Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.399
Filtrer
1.
Arterioscler Thromb Vasc Biol ; 44(7): 1523-1536, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38695171

RÉSUMÉ

The implementation of human induced pluripotent stem cell (hiPSC) models has introduced an additional tool for identifying molecular mechanisms of disease that complement animal models. Patient-derived or CRISPR/Cas9-edited induced pluripotent stem cells differentiated into smooth muscle cells (SMCs) have been leveraged to discover novel mechanisms, screen potential therapeutic strategies, and model in vivo development. The field has evolved over almost 15 years of research using hiPSC-SMCs and has made significant strides toward overcoming initial challenges such as the lineage specificity of SMC phenotypes. However, challenges both specific (eg, the lack of specific markers to thoroughly validate hiPSC-SMCs) and general (eg, a lack of transparency and consensus around methodology in the field) remain. In this review, we highlight the recent successes and remaining challenges of the hiPSC-SMC model.


Sujet(s)
Différenciation cellulaire , Cellules souches pluripotentes induites , Myocytes du muscle lisse , Humains , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/anatomopathologie , Myocytes du muscle lisse/anatomopathologie , Myocytes du muscle lisse/métabolisme , Animaux , Phénotype , Muscles lisses vasculaires/anatomopathologie , Muscles lisses vasculaires/métabolisme , Lignage cellulaire
2.
Stem Cell Res ; 77: 103444, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38761686

RÉSUMÉ

The NF1 gene is related to neurofibromatosis type 1 (NF1), which is an autosomal dominant disorder associated with multisystem involvement and epilepsy susceptibility. A human induced pluripotent stem cell (iPSC) line was derived from a pediatric patient with NF1 and epilepsy, harboring a heterozygous NF1 gene mutation. The iPSC line exhibits high levels of pluripotency markers, maintains the NF1 gene mutation, and demonstrates the capacity to undergo differentiation potential in vitro into three germ layers. The iPSC line will serve as a valuable resource for investigating the underlying mechanisms and conducting drug screening related to NF1 and NF1-associated epilepsy.


Sujet(s)
Épilepsie , Hétérozygote , Cellules souches pluripotentes induites , Mutation , Neurofibromatose de type 1 , Humains , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/anatomopathologie , Neurofibromatose de type 1/génétique , Neurofibromatose de type 1/anatomopathologie , Épilepsie/génétique , Épilepsie/anatomopathologie , Neurofibromine-1/génétique , Lignée cellulaire , Différenciation cellulaire , Mâle , Gènes nf1
3.
Biochem Biophys Res Commun ; 721: 150124, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-38776833

RÉSUMÉ

Prader-Willi syndrome (PWS) is a complex epigenetic disorder caused by the deficiency of paternally expressed genes in chromosome 15q11-q13. This syndrome also includes endocrine dysfunction, leading to short stature, hypogonadism, and obscure hyperphagia. Although recent progress has been made toward understanding the genetic basis for PWS, the molecular mechanisms underlying its pathology in obesity remain unclear. In this study, we examined the adipocytic characteristics of two PWS-induced pluripotent stem cell (iPSC) lines: those with the 15q11-q13 gene deletion (iPWS cells) and those with 15q11-q13 abnormal methylation (M-iPWS cells). The transcript levels of the lipid-binding protein aP2 were decreased in iPWS and M-iPWS adipocytes. Flow-cytometry analysis showed that PWS adipocytes accumulated more lipid droplets than did normal individual adipocytes. Furthermore, glucose uptake upon insulin stimulation was attenuated compared to that in normal adipocytes. Overall, our results suggest a significantly increased lipid content and defective in glucose metabolism in PWS adipocytes.


Sujet(s)
Adipocytes , Cellules souches pluripotentes induites , Syndrome de Prader-Willi , Syndrome de Prader-Willi/anatomopathologie , Syndrome de Prader-Willi/métabolisme , Syndrome de Prader-Willi/génétique , Adipocytes/métabolisme , Adipocytes/anatomopathologie , Humains , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/anatomopathologie , Glucose/métabolisme , Chromosomes humains de la paire 15/génétique , Chromosomes humains de la paire 15/métabolisme , Protéines de liaison aux acides gras/métabolisme , Protéines de liaison aux acides gras/génétique , Lignée cellulaire , Méthylation de l'ADN , Délétion de gène , Métabolisme lipidique , Insuline/métabolisme
4.
Mol Genet Metab ; 142(2): 108492, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38759397

RÉSUMÉ

Pathogenic variants in the O-GlcNAc transferase gene (OGT) have been associated with a congenital disorder of glycosylation (OGT-CDG), presenting with intellectual disability which may be of neuroectodermal origin. To test the hypothesis that pathology is linked to defects in differentiation during early embryogenesis, we developed an OGT-CDG induced pluripotent stem cell line together with isogenic control generated by CRISPR/Cas9 gene-editing. Although the OGT-CDG variant leads to a significant decrease in OGT and O-GlcNAcase protein levels, there were no changes in differentiation potential or stemness. However, differentiation into ectoderm resulted in significant differences in O-GlcNAc homeostasis. Further differentiation to neuronal stem cells revealed differences in morphology between patient and control lines, accompanied by disruption of the O-GlcNAc pathway. This suggests a critical role for O-GlcNAcylation in early neuroectoderm architecture, with robust compensatory mechanisms in the earliest stages of stem cell differentiation.


Sujet(s)
Différenciation cellulaire , Cellules souches pluripotentes induites , Déficience intellectuelle , N-acetylglucosaminyltransferase , Plaque neurale , Phénotype , Humains , N-acetylglucosaminyltransferase/génétique , N-acetylglucosaminyltransferase/métabolisme , Déficience intellectuelle/génétique , Déficience intellectuelle/anatomopathologie , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/anatomopathologie , Plaque neurale/métabolisme , Troubles congénitaux de la glycosylation/génétique , Troubles congénitaux de la glycosylation/anatomopathologie , Troubles congénitaux de la glycosylation/métabolisme , Systèmes CRISPR-Cas , Glycosylation , Édition de gène , Cellules souches neurales/métabolisme , Cellules souches neurales/anatomopathologie
5.
Acta Neuropathol Commun ; 12(1): 69, 2024 04 25.
Article de Anglais | MEDLINE | ID: mdl-38664831

RÉSUMÉ

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder that primarily affects motor neurons, leading to progressive muscle weakness and loss of voluntary muscle control. While the exact cause of ALS is not fully understood, emerging research suggests that dysfunction of the nuclear envelope (NE) may contribute to disease pathogenesis and progression. The NE plays a role in ALS through several mechanisms, including nuclear pore defects, nucleocytoplasmic transport impairment, accumulation of mislocalized proteins, and nuclear morphology abnormalities. The LINC complex is the second biggest multi-protein complex in the NE and consists of the SUN1/2 proteins spanning the inner nuclear membrane and Nesprin proteins embedded in the outer membrane. The LINC complex, by interacting with both the nuclear lamina and the cytoskeleton, transmits mechanical forces to the nucleus regulating its morphology and functional homeostasis. In this study we show extensive alterations to the LINC complex in motor and cortical iPSC-derived neurons and spinal cord organoids carrying the ALS causative mutation in the C9ORF72 gene (C9). Importantly, we show that such alterations are present in vivo in a cohort of sporadic ALS and C9-ALS postmortem spinal cord and motor cortex specimens. We also found that LINC complex disruption strongly correlated with nuclear morphological alterations occurring in ALS neurons, independently of TDP43 mislocalization. Altogether, our data establish morphological and functional alterations to the LINC complex as important events in ALS pathogenic cascade, making this pathway a possible target for both biomarker and therapy development.


Sujet(s)
Sclérose latérale amyotrophique , Protéine C9orf72 , Démence frontotemporale , Sclérose latérale amyotrophique/génétique , Sclérose latérale amyotrophique/anatomopathologie , Sclérose latérale amyotrophique/métabolisme , Humains , Protéine C9orf72/génétique , Protéine C9orf72/métabolisme , Démence frontotemporale/génétique , Démence frontotemporale/anatomopathologie , Démence frontotemporale/métabolisme , Mâle , Motoneurones/anatomopathologie , Motoneurones/métabolisme , Moelle spinale/anatomopathologie , Moelle spinale/métabolisme , Enveloppe nucléaire/métabolisme , Enveloppe nucléaire/anatomopathologie , Femelle , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/anatomopathologie , Adulte d'âge moyen , Sujet âgé , Cortex moteur/anatomopathologie , Cortex moteur/métabolisme
6.
Article de Anglais | MEDLINE | ID: mdl-38571695

RÉSUMÉ

In rheumatoid arthritis, dysregulated cytokine signaling has been implicated as a primary factor in chronic inflammation. Many antirheumatic and biological therapies are used to suppress joint inflammation, but despite these advances, effectiveness is not universal, and delivery is often at high doses, which can predispose patients to significant off-target effects. During chronic inflammation, the inappropriate regulation of signaling factors by macrophages accelerates progression of disease by driving an imbalance of inflammatory cytokines, making macrophages an ideal cellular target. To develop a macrophage-based therapy to treat chronic inflammation, we engineered a novel induced pluripotent stem cell (iPSC)-derived macrophage capable of delivering soluble TNF receptor 1 (TNFR1), an anti-inflammatory biologic inhibitor of tumor necrosis factor alpha (TNF-α), in an auto-regulated manner in response to TNF-α. Murine iPSCs were differentiated into macrophages (iMACs) over a 17-day optimized protocol with continued successful differentiation confirmed at key timepoints. Varying inflammatory and immunomodulatory stimuli demonstrated traditional macrophage function and phenotypes. In response to TNF-α, therapeutic iMACs produced high levels of sTNFR1 in an autoregulated manner, which inhibited inflammatory signaling. This self-regulating iMAC system demonstrated the potential for macrophage-based drug delivery as a novel therapeutic approach for a variety of chronic inflammatory diseases.


Sujet(s)
Produits biologiques , Cellules souches pluripotentes induites , Humains , Souris , Animaux , Facteur de nécrose tumorale alpha/pharmacologie , Cellules souches pluripotentes induites/anatomopathologie , Cytokines/pharmacologie , Macrophages , Inflammation/anatomopathologie , Anti-inflammatoires/pharmacologie , Produits biologiques/usage thérapeutique
7.
Biol Chem ; 405(6): 427-439, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38651266

RÉSUMÉ

Integration of multiple data sources presents a challenge for accurate prediction of molecular patho-phenotypic features in automated analysis of data from human model systems. Here, we applied a machine learning-based data integration to distinguish patho-phenotypic features at the subcellular level for dilated cardiomyopathy (DCM). We employed a human induced pluripotent stem cell-derived cardiomyocyte (iPSC-CM) model of a DCM mutation in the sarcomere protein troponin T (TnT), TnT-R141W, compared to isogenic healthy (WT) control iPSC-CMs. We established a multimodal data fusion (MDF)-based analysis to integrate source datasets for Ca2+ transients, force measurements, and contractility recordings. Data were acquired for three additional layer types, single cells, cell monolayers, and 3D spheroid iPSC-CM models. For data analysis, numerical conversion as well as fusion of data from Ca2+ transients, force measurements, and contractility recordings, a non-negative blind deconvolution (NNBD)-based method was applied. Using an XGBoost algorithm, we found a high prediction accuracy for fused single cell, monolayer, and 3D spheroid iPSC-CM models (≥92 ± 0.08 %), as well as for fused Ca2+ transient, beating force, and contractility models (>96 ± 0.04 %). Integrating MDF and XGBoost provides a highly effective analysis tool for prediction of patho-phenotypic features in complex human disease models such as DCM iPSC-CMs.


Sujet(s)
Cardiomyopathie dilatée , Cellules souches pluripotentes induites , Apprentissage machine , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/anatomopathologie , Cardiomyopathie dilatée/anatomopathologie , Cardiomyopathie dilatée/métabolisme , Humains , Phénotype , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Troponine T/métabolisme , Calcium/métabolisme
8.
Commun Biol ; 7(1): 413, 2024 Apr 09.
Article de Anglais | MEDLINE | ID: mdl-38594382

RÉSUMÉ

Better understanding of the earliest molecular pathologies of all neurodegenerative diseases is expected to improve human therapeutics. We investigated the earliest molecular pathology of spinocerebellar ataxia type 1 (SCA1), a rare familial neurodegenerative disease that primarily induces death and dysfunction of cerebellum Purkinje cells. Extensive prior studies have identified involvement of transcription or RNA-splicing factors in the molecular pathology of SCA1. However, the regulatory network of SCA1 pathology, especially central regulators of the earliest developmental stages and inflammatory events, remains incompletely understood. Here, we elucidated the earliest developmental pathology of SCA1 using originally developed dynamic molecular network analyses of sequentially acquired RNA-seq data during differentiation of SCA1 patient-derived induced pluripotent stem cells (iPSCs) to Purkinje cells. Dynamic molecular network analysis implicated histone genes and cytokine-relevant immune response genes at the earliest stages of development, and revealed relevance of ISG15 to the following degradation and accumulation of mutant ataxin-1 in Purkinje cells of SCA1 model mice and human patients.


Sujet(s)
Cellules souches pluripotentes induites , Ataxies spinocérébelleuses , Animaux , Humains , Souris , Cytokines , Cellules souches pluripotentes induites/anatomopathologie , Souris transgéniques , Cellules de Purkinje/physiologie , Ataxies spinocérébelleuses/génétique , Ataxies spinocérébelleuses/anatomopathologie , Ubiquitines
9.
Stem Cell Res ; 77: 103381, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38493608

RÉSUMÉ

Congenital disorder of glycosylation (CDG) is inherited metabolicdiseasecaused by defects in the genes important for the process of protein and lipidglycosylation. We established an induced pluripotent stem cell (iPSC) line from peripheral blood mononuclear cells of a 6-month-old boy with congenital disorder of glycosylation carrying heterozygous mutations c.1193 T > C (p.I398T) and c.376_384dup CCGCAGCAC (p.P126_H128 dupPQH) in MPI gene. This iPSC line was free of exogenous gene, expressed pluripotency markers, has normal karyotype, exhibited differentiation potential and harbored the same mutations found in the patient. This cell line will provide a reliable cell model for further studies on the potential therapeutic targets of CDG.


Sujet(s)
Troubles congénitaux de la glycosylation , Hétérozygote , Cellules souches pluripotentes induites , Mutation , Humains , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/anatomopathologie , Troubles congénitaux de la glycosylation/génétique , Troubles congénitaux de la glycosylation/anatomopathologie , Mâle , Nourrisson , Lignée cellulaire , Phosphotransferases phosphomutases/génétique , Phosphotransferases phosphomutases/déficit , Différenciation cellulaire , Glycosylation
10.
Stem Cell Res ; 77: 103401, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38537501

RÉSUMÉ

The CMT1A variant accounts for over 60% of cases of Charcot-Marie-Tooth disease (CMT), one of the most common human neuropathies. The cause of CMT1A has been identified as the duplication of PMP22, a myelin protein expressed in Schwann cells. Yet, the pathological mechanisms have not been elucidated, and no treatment is currently available. In our study, we established an iPS cell line from a CMT1A patient with PMP22 duplication. The generated iPSCs maintain pluripotency and in vitro differentiation potency.


Sujet(s)
Maladie de Charcot-Marie-Tooth , Cellules souches pluripotentes induites , Protéines de la myéline , Maladie de Charcot-Marie-Tooth/génétique , Maladie de Charcot-Marie-Tooth/anatomopathologie , Humains , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/anatomopathologie , Protéines de la myéline/génétique , Protéines de la myéline/métabolisme , Lignée cellulaire , Différenciation cellulaire , Duplication de gène , Mâle
11.
Stem Cell Res ; 76: 103379, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38458030

RÉSUMÉ

Leigh syndrome is a rare autosomal recessive disorder showcasing a diverse range of neurological symptoms. Classical Leigh syndrome is associated with mitochondrial complex I deficiency, primarily resulting from biallelic mutations in the NDUFAF5 gene, encoding the NADH:ubiquinone oxidoreductase complex assembly factor 5. Using the Sendai virus delivery system, we generated an induced pluripotent stem cell line from peripheral blood mononuclear cells of a 47-years-old female patient who carried a homozygous NDUFAF5 c.836 T > G (p.Met279Arg) mutation. This cellular model serves as a tool for investigating the underlying pathogenic mechanisms and for the development of potential treatments for Leigh syndrome.


Sujet(s)
Cellules souches pluripotentes induites , Maladie de Leigh , Maladies mitochondriales , Humains , Femelle , Adulte d'âge moyen , Maladie de Leigh/génétique , Mutation faux-sens , Cellules souches pluripotentes induites/anatomopathologie , Agranulocytes/anatomopathologie , Maladies mitochondriales/génétique , Maladies mitochondriales/anatomopathologie , Mutation , Methyltransferases/génétique , Protéines mitochondriales/génétique
12.
Front Biosci (Landmark Ed) ; 29(3): 114, 2024 Mar 20.
Article de Anglais | MEDLINE | ID: mdl-38538275

RÉSUMÉ

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.


Sujet(s)
Sclérose latérale amyotrophique , Cellules souches pluripotentes induites , Cellules souches pluripotentes , Humains , Sclérose latérale amyotrophique/génétique , Sclérose latérale amyotrophique/thérapie , Sclérose latérale amyotrophique/métabolisme , Motoneurones/métabolisme , Cellules souches pluripotentes/métabolisme , Cellules souches pluripotentes/anatomopathologie , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/anatomopathologie
13.
Int J Mol Sci ; 25(5)2024 Feb 28.
Article de Anglais | MEDLINE | ID: mdl-38474060

RÉSUMÉ

The pathophysiology of nonketotic hyperglycinemia (NKH), a rare neuro-metabolic disorder associated with severe brain malformations and life-threatening neurological manifestations, remains incompletely understood. Therefore, a valid human neural model is essential. We aimed to investigate the impact of GLDC gene variants, which cause NKH, on cellular fitness during the differentiation process of human induced pluripotent stem cells (iPSCs) into iPSC-derived astrocytes and to identify sustainable mechanisms capable of overcoming GLDC deficiency. We developed the GLDC27-FiPS4F-1 line and performed metabolomic, mRNA abundance, and protein analyses. This study showed that although GLDC27-FiPS4F-1 maintained the parental genetic profile, it underwent a metabolic switch to an altered serine-glycine-one-carbon metabolism with a coordinated cell growth and cell cycle proliferation response. We then differentiated the iPSCs into neural progenitor cells (NPCs) and astrocyte-lineage cells. Our analysis showed that GLDC-deficient NPCs had shifted towards a more heterogeneous astrocyte lineage with increased expression of the radial glial markers GFAP and GLAST and the neuronal markers MAP2 and NeuN. In addition, we detected changes in other genes related to serine and glycine metabolism and transport, all consistent with the need to maintain glycine at physiological levels. These findings improve our understanding of the pathology of nonketotic hyperglycinemia and offer new perspectives for therapeutic options.


Sujet(s)
Hyperglycinémie non cétosique , Cellules souches pluripotentes induites , Humains , Hyperglycinémie non cétosique/génétique , Hyperglycinémie non cétosique/anatomopathologie , Glycine dehydrogenase (decarboxylating)/génétique , Astrocytes/anatomopathologie , Cellules souches pluripotentes induites/anatomopathologie , Glycine , Sérine
14.
Stem Cell Reports ; 19(3): 383-398, 2024 Mar 12.
Article de Anglais | MEDLINE | ID: mdl-38366597

RÉSUMÉ

The transplantation of neural stem/progenitor cells (NS/PCs) derived from human induced pluripotent stem cells (hiPSCs) has shown promise in spinal cord injury (SCI) model animals. Establishing a functional synaptic connection between the transplanted and host neurons is crucial for motor function recovery. To boost therapeutic outcomes, we developed an ex vivo gene therapy aimed at promoting synapse formation by expressing the synthetic excitatory synapse organizer CPTX in hiPSC-NS/PCs. Using an immunocompromised transgenic rat model of SCI, we evaluated the effects of transplanting CPTX-expressing hiPSC-NS/PCs using histological and functional analyses. Our findings revealed a significant increase in excitatory synapse formation at the transplantation site. Retrograde monosynaptic tracing indicated extensive integration of transplanted neurons into the surrounding neuronal tracts facilitated by CPTX. Consequently, locomotion and spinal cord conduction significantly improved. Thus, ex vivo gene therapy targeting synapse formation holds promise for future clinical applications and offers potential benefits to individuals with SCI.


Sujet(s)
Cellules souches pluripotentes induites , Traumatismes de la moelle épinière , Humains , Rats , Animaux , Cellules souches pluripotentes induites/anatomopathologie , Différenciation cellulaire/génétique , Transplantation de cellules souches , Traumatismes de la moelle épinière/génétique , Traumatismes de la moelle épinière/thérapie , Traumatismes de la moelle épinière/anatomopathologie , Moelle spinale , Thérapie génétique , Récupération fonctionnelle/physiologie
15.
Eur J Med Res ; 29(1): 101, 2024 Feb 06.
Article de Anglais | MEDLINE | ID: mdl-38321571

RÉSUMÉ

Iron metabolism disorders are implicated in the pathogenesis of Alzheimer's disease (AD). It was previously reported that transferrin receptor (TFR1) expression was upregulated in AD mouse model. However, the precise biological functions of TFR1 in AD progression remains unclear. Herein, we observed a gradual increase in TFR1 protein expression during the differentiation of AD patient-derived induced pluripotent stem cells (AD-iPS). TFR1 knockdown inhibited the protein expression of ferritin and ferritin heavy chain 1 (FTH1), enhanced the expression of ferroportin 1 (FPN1), and decreased intracellular levels of total iron, labile iron, and reactive oxygen species (ROS). Moreover, TFR1 knockdown improved mitochondrial membrane potential (MMP), increased adenosine triphosphate (ATP) content, downregulated mitochondrial fission proteins, and upregulated mitochondrial fusion proteins. TFR1 knockdown alleviated iron overload and mitochondrial dysfunction in neural cells differentiated from AD-iPS, while TFR1 overexpression showed the opposite results. Additionally, TFR1interacted with glycogen synthase kinase 3 beta (GSK3B) and promoted GSK3B expression. GSK3B overexpression reversed the inhibitory effects of TFR1 knockdown on iron overload and mitochondrial dysfunction in AD-iPS differentiated neural cells. In conclusion, TFR1 knockdown alleviated iron overload and mitochondrial dysfunction in neural cells differentiated from AD-iPS by promoting GSK3B expression. Our findings provide a potential therapeutic target for the treatment of AD.


Sujet(s)
Maladie d'Alzheimer , Cellules souches pluripotentes induites , Surcharge en fer , Maladies mitochondriales , Humains , Souris , Animaux , Maladie d'Alzheimer/anatomopathologie , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/anatomopathologie , Glycogen synthase kinase 3 beta/métabolisme , Fer/métabolisme , Récepteurs à la transferrine/métabolisme , Surcharge en fer/métabolisme
16.
Dis Model Mech ; 17(2)2024 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-38353122

RÉSUMÉ

Nervous system tumors, particularly brain tumors, represent the most common tumors in children and one of the most lethal tumors in adults. Despite decades of research, there are few effective therapies for these cancers. Although human nervous system tumor cells and genetically engineered mouse models have served as excellent platforms for drug discovery and preclinical testing, they have limitations with respect to accurately recapitulating important aspects of the pathobiology of spontaneously arising human tumors. For this reason, attention has turned to the deployment of human stem cell engineering involving human embryonic or induced pluripotent stem cells, in which genetic alterations associated with nervous system cancers can be introduced. These stem cells can be used to create self-assembling three-dimensional cerebral organoids that preserve key features of the developing human brain. Moreover, stem cell-engineered lines are amenable to xenotransplantation into mice as a platform to investigate the tumor cell of origin, discover cancer evolutionary trajectories and identify therapeutic vulnerabilities. In this article, we review the current state of human stem cell models of nervous system tumors, discuss their advantages and disadvantages, and provide consensus recommendations for future research.


Sujet(s)
Tumeurs du cerveau , Cellules souches pluripotentes induites , Enfant , Humains , Animaux , Souris , Différenciation cellulaire , Cellules souches pluripotentes induites/anatomopathologie , Tumeurs du cerveau/anatomopathologie , Encéphale/anatomopathologie , Mutation
17.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(1): 38-41, 2024 Jan 10.
Article de Chinois | MEDLINE | ID: mdl-38171557

RÉSUMÉ

OBJECTIVE: To obtain skin-derived induced pluripotent stem cells (iPSCs) from an Osteogenesis imperfecta (OI) patient carrying WNT1c.677C>T mutation in order to provide a new cell model for investigating the underlying molecular mechanism and stem cell therapy for OI. METHODS: The pathogenic variant of the patient was identified by Sanger sequencing. With informed consent from the patient, skin tissue was biopsied, and primary skin fibroblasts were cultured. Skin fibroblasts were induced into iPSCs using Sendai virus-mediated non-genomic integration reprogramming method. The iPSC cell lines were characterized for pluripotency, differentiation capacity, and karyotyping assay. RESULTS: The patient was found to carry homozygous missense c.677C>T (p.Ser226Leu) mutation of the WNT1 gene. The established iPSC lines possessed self-renewal and capacity for in vitro differentiation. It also has a diploid karyotype (46,XX). CONCLUSION: A patient-specific WNT1 gene mutation (WNT1c.677C>T) iPSC line was established, which can provide a cell model for the study of OI caused by the mutation.


Sujet(s)
Cellules souches pluripotentes induites , Ostéogenèse imparfaite , Humains , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/anatomopathologie , Ostéogenèse imparfaite/génétique , Mutation , Différenciation cellulaire/génétique , Lignée cellulaire
18.
Biochem Soc Trans ; 52(1): 163-176, 2024 Feb 28.
Article de Anglais | MEDLINE | ID: mdl-38288874

RÉSUMÉ

The investigation of neurodegenerative diseases advanced significantly with the advent of cell-reprogramming technology, leading to the creation of new models of human illness. These models, derived from induced pluripotent stem cells (iPSCs), facilitate the study of sporadic as well as hereditary diseases and provide a comprehensive understanding of the molecular mechanisms involved with neurodegeneration. Through proteomics, a quantitative tool capable of identifying thousands of proteins from small sample volumes, researchers have attempted to identify disease mechanisms by detecting differentially expressed proteins and proteoforms in disease models, biofluids, and postmortem brain tissue. The integration of these two technologies allows for the identification of novel pathological targets within the realm of neurodegenerative diseases. Here, we highlight studies from the past 5 years on the contributions of iPSCs within neuroproteomic investigations, which uncover the molecular mechanisms behind these illnesses.


Sujet(s)
Cellules souches pluripotentes induites , Maladies neurodégénératives , Humains , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/anatomopathologie , Reprogrammation cellulaire , Maladies neurodégénératives/métabolisme
19.
Free Radic Biol Med ; 213: 164-173, 2024 03.
Article de Anglais | MEDLINE | ID: mdl-38246514

RÉSUMÉ

Peripheral autonomic nervous system (P-ANS) dysfunction is a critical non-motor phenotype of Parkinson's disease (PD). The majority of PD cases are sporadic and lack identified PD-associated genes involved. Epidemiological and animal model studies suggest an association with pesticides and other environmental toxins. However, the cellular mechanisms underlying toxin induced P-ANS dysfunctions remain unclear. Here, we mapped the global transcriptome changes in human induced pluripotent stem cell (iPSC) derived P-ANS sympathetic neurons during inhibition of the mitochondrial respiratory chain by the PD-related pesticide, rotenone. We revealed distinct transcriptome profiles between acute and chronic exposure to rotenone. In the acute stage, there was a down regulation of specific cation channel genes, known to mediate electrophysiological activity, while in the chronic stage, the human P-ANS neurons exhibited dysregulation of anti-apoptotic and Golgi apparatus-related pathways. Moreover, we identified the sodium voltage-gated channel subunit SCN3A/Nav1.3 as a potential biomarker in human P-ANS neurons associated with PD. Our analysis of the rotenone-altered coding and non-coding transcriptome of human P-ANS neurons may thus provide insight into the pathological signaling events in the sympathetic neurons during PD progression.


Sujet(s)
Cellules souches pluripotentes induites , Maladie de Parkinson , Animaux , Humains , Maladie de Parkinson/métabolisme , Roténone/toxicité , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/anatomopathologie , Neurones/métabolisme , Phénotype
20.
Dokl Biol Sci ; 515(1): 15-19, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38190040

RÉSUMÉ

A new in vitro model of Huntington's disease (HD) was developed via a direct reprogramming of dermal fibroblasts from HD patients into striatal neurons. A reprogramming into induced pluripotent stem (iPS) cells is obviated in the case of direct reprogramming, which thus yields neurons that preserve the epigenetic information inherent in cells of a particular donor and, consequently, the age-associated disease phenotype. A main histopathological feature of HD was reproduced in the new model; i.e., aggregates of mutant huntingtin accumulated in striatal neurons derived from a patient's fibroblasts. Experiments with cultured neurons obtained via direct reprogramming make it possible to individually assess the progression of neuropathology and to implement a personalized approach to choosing the treatment strategy and drugs for therapy. The in vitro model of HD can be used in preclinical drug studies.


Sujet(s)
Maladie de Huntington , Cellules souches pluripotentes induites , Humains , Animaux , Maladie de Huntington/génétique , Maladie de Huntington/anatomopathologie , Neurones , Corps strié/anatomopathologie , Fibroblastes , Cellules souches pluripotentes induites/anatomopathologie , Modèles animaux de maladie humaine
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...