Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Stem Cell Reports ; 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38701777

RESUMEN

Embryonic stem cells (ESCs) are defined as stem cells with self-renewing and differentiation capabilities. These unique properties are tightly regulated and controlled by complex genetic and molecular mechanisms, whose understanding is essential for both basic and translational research. A large number of studies have mostly focused on understanding the molecular mechanisms governing pluripotency and differentiation of ESCs, while the regulation of proliferation has received comparably less attention. Here, we investigate the role of ZZZ3 (zinc finger ZZ-type containing 3) in human ESCs homeostasis. We found that knockdown of ZZZ3 negatively impacts ribosome biogenesis, translation, and mTOR signaling, leading to a significant reduction in cell proliferation. This process occurs without affecting pluripotency, suggesting that ZZZ3-depleted ESCs enter a "dormant-like" state and that proliferation and pluripotency can be uncoupled also in human ESCs.

2.
Parkinsonism Relat Disord ; 123: 106978, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38678852

RESUMEN

INTRODUCTION: Differentiating Progressive Supranuclear Palsy (PSP) from Parkinson's Disease (PD) may be clinically challenging. In this study, we explored the performance of machine learning models based on MR imaging and blood molecular biomarkers in distinguishing between these two neurodegenerative diseases. METHODS: Twenty-eight PSP patients, 46 PD patients and 60 control subjects (HC) were consecutively enrolled in the study. Serum concentration of neurofilament light chain protein (Nf-L) was assessed by single molecule array (SIMOA), while an automatic segmentation algorithm was employed for T1-weighted measurements of third ventricle width/intracranial diameter ratio (3rdV/ID). Machine learning (ML) models with Logistic Regression (LR), Random Forest (RF), and XGBoost algorithms based on 3rdV/ID and serum Nf-L levels were tested in distinguishing among PSP, PD and HC. RESULTS: PSP patients showed higher serum Nf-L levels and larger 3rdV/ID ratio in comparison with both PD and HC groups (p < 0.005). All ML algorithms (LR, RF and XGBoost) showed that the combination of MRI and blood biomarkers had excellent classification performances in differentiating PSP from PD (AUC ≥0.92), outperforming each biomarker used alone (AUC: 0.85-0.90). Among the different algorithms, XGBoost was slightly more powerful than LR and RF in distinguishing PSP from PD patients, reaching AUC of 0.94 ± 0.04. CONCLUSION: Our findings highlight the usefulness of combining blood and simple linear MRI biomarkers to accurately distinguish between PSP and PD patients. This multimodal approach may play a pivotal role in patient management and clinical decision-making, paving the way for more effective and timely interventions in these neurodegenerative diseases.

3.
Sci Rep ; 14(1): 5941, 2024 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-38467734

RESUMEN

Prolonged febrile seizures (FS) in children are linked to the development of temporal lobe epilepsy (MTLE). The association between these two pathologies may be ascribed to the long-term effects that FS exert on neural stem cells, negatively affecting the generation of new neurons. Among the insults associated with FS, oxidative stress is noteworthy. Here, we investigated the consequences of exposure to hydrogen peroxide (H2O2) in an induced pluripotent stem cell-derived neural stem cells (iNSCs) model of a patient affected by FS and MTLE. In our study, we compare the findings from the MTLE patient with those derived from iNSCs of a sibling exhibiting a milder phenotype defined only by FS, as well as a healthy individual. In response to H2O2 treatment, iNSCs derived from MTLE patients demonstrated an elevated production of reactive oxygen species and increased apoptosis, despite the higher expression levels of antioxidant genes and proteins compared to other cell lines analysed. Among the potential causative mechanisms of enhanced vulnerability of MTLE patient iNSCs to oxidative stress, we found that these cells express low levels of the heat shock protein HSPB1 and of the autophagy adaptor SQSTM1/p62. Pre-treatment of diseased iNSCs with the antioxidant molecule ascorbic acid restored HSBP1 and p62 expression and simultaneously reduced the levels of ROS and apoptosis. Our findings suggest the potential for rescuing the impaired oxidative stress response in diseased iNSCs through antioxidant treatment, offering a promising mechanism to prevent FS degeneration in MTLE.


Asunto(s)
Epilepsia del Lóbulo Temporal , Convulsiones Febriles , Niño , Humanos , Epilepsia del Lóbulo Temporal/tratamiento farmacológico , Epilepsia del Lóbulo Temporal/metabolismo , Convulsiones Febriles/tratamiento farmacológico , Convulsiones Febriles/genética , Ácido Ascórbico/farmacología , Ácido Ascórbico/uso terapéutico , Ácido Ascórbico/metabolismo , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Antioxidantes/metabolismo , Peróxido de Hidrógeno/metabolismo , Estrés Oxidativo , Hipocampo/metabolismo , Proteínas de Choque Térmico/metabolismo
4.
Cells ; 11(21)2022 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-36359887

RESUMEN

Unverricht-Lundborg disease (ULD), also known as progressive myoclonic epilepsy 1 (EPM1), is a rare autosomal recessive neurodegenerative disorder characterized by a complex symptomatology that includes action- and stimulus-sensitive myoclonus and tonic-clonic seizures. The main cause of the onset and development of ULD is a repeat expansion of a dodecamer sequence localized in the promoter region of the gene encoding cystatin B (CSTB), an inhibitor of lysosomal proteases. Although this is the predominant mutation found in most patients, the physio-pathological mechanisms underlying the disease complexity remain largely unknown. In this work, we used patient-specific iPSCs and their neuronal derivatives to gain insight into the molecular and genetic machinery responsible for the disease in two Italian siblings affected by different phenotypes of ULD. Specifically, fragment length analysis on amplified CSTB promoters found homozygous status for dodecamer expansion in both patients and showed that the number of dodecamer repeats is the same in both. Furthermore, the luciferase reporter assay showed that the CSTB promoter activity was similarly reduced in both lines compared to the control. This information allowed us to draw important conclusions: (1) the phenotypic differences of the patients do not seem to be strictly dependent on the genetic mutation around the CSTB gene, and (2) that some other molecular mechanisms, not yet clearly identified, might be taken into account. In line with the inhibitory role of cystatin B on cathepsins, molecular investigations performed on iPSCs-derived neurons showed an increased expression of lysosomal cathepsins (B, D, and L) and a reduced expression of CSTB protein. Intriguingly, the increase in cathepsin expression does not appear to be correlated with the residual amount of CSTB, suggesting that other mechanisms, in addition to the regulation of cathepsins, could be involved in the pathological complexity of the disease.


Asunto(s)
Síndrome de Unverricht-Lundborg , Humanos , Síndrome de Unverricht-Lundborg/genética , Cistatina B/genética , Hermanos , Perfil Genético , Catepsinas/genética
5.
Biomedicines ; 10(5)2022 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-35625812

RESUMEN

Mutations in SCN1A gene, encoding the voltage-gated sodium channel (VGSC) NaV1.1, are widely recognized as a leading cause of genetic febrile seizures (FS), due to the decrease in the Na+ current density, mainly affecting the inhibitory neuronal transmission. Here, we generated induced pluripotent stem cells (iPSCs)-derived neurons (idNs) from a patient belonging to a genetically well-characterized Italian family, carrying the c.434T > C mutation in SCN1A gene (hereafter SCN1AM145T). A side-by-side comparison of diseased and healthy idNs revealed an overall maturation delay of SCN1AM145T cells. Membranes isolated from both diseased and control idNs were injected into Xenopus oocytes and both GABA and AMPA currents were successfully recorded. Patch-clamp measurements on idNs revealed depolarized action potential for SCN1AM145T, suggesting a reduced excitability. Expression analyses of VGSCs and chloride co-transporters NKCC1 and KCC2 showed a cellular "dysmaturity" of mutated idNs, strengthened by the high expression of SCN3A, a more fetal-like VGSC isoform, and a high NKCC1/KCC2 ratio, in mutated cells. Overall, we provide strong evidence for an intrinsic cellular immaturity, underscoring the role of mutant NaV1.1 in the development of FS. Furthermore, our data are strengthening previous findings obtained using transfected cells and recordings on human slices, demonstrating that diseased idNs represent a powerful tool for personalized therapy and ex vivo drug screening for human epileptic disorders.

6.
Commun Biol ; 4(1): 1146, 2021 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-34593953

RESUMEN

miRNAs modulate cardiomyocyte specification by targeting mRNAs of cell cycle regulators and acting in cardiac muscle lineage gene regulatory loops. It is unknown if or to-what-extent these miRNA/mRNA networks are operative during cardiomyocyte differentiation of adult cardiac stem/progenitor cells (CSCs). Clonally-derived mouse CSCs differentiated into contracting cardiomyocytes in vitro (iCMs). Comparison of "CSCs vs. iCMs" mRNome and microRNome showed a balanced up-regulation of CM-related mRNAs together with a down-regulation of cell cycle and DNA replication mRNAs. The down-regulation of cell cycle genes and the up-regulation of the mature myofilament genes in iCMs reached intermediate levels between those of fetal and neonatal cardiomyocytes. Cardiomyo-miRs were up-regulated in iCMs. The specific networks of miRNA/mRNAs operative in iCMs closely resembled those of adult CMs (aCMs). miR-1 and miR-499 enhanced myogenic commitment toward terminal differentiation of iCMs. In conclusions, CSC specification/differentiation into contracting iCMs follows known cardiomyo-MiR-dependent developmental cardiomyocyte differentiation trajectories and iCMs transcriptome/miRNome resembles that of CMs.


Asunto(s)
Diferenciación Celular/genética , MicroARNs/genética , Miocitos Cardíacos/metabolismo , ARN Mensajero/genética , Células Madre/metabolismo , Animales , Ciclo Celular , Ratones , MicroARNs/metabolismo , Desarrollo de Músculos/genética , ARN Mensajero/metabolismo , Transcriptoma , Regulación hacia Arriba
7.
Cells ; 10(9)2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34572080

RESUMEN

Embryonic stem cells (ESCs) are pluripotent cells with indefinite self-renewal ability and differentiation properties. To function properly and maintain genomic stability, ESCs need to be endowed with an efficient repair system as well as effective redox homeostasis. In this study, we investigated different aspects involved in ESCs' response to iron accumulation following stable knockdown of the ferritin heavy chain (FTH1) gene, which encodes for a major iron storage protein with ferroxidase activity. Experimental findings highlight unexpected and, to a certain extent, paradoxical results. If on one hand FTH1 silencing does not correlate with increased ROS production nor with changes in the redox status, strengthening the concept that hESCs are extremely resistant and, to a certain extent, even refractory to intracellular iron imbalance, on the other, the differentiation potential of hESCs seems to be affected and apoptosis is observed. Interestingly, we found that FTH1 silencing is accompanied by a significant activation of the nuclear factor (erythroid-derived-2)-like 2 (Nrf2) signaling pathway and pentose phosphate pathway (PPP), which crosstalk in driving hESCs antioxidant cascade events. These findings shed new light on how hESCs perform under oxidative stress, dissecting the molecular mechanisms through which Nrf2, in combination with PPP, counteracts oxidative injury triggered by FTH1 knockdown.


Asunto(s)
Ferritinas/genética , Células Madre Embrionarias Humanas/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Oxidorreductasas/genética , Elementos de Respuesta Antioxidante , Apoptosis , Diferenciación Celular , Células Cultivadas , Ferritinas/farmacología , Silenciador del Gen , Humanos , Oxidación-Reducción , Oxidorreductasas/metabolismo , Vía de Pentosa Fosfato , Transducción de Señal
8.
Stem Cell Res ; 53: 102329, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33865103

RESUMEN

Unverricht-Lundborg disease (ULD) is an inherited form of progressive myoclonus epilepsy caused by mutations in the gene encoding Cystatin B (CSTB), an inhibitor of lysosomal proteases. The most common mutation described in ULD patients is an unstable expansion of a dodecamer sequence located in the CSTB gene promoter. This expansion is causative of the downregulation of CSTB gene expression and, consequently, of its inhibitory activity. Here we report the generation of induced pluripotent stem cell (iPSC) lines from two Italian siblings having a family history of ULD and affected by different clinical and pathological phenotypes of the disease.


Asunto(s)
Células Madre Pluripotentes Inducidas , Síndrome de Unverricht-Lundborg , Cistatina B/genética , Humanos , Italia , Hermanos
9.
Int J Mol Sci ; 22(4)2021 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-33670616

RESUMEN

Arrhythmogenic Right Ventricular cardiomyopathy (ARVC) is an inherited cardiac muscle disease linked to genetic deficiency in components of the desmosomes. The disease is characterized by progressive fibro-fatty replacement of the right ventricle, which acts as a substrate for arrhythmias and sudden cardiac death. The molecular mechanisms underpinning ARVC are largely unknown. Here we propose a mathematical model for investigating the molecular dynamics underlying heart remodeling and the loss of cardiac myocytes identity during ARVC. Our methodology is based on three computational models: firstly, in the context of the Wnt pathway, we examined two different competition mechanisms between ß-catenin and Plakoglobin (PG) and their role in the expression of adipogenic program. Secondly, we investigated the role of RhoA-ROCK pathway in ARVC pathogenesis, and thirdly we analyzed the interplay between Wnt and RhoA-ROCK pathways in the context of the ARVC phenotype. We conclude with the following remark: both Wnt/ß-catenin and RhoA-ROCK pathways must be inactive for a significant increase of PPARγ expression, suggesting that a crosstalk mechanism might be responsible for mediating ARVC pathogenesis.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Adipogénesis/genética , Algoritmos , Displasia Ventricular Derecha Arritmogénica/genética , Displasia Ventricular Derecha Arritmogénica/metabolismo , Displasia Ventricular Derecha Arritmogénica/patología , Células Cultivadas , Simulación por Computador , Regulación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/citología , Modelos Teóricos , PPAR gamma/genética , PPAR gamma/metabolismo , gamma Catenina/metabolismo
10.
Stem Cell Res ; 49: 102083, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33370868

RESUMEN

Here, we described the generation of human induced pluripotent stem cell lines (hiPSCs) from fibroblasts isolated by punch biopsies of two siblings carrying inherited mutation (c.434 T > C) in the SCN1A gene, encoding for the neuronal voltage gated sodium channel NaV1.1. The mutation leads to the substitution of a highly conserved methionine with a threonine (M145T) in the protein sequence, leading to infant febrile seizures (FS). The older brother, affected by complex FS, also developed temporal lobe epilepsy (TLE) during adolescence.


Asunto(s)
Línea Celular , Células Madre Pluripotentes Inducidas , Convulsiones Febriles , Adolescente , Humanos , Lactante , Masculino , Mutación , Mutación Missense/genética , Canal de Sodio Activado por Voltaje NAV1.1/genética , Convulsiones Febriles/genética
11.
Int J Mol Sci ; 21(21)2020 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-33114544

RESUMEN

The 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors (statins) exert pleiotropic effects on cardiac cell biology which are not yet fully understood. Here we tested whether statin treatment affects resident endogenous cardiac stem/progenitor cell (CSC) activation in vitro and in vivo after myocardial infarction (MI). Statins (Rosuvastatin, Simvastatin and Pravastatin) significantly increased CSC expansion in vitro as measured by both BrdU incorporation and cell growth curve. Additionally, statins increased CSC clonal expansion and cardiosphere formation. The effects of statins on CSC growth and differentiation depended on Akt phosphorylation. Twenty-eight days after myocardial infarction by permanent coronary ligation in rats, the number of endogenous CSCs in the infarct border zone was significantly increased by Rosuvastatin-treatment as compared to untreated controls. Additionally, commitment of the activated CSCs into the myogenic lineage (c-kitpos/Gata4pos CSCs) was increased by Rosuvastatin administration. Accordingly, Rosuvastatin fostered new cardiomyocyte formation after MI. Finally, Rosuvastatin treatment reversed the cardiomyogenic defects of CSCs in c-kit haploinsufficient mice, increasing new cardiomyocyte formation by endogenous CSCs in these mice after myocardial infarction. In summary, statins, by sustaining Akt activation, foster CSC growth and differentiation in vitro and in vivo. The activation and differentiation of the endogenous CSC pool and consequent new myocyte formation by statins improve myocardial remodeling after coronary occlusion in rodents. Similar effects might contribute to the beneficial effects of statins on human cardiovascular diseases.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Células Musculares/citología , Infarto del Miocardio/tratamiento farmacológico , Miocardio/citología , Células Madre/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Ratones , Células Musculares/efectos de los fármacos , Células Musculares/metabolismo , Infarto del Miocardio/etiología , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Fosforilación/efectos de los fármacos , Pravastatina/administración & dosificación , Pravastatina/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Rosuvastatina Cálcica/administración & dosificación , Rosuvastatina Cálcica/farmacología , Simvastatina/administración & dosificación , Simvastatina/farmacología , Células Madre/citología , Células Madre/metabolismo
12.
Int J Mol Sci ; 21(12)2020 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-32575374

RESUMEN

Cardiovascular diseases (CVDs) are a class of disorders affecting the heart or blood vessels. Despite progress in clinical research and therapy, CVDs still represent the leading cause of mortality and morbidity worldwide. The hallmarks of cardiac diseases include heart dysfunction and cardiomyocyte death, inflammation, fibrosis, scar tissue, hyperplasia, hypertrophy, and abnormal ventricular remodeling. The loss of cardiomyocytes is an irreversible process that leads to fibrosis and scar formation, which, in turn, induce heart failure with progressive and dramatic consequences. Both genetic and environmental factors pathologically contribute to the development of CVDs, but the precise causes that trigger cardiac diseases and their progression are still largely unknown. The lack of reliable human model systems for such diseases has hampered the unraveling of the underlying molecular mechanisms and cellular processes involved in heart diseases at their initial stage and during their progression. Over the past decade, significant scientific advances in the field of stem cell biology have literally revolutionized the study of human disease in vitro. Remarkably, the possibility to generate disease-relevant cell types from induced pluripotent stem cells (iPSCs) has developed into an unprecedented and powerful opportunity to achieve the long-standing ambition to investigate human diseases at a cellular level, uncovering their molecular mechanisms, and finally to translate bench discoveries into potential new therapeutic strategies. This review provides an update on previous and current research in the field of iPSC-driven cardiovascular disease modeling, with the aim of underlining the potential of stem-cell biology-based approaches in the elucidation of the pathophysiology of these life-threatening diseases.


Asunto(s)
Cardiopatías/patología , Células Madre Pluripotentes Inducidas/citología , Diferenciación Celular , Progresión de la Enfermedad , Cardiopatías/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Modelos Biológicos , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Transducción de Señal
13.
Biosensors (Basel) ; 10(3)2020 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-32121446

RESUMEN

In this work, a disposable passive microfluidic device for cell culturing that does not require any additional/external pressure sources is introduced. By regulating the height of fluidic columns and the aperture and closure of the source wells, the device can provide different media and/or drug flows, thereby allowing different flow patterns with respect to time. The device is made of two Polymethylmethacrylate (PMMA) layers fabricated by micro-milling and solvent assisted bonding and allows us to ensure a flow rate of 18.6 µl/ℎ - 7%/day, due to a decrease of the fluid height while the liquid is driven from the reservoirs into the channels. Simulations and experiments were conducted to characterize flows and diffusion in the culture chamber. Melanoma tumor cells were used to test the device and carry out cell culturing experiments for 48 hours. Moreover, HeLa, Jurkat, A549 and HEK293T cell lines were cultivated successfully inside the microfluidic device for 72 hours.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Microfluídica/métodos , Humanos
14.
J Assist Reprod Genet ; 37(1): 149-158, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31701304

RESUMEN

PURPOSE: To detect putative differences in the miRNomic profile of follicular fluids collected after follicular-phase-stimulation (FPS-FFs) and paired luteal-phase-stimulation (LPS-FFs) in the same ovarian cycles (DuoStim). METHODS: Exploratory study at a private IVF center and University involving FPS-FFs and paired-LPS-FFs collected from 15 reduced ovarian reserve and advanced maternal age women undergoing DuoStim (n = 30 paired samples). The samples were combined in 6 paired pools (5 samples each) and balanced according to maternal age and number of cumulus-oocyte-complexes. Micro-RNAs were isolated and sequenced. Four miRNAs were then selected for further validation on 6 single pairs of FPS-FFs and LPS-FFs by qPCR. RESULTS: Forty-three miRNAs were detected in both FPS-FFs and paired-LPS-FFs after sequencing and no statistically significant differences were reported. Thirty-three KEGG pathways were identified as regulated from the detected miRNAs. Four miRNAs (miR-146b, miR-191, miR-320a, and miR-483) were selected for qPCR validation since consistently expressed in our samples and possibly involved in the regulation/establishment of a healthy follicular environment. Again, no significant differences were reported between FPS-FFs and paired-LPS-FFs, also when the analysis was corrected for maternal age and number of cumulus-oocyte-complexes in generalized linear models. CONCLUSIONS: These data complement the embryological, chromosomal, and clinical evidence of equivalence between FPS and LPS published to date.


Asunto(s)
Líquido Folicular/metabolismo , Fase Folicular/genética , Infertilidad Femenina/genética , Fase Luteínica/genética , Ciclo Menstrual/genética , MicroARNs/genética , Inducción de la Ovulación/métodos , Adulto , Femenino , Fase Folicular/metabolismo , Perfilación de la Expresión Génica , Humanos , Fase Luteínica/metabolismo
15.
Int J Mol Sci ; 20(22)2019 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-31744081

RESUMEN

A comprehensive understanding of the molecular basis and mechanisms underlying cardiac diseases is mandatory for the development of new and effective therapeutic strategies. The lack of appropriate in vitro cell models that faithfully mirror the human disease phenotypes has hampered the understanding of molecular insights responsible of heart injury and disease development. Over the past decade, important scientific advances have revolutionized the field of stem cell biology through the remarkable discovery of reprogramming somatic cells into induced pluripotent stem cells (iPSCs). These advances allowed to achieve the long-standing ambition of modelling human disease in a dish and, more interestingly, paved the way for unprecedented opportunities to translate bench discoveries into new therapies and to come closer to a real and effective stem cell-based medicine. The possibility to generate patient-specific iPSCs, together with the new advances in stem cell differentiation procedures and the availability of novel gene editing approaches and tissue engineering, has proven to be a powerful combination for the generation of phenotypically complex, pluripotent stem cell-based cellular disease models with potential use for early diagnosis, drug screening, and personalized therapy. This review will focus on recent progress and future outcome of iPSCs technology toward a customized medicine and new therapeutic options.


Asunto(s)
Cardiopatías/terapia , Medicina Regenerativa , Trasplante de Células Madre , Técnicas de Cultivo de Célula , Tratamiento Basado en Trasplante de Células y Tejidos , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Andamios del Tejido/química
16.
J Cell Mol Med ; 23(11): 7382-7394, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31536674

RESUMEN

Involvement of the central nervous system (CNS) is an uncommon feature in systemic lupus erythematosus (SLE), making diagnosis rather difficult and challenging due to the poor specificity of neuropathic symptoms and neurological symptoms. In this work, we used human-induced pluripotent stem cells (hiPSCs) derived from CNS-SLE patient, with the aim to dissect the molecular insights underlying the disease by gene expression analysis and modulation of implicated pathways. CNS-SLE-derived hiPSCs allowed us to provide evidence of Erk and Akt pathways involvement and to identify a novel cohort of potential biomarkers, namely CHCHD2, IDO1, S100A10, EPHA4 and LEFTY1, never reported so far. We further extended the study analysing a panel of oxidative stress-related miRNAs and demonstrated, under normal or stress conditions, a strong dysregulation of several miRNAs in CNS-SLE-derived compared to control hiPSCs. In conclusion, we provide evidence that iPSCs reprogrammed from CNS-SLE patient are a powerful useful tool to investigate the molecular mechanisms underlying the disease and to eventually develop innovative therapeutic approaches.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Lupus Eritematoso Sistémico/fisiopatología , Vasculitis por Lupus del Sistema Nervioso Central/fisiopatología , Biomarcadores/metabolismo , Femenino , Expresión Génica/fisiología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Vasculitis por Lupus del Sistema Nervioso Central/metabolismo , MicroARNs/metabolismo , Estrés Oxidativo/fisiología , Transducción de Señal/fisiología
17.
J Cell Mol Med ; 23(8): 5440-5453, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31237115

RESUMEN

Although the concepts of somatic cell reprogramming and human-induced pluripotent stem cells (hiPSCs) generation have undergone several analyses to validate the usefulness of these cells in research and clinic, it remains still controversial whether the hiPSCs are equivalent to human embryonic stem cells (hESCs), pointing to the need of further characterization for a more comprehensive understanding of pluripotency. Most of the experimental evidence comes from the transcriptome analysis, while a little is available on protein data, and even less is known about the post-translational modifications. Here, we report a combined strategy of mass spectrometry and gene expression profiling for proteogenomic analysis of reprogrammed and embryonic stem cells. The data obtained through this integrated, multi-"omics" approach indicate that a small, but still significant, number of distinct pathways is enriched in reprogrammed versus embryonic stem cells, supporting the view that pluripotency is an extremely complex, multifaceted phenomenon, with peculiarities that are characteristic of each cell type.


Asunto(s)
Células Madre Embrionarias Humanas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Células Cultivadas , Reprogramación Celular/genética , Fibroblastos/metabolismo , Perfilación de la Expresión Génica/métodos , Humanos , Espectrometría de Masas/métodos , Procesamiento Proteico-Postraduccional/genética , Proteogenómica/métodos , Transcriptoma/genética
18.
Biosensors (Basel) ; 9(1)2019 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-30832416

RESUMEN

Biomarkers detection at an ultra-low concentration in biofluids (blood, serum, saliva, etc.) is a key point for the early diagnosis success and the development of personalized therapies. However, it remains a challenge due to limiting factors like (i) the complexity of analyzed media, and (ii) the aspecificity detection and the poor sensitivity of the conventional methods. In addition, several applications require the integration of the primary sensors with other devices (microfluidic devices, capillaries, flasks, vials, etc.) where transducing the signal might be difficult, reducing performances and applicability. In the present work, we demonstrate a new class of optical biosensor we have developed integrating an optical waveguide (OWG) with specific plasmonic surfaces. Exploiting the plasmonic resonance, the devices give consistent results in surface enhanced Raman spectroscopy (SERS) for continuous and label-free detection of biological compounds. The OWG allows driving optical signals in the proximity of SERS surfaces (detection area) overcoming spatial constraints, in order to reach places previously optically inaccessible. A rutile prism couples the remote laser source to the OWG, while a Raman spectrometer collects the SERS far field scattering. The present biosensors were implemented by a simple fabrication process, which includes photolithography and nanofabrication. By using such devices, it was possible to detect cell metabolites like Phenylalanine (Phe), Adenosine 5-triphosphate sodium hydrate (ATP), Sodium Lactate, Human Interleukin 6 (IL6), and relate them to possible metabolic pathway variation.


Asunto(s)
Técnicas Biosensibles/métodos , Óptica y Fotónica/métodos , Espectrometría Raman/métodos , Adenosina/química , Adenosina/aislamiento & purificación , Adenosina Trifosfato/química , Adenosina Trifosfato/aislamiento & purificación , Humanos , Interleucina-6/química , Interleucina-6/aislamiento & purificación , Dispositivos Laboratorio en un Chip , Límite de Detección , Fenilalanina/química , Fenilalanina/aislamiento & purificación , Lactato de Sodio/química , Lactato de Sodio/aislamiento & purificación , Resonancia por Plasmón de Superficie , Propiedades de Superficie
19.
RSC Adv ; 9(8): 4246-4257, 2019 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-35520194

RESUMEN

The aim of this study was the design of a 3D scaffold composed of poly(vinyl) alcohol (PVA) for cardiac tissue engineering (CTE) applications. The PVA scaffold was fabricated using a combination of gas foaming and freeze-drying processes that did not need any cross-linking agents. We obtained a biocompatible porous matrix with excellent mechanical properties. We measured the stress-strain curves of the PVA scaffolds and we showed that the elastic behavior is similar to that of the extracellular matrix of muscles. The SEM observations revealed that the scaffolds possess micro pores having diameters ranging from 10 µm to 370 µm that fit to the dimensions of the cells. A further purpose of this study was to test scaffolds ability to support human induced pluripotent stem cells growth and differentiation into cardiomyocytes. As the proliferation tests show, the number of live stem cells on the scaffold after 12 days was increased with respect to the initial number of cells, revealing the cytocompatibility of the substrate. In addition, the differentiated cells on the PVA scaffolds expressed anti-troponin T, a marker specific of the cardiac sarcomere. We demonstrated the ability of the cardiomyocytes to pulse within the scaffolds. In conclusion, the developed scaffold show the potential to be used as a biomaterial for CTE applications.

20.
Cell Death Dis ; 9(1): 6, 2018 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-29305588

RESUMEN

Human pluripotent stem cells (hPSCs), including human embryonic stem cells (hESCs) derived from blastocyst and human induced pluripotent stem cells (hiPSCs) generated from somatic cells by ectopic expression of defined transcriptional factors, have both the ability to self-renew and to differentiate into all cell types. Here we explored the two antagonistic effects of retinoic acid (RA) on hiPSCs. Although RA has been widely described as a pharmacological agent with a critical role in initiating differentiation of pluripotent stem cells, we demonstrate that short-term RA exposure not only antagonizes cell differentiation and sustains pluripotency of hiPSCs, but it also boosts and improves their properties and characteristics. To shed light on the mechanistic insights involved in the resistance to differentiation of hiPSCs cultured in RA conditions, as well as their improved pluripotency state, we focused our attention on the Wnt pathway. Our findings show that RA inhibits the Wnt canonical pathway and positively modulates the Akt/mTOR signaling, explaining why such perturbations, under our experimental conditions, do not lead to hiPSCs differentiation. Altogether, these data uncover a novel role for RA in favouring the maintenance of ground-state pluripotency, supporting its bivalent role, dose- and time-dependent, for hiPSCs differentiation and self-renewal processes.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Madre Pluripotentes Inducidas/citología , Tretinoina/farmacología , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Fibroblastos/citología , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Linfocitos T/citología , Serina-Treonina Quinasas TOR/metabolismo , Telomerasa/genética , Telomerasa/metabolismo , Telómero/genética , Telómero/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...