Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Front Mol Neurosci ; 16: 1280546, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38125008

RESUMEN

Spinocerebellar ataxia type 1 (SCA1) is an autosomal dominant neurodegenerative disease caused by a trinucleotide (CAG) repeat expansion in the ATXN1 gene. It is characterized by the presence of polyglutamine (polyQ) intranuclear inclusion bodies (IIBs) within affected neurons. In order to investigate the impact of polyQ IIBs in SCA1 pathogenesis, we generated a novel protein aggregation model by inducible overexpression of the mutant ATXN1(Q82) isoform in human neuroblastoma SH-SY5Y cells. Moreover, we developed a simple and reproducible protocol for the efficient isolation of insoluble IIBs. Biophysical characterization showed that polyQ IIBs are enriched in RNA molecules which were further identified by next-generation sequencing. Finally, a protein interaction network analysis indicated that sequestration of essential RNA transcripts within ATXN1(Q82) IIBs may affect the ribosome resulting in error-prone protein synthesis and global proteome instability. These findings provide novel insights into the molecular pathogenesis of SCA1, highlighting the role of polyQ IIBs and their impact on critical cellular processes.

2.
Micromachines (Basel) ; 14(10)2023 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-37893266

RESUMEN

In the last decade, there has been a notable advancement in diverse bioreactor types catering to various applications. However, conventional bioreactors often exhibit bulkiness and high costs, making them less accessible to many researchers and laboratory facilities. In light of these challenges, this article aims to introduce and evaluate the development of a do-it-yourself (DIY) 3D printed smart bioreactor, offering a cost-effective and user-friendly solution for the proliferation of various bioentities, including bacteria and human organoids, among others. The customized bioreactor was fabricated under an ergonomic design and assembled with 3D printed mechanical parts combined with electronic components, under 3D printed housing. The 3D printed parts were designed using SOLIDWORKS® CAD Software (2022 SP2.0 Professional version) and fabricated via the fused filament fabrication (FFF) technique. All parts were 3D printed with acrylonitrile butadiene styrene (ABS) in order for the bioreactor to be used under sterile conditions. The printed low-cost bioreactor integrates Internet-of-things (IoT) functionalities, since it provides the operator with the ability to change its operational parameters (sampling frequency, rotor speed, and duty cycle) remotely, via a user-friendly developed mobile application and to save the user history locally on the device. Using this bioreactor, which is adjusted to a standard commercial 12-well plate, proof of concept of a successful operation of the bioreactor during a 2-day culture of Escherichia coli bacteria (Mach1 strain) is presented. This study paves the way for more in-depth investigation of bacterial and various biological-entity growth cultures, utilizing 3D printing technology to create customized low-cost bioreactors.

3.
J Funct Biomater ; 14(9)2023 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-37754879

RESUMEN

The latest developments in tissue engineering scaffolds have sparked a growing interest in the creation of controlled 3D cellular structures that emulate the intricate biophysical and biochemical elements found within versatile in vivo microenvironments. The objective of this study was to 3D-print a monolithic silica scaffold specifically designed for the cultivation of neural precursor cells. Initially, a preliminary investigation was conducted to identify the critical parameters pertaining to calcination. This investigation aimed to produce sturdy and uniform scaffolds with a minimal wall-thickness of 0.5 mm in order to mitigate the formation of cracks. Four cubic specimens, with different wall-thicknesses of 0.5, 1, 2, and 4 mm, were 3D-printed and subjected to two distinct calcination profiles. Thermogravimetric analysis was employed to examine the freshly printed material, revealing critical temperatures associated with increased mass loss. Isothermal steps were subsequently introduced to facilitate controlled phase transitions and reduce crack formation even at the minimum wall thickness of 0.5 mm. The optimized structure stability was obtained for the slow calcination profile (160 min) then the fast calcination profile (60 min) for temperatures up to 900 °C. In situ X-ray diffraction analysis was also employed to assess the crystal phases of the silicate based material throughout various temperature profiles up to 1200 °C, while scanning electron microscopy was utilized to observe micro-scale crack formation. Then, ceramic scaffolds were 3D-printed, adopting a hexagonal and spherical channel structures with channel opening of 2 mm, and subsequently calcined using the optimized slow profile. Finally, the scaffolds were evaluated in terms of biocompatibility, cell proliferation, and differentiation using neural precursor cells (NPCs). These experiments indicated proliferation of NPCs (for 13 days) and differentiation into neurons which remained viable (up to 50 days in culture). In parallel, functionality was verified by expression of pre- (SYN1) and post-synaptic (GRIP1) markers, suggesting that 3D-printed scaffolds are a promising system for biotechnological applications using NPCs.

5.
Adv Exp Med Biol ; 1423: 149-160, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37525039

RESUMEN

Parkinson's disease (PD) is the second most common neurodegenerative disorder, affecting millions of people worldwide. Despite considerable efforts, the underlying pathological mechanisms remain elusive, and yet, no treatment has been developed to efficiently reverse or modify disease progression. Thus, new experimental models are required to provide insights into the pathology of PD. Small-molecule neural precursor cells (smNPCs) are ideal for the study of neurodegenerative disorders due to their neural identity and stem cell properties. Cytoplasmic aggregates of α-synuclein (αSyn) are considered a hallmark of PD and a point mutation in the gene encoding p.A53T is responsible for a familial PD form with earlier and robust symptom onset. In order to study the cellular pathology of PD, we genetically modified smNPCs to inducibly overexpress EYFP-SNCA A53T. This cellular model was biochemically characterized, while dysregulated biological pathways and key regulators of PD pathology were identified by computational analyses. Our study indicates three novel genes, UBA52, PIP5K1A, and RPS2, which may mediate PD cellular pathology.


Asunto(s)
Células-Madre Neurales , Enfermedades Neurodegenerativas , Enfermedad de Parkinson , Humanos , Enfermedad de Parkinson/metabolismo , Células-Madre Neurales/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Neuronas/metabolismo
6.
Mov Disord ; 38(8): 1428-1442, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37278528

RESUMEN

BACKGROUND: Spinocerebellar ataxia type 1 (SCA1) is a neurodegenerative disease caused by a polyglutamine expansion in the ataxin-1 protein resulting in neuropathology including mutant ataxin-1 protein aggregation, aberrant neurodevelopment, and mitochondrial dysfunction. OBJECTIVES: Identify SCA1-relevant phenotypes in patient-specific fibroblasts and SCA1 induced pluripotent stem cells (iPSCs) neuronal cultures. METHODS: SCA1 iPSCs were generated and differentiated into neuronal cultures. Protein aggregation and neuronal morphology were evaluated using fluorescent microscopy. Mitochondrial respiration was measured using the Seahorse Analyzer. The multi-electrode array (MEA) was used to identify network activity. Finally, gene expression changes were studied using RNA-seq to identify disease-specific mechanisms. RESULTS: Bioenergetics deficits in patient-derived fibroblasts and SCA1 neuronal cultures showed altered oxygen consumption rate, suggesting involvement of mitochondrial dysfunction in SCA1. In SCA1 hiPSC-derived neuronal cells, nuclear and cytoplasmic aggregates were identified similar in localization as aggregates in SCA1 postmortem brain tissue. SCA1 hiPSC-derived neuronal cells showed reduced dendrite length and number of branching points while MEA recordings identified delayed development in network activity in SCA1 hiPSC-derived neuronal cells. Transcriptome analysis identified 1050 differentially expressed genes in SCA1 hiPSC-derived neuronal cells associated with synapse organization and neuron projection guidance, where a subgroup of 151 genes was highly associated with SCA1 phenotypes and linked to SCA1 relevant signaling pathways. CONCLUSIONS: Patient-derived cells recapitulate key pathological features of SCA1 pathogenesis providing a valuable tool for the identification of novel disease-specific processes. This model can be used for high throughput screenings to identify compounds, which may prevent or rescue neurodegeneration in this devastating disease. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Asunto(s)
Células Madre Pluripotentes Inducidas , Ataxias Espinocerebelosas , Ratones , Animales , Ataxinas/metabolismo , Agregado de Proteínas , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Ratones Transgénicos , Células de Purkinje/metabolismo , Células de Purkinje/patología , Ataxias Espinocerebelosas/metabolismo , Fibroblastos/metabolismo
7.
Mol Biol Evol ; 39(10)2022 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-36205081

RESUMEN

Although new genes can arrive from modes other than duplication, few examples are well characterized. Given high expression in some human brain subregions and a putative link to psychological disorders [e.g., schizophrenia (SCZ)], suggestive of brain functionality, here we characterize piggyBac transposable element-derived 1 (PGBD1). PGBD1 is nonmonotreme mammal-specific and under purifying selection, consistent with functionality. The gene body of human PGBD1 retains much of the original DNA transposon but has additionally captured SCAN and KRAB domains. Despite gene body retention, PGBD1 has lost transposition abilities, thus transposase functionality is absent. PGBD1 no longer recognizes piggyBac transposon-like inverted repeats, nonetheless PGBD1 has DNA binding activity. Genome scale analysis identifies enrichment of binding sites in and around genes involved in neuronal development, with association with both histone activating and repressing marks. We focus on one of the repressed genes, the long noncoding RNA NEAT1, also dysregulated in SCZ, the core structural RNA of paraspeckles. DNA binding assays confirm specific binding of PGBD1 both in the NEAT1 promoter and in the gene body. Depletion of PGBD1 in neuronal progenitor cells (NPCs) results in increased NEAT1/paraspeckles and differentiation. We conclude that PGBD1 has evolved core regulatory functionality for the maintenance of NPCs. As paraspeckles are a mammal-specific structure, the results presented here show a rare example of the evolution of a novel gene coupled to the evolution of a contemporaneous new structure.


Asunto(s)
Elementos Transponibles de ADN , ARN Largo no Codificante , Animales , Núcleo Celular/genética , Histonas/metabolismo , Humanos , Mamíferos/genética , Mamíferos/metabolismo , Proteínas del Tejido Nervioso , Paraspeckles , ARN Largo no Codificante/metabolismo , Transposasas/genética , Transposasas/metabolismo
8.
Int J Mol Sci ; 23(10)2022 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-35628660

RESUMEN

Huntington's disease (HD) is caused by the production of a mutant huntingtin (HTT) with an abnormally long poly-glutamine (polyQ) tract, forming aggregates and inclusions in neurons. Previous work by us and others has shown that an increase or decrease in polyQ-triggered aggregates can be passive simply due to the interaction of proteins with the aggregates. To search for proteins with active (functional) effects, which might be more effective in finding therapies and mechanisms of HD, we selected among the proteins that interact with HTT a total of 49 pairs of proteins that, while being paralogous to each other (and thus expected to have similar passive interaction with HTT), are located in different regions of the protein interaction network (suggesting participation in different pathways or complexes). Three of these 49 pairs contained members with opposite effects on HD, according to the literature. The negative members of the three pairs, MID1, IKBKG, and IKBKB, interact with PPP2CA and TUBB, which are known negative factors in HD, as well as with HSP90AA1 and RPS3. The positive members of the three pairs interact with HSPA9. Our results provide potential HD modifiers of functional relevance and reveal the dynamic aspect of paralog evolution within the interaction network.


Asunto(s)
Enfermedad de Huntington , Humanos , Enfermedad de Huntington/metabolismo , Quinasa I-kappa B/metabolismo , Cuerpos de Inclusión/metabolismo , Neuronas/metabolismo , Mapas de Interacción de Proteínas
9.
Antioxidants (Basel) ; 10(9)2021 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-34573082

RESUMEN

Neurodegenerative polyglutamine (polyQ) disorders are caused by trinucleotide repeat expansions within the coding region of disease-causing genes. PolyQ-expanded proteins undergo conformational changes leading to the formation of protein inclusions which are associated with selective neuronal degeneration. Several lines of evidence indicate that these mutant proteins are associated with oxidative stress, proteasome impairment and microglia activation. These events may correlate with the induction of inflammation in the nervous system and disease progression. Here, we review the effect of polyQ-induced oxidative stress in cellular and animal models of polyQ diseases. Furthermore, we discuss the interplay between oxidative stress, neurodegeneration and neuroinflammation using as an example the well-known neuroinflammatory disease, Multiple Sclerosis. Finally, we review some of the pharmaceutical interventions which may delay the onset and progression of polyQ disorders by targeting disease-associated mechanisms.

10.
Microorganisms ; 9(6)2021 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-34199815

RESUMEN

Recently, metal oxides and magnesium hydroxide nanoparticles (NPs) with high surface-to-volume ratios were shown to possess antibacterial properties with applications in biomedicine and agriculture. To assess recent observations from field trials on tomatoes showing resistance to pathogen attacks, porous micron-scale particles composed of nano-grains of MgO were hydrated and sprayed on the leaves of healthy tomato (Solanum lycopersicum) plants in a 20-day program. The results showed that the spray induced (a) a modest and selective stress gene response that was consistent with the absence of phytotoxicity and the production of salicylic acid as a signalling response to pathogens; (b) a shift of the phylloplane microbiota from near 100% dominance by Gram (-) bacteria, leaving extremophiles and cyanobacteria to cover the void; and (c) a response of the fungal leaf phylloplane that showed that the leaf epiphytome was unchanged but the fungal load was reduced by about 70%. The direct microbiome changes together with the low level priming of the plant's immune system may explain the previously observed resistance to pathogen assaults in field tomato plants sprayed with the same hydrated porous micron-scale particles.

11.
Genes (Basel) ; 11(10)2020 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-32992839

RESUMEN

BACKGROUND: Several experimental models of polyglutamine (polyQ) diseases have been previously developed that are useful for studying disease progression in the primarily affected central nervous system. However, there is a missing link between cellular and animal models that would indicate the molecular defects occurring in neurons and are responsible for the disease phenotype in vivo. METHODS: Here, we used a computational approach to identify dysregulated pathways shared by an in vitro and an in vivo model of ATXN1(Q82) protein aggregation, the mutant protein that causes the neurodegenerative polyQ disease spinocerebellar ataxia type-1 (SCA1). RESULTS: A set of common dysregulated pathways were identified, which were utilized to construct cerebellum-specific protein-protein interaction (PPI) networks at various time-points of protein aggregation. Analysis of a SCA1 network indicated important nodes which regulate its function and might represent potential pharmacological targets. Furthermore, a set of drugs interacting with these nodes and predicted to enter the blood-brain barrier (BBB) was identified. CONCLUSIONS: Our study points to molecular mechanisms of SCA1 linked from both cellular and animal models and suggests drugs that could be tested to determine whether they affect the aggregation of pathogenic ATXN1 and SCA1 disease progression.


Asunto(s)
Ataxina-1/metabolismo , Cerebelo/metabolismo , Redes Reguladoras de Genes , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Péptidos/metabolismo , Mapas de Interacción de Proteínas , Animales , Ataxina-1/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Péptidos/genética
12.
J Funct Biomater ; 11(3)2020 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-32947990

RESUMEN

BACKGROUND: Biocompatible materials-topography could be used for the construction of scaffolds allowing the three-dimensional (3D) organization of human stem cells into functional tissue-like structures with a defined architecture. METHODS: Structural characterization of an alumina-based substrate was performed through XRD, Brunauer-Emmett-Teller (BET) analysis, scanning electron microscopy (SEM), and wettability measurements. Biocompatibility of the substrate was assessed by measuring the proliferation and differentiation of human neural precursor stem cells (NPCs). RESULTS: α-Al2O3 is a ceramic material with crystallite size of 40 nm; its surface consists of aggregates in the range of 8-22 µm which forms a rough surface in the microscale with 1-8 µm cavities. The non-calcined material has a surface area of 5.5 m2/gr and pore size distribution of 20 nm, which is eliminated in the calcined structure. Thus, the pore network on the surface and the body of the ceramic becomes more water proof, as indicated by wettability measurements. The alumina-based substrate supported the proliferation of human NPCs and their differentiation into functional neurons. CONCLUSIONS: Our work indicates the potential use of alumina for the construction of 3D engineered biosystems utilizing human neurons. Such systems may be useful for diagnostic purposes, drug testing, or biotechnological applications.

13.
Mol Autism ; 11(1): 42, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32487215

RESUMEN

Patients diagnosed with chromosome microdeletions or duplications, known as copy number variants (CNVs), present a unique opportunity to investigate the relationship between patient genotype and cell phenotype. CNVs have high genetic penetrance and give a good correlation between gene locus and patient clinical phenotype. This is especially effective for the study of patients with neurodevelopmental disorders (NDD), including those falling within the autism spectrum disorders (ASD). A key question is whether this correlation between genetics and clinical presentation at the level of the patient can be translated to the cell phenotypes arising from the neurodevelopment of patient induced pluripotent stem cells (iPSCs).Here, we examine how iPSCs derived from ASD patients with an associated CNV inform our understanding of the genetic and biological mechanisms underlying the aetiology of ASD. We consider selection of genetically characterised patient iPSCs; use of appropriate control lines; aspects of human neurocellular biology that can capture in vitro the patient clinical phenotype; and current limitations of patient iPSC-based studies. Finally, we consider how future research may be enhanced to maximise the utility of CNV patients for research of pathological mechanisms or therapeutic targets.


Asunto(s)
Trastorno del Espectro Autista/etiología , Trastorno del Espectro Autista/metabolismo , Variaciones en el Número de Copia de ADN , Susceptibilidad a Enfermedades , Células Madre Pluripotentes Inducidas/metabolismo , Animales , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Genómica/métodos , Humanos , Trastornos del Neurodesarrollo/etiología , Trastornos del Neurodesarrollo/metabolismo , Neuronas/metabolismo , Sinapsis/metabolismo
14.
Redox Biol ; 32: 101458, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32145456

RESUMEN

Spinocerebellar ataxia type-1 (SCA1) is caused by an abnormally expanded polyglutamine (polyQ) tract in ataxin-1. These expansions are responsible for protein misfolding and self-assembly into intranuclear inclusion bodies (IIBs) that are somehow linked to neuronal death. However, owing to lack of a suitable cellular model, the downstream consequences of IIB formation are yet to be resolved. Here, we describe a nuclear protein aggregation model of pathogenic human ataxin-1 and characterize IIB effects. Using an inducible Sleeping Beauty transposon system, we overexpressed the ATXN1(Q82) gene in human mesenchymal stem cells that are resistant to the early cytotoxic effects caused by the expression of the mutant protein. We characterized the structure and the protein composition of insoluble polyQ IIBs which gradually occupy the nuclei and are responsible for the generation of reactive oxygen species. In response to their formation, our transcriptome analysis reveals a cerebellum-specific perturbed protein interaction network, primarily affecting protein synthesis. We propose that insoluble polyQ IIBs cause oxidative and nucleolar stress and affect the assembly of the ribosome by capturing or down-regulating essential components. The inducible cell system can be utilized to decipher the cellular consequences of polyQ protein aggregation. Our strategy provides a broadly applicable methodology for studying polyQ diseases.


Asunto(s)
Cuerpos de Inclusión Intranucleares , Proteínas del Tejido Nervioso , Ataxina-1/genética , Ataxina-1/metabolismo , Humanos , Cuerpos de Inclusión Intranucleares/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Estrés Oxidativo
15.
PLoS One ; 12(9): e0185498, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28949988

RESUMEN

Diabetes is a chronic disease characterized by high levels of blood glucose. Diabetic patients should normalize these levels in order to avoid short and long term clinical complications. Presently, blood glucose monitoring is dependent on frequent finger pricking and enzyme based systems that analyze the drawn blood. Continuous blood glucose monitors are already on market but suffer from technical problems, inaccuracy and short operation time. A novel approach for continuous glucose monitoring is the development of implantable cell-based biosensors that emit light signals corresponding to glucose concentrations. Such devices use genetically modified cells expressing chimeric genes with glucose binding properties. MSCs are good candidates as carrier cells, as they can be genetically engineered and expanded into large numbers. They also possess immunomodulatory properties that, by reducing local inflammation, may assist long operation time. Here, we generated a novel immortalized human MSC line co-expressing hTERT and a secreted glucose biosensor transgene using the Sleeping Beauty transposon technology. Genetically modified hMSCs retained their mesenchymal characteristics. Stable transgene expression was validated biochemically. Increased activity of hTERT was accompanied by elevated and constant level of stem cell pluripotency markers and subsequently, by MSC immortalization. Furthermore, these cells efficiently suppressed PBMC proliferation in MLR transwell assays, indicating that they possess immunomodulatory properties. Finally, biosensor protein produced by MSCs was used to quantify glucose in cell-free assays. Our results indicate that our immortalized MSCs are suitable for measuring glucose concentrations in a physiological range. Thus, they are appropriate for incorporation into a cell-based, immune-privileged, glucose-monitoring medical device.


Asunto(s)
Técnicas Biosensibles , Glucemia/metabolismo , Células Madre Mesenquimatosas/metabolismo , Línea Celular Transformada , Proliferación Celular , Humanos , Células Madre Mesenquimatosas/citología
16.
Cytotherapy ; 19(7): 808-820, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28454681

RESUMEN

BACKGROUND: Human mesenchymal stem cells (MSC) are important tools for several cell-based therapies. However, their use in such therapies requires in vitro expansion during which MSCs quickly reach replicative senescence. Replicative senescence has been linked to macromolecular damage, and especially oxidative stress-induced DNA damage. Recent studies on the other hand, have implicated telomerase in the cellular response to oxidative damage, suggesting that telomerase has a telomere-length independent function that promotes survival. METHODS: Here, we studied the DNA damage accumulation and repair during in vitro expansion as well as after acute external oxidative exposure of control MSCs and MSCs that overexpress the catalytic subunit of telomerase (hTERT MSCs). RESULTS: We showed that hTERT MSCs at high passages have a significant lower percentage of DNA lesions as compared to control cells of the same passages. Additionally, less damage was accumulated due to external oxidative insult in the nuclei of hTERT overexpressing cells as compared to the control cells. Moreover, we demonstrated that oxidative stress leads to diverse nucleus malformations, such as multillobular nuclei or donut-shaped nuclei, in the control cells whereas hTERT MSCs showed significant resistance to the formation of such defects. Finally, hTERT MSCs were found to possess higher activities of the basic antioxidant enzymes, superoxide dismutase and catalase, than control MSCs. DISCUSSION: On the basis of these results, we propose that hTERT enhancement confers resistance to genomic damage due to the amelioration of the cell's basic antioxidant machinery.


Asunto(s)
Antioxidantes/metabolismo , Daño del ADN , Células Madre Mesenquimatosas/fisiología , Estrés Oxidativo , Telomerasa/metabolismo , Catalasa/metabolismo , Células Cultivadas , Senescencia Celular/fisiología , Humanos , Peróxido de Hidrógeno/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Subunidades de Proteína , Superóxido Dismutasa/metabolismo , Telomerasa/genética , Telómero , Homeostasis del Telómero
17.
Free Radic Biol Med ; 103: 226-235, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28034832

RESUMEN

The age-associated decline of adult stem cell function contributes to the physiological failure of homeostasis during aging. The proteasome plays a key role in the maintenance of proteostasis and its failure is associated with various biological phenomena including senescence and aging. Although stem cell biology has attracted intense attention, the role of proteasome in stemness and its age-dependent deterioration remains largely unclear. By employing both Wharton's-Jelly- and Adipose-derived human adult mesenchymal stem cells (hMSCs), we reveal a significant age-related decline in proteasome content and peptidase activities, accompanied by alterations of proteasomal complexes. Additionally, we show that senescence and the concomitant failure of proteostasis negatively affects stemness. Remarkably, the loss of proliferative capacity and stemness of hMSCs can be counteracted through proteasome activation. At the mechanistic level, we demonstrate for the first time that Oct4 binds at the promoter region of ß2 and ß5 proteasome subunits and thus possibly regulates their expression. A firm understanding of the mechanisms regulating proteostasis in stem cells will pave the way to innovative stem cell-based interventions to improve healthspan and lifespan.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proliferación Celular , Células Cultivadas , Senescencia Celular , Activación Enzimática , Expresión Génica , Humanos , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo
18.
Histol Histopathol ; 32(10): 1041-1055, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28035654

RESUMEN

Mesenchymal stromal cells (MSC) have been suggested to have beneficial effects on animal models of traumatic brain injury (TBI), owing to their neurotrophic and immunomodulatory properties. Adipose tissue-derived stromal cells (ASCs) are multipotent MSC that can be harvested with minimally invasive methods, show a high proliferative capacity, low immunogenicity if allogeneic, and can be used in autologous or heterologous settings. In the present study ASCs were genetically labelled using the Sleeping Beauty transposon to express the fluorescent protein Venus. Venus+ASCs were transplanted intra-cerebroventricularly (ICV), on a rat TBI model and their survival, fate and effects on host brain responses were examined at seven days post-injury (7dPI). We provide evidence that Venus+ASCs survived, migrated into the periventricular striatum and were negative for neuronal or glial lineage differentiation markers. Venus+ASCs stimulated the proliferation of endogenous neural stem cells (NSCs) in the brain neurogenic niches, the subventricular zone (SVZ) and the hippocampal dentate gyrus (DG). It was also evident that Venus+ASCs modify the host brain's cellular microenvironment both at the injury site and at their localization area by promoting a significant reduction of the lesion area, as well as altering the post-injury, pro-inflammatory profile of microglial and astrocytic cell populations. Our data support the view that ICV transplantation of ASCs induces alterations in the host brain's cellular response to injury that may be correlated to a reversal from a detrimental to a beneficial state which is permissive for regeneration and repair.


Asunto(s)
Adipocitos/fisiología , Adipocitos/trasplante , Lesiones Traumáticas del Encéfalo/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Animales , Lesiones Traumáticas del Encéfalo/patología , Diferenciación Celular , Proliferación Celular , Microambiente Celular , Corteza Cerebral/lesiones , Corteza Cerebral/patología , Citometría de Flujo , Infusiones Intraventriculares , Masculino , Células-Madre Neurales , Neurogénesis , Ratas , Ratas Wistar , Resultado del Tratamiento
20.
Front Neurosci ; 9: 247, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26236184

RESUMEN

Polyglutamine diseases are fatal neurological disorders that affect the central nervous system. They are caused by mutations in disease genes that contain CAG trinucleotide expansions in their coding regions. These mutations are translated into expanded glutamine chains in pathological proteins. Mutant proteins induce cytotoxicity, form intranuclear aggregates and cause neuronal cell death in specific brain regions. At the moment there is no cure for these diseases and only symptomatic treatments are available. Here, we discuss novel therapeutic approaches that aim in neuronal cell replacement using induced pluripotent or adult stem cells. Additionally, we present the beneficial effect of genetically engineered mesenchymal stem cells and their use as disease models or RNAi/gene delivery vehicles. In combination with their paracrine and cell-trophic properties, such cells may prove useful for the development of novel therapies against polyglutamine diseases.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA