Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 89
Filtrer
1.
Sci Rep ; 14(1): 18002, 2024 08 03.
Article de Anglais | MEDLINE | ID: mdl-39097642

RÉSUMÉ

Zika virus (ZIKV) infection was first reported in 2015 in Brazil as causing microcephaly and other developmental abnormalities in newborns, leading to the identification of Congenital Zika Syndrome (CZS). Viral infections have been considered an environmental risk factor for neurodevelopmental disorders outcome, such as Autism Spectrum Disorder (ASD). Moreover, not only the infection per se, but maternal immune system activation during pregnancy, has been linked to fetal neurodevelopmental disorders. To understand the impact of ZIKV vertical infection on brain development, we derived induced pluripotent stem cells (iPSC) from Brazilian children born with CZS, some of the patients also being diagnosed with ASD. Comparing iPSC-derived neurons from CZS with a control group, we found lower levels of pre- and postsynaptic proteins and reduced functional synapses by puncta co-localization. Furthermore, neurons and astrocytes derived from the CZS group showed decreased glutamate levels. Additionally, the CZS group exhibited elevated levels of cytokine production, one of which being IL-6, already associated with the ASD phenotype. These preliminary findings suggest that ZIKV vertical infection may cause long-lasting disruptions in brain development during fetal stages, even in the absence of the virus after birth. These disruptions could contribute to neurodevelopmental disorders manifestations such as ASD. Our study contributes with novel knowledge of the CZS outcomes and paves the way for clinical validation and the development of potential interventions to mitigate the impact of ZIKV vertical infection on neurodevelopment.


Sujet(s)
Encéphale , Cellules souches pluripotentes induites , Transmission verticale de maladie infectieuse , Synapses , Infection par le virus Zika , Virus Zika , Humains , Infection par le virus Zika/virologie , Infection par le virus Zika/anatomopathologie , Femelle , Virus Zika/pathogénicité , Synapses/anatomopathologie , Synapses/métabolisme , Encéphale/virologie , Encéphale/anatomopathologie , Grossesse , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/virologie , Neurones/virologie , Neurones/métabolisme , Neurones/anatomopathologie , Mâle , Astrocytes/virologie , Astrocytes/métabolisme , Maladies neuro-inflammatoires/virologie , Maladies neuro-inflammatoires/anatomopathologie , Maladies neuro-inflammatoires/métabolisme , Complications infectieuses de la grossesse/virologie , Complications infectieuses de la grossesse/anatomopathologie , Brésil , Nouveau-né , Trouble du spectre autistique/virologie , Enfant
2.
Cell Reprogram ; 26(3): 107-115, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38917437

RÉSUMÉ

Our group generated two induced pluripotent stem cell (iPSC) lines for in vitro red blood cell (RBC) production from blood donors with extensively known erythrocyte antigen profiles. One line was intended to give rise to RBCs for transfusions in patients with sickle cell disease (SCD), while the other was developed to create RBC panel reagents. Two blood donors were selected based on their RBC phenotypes, further complemented by high-throughput DNA array analysis to obtain a more comprehensive erythrocyte antigen profile. Enriched erythroblast populations from the donors' peripheral blood mononuclear cells were reprogrammed into iPSCs using nonintegrative plasmid vectors. The iPSC lines were characterized and subsequently subjected to hematopoietic differentiation. iPSC PB02 and iPSC PB12 demonstrated in vitro and in vivo iPSC features and retained the genotype of each blood donor's RBC antigen profile. Colony-forming cell assays confirmed that iPSC PB02 and iPSC PB12 generated hematopoietic progenitors. These two iPSC lines were generated with defined erythrocyte antigen profiles, self-renewal capacity, and hematopoietic differentiation potential. With improvements in hematopoietic differentiation, these cells could potentially be more efficiently differentiated into RBCs in the future. They could serve as a complementary approach for obtaining donor-independent RBCs and addressing specific demands for blood transfusions.


Sujet(s)
Donneurs de sang , Différenciation cellulaire , Érythrocytes , Cellules souches pluripotentes induites , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Humains , Érythrocytes/métabolisme , Érythrocytes/cytologie , Lignée cellulaire , Animaux , Antigènes de groupe sanguin , Souris , Drépanocytose/thérapie , Drépanocytose/sang
3.
J Mater Chem B ; 12(21): 5085-5097, 2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38713059

RÉSUMÉ

Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by cognitive impairment associated with the accumulation of beta-amyloid protein (Aß). Aß activates glial cells in the brain, increasing the secretion of proinflammatory cytokines, which leads to neuroinflammation and neuronal death. Currently, there are no effective treatments that cure or stop its progression; therefore, AD is considered a global health priority. The main limitations are the low drug bioavailability and impermeability of the blood-brain barrier (BBB). Fortunately, nanomedicine has emerged as a promising field for the development of new nanosystems for the controlled and targeted delivery of drugs to the brain. Therefore, in this work, lipid-polymer hybrid nanoparticles (LPHNPs) conjugated with transferrin (Tf) to facilitate crossing the BBB and loaded with N-acetylcysteine (NAC) for its anti-inflammatory effect were synthesized, and their physicochemical characterization was carried out. Subsequently, an in vitro model involving human astrocytes derived from induced pluripotent stem cells (iPSC) from an AD-diagnosed patient was developed, which was brought to a reactive state by stimulation with lipopolysaccharides (LPSs). The cell culture was treated with either Tf-conjugated LPHNPs loaded with NAC (NAC-Tf-LPHNPs) at 0.25 mg mL-1, or free NAC at 5 mM. The results showed that NAC-Tf-LPHNPs favorably modulated the expression of proinflammatory genes such as interleukin-1ß (IL-1ß), amyloid precursor protein (APP) and glial fibrillary acidic protein (GFAP). In addition, they reduced the secretion of the proinflammatory cytokines interleukin 6 (IL-6), IL-1ß and interferon-gamma (INF-γ). Results for both cases were compared to the group of cells that did not receive any treatment. In contrast, free NAC only had this effect on the expression of IL-1ß and the secretion of the cytokines IL-6 and INF-γ. These results indicate the potential of NAC-Tf-LPHNPs for AD treatment.


Sujet(s)
Acétylcystéine , Maladie d'Alzheimer , Astrocytes , Cellules souches pluripotentes induites , Nanoparticules , Humains , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Nanoparticules/composition chimique , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/effets des médicaments et des substances chimiques , Acétylcystéine/composition chimique , Acétylcystéine/pharmacologie , Astrocytes/effets des médicaments et des substances chimiques , Astrocytes/métabolisme , Polymères/composition chimique , Polymères/pharmacologie , Lipides/composition chimique , Marqueurs biologiques/métabolisme , Taille de particule , Maladies neuro-inflammatoires/traitement médicamenteux
4.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167097, 2024 06.
Article de Anglais | MEDLINE | ID: mdl-38408544

RÉSUMÉ

Zika virus (ZIKV) infection was first associated with Central Nervous System (CNS) infections in Brazil in 2015, correlated with an increased number of newborns with microcephaly, which ended up characterizing the Congenital Zika Syndrome (CZS). Here, we investigated the impact of ZIKV infection on the functionality of iPSC-derived astrocytes. Besides, we extrapolated our findings to a Brazilian cohort of 136 CZS children and validated our results using a mouse model. Interestingly, ZIKV infection in neuroprogenitor cells compromises cell migration and causes apoptosis but does not interfere in astrocyte generation. Moreover, infected astrocytes lost their ability to uptake glutamate while expressing more glutamate transporters and secreted higher levels of IL-6. Besides, infected astrocytes secreted factors that impaired neuronal synaptogenesis. Since these biological endophenotypes were already related to Autism Spectrum Disorder (ASD), we extrapolated these results to a cohort of children, now 6-7 years old, and found seven children with ASD diagnosis (5.14 %). Additionally, mice infected by ZIKV revealed autistic-like behaviors, with a significant increase of IL-6 mRNA levels in the brain. Considering these evidence, we inferred that ZIKV infection during pregnancy might lead to synaptogenesis impairment and neuroinflammation, which could increase the risk for ASD.


Sujet(s)
Astrocytes , Trouble du spectre autistique , Maladies neuro-inflammatoires , Synapses , Infection par le virus Zika , Virus Zika , Infection par le virus Zika/anatomopathologie , Infection par le virus Zika/métabolisme , Infection par le virus Zika/virologie , Infection par le virus Zika/complications , Trouble du spectre autistique/virologie , Trouble du spectre autistique/métabolisme , Trouble du spectre autistique/étiologie , Trouble du spectre autistique/anatomopathologie , Humains , Animaux , Souris , Virus Zika/physiologie , Femelle , Enfant , Synapses/métabolisme , Synapses/anatomopathologie , Maladies neuro-inflammatoires/virologie , Maladies neuro-inflammatoires/métabolisme , Maladies neuro-inflammatoires/anatomopathologie , Maladies neuro-inflammatoires/étiologie , Astrocytes/virologie , Astrocytes/métabolisme , Astrocytes/anatomopathologie , Mâle , Interleukine-6/métabolisme , Interleukine-6/génétique , Grossesse , Facteurs de risque , Cellules souches pluripotentes induites/virologie , Cellules souches pluripotentes induites/métabolisme , Brésil/épidémiologie , Modèles animaux de maladie humaine , Neurogenèse
5.
Stem Cell Res ; 76: 103337, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38359473

RÉSUMÉ

Parkinson's disease (PD) is characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta, which results in a prominent reduction of striatal dopamine levels leading to motor alterations. The mechanisms underlying neurodegeneration in PD remain unknown. Here, we generated an induced pluripotent stem cell line from dermal fibroblasts of a Mexican patient diagnosed with sporadic PD (UNAMi002-A) and another cell line from dermal fibroblasts of a patient carrying the point mutation c.1423delC in PINK1 (UNAMi003-A). These patient-derived iPS cell lines offer the possibility of modeling PD and understanding the mechanisms that contribute to dopamine neuron loss.


Sujet(s)
Cellules souches pluripotentes induites , Maladie de Parkinson , Humains , Maladie de Parkinson/génétique , Maladie de Parkinson/métabolisme , Cellules souches pluripotentes induites/métabolisme , Neurones dopaminergiques/métabolisme , Dopamine/métabolisme , Protein kinases/génétique , Mutation/génétique
6.
Biochem Soc Trans ; 52(1): 163-176, 2024 02 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
7.
Stem Cell Res ; 75: 103307, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38244535

RÉSUMÉ

FHL1 gene locates in the Xq26 region and encodes for four and half LIM domain protein 1. It plays a crucial role in muscle cells and mutations in FHL1 are related to muscular dystrophy (MD). Peripheral blood mononuclear cells (PBMCs) were obtained from 2 family patients with MD that carry a pathogenic missense mutation in FHL1 (c.377G > A, p.C126Y). Induced pluripotent stem cells (iPSCs) were generated by PBMCs reprogramming using the lentiviral-hSTEMCCA-loxP vector, obtaining FHL1-T and FHL1-V iPSCs lines from patients. FHL1 genotype was maintained, and stemness and pluripotency were confirmed in both iPSCs lines.


Sujet(s)
Cellules souches pluripotentes induites , Dystrophies musculaires , Humains , Mutation faux-sens , Cellules souches pluripotentes induites/métabolisme , Agranulocytes/métabolisme , Protéines du muscle/génétique , Mutation , Protéines et peptides de signalisation intracellulaire/génétique , Protéines à domaine LIM/génétique
8.
Stem Cell Res ; 75: 103309, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38217995

RÉSUMÉ

Retinitis pigmentosa (RP) is the most common retinal degeneration in humans and is characterized by the progressive degeneration of rods and cones and retinal pigment epithelium. We generated the IOCVi001-A induced pluripotent stem cell (iPSC) line from dermal fibroblast of a patient with a homozygous c.498_499insC (p.(Asn167Glnfs⁎34) variant in the Membrane-type frizzled related protein (MFRP) gene, a genetic defect causing a syndrome characterized by RP and small eye size (nanophthalmos). IOCVi001-A displayed normal stemness, expressed pluripotent stem cell markers and displayed a normal karyotype. This iPSC line can be used for in vitro disease modeling for complex forms of RP.


Sujet(s)
Hypopituitarisme , Cellules souches pluripotentes induites , Microphtalmie , Rétinite pigmentaire , Humains , Microphtalmie/génétique , Microphtalmie/métabolisme , Cellules souches pluripotentes induites/métabolisme , Protéines membranaires/génétique , Rétinite pigmentaire/génétique , Rétinite pigmentaire/métabolisme , Mutation
9.
Blood ; 143(6): 548-560, 2024 Feb 08.
Article de Anglais | MEDLINE | ID: mdl-37944157

RÉSUMÉ

ABSTRACT: Nonmuscle cell contractility is an essential feature underlying diverse cellular processes such as motility, morphogenesis, division and genome replication, intracellular transport, and secretion. Blood clot contraction is a well-studied process driven by contracting platelets. Megakaryocytes (MKs), which are the precursors to platelets, can be found in bone marrow and lungs. Although they express many of the same proteins and structures found in platelets, little is known about their ability to engage with extracellular proteins such as fibrin and contract. Here, we have measured the ability of MKs to compress plasma clots. Megakaryocytes derived from human induced pluripotent stem cells (iPSCs) were suspended in human platelet-free blood plasma and stimulated with thrombin. Using real-time macroscale optical tracking, confocal microscopy, and biomechanical measurements, we found that activated iPSC-derived MKs (iMKs) caused macroscopic volumetric clot shrinkage, as well as densification and stiffening of the fibrin network via fibrin-attached plasma membrane protrusions undergoing extension-retraction cycles that cause shortening and bending of fibrin fibers. Contraction induced by iMKs involved 2 kinetic phases with distinct rates and durations. It was suppressed by inhibitors of nonmuscle myosin IIA, actin polymerization, and integrin αIIbß3-fibrin interactions, indicating that the molecular mechanisms of iMK contractility were similar or identical to those in activated platelets. Our findings provide new insights into MK biomechanics and suggest that iMKs can be used as a model system to study platelet contractility. Physiologically, the ability of MKs to contract plasma clots may play a role in the mechanical remodeling of intravascular blood clots and thrombi.


Sujet(s)
Cellules souches pluripotentes induites , Thrombose , Humains , Mégacaryocytes/métabolisme , Cellules souches pluripotentes induites/métabolisme , Plaquettes/métabolisme , Thrombose/métabolisme , Fibrine/métabolisme , Plasma sanguin
10.
Nat Rev Endocrinol ; 20(2): 77-92, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38102391

RÉSUMÉ

Pituitary cells that express the transcription factor SOX2 are stem cells because they can self-renew and differentiate into multiple pituitary hormone-producing cell types as organoids. Wounding and physiological challenges can activate pituitary stem cells, but cell numbers are not fully restored, and the ability to mobilize stem cells decreases with increasing age. The basis of these limitations is still unknown. The regulation of stem cell quiescence and activation involves many different signalling pathways, including those mediated by WNT, Hippo and several cytokines; more research is needed to understand the interactions between these pathways. Pituitary organoids can be formed from human or mouse embryonic stem cells, or from human induced pluripotent stem cells. Human pituitary organoid transplantation is sufficient to induce corticosterone release in hypophysectomized mice, raising the possibility of therapeutic applications. Today, pituitary organoids have the potential to assess the role of individual genes and genetic variants on hormone production ex vivo, providing an important tool for the advancement of exciting frontiers in pituitary stem cell biology and pituitary organogenesis. In this article, we provide an overview of notable discoveries in pituitary stem cell function and highlight important areas for future research.


Sujet(s)
Cellules souches pluripotentes induites , Humains , Animaux , Souris , Cellules souches pluripotentes induites/métabolisme , Hypophyse/métabolisme , Facteurs de transcription/métabolisme , Transduction du signal , Différenciation cellulaire
11.
Front Immunol ; 14: 1283331, 2023.
Article de Anglais | MEDLINE | ID: mdl-38146365

RÉSUMÉ

TNF-α is essential for induction and maintenance of inflammatory responses and its dysregulation is associated with susceptibility to various pathogens that infect the central nervous system. Activation of both microglia and astrocytes leads to TNF-α production, which in turn triggers further activation of these cells. Astrocytes have been implicated in the pathophysiology of a wide range of neurodegenerative diseases with either harmful or protective roles, as these cells are capable of secreting several inflammatory factors and also promote synapse elimination and remodeling. These responses are possible because they sense their surroundings via several receptors, including the metabotropic glutamate receptor 5 (mGluR5). Under neuroinflammatory conditions, mGluR5 activation in astrocytes can be neuroprotective or have the opposite effect. In the current study, we investigated the role of mGluR5 in hiPSC-derived astrocytes subjected to pro-inflammatory stimulation by recombinant TNF-α (rTNF-α). Our results show that mGluR5 blockade by CTEP decreases the secreted levels of pro-inflammatory cytokines (IL-6 and IL-8) following short rTNF-α stimulation, although this effect subsides with time. Additionally, CTEP enhances synaptoneurosome phagocytosis by astrocytes in both non-stimulated and rTNF-α-stimulated conditions, indicating that mGluR5 blockade alone is enough to drive synaptic material engulfment. Finally, mGluR5 antagonism as well as rTNF-α stimulation augment the expression of the reactivity marker SERPINA3 and reduces the expression of synaptogenic molecules. Altogether, these data suggest a complex role for mGluR5 in human astrocytes, since its blockade may have beneficial and detrimental effects under inflammatory conditions.


Sujet(s)
Astrocytes , Cellules souches pluripotentes induites , Phagocytose , Récepteur-5 métabotropique du glutamate , Humains , Astrocytes/métabolisme , Cellules souches pluripotentes induites/métabolisme , Facteur de nécrose tumorale alpha/métabolisme
12.
Stem Cell Reports ; 18(10): 1905-1912, 2023 10 10.
Article de Anglais | MEDLINE | ID: mdl-37774702

RÉSUMÉ

Identifying human leukocyte antigen (HLA) haplotype-homozygous donors for the generation of induced pluripotent stem (iPS) cell lines permits the construction of biobanks immunologically compatible with significant numbers of individuals for use in therapy. However, two questions must be addressed to create such a bank: how many cell lines are necessary to match most of the recipient population and how many people should be tested to find these donors? In Japan and the UK, 50 and 100 distinct HLA-A, -B, and -DRB1 triple-homozygous haplotypes would cover 90% of those populations, respectively. Using data from the Brazilian National Registry of Bone Marrow Donors (REDOME), encompassing 4,017,239 individuals, we identified 1,906 distinct triple-homozygous HLA haplotypes. In Brazil, 559 triple-homozygous cell lines cover 95% of the population, and 3.8 million people would have to be screened. Finally, we show the contribution of the 30 most frequent triple-homozygous HLA haplotypes in Brazil to populations of different countries.


Sujet(s)
Cellules souches pluripotentes induites , Humains , Brésil , Cellules souches pluripotentes induites/métabolisme , Antigènes HLA/métabolisme , Antigènes HLA-A/génétique , Antigènes HLA-A/métabolisme , Donneurs de tissus , Antigènes d'histocompatibilité de classe I/métabolisme , Haplotypes/génétique , Antigènes d'histocompatibilité de classe II/métabolisme , Allèles , Fréquence d'allèle
13.
Stem Cell Res ; 71: 103157, 2023 09.
Article de Anglais | MEDLINE | ID: mdl-37393721

RÉSUMÉ

The arrhythmogenic cardiomyopathy (ACM) is an inherited heart muscle disease characterized by the progressive replacement of contractile myocardium by fibro-fatty adipose tissue, that generates ventricular arrhythmias and sudden death in patients. The ACM has a genetic origin with alterations in desmosomal genes with the most commonly mutated being the PKP2 gene. We generated two CRISPR/Cas9 edited iPSCs lines, one iPSC line with a point mutation in PKP2 reported in patients with ACM and another iPSC line with a premature stop codon to knock-out the same gene.


Sujet(s)
Dysplasie ventriculaire droite arythmogène , Cardiomyopathies , Cellules souches pluripotentes induites , Humains , Mutation ponctuelle , Cellules souches pluripotentes induites/métabolisme , Dysplasie ventriculaire droite arythmogène/génétique , Systèmes CRISPR-Cas/génétique , Cardiomyopathies/génétique , Mutation/génétique , Plakophilines/génétique , Plakophilines/métabolisme
14.
Int J Mol Sci ; 24(10)2023 May 18.
Article de Anglais | MEDLINE | ID: mdl-37240306

RÉSUMÉ

Alzheimer's disease (AD) is a chronic neurological condition characterized by the severe loss of cholinergic neurons. Currently, the incomplete understanding of the loss of neurons has prevented curative treatments for familial AD (FAD). Therefore, modeling FAD in vitro is essential for studying cholinergic vulnerability. Moreover, to expedite the discovery of disease-modifying therapies that delay the onset and slow the progression of AD, we depend on trustworthy disease models. Although highly informative, induced pluripotent stem cell (iPSCs)-derived cholinergic neurons (ChNs) are time-consuming, not cost-effective, and labor-intensive. Other sources for AD modeling are urgently needed. Wild-type and presenilin (PSEN)1 p.E280A fibroblast-derived iPSCs, menstrual blood-derived menstrual stromal cells (MenSCs), and umbilical cord-derived Wharton Jelly's mesenchymal stromal cells (WJ-MSCs) were cultured in Cholinergic-N-Run and Fast-N-Spheres V2 medium to obtain WT and PSEN 1 E280A cholinergic-like neurons (ChLNs, 2D) and cerebroid spheroids (CSs, 3D), respectively, and to evaluate whether ChLNs/CSs can reproduce FAD pathology. We found that irrespective of tissue source, ChLNs/CSs successfully recapitulated the AD phenotype. PSEN 1 E280A ChLNs/CSs show accumulation of iAPPß fragments, produce eAß42, present TAU phosphorylation, display OS markers (e.g., oxDJ-1, p-JUN), show loss of ΔΨm, exhibit cell death markers (e.g., TP53, PUMA, CASP3), and demonstrate dysfunctional Ca2+ influx response to ACh stimuli. However, PSEN 1 E280A 2D and 3D cells derived from MenSCs and WJ-MSCs can reproduce FAD neuropathology more efficiently and faster (11 days) than ChLNs derived from mutant iPSCs (35 days). Mechanistically, MenSCs and WJ-MSCs are equivalent cell types to iPSCs for reproducing FAD in vitro.


Sujet(s)
Maladie d'Alzheimer , Cellules souches pluripotentes induites , Cellules souches mésenchymateuses , Humains , Cellules souches pluripotentes induites/métabolisme , Maladie d'Alzheimer/métabolisme , Neurones cholinergiques/métabolisme , Cellules souches mésenchymateuses/métabolisme , Agents cholinergiques/métabolisme , Préséniline-1/génétique , Préséniline-1/métabolisme
15.
Pigment Cell Melanoma Res ; 36(5): 330-347, 2023 09.
Article de Anglais | MEDLINE | ID: mdl-37132530

RÉSUMÉ

Melanoma, a lethal malignancy that arises from melanocytes, exhibits a multiplicity of clinico-pathologically distinct subtypes in sun-exposed and non-sun-exposed areas. Melanocytes are derived from multipotent neural crest cells and are present in diverse anatomical locations, including skin, eyes, and various mucosal membranes. Tissue-resident melanocyte stem cells and melanocyte precursors contribute to melanocyte renewal. Elegant studies using mouse genetic models have shown that melanoma can arise from either melanocyte stem cells or differentiated pigment-producing melanocytes depending on a combination of tissue and anatomical site of origin and activation of oncogenic mutations (or overexpression) and/or the repression in expression or inactivating mutations in tumor suppressors. This variation raises the possibility that different subtypes of human melanomas (even subsets within each subtype) may also be a manifestation of malignancies of distinct cells of origin. Melanoma is known to exhibit phenotypic plasticity and trans-differentiation (defined as a tendency to differentiate into cell lineages other than the original lineage from which the tumor arose) along vascular and neural lineages. Additionally, stem cell-like properties such as pseudo-epithelial-to-mesenchymal (EMT-like) transition and expression of stem cell-related genes have also been associated with the development of melanoma drug resistance. Recent studies that employed reprogramming melanoma cells to induced pluripotent stem cells have uncovered potential relationships between melanoma plasticity, trans-differentiation, and drug resistance and implications for cell or origin of human cutaneous melanoma. This review provides a comprehensive summary of the current state of knowledge on melanoma cell of origin and the relationship between tumor cell plasticity and drug resistance.


Sujet(s)
Cellules souches pluripotentes induites , Mélanome , Tumeurs cutanées , Animaux , Souris , Humains , Mélanome/anatomopathologie , Tumeurs cutanées/anatomopathologie , Plasticité cellulaire , Mélanocytes/métabolisme , Différenciation cellulaire , Résistance aux substances , Cellules souches pluripotentes induites/métabolisme , Crête neurale/métabolisme
16.
Eur Arch Psychiatry Clin Neurosci ; 273(8): 1649-1664, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37039888

RÉSUMÉ

Schizophrenia is a severe psychiatric disorder of neurodevelopmental origin that affects around 1% of the world's population. Proteomic studies and other approaches have provided evidence of compromised cellular processes in the disorder, including mitochondrial function. Most of the studies so far have been conducted on postmortem brain tissue from patients, and therefore, do not allow the evaluation of the neurodevelopmental aspect of the disorder. To circumvent that, we studied the mitochondrial and nuclear proteomes of neural stem cells (NSCs) and neurons derived from induced pluripotent stem cells (iPSCs) from schizophrenia patients versus healthy controls to assess possible alterations related to energy metabolism and mitochondrial function during neurodevelopment in the disorder. Our results revealed differentially expressed proteins in pathways related to mitochondrial function, cell cycle control, DNA repair and neuritogenesis and their possible implication in key process of neurodevelopment, such as neuronal differentiation and axonal guidance signaling. Moreover, functional analysis of NSCs revealed alterations in mitochondrial oxygen consumption in schizophrenia-derived cells and a tendency of higher levels of intracellular reactive oxygen species (ROS). Hence, this study shows evidence that alterations in important cellular processes are present during neurodevelopment and could be involved with the establishment of schizophrenia, as well as the phenotypic traits observed in adult patients. Neural stem cells (NSCs) and neurons were derived from induced pluripotent stem cells (iPSCs) from schizophrenia patients and controls. Proteomic analyses were performed on the enriched mitochondrial and nuclear fractions of NSCs and neurons. Whole-cell proteomic analysis was also performed in neurons. Our results revealed alteration in proteins related to mitochondrial function, cell cycle control, among others. We also performed energy pathway analysis and reactive oxygen species (ROS) analysis of NSCs, which revealed alterations in mitochondrial oxygen consumption and a tendency of higher levels of intracellular ROS in schizophrenia-derived cells.


Sujet(s)
Cellules souches pluripotentes induites , Schizophrénie , Adulte , Humains , Schizophrénie/métabolisme , Cellules souches pluripotentes induites/métabolisme , Différenciation cellulaire/génétique , Espèces réactives de l'oxygène/métabolisme , Protéomique , Points de contrôle du cycle cellulaire , Mitochondries/métabolisme
17.
Gene ; 871: 147424, 2023 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-37054903

RÉSUMÉ

Xia-Gibbs syndrome (XGS) is a syndromic form of intellectual disability caused by heterozygous AHDC1 variants, but the pathophysiological mechanisms underlying this syndrome are still unclear. In this manuscript, we describe the development of two different functional models: three induced pluripotent stem cell (iPSC) lines with different loss-of-function (LoF) AHDC1 variants, derived by reprogramming peripheral blood mononuclear cells from XGS patients, and a zebrafish strain with a LoF variant in the ortholog gene (ahdc1) obtained through CRISPR/Cas9-mediated editing. The three iPSC lines showed expression of pluripotency factors (SOX2, SSEA-4, OCT3/4, and NANOG). To verify the capacity of iPSC to differentiate into the three germ layers, we obtained embryoid bodies (EBs), induced their differentiation, and confirmed the mRNA expression of ectodermal, mesodermal, and endodermal markers using the TaqMan hPSC Scorecard. The iPSC lines were also approved for the following quality tests: chromosomal microarray analysis (CMA), mycoplasma testing, and short tandem repeat (STR) DNA profiling. The zebrafish model has an insertion of four base pairs in the ahdc1 gene, is fertile, and breeding between heterozygous and wild-type (WT) animals generated offspring in a genotypic proportion in agreement with Mendelian law. The established iPSC and zebrafish lines were deposited on the hpscreg.eu and zfin.org platforms, respectively. These biological models are the first for XGS and will be used in future studies that investigate the pathophysiology of this syndrome, unraveling its underlying molecular mechanisms.


Sujet(s)
Malformations multiples , Cellules souches pluripotentes induites , Déficience intellectuelle , Animaux , Déficience intellectuelle/génétique , Cellules souches pluripotentes induites/métabolisme , Danio zébré/génétique , Agranulocytes , Malformations multiples/génétique , Différenciation cellulaire/génétique , Syndrome
18.
Stem Cell Res ; 69: 103076, 2023 06.
Article de Anglais | MEDLINE | ID: mdl-36963214

RÉSUMÉ

Human induced pluripotent stem cell (hiPSC) line INEUi001-A was reprogrammed from peripheral blood mononuclear cells (PBMC) using the lentiviral-hSTEMCCA-loxP vector. PBMCs were obtained from a 75- year-old female ALS/FTD disease patient carrying a heterozygous deletion within the C9ORF72 hexanucleotide repeat region resulting in a GGGGCCG sequence (∼1.16 repeats). C9ORF72 genotype was maintained and stemness and pluripotency confirmed in INEUi001-A hiPSC line.


Sujet(s)
Sclérose latérale amyotrophique , Démence frontotemporale , Cellules souches pluripotentes induites , Femelle , Humains , Sujet âgé , Sclérose latérale amyotrophique/génétique , Sclérose latérale amyotrophique/métabolisme , Démence frontotemporale/génétique , Cellules souches pluripotentes induites/métabolisme , Protéine C9orf72/génétique , Agranulocytes/métabolisme , Génotype
19.
Stem Cell Res Ther ; 14(1): 42, 2023 03 16.
Article de Anglais | MEDLINE | ID: mdl-36927767

RÉSUMÉ

BACKGROUND: The generation of induced pluripotent stem cells has opened the field of study for stem cell research, disease modeling and drug development. However, the epigenetic signatures present in somatic cells make cell reprogramming still an inefficient process. This epigenetic memory constitutes an obstacle in cellular reprogramming. Here, we report the effect of hydralazine (HYD) and valproic acid (VPA), two small molecules with proven epigenetic activity, on the expression of pluripotency genes in adult (aHF) and neonatal (nbHF) human fibroblasts. METHODS: aHF and nbHF were treated with HYD and/or VPA, and viability and gene expression assays for OCT4, NANOG, c-MYC, KLF4, DNMT1, TET3, ARID1A and ARID2 by quantitative PCR were performed. aHF and nbHF were transfected with episomal plasmid bearing Yamanaka factors (OCT4, SOX2, KLF4 and c-MYC) and exposed to HYD and VPA to determine the reprogramming efficiency. Methylation sensitive restriction enzyme (MSRE) qPCR assays were performed on OCT4 and NANOG promoter regions. Immunofluorescence assays were carried out for pluripotency genes on iPSC derived from aHF and nbHF. RESULTS: HYD upregulated the expression of OCT4 (2.5-fold) and NANOG (fourfold) genes but not c-Myc or KLF4 in aHF and had no significant effect on the expression of all these genes in nbHF. VPA upregulated the expression of NANOG (twofold) in aHF and c-MYC in nbHF, while it downregulated the expression of NANOG in nbHF. The combination of HYD and VPA canceled the OCT4 and NANOG overexpression induced by HYD in aHF, while it reinforced the effects of VPA on c-Myc expression in nbHF. The HYD-induced overexpression of OCT4 and NANOG in aHDF was not dependent on demethylation of gene promoters, and no changes in the reprogramming efficiency were observed in both cell populations despite the downregulation of epigenetic genes DNMT1, ARID1A, and ARID2 in nbHF. CONCLUSIONS: Our data provide evidence that HYD regulates the expression of OCT4 and NANOG pluripotency genes as well as ARID1A and ARID2 genes, two members of the SWI/SNF chromatin remodeling complex family, in normal human dermal fibroblasts.


Sujet(s)
Assemblage et désassemblage de la chromatine , Cellules souches pluripotentes induites , Nouveau-né , Humains , Facteur-4 de type Kruppel , Reprogrammation cellulaire/génétique , Cellules souches pluripotentes induites/métabolisme , Fibroblastes/métabolisme , Facteur de transcription Oct-3/génétique , Facteur de transcription Oct-3/métabolisme
20.
Semin Cell Dev Biol ; 144: 77-86, 2023 07 30.
Article de Anglais | MEDLINE | ID: mdl-36210260

RÉSUMÉ

Human-induced pluripotent stem cells (hiPSCs) have provided new methods to study neurodegenerative diseases. In addition to their wide application in neuronal disorders, hiPSCs technology can also encompass specific conditions, such as inherited retinal dystrophies. The possibility of evaluating alterations related to retinal disorders in 3D organoids increases the truthfulness of in vitro models. Moreover, both Alzheimer's (AD) and Parkinson's disease (PD) have been described as causing early retinal alterations, generating beta-amyloid protein accumulation, or affecting dopaminergic amacrine cells. This review addresses recent advances and future perspectives obtained from in vitro modeling of retinal diseases, focusing on retinitis pigmentosa (RP). Additionally, we depicted the possibility of evaluating changes related to AD and PD in retinal organoids obtained from potential patients long before the onset of the disease, constituting a valuable tool in early diagnosis. With this, we pointed out prospects in the study of retinal dystrophies and early diagnosis of AD and PD.


Sujet(s)
Maladie d'Alzheimer , Cellules souches pluripotentes induites , Maladie de Parkinson , Rétinite pigmentaire , Humains , Cellules souches pluripotentes induites/métabolisme , Maladie de Parkinson/diagnostic , Maladie de Parkinson/métabolisme , Maladie d'Alzheimer/diagnostic , Maladie d'Alzheimer/métabolisme , Rétinite pigmentaire/métabolisme , Organoïdes , Diagnostic précoce
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE