spheroids, with multiple cell dosages. The recipient animals were sacrificed at 4 weeks post-transplantation for histological analysis. Intracoronary administration of dispersed human PSC-CMs in the cynomolgus monkeys did not lead to coronary embolism or graft survival. Although the transplanted cardiac spheroids became partially engrafted, they also induced scar formation due to cardiac ischemic injury. Cardiac engraftment and scar formation were reasonably consistent with the spheroid size or cell dosage. These findings indicate that intracoronary transplantation of PSC-CMs is an inefficient therapeutic approach.


Subject(s)
Myocytes, Cardiac , Pluripotent Stem Cells , Animals , Humans , Male , Cicatrix/pathology , Macaca fascicularis , Myocytes, Cardiac/pathology , Pluripotent Stem Cells/pathology
5.
Neuromuscul Disord ; 32(11-12): 908-922, 2022 12.
Article in English | MEDLINE | ID: mdl-36418198

ABSTRACT

Mutations in the dystrophin gene cause the most common and currently incurable Duchenne muscular dystrophy (DMD) characterized by progressive muscle wasting. Although abnormal Ca2+ handling is a pathological feature of DMD, mechanisms underlying defective Ca2+ homeostasis remain unclear. Here we generate a novel DMD patient-derived pluripotent stem cell (PSC) model of skeletal muscle with an isogenic control using clustered regularly interspaced short palindromic repeat (CRISPR)-mediated precise gene correction. Transcriptome analysis identifies dysregulated gene sets in the absence of dystrophin, including genes involved in Ca2+ handling, excitation-contraction coupling and muscle contraction. Specifically, analysis of intracellular Ca2+ transients and mathematical modeling of Ca2+ dynamics reveal significantly reduced cytosolic Ca2+ clearance rates in DMD-PSC derived myotubes. Pharmacological assays demonstrate Ca2+ flux in myotubes is determined by both intracellular and extracellular sources. DMD-PSC derived myotubes display significantly reduced velocity of contractility. Compared with a non-isogenic wildtype PSC line, these pathophysiological defects could be rescued by CRISPR-mediated precise gene correction. Our study provides new insights into abnormal Ca2+ homeostasis in DMD and suggests that Ca2+ signaling pathways amenable to pharmacological modulation are potential therapeutic targets. Importantly, we have established a human physiology-relevant in vitro model enabling rapid pre-clinical testing of potential therapies for DMD.


Subject(s)
Muscular Dystrophy, Duchenne , Pluripotent Stem Cells , Humans , Dystrophin/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , CRISPR-Cas Systems , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Muscular Dystrophy, Duchenne/pathology , Muscle, Skeletal/pathology , Muscle Fibers, Skeletal/pathology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology
6.
Int J Mol Sci ; 23(14)2022 Jul 14.
Article in English | MEDLINE | ID: mdl-35887140

ABSTRACT

Previous studies have shown that early therapeutic events of neural precursor cells (NPCs) transplantation to animals with acute ischemic stroke readily protected neuronal cell damage and improved behavioral recovery through paracrine mechanisms. In this study, we tested the hypothesis that administration of conditioned medium from NPCs (NPC-CMs) could recapitulate the beneficial effects of cell transplantation. Rats with permanent middle cerebral artery occlusion (pMCAO) were randomly assigned to one of the following groups: PBS control, Vehicle (medium) controls, single (NPC-CM(S)) or multiple injections of NPC-CM(NPC-CM(M)) groups. A single intravenous injection of NPC-CM exhibited strong neuroregenerative potential to induce behavioral recovery, and multiple injections enhanced this activity further by suppressing inflammatory damage and inducing endogenous neurogenesis leading to histopathological and functional recovery. Proteome analysis of NPC-CM identified a number of proteins that are known to be associated with nervous system development, neurogenesis, and angiogenesis. In addition, transcriptome analysis revealed the importance of the inflammatory response during stroke recovery and some of the key hub genes in the interaction network were validated. Thus, our findings demonstrated that NPC-CM promoted functional recovery and reduced cerebral infarct and inflammation with enhanced endogenous neurogenesis, and the results highlighted the potency of NPC-CM in stroke therapy.


Subject(s)
Ischemic Stroke , Neural Stem Cells , Pluripotent Stem Cells , Stroke , Animals , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Humans , Neurogenesis , Neurons , Pluripotent Stem Cells/pathology , Rats , Recovery of Function , Stroke/pathology , Stroke/therapy
7.
Phytomedicine ; 103: 154234, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35689903

ABSTRACT

BACKGROUND: The Modified Shenlingbaizhu Decoction (MSD) utilizes various phytomedicines has been applied to treat colorectal cancer (CRC). Colorectal cancer stem cells (CSCs) have proven to be tightly associated with CRC progression and metastasis. The mechanism of MSD's inhibitory effect on CSCs has not been determined. PURPOSE: To figure out how MSD inhibits the pluripotency of CSCs and impedes the EMT program. METHODS: The ingredients of MSD extracts were characterized by high-performance liquid chromatography (HPLC). BALB/c-nu mice were transplanted into EGFP labeled SW480 CRC cells and the tumor weight and volume were recorded before and after various doses of MSD treatment. The concentration of TGF-ß1 was quantified with an Enzyme-linked immunosorbent assay. To delineate the logical relationship between EMT and CSCs regulated by MSD, TGF-ß/Smad inhibitor and activator were adopted in tumor-bearing mice and diverse CRC cell lines. Cancer stem cell markers were analyzed by flow cytometry. In vitro analysis of cell motility and viability were done using CCK-8, wound healing, and invasion assay. Immunohistochemistry (IHC) and western blotting (WB) were used for detecting protein expression. The collected results were statistically analyzed with GraphPad Prism 8.0. RESULTS: MSD treatment significantly reduced the size of colorectal cancer tumors and lowered the serum content of TGF-ß1 in mice. Importantly, MSD markedly reduced the expression of pluripotent factors and depressed CD133+ stem cells in the tumor tissues. The TGF-ß/Smad inhibitor neutralized the EMT signaling and lowered the pluripotency by dephosphorylation of SMAD2/3. Similarly, MSD attenuated the pluripotency by limiting TGF-ß/Smad signaling-induced EMT in vivo. MSD inhibited colorectal cancer cell proliferation, migration, and invasion. CONCLUSIONS: MSD inhibits the growth of colorectal cancer. It dampens the pluripotency of CSCs by repressing the TGF-ß-induced EMT program.


Subject(s)
Colorectal Neoplasms , Drugs, Chinese Herbal , Neoplastic Stem Cells , Pluripotent Stem Cells , Transforming Growth Factor beta1 , Animals , Cell Line, Tumor , Cell Movement/drug effects , Colorectal Neoplasms/blood , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drugs, Chinese Herbal/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phytotherapy , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/blood
8.
Nat Cell Biol ; 24(5): 659-671, 2022 05.
Article in English | MEDLINE | ID: mdl-35550611

ABSTRACT

Heart regeneration is an unmet clinical need, hampered by limited renewal of adult cardiomyocytes and fibrotic scarring. Pluripotent stem cell-based strategies are emerging, but unravelling cellular dynamics of host-graft crosstalk remains elusive. Here, by combining lineage tracing and single-cell transcriptomics in injured non-human primate heart biomimics, we uncover the coordinated action modes of human progenitor-mediated muscle repair. Chemoattraction via CXCL12/CXCR4 directs cellular migration to injury sites. Activated fibroblast repulsion targets fibrosis by SLIT2/ROBO1 guidance in organizing cytoskeletal dynamics. Ultimately, differentiation and electromechanical integration lead to functional restoration of damaged heart muscle. In vivo transplantation into acutely and chronically injured porcine hearts illustrated CXCR4-dependent homing, de novo formation of heart muscle, scar-volume reduction and prevention of heart failure progression. Concurrent endothelial differentiation contributed to graft neovascularization. Our study demonstrates that inherent developmental programmes within cardiac progenitors are sequentially activated in disease, enabling the cells to sense and counteract acute and chronic injury.


Subject(s)
Nerve Tissue Proteins , Pluripotent Stem Cells , Animals , Cell Differentiation , Cicatrix/pathology , Cicatrix/prevention & control , Fibrosis , Humans , Myocardium/pathology , Myocytes, Cardiac/pathology , Pluripotent Stem Cells/pathology , Receptors, Immunologic , Swine
9.
Cells ; 10(11)2021 11 19.
Article in English | MEDLINE | ID: mdl-34831467

ABSTRACT

Human pluripotent stem cells (hPSC) are known to acquire chromosomal abnormalities, which range from point mutations to large copy number changes, including full chromosome aneuploidy. These aberrations have a wide-ranging influence on the state of cells, in both the undifferentiated and differentiated state. Currently, very little is known on how these abnormalities will impact the clinical translation of hPSC, and particularly their potential to prime cells for oncogenic transformation. A further complication is that many of these abnormalities exist in a mosaic state in culture, which complicates their detection with conventional karyotyping methods. In this review we discuss current knowledge on how these aberrations influence the cell state and how this may impact the future of research and the cells' clinical potential.


Subject(s)
Chromosome Aberrations , Pluripotent Stem Cells/pathology , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Differentiation/genetics , DNA Copy Number Variations/genetics , Humans , Models, Biological , Pluripotent Stem Cells/metabolism
10.
Int J Mol Sci ; 22(17)2021 Sep 05.
Article in English | MEDLINE | ID: mdl-34502527

ABSTRACT

Glucocorticoids are amongst the most used drugs to treat retinal diseases of various origins. Yet, the transcriptional regulations induced by glucocorticoid receptor (GR) and mineralocorticoid receptor (MR) activation in retinal pigment epithelium cells (RPE) that form the outer blood-retina barrier are unknown. Levels of endogenous corticoids, ligands for MR and GR, were measured in human ocular media. Human RPE cells derived from induced pluripotent stem cells (iRPE) were used to analyze the pan-transcriptional regulations induced by aldosterone-an MR-specific agonist, or cortisol or cortisol + RU486-a GR antagonist. The retinal phenotype of transgenic mice that overexpress the human MR (P1.hMR) was analyzed. In the human eye, the main ligand for GR and MR is cortisol. The iRPE cells express functional GR and MR. The subset of genes regulated by aldosterone and by cortisol + RU-486, and not by cortisol alone, mimics an imbalance toward MR activation. They are involved in extracellular matrix remodeling (CNN1, MGP, AMTN), epithelial-mesenchymal transition, RPE cell proliferation and migration (ITGB3, PLAUR and FOSL1) and immune balance (TNFSF18 and PTX3). The P1.hMR mice showed choroidal vasodilation, focal alteration of the RPE/choroid interface and migration of RPE cells together with RPE barrier function alteration, similar to human retinal diseases within the pachychoroid spectrum. RPE is a corticosteroid-sensitive epithelium. MR pathway activation in the RPE regulates genes involved in barrier function, extracellular matrix, neural regulation and epithelial differentiation, which could contribute to retinal pathology.


Subject(s)
Aldosterone/metabolism , Hydrocortisone/metabolism , Pluripotent Stem Cells/metabolism , Receptors, Mineralocorticoid/metabolism , Retinal Diseases/metabolism , Retinal Pigment Epithelium/metabolism , Animals , Epithelial-Mesenchymal Transition , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Humans , Mice , Mice, Transgenic , Pluripotent Stem Cells/pathology , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/genetics , Retinal Diseases/genetics , Retinal Diseases/pathology , Retinal Pigment Epithelium/pathology
11.
Science ; 373(6559): eabc1048, 2021 Sep 03.
Article in English | MEDLINE | ID: mdl-34516843

ABSTRACT

Oncogenes only transform cells under certain cellular contexts, a phenomenon called oncogenic competence. Using a combination of a human pluripotent stem cell­derived cancer model along with zebrafish transgenesis, we demonstrate that the transforming ability of BRAFV600E along with additional mutations depends on the intrinsic transcriptional program present in the cell of origin. In both systems, melanocytes are less responsive to mutations, whereas both neural crest and melanoblast populations are readily transformed. Profiling reveals that progenitors have higher expression of chromatin-modifying enzymes such as ATAD2, a melanoma competence factor that forms a complex with SOX10 and allows for expression of downstream oncogenic and neural crest programs. These data suggest that oncogenic competence is mediated by regulation of developmental chromatin factors, which then allow for proper response to those oncogenes.


Subject(s)
Carcinogenesis/genetics , Carcinogenesis/pathology , Chromatin/metabolism , Melanoma/genetics , Melanoma/pathology , Neural Crest/pathology , ATPases Associated with Diverse Cellular Activities/genetics , ATPases Associated with Diverse Cellular Activities/metabolism , Animals , Animals, Genetically Modified , Chromatin/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Melanocytes/metabolism , Melanocytes/pathology , Mice , Neoplasms, Experimental , Neoplastic Stem Cells/pathology , Neural Crest/metabolism , Pluripotent Stem Cells/pathology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Transcription, Genetic , Zebrafish
12.
Int J Mol Sci ; 22(16)2021 Aug 20.
Article in English | MEDLINE | ID: mdl-34445681

ABSTRACT

Parthenogenetic embryos have been widely studied as an effective tool related to paternal and maternal imprinting genes and reproductive problems for a long time. In this study, we established a parthenogenetic epiblast-like stem cell line through culturing parthenogenetic diploid blastocysts in a chemically defined medium containing activin A and bFGF named paAFSCs. The paAFSCs expressed pluripotent marker genes and germ-layer-related genes, as well as being alkaline-phosphatase-positive, which is similar to epiblast stem cells (EpiSCs). We previously showed that advanced embryonic stem cells (ASCs) represent hypermethylated naive pluripotent embryonic stem cells (ESCs). Here, we converted paAFSCs to ASCs by replacing bFGF with bone morphogenetic protein 4 (BMP4), CHIR99021, and leukemia inhibitory factor (LIF) in a culture medium, and we obtained parthenogenetic advanced stem cells (paASCs). The paASCs showed similar morphology with ESCs and also displayed a stronger developmental potential than paAFSCs in vivo by producing chimaeras. Our study demonstrates that maternal genes could support parthenogenetic EpiSCs derived from blastocysts and also have the potential to convert primed state paAFSCs to naive state paASCs.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Parthenogenesis/physiology , Activins/metabolism , Animals , Blastocyst/metabolism , Bone Morphogenetic Protein 4/pharmacology , Cell Culture Techniques/methods , Cell Differentiation/drug effects , DNA Methylation/drug effects , Embryo Culture Techniques/methods , Female , Fibroblast Growth Factors/pharmacology , Germ Layers/metabolism , Germ Layers/physiology , Leukemia Inhibitory Factor/pharmacology , Mice , Mice, 129 Strain , Mice, Inbred ICR , Mouse Embryonic Stem Cells/cytology , Parthenogenesis/genetics , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology
13.
Int J Mol Sci ; 22(14)2021 Jul 20.
Article in English | MEDLINE | ID: mdl-34299348

ABSTRACT

Mitochondrial diseases disrupt cellular energy production and are among the most complex group of inherited genetic disorders. Affecting approximately 1 in 5000 live births, they are both clinically and genetically heterogeneous, and can be highly tissue specific, but most often affect cell types with high energy demands in the brain, heart, and kidneys. There are currently no clinically validated treatment options available, despite several agents showing therapeutic promise. However, modelling these disorders is challenging as many non-human models of mitochondrial disease do not completely recapitulate human phenotypes for known disease genes. Additionally, access to disease-relevant cell or tissue types from patients is often limited. To overcome these difficulties, many groups have turned to human pluripotent stem cells (hPSCs) to model mitochondrial disease for both nuclear-DNA (nDNA) and mitochondrial-DNA (mtDNA) contexts. Leveraging the capacity of hPSCs to differentiate into clinically relevant cell types, these models permit both detailed investigation of cellular pathomechanisms and validation of promising treatment options. Here we catalogue hPSC models of mitochondrial disease that have been generated to date, summarise approaches and key outcomes of phenotypic profiling using these models, and discuss key criteria to guide future investigations using hPSC models of mitochondrial disease.


Subject(s)
Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Pluripotent Stem Cells/pathology , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , DNA, Mitochondrial/genetics , Humans , Phenotype
14.
Cells ; 10(6)2021 06 01.
Article in English | MEDLINE | ID: mdl-34206038

ABSTRACT

Human midbrain dopamine (DA) neurons are a heterogeneous group of cells that share a common neurotransmitter phenotype and are in close anatomical proximity but display different functions, sensitivity to degeneration, and axonal innervation targets. The A9 DA neuron subtype controls motor function and is primarily degenerated in Parkinson's disease (PD), whereas A10 neurons are largely unaffected by the condition, and their dysfunction is associated with neuropsychiatric disorders. Currently, DA neurons can only be reliably classified on the basis of topographical features, including anatomical location in the midbrain and projection targets in the forebrain. No systematic molecular classification at the genome-wide level has been proposed to date. Although many years of scientific efforts in embryonic and adult mouse brain have positioned us to better understand the complexity of DA neuron biology, many biological phenomena specific to humans are not amenable to being reproduced in animal models. The establishment of human cell-based systems combined with advanced computational single-cell transcriptomics holds great promise for decoding the mechanisms underlying maturation and diversification of human DA neurons, and linking their molecular heterogeneity to functions in the midbrain. Human pluripotent stem cells have emerged as a useful tool to recapitulate key molecular features of mature DA neuron subtypes. Here, we review some of the most recent advances and discuss the current challenges in using stem cells, to model human DA biology. We also describe how single cell RNA sequencing may provide key insights into the molecular programs driving DA progenitor specification into mature DA neuron subtypes. Exploiting the state-of-the-art approaches will lead to a better understanding of stem cell-derived DA neurons and their use in disease modeling and regenerative medicine.


Subject(s)
Dopaminergic Neurons/metabolism , Mesencephalon/metabolism , Parkinson Disease , Pluripotent Stem Cells/metabolism , RNA-Seq , Single-Cell Analysis , Animals , Dopaminergic Neurons/pathology , Humans , Mesencephalon/pathology , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Pluripotent Stem Cells/pathology
15.
Cells ; 10(7)2021 06 22.
Article in English | MEDLINE | ID: mdl-34206228

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder characterized by the progressive decline of motor, cognitive, and psychiatric functions. HD results from an autosomal dominant mutation that causes a trinucleotide CAG repeat expansion and the production of mutant Huntingtin protein (mHTT). This results in the initial selective and progressive loss of medium spiny neurons (MSNs) in the striatum before progressing to involve the whole brain. There are currently no effective treatments to prevent or delay the progression of HD as knowledge into the mechanisms driving the selective degeneration of MSNs has been hindered by a lack of access to live neurons from individuals with HD. The invention of cell reprogramming provides a revolutionary technique for the study, and potential treatment, of neurological conditions. Cell reprogramming technologies allow for the generation of live disease-affected neurons from patients with neurological conditions, becoming a primary technique for modelling these conditions in vitro. The ability to generate HD-affected neurons has widespread applications for investigating the pathogenesis of HD, the identification of new therapeutic targets, and for high-throughput drug screening. Cell reprogramming also offers a potential autologous source of cells for HD cell replacement therapy. This review provides a comprehensive analysis of the use of cell reprogramming to model HD and a discussion on recent advancements in cell reprogramming technologies that will benefit the HD field.


Subject(s)
Cellular Reprogramming , Huntington Disease/pathology , Models, Biological , Animals , Disease Models, Animal , Humans , Phenotype , Pluripotent Stem Cells/pathology
16.
Methods Mol Biol ; 2299: 435-445, 2021.
Article in English | MEDLINE | ID: mdl-34028759

ABSTRACT

The mechanisms of kidney injury and fibrosis can now be studied using kidney organoids derived from human pluripotent stem cells (hPSCs). Mature kidney organoids contain nephrons and stromal cells with fibrogenic potential, spatially organized in a manner that resembles the anatomy of the kidney. Organoid nephron damage and interstitial fibrosis can be induced under well-controlled experimental conditions in vitro, making this an ideal system for the study of tissue-intrinsic cell signaling and intercellular crosstalk mechanisms in the absence of systemic signals and immune cells that are present in vivo. Here we describe methods for the generation of kidney organoids from a widely used hPSC line, and for the induction and analysis of nephron damage and interstitial fibrosis.


Subject(s)
Cell Culture Techniques/methods , Kidney/pathology , Organoids/pathology , Pluripotent Stem Cells/cytology , Cell Communication , Cell Line , Fibrosis , Genetic Markers , Humans , Kidney/chemistry , Microscopy, Fluorescence , Organoids/chemistry , Organoids/cytology , Pluripotent Stem Cells/chemistry , Pluripotent Stem Cells/pathology , Real-Time Polymerase Chain Reaction , Signal Transduction
17.
STAR Protoc ; 2(2): 100493, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33997813

ABSTRACT

This protocol describes the production of hepatocyte-like cells (HLCs) from human pluripotent stem cells and how to induce hepatic steatosis, a condition characterized by intracellular lipid accumulation. Following differentiation to an HLC phenotype, intracellular lipid accumulation is induced with a steatosis induction cocktail, allowing the user to examine the cellular processes that underpin hepatic steatosis. Furthermore, the renewable nature of our system, on a defined genetic background, permits in-depth mechanistic analysis, which may facilitate therapeutic target identification in the future. For complete details on the use and execution of this protocol, please refer to Sinton et al. (2021).


Subject(s)
Cell Differentiation , Fatty Liver/metabolism , Hepatocytes/metabolism , Models, Biological , Pluripotent Stem Cells/metabolism , Fatty Liver/pathology , Hepatocytes/pathology , Humans , Pluripotent Stem Cells/pathology
18.
J Cell Biol ; 220(6)2021 06 07.
Article in English | MEDLINE | ID: mdl-33851958

ABSTRACT

It is well established that short telomeres activate an ATM-driven DNA damage response that leads to senescence in terminally differentiated cells. However, technical limitations have hampered our understanding of how telomere shortening is signaled in human stem cells. Here, we show that telomere attrition induces ssDNA accumulation (G-strand) at telomeres in human pluripotent stem cells (hPSCs), but not in their differentiated progeny. This led to a unique role for ATR in the response of hPSCs to telomere shortening that culminated in an extended S/G2 cell cycle phase and a longer period of mitosis, which was associated with aneuploidy and mitotic catastrophe. Loss of p53 increased resistance to death, at the expense of increased mitotic abnormalities in hPSCs. Taken together, our data reveal an unexpected dominant role of ATR in hPSCs, combined with unique cell cycle abnormalities and, ultimately, consequences distinct from those observed in their isogenic differentiated counterparts.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle , Mitosis , Pluripotent Stem Cells/pathology , Telomere/physiology , Tumor Suppressor Protein p53/metabolism , Aneuploidy , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Cycle Proteins/genetics , DNA Damage , Humans , Pluripotent Stem Cells/metabolism , Tumor Suppressor Protein p53/genetics
19.
Mol Cell ; 81(11): 2349-2360.e6, 2021 06 03.
Article in English | MEDLINE | ID: mdl-33852895

ABSTRACT

Telomere length control is critical for cellular lifespan and tumor suppression. Telomerase is transiently activated in the inner cell mass of the developing blastocyst to reset telomere reserves. Its silencing upon differentiation leads to gradual telomere shortening in somatic cells. Here, we report that transcriptional regulation through cis-regulatory elements only partially accounts for telomerase activation in pluripotent cells. Instead, developmental control of telomerase is primarily driven by an alternative splicing event, centered around hTERT exon 2. Skipping of exon 2 triggers hTERT mRNA decay in differentiated cells, and conversely, its retention promotes telomerase accumulation in pluripotent cells. We identify SON as a regulator of exon 2 alternative splicing and report a patient carrying a SON mutation and suffering from insufficient telomerase and short telomeres. In summary, our study highlights a critical role for hTERT alternative splicing in the developmental regulation of telomerase and implicates defective splicing in telomere biology disorders.


Subject(s)
Alternative Splicing , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic , Minor Histocompatibility Antigens/genetics , Telomerase/genetics , Telomere Homeostasis , Telomere/metabolism , Blastocyst/metabolism , Blastocyst/pathology , Cell Differentiation , Child, Preschool , DNA-Binding Proteins/deficiency , Female , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/pathology , Humans , Pedigree , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology , Primary Cell Culture , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Telomerase/deficiency , Telomere/pathology
20.
Cells ; 10(4)2021 04 20.
Article in English | MEDLINE | ID: mdl-33924240

ABSTRACT

Muse cells are non-tumorigenic endogenous reparative pluripotent cells with high therapeutic potential. They are identified as cells positive for the pluripotent surface marker SSEA-3 in the bone marrow, peripheral blood, and connective tissue. Muse cells also express other pluripotent stem cell markers, are able to differentiate into cells representative of all three germ layers, self-renew from a single cell, and are stress tolerant. They express receptors for sphingosine-1-phosphate (S1P), which is actively produced by damaged cells, allowing circulating cells to selectively home to damaged tissue. Muse cells spontaneously differentiate on-site into multiple tissue-constituent cells with few errors and replace damaged/apoptotic cells with functional cells, thereby contributing to tissue repair. Intravenous injection of exogenous Muse cells to increase the number of circulating Muse cells enhances their reparative activity. Muse cells also have a specific immunomodulatory system, represented by HLA-G expression, allowing them to be directly administered without HLA-matching or immunosuppressant treatment. Owing to these unique characteristics, clinical trials using intravenously administered donor-Muse cells have been conducted for myocardial infarction, stroke, epidermolysis bullosa, spinal cord injury, perinatal hypoxic ischemic encephalopathy, and amyotrophic lateral sclerosis. Muse cells have the potential to break through the limitations of current cell therapies for neurologic diseases, including amyotrophic lateral sclerosis. Muse cells provide a new therapeutic strategy that requires no HLA-matching or immunosuppressant treatment for administering donor-derived cells, no gene introduction or differentiation induction for cell preparation, and no surgery for delivering the cells to patients.


Subject(s)
Neoplasms/pathology , Nervous System Diseases/therapy , Pluripotent Stem Cells/pathology , Animals , Apoptosis , Clinical Trials as Topic , Humans , Mesenchymal Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...