Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 109
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 723: 150187, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-38850809

RESUMEN

This study investigated the effects of far-infrared (FIR) irradiation on low-density lipoprotein cholesterol (LDL-C) uptake by human hepatocellular carcinoma G2 (HepG2) cells via the regulation of proprotein convertase subtilisin/kexin type 9 (PCSK9). FIR irradiation for 30 min significantly decreased PCSK9 expression (p < 0.01) in HepG2 cells. FIR irradiation substantially increased the low-density lipoprotein receptor (p < 0.0001) and LDL-C uptake (p < 0.01). Activation of transient receptor potential vanilloid (TRPV) channels mimicked the effects of FIR irradiation, significantly decreasing the protein expression of PCSK9 (p < 0.05). Conversely, inhibition of TRP channels using ruthenium red reversed the reduction in PCSK9 protein expression following FIR irradiation (p < 0.01). The specific activation of TRPV4 using 4α-PDD mimicked the effect of FIR irradiation (p < 0.01), whereas PCSK9 reduction by FIR irradiation was significantly reversed by the inhibition of TRPV4 using RN1734 (p < 0.05). These findings implied that FIR irradiation emitted from a ceramic lamp specifically increased TRPV4 activity. These findings provide insights into a novel therapeutic approach using FIR irradiation for LDL-C regulation and its implications for cardiovascular health.


Asunto(s)
LDL-Colesterol , Regulación hacia Abajo , Rayos Infrarrojos , Proproteína Convertasa 9 , Canales Catiónicos TRPV , Humanos , Proproteína Convertasa 9/metabolismo , Proproteína Convertasa 9/genética , Células Hep G2 , Canales Catiónicos TRPV/metabolismo , LDL-Colesterol/metabolismo , Regulación hacia Abajo/efectos de la radiación
2.
Muscle Nerve ; 68(2): 219-229, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37243484

RESUMEN

INTRODUCTION/AIMS: Human tonsils are a readily accessible source of stem cells for the potential treatment of skeletal muscle disorders. We reported previously that tonsil-derived mesenchymal stem cells (TMSCs) can differentiate into skeletal muscle cells (SKMCs), which renders TMSCs promising candidates for cell therapy for skeletal muscle disorders. However, the functional properties of the myocytes differentiated from mesenchymal stem cells have not been clearly evaluated. In this study we investigated whether myocytes differentiated from TMSCs (skeletal muscle cells derived from tonsil mesenchymal stem cells [TMSC-SKMCs]) exhibit the functional characteristics of SKMCs. METHODS: To test the insulin reactivity of TMSC-SKMCs, the expression of glucose transporter 4 (GLUT4) and phosphatidylinositol 3-kinase/Akt was analyzed after the cells were treated for 30 minutes with 100 nmol/L insulin in normal or high-glucose medium. We also examined whether these cells formed a neuromuscular junction (NMJ) when cocultured with motor neurons, and whether they were stimulated by electrical signals using whole-cell patch clamping. RESULTS: Skeletal muscle cells derived from tonsil mesenchymal stem cells expressed SKMC markers, such as MYOD, MYH3, MYH8, TNNI1, and TTN, at high levels, and exhibited a multinucleated cell morphology and a myotube-like shape. The expression of the acetylcholine receptor and GLUT4 was confirmed in TMSC-SKMCs. In addition, these cells exhibited insulin-mediated glucose uptake, NMJ formation, and transient changes in cell membrane action potential, all of which are representative functions of human SKMCs. DISCUSSION: Tonsil-derived mesenchymal stem cells can be functionally differentiated into SKMCs and may have potential for clinical application for the treatment of skeletal muscle disorders.


Asunto(s)
Células Madre Mesenquimatosas , Tonsila Palatina , Humanos , Diferenciación Celular/fisiología , Fibras Musculares Esqueléticas/metabolismo , Insulina , Músculo Esquelético
3.
Int J Mol Sci ; 24(4)2023 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36835347

RESUMEN

For the clinical application of mesenchymal stem cells (MSCs), the optimization of biological products (e [...].


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Diferenciación Celular
4.
Int J Mol Sci ; 23(2)2022 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-35054901

RESUMEN

Mesenchymal stem cells (MSCs) can differentiate into endoderm lineages, especially parathyroid-hormone (PTH)-releasing cells. We have previously reported that tonsil-derived MSC (T-MSC) can differentiate into PTH-releasing cells (T-MSC-PTHCs), which restored the parathyroid functions in parathyroidectomy (PTX) rats. In this study, we demonstrate quality optimization by standardizing the differentiation rate for a better clinical application of T-MSC-PTHCs to overcome donor-dependent variation of T-MSCs. Quantitation results of PTH mRNA copy number in the differentiated cells and the PTH concentration in the conditioned medium confirmed that the differentiation efficiency largely varied depending on the cells from each donor. In addition, the differentiation rate of the cells from all the donors greatly improved when differentiation was started at a high cell density (100% confluence). The large-scale expression profiling of T-MSC-PTHCs by RNA sequencing indicated that those genes involved in exiting the differentiation and the cell cycle were the major pathways for the differentiation of T-MSC-PTHCs. Furthermore, the implantation of the T-MSC-PTHCs, which were differentiated at a high cell density embedded in hyaluronic acid, resulted in a higher serum PTH in the PTX model. This standardized efficiency of differentiation into PTHC was achieved by initiating differentiation at a high cell density. Our findings provide a potential solution to overcome the limitations due to donor-dependent variation by establishing a standardized differentiation protocol for the clinical application of T-MSC therapy in treating hypoparathyroidism.


Asunto(s)
Diferenciación Celular , Células Madre Mesenquimatosas/metabolismo , Tonsila Palatina/citología , Hormona Paratiroidea/biosíntesis , Biomarcadores , Calcio/metabolismo , Técnicas de Cultivo de Célula , Células Cultivadas , Inhibición de Contacto , Espacio Extracelular/metabolismo , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Células Madre Mesenquimatosas/citología
6.
Stem Cells ; 37(10): 1252-1260, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31287931

RESUMEN

Since the discovery of stem cells and multipotency characteristics of mesenchymal stem cells (MSCs), there has been tremendous development in regenerative medicine. MSCs derived from bone marrow have been widely used in various research applications, yet there are limitations such as invasiveness of obtaining samples, low yield and proliferation rate, and questions regarding their practicality in clinical applications. Some have suggested that MSCs from other sources, specifically those derived from palatine tonsil tissues, that is, tonsil-derived MSCs (TMSCs), could be considered as a new potential therapeutic tool in regenerative medicine due to their superior proliferation rate and differentiation capabilities with low immunogenicity and ease of obtaining. Several studies have determined that TMSCs have differentiation potential not only into the mesodermal lineage but also into the endodermal as well as ectodermal lineages, expanding their potential usage and placing them as an appealing option to consider for future studies in regenerative medicine. In this review, the differentiation capacities of TMSCs and their therapeutic competencies from past studies are addressed. Stem Cells 2019;37:1252-1260.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Tonsila Palatina/metabolismo , Medicina Regenerativa/métodos , Ingeniería de Tejidos/métodos , Humanos , Tonsila Palatina/citología
7.
Cell Physiol Biochem ; 52(2): 240-253, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30816672

RESUMEN

BACKGROUND/AIMS: Far-infrared (FIR) irradiation has been reported to exhibit various biological effects including improvement of cardiovascular function. However, its effect on the differentiation of stem cells has not been studied. Using tonsil-derived mesenchymal stem cells (TMSC), we examined whether and how FIR irradiation affects adipogenic or osteogenic differentiation. METHODS: TMSC were exposed to FIR irradiation (3-25 µm wavelength) for various times (0, 30, or 60 min), and then adipogenic or osteogenic differentiation was induced for 14 days with its respective commercially available differentiation medium. At the end of the differentiation, the cells were stained using Oil red O or Alizarin red S solution, and the expression of differentiation-specific proteins was analyzed by western blotting. RESULTS: FIR irradiation did not alter cell viability or the expression of MSC-specific surface antigens (CD14, CD34, CD45, CD73, CD90, and CD105) in TMSC. However, FIR irradiation significantly inhibited adipogenic differentiation of TMSC, as evidenced by decreased Oil red O staining as well as protein expression of peroxisome proliferator-activated receptor γ and fatty acid binding protein 4. In contrast, FIR irradiation induced osteogenic differentiation, as evidenced by increased Alizarin red S staining as well as protein expression of osteocalcin and alkaline phosphatase. Treatment with heat alone did not inhibit the adipogenic differentiation of TMSC, suggesting that the inhibitory effect on adipogenic differentiation was not due to heat induced by FIR irradiation. However, heat alone did stimulate osteogenic differentiation, but to a lesser extent than FIR irradiation. Furthermore, FIR irradiation increased intracellular Ca²âº levels and the activity of protein phosphatase 2B (PP2B) in TMSC. Treatment with cyclosporin A, a specific PP2B inhibitor, reversed the inhibitory effect of FIR irradiation on adipogenic differentiation of TMSC, but had no effect on osteogenic differentiation. CONCLUSION: Our data demonstrate that FIR irradiation inhibits adipogenic differentiation but enhances osteogenic differentiation of TMSC; the inhibitory effect on adipogenic differentiation is non-thermal and mediated at least in part by activation of Ca²âº-dependent PP2B.


Asunto(s)
Adipogénesis , Calcineurina/metabolismo , Diferenciación Celular , Rayos Infrarrojos , Células Madre Mesenquimatosas/enzimología , Osteogénesis , Tonsila Palatina/enzimología , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Células Madre Mesenquimatosas/citología , Tonsila Palatina/citología
8.
Int J Mol Sci ; 20(11)2019 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-31159418

RESUMEN

Human tonsil-derived mesenchymal stem cells (T-MSCs) are newly identified MSCs and present typical features of MSCs, including having the differentiation capacity into the three germ layers and excellent proliferation capacity. They are easily sourced and are useful for stem cell therapy in various disease states. We previously reported that T-MSCs could be differentiated into skeletal myocytes and Schwann-like cells; therefore, they are a promising candidate for cell therapies for neuromuscular disease. Motor neurons (MNs), which regulate spontaneous behavior, are affected by a wide range of MN diseases (MNDs) for which there are no effective remedies. We investigated the differentiation potential of MN-like cells derived from T-MSCs (T-MSC-MNCs) for application to therapy of MNDs. After the process of MN differentiation, the expression of MN-related markers, including Islet 1, HB9/HLXB9 (HB9), and choline acetyltransferase (ChAT), was increased when compared with undifferentiated T-MSCs. The secretion of acetylcholine to the conditioned medium was significantly increased after MN differentiation. We cocultured T-MSC-MNCs and human skeletal muscle cells, and confirmed the presence of the acetylcholine receptor clusters, which demonstrated the formation of neuromuscular junctions. The potential functional improvements afforded by these T-MSC-MNCs could be useful in the treatment of MNDs caused by genetic mutation, viral infection, or environmental problems.


Asunto(s)
Diferenciación Celular , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Neuronas Motoras/citología , Neuronas Motoras/fisiología , Unión Neuromuscular/fisiología , Tonsila Palatina/citología , Acetilcolina/metabolismo , Biomarcadores , Células Cultivadas , Expresión Génica , Humanos , Inmunohistoquímica , Fibras Musculares Esqueléticas/metabolismo , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo
9.
Muscle Nerve ; 57(3): 478-486, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28796340

RESUMEN

INTRODUCTION: Mesenchymal stem cells (MSCs) can differentiate into various cell types. METHODS: In this study we investigated the potential of human tonsil-derived MSCs (T-MSCs) for neuromuscular regeneration in trembler-J (Tr-J) mice, a model for Charcot-Marie-Tooth disease type 1A (CMT1A). RESULTS: T-MSCs differentiated toward skeletal myocytes with increased expression of skeletal muscle-related markers (including troponin I type 1, and myogenin), and the formation of myotubes in vitro. In-situ transplantation of T-MSC-derived myocytes (T-MSC myocytes) into the gastrocnemius muscle in Tr-J mice enhanced motor function, with recovery of compound muscle action potential amplitudes. Morphology of the sciatic nerve and skeletal muscle recovered without the formation of teratomas, and the expression levels of nerve growth factor and glial-cell-line-derived neurotrophic factor were increased significantly in T-MSC myocytes compared with T-MSCs in vitro. DISCUSSION: Transplantation of T-MSC myocytes could enable neuromuscular regeneration in patients with CMT1A. Muscle Nerve 57: 478-486, 2018.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Músculo Esquelético/fisiopatología , Tonsila Palatina/citología , Potenciales de Acción/fisiología , Animales , Diferenciación Celular/fisiología , Enfermedad de Charcot-Marie-Tooth/fisiopatología , Modelos Animales de Enfermedad , Masculino , Ratones
10.
J Med Genet ; 54(11): 771-780, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28835480

RESUMEN

BACKGROUND: Fabry disease is characterised by the progressive accumulation of globotriaosylceramide (Gb3) and related glycosphingolipids in vascular endothelial cells. Enzyme replacement therapy (ERT) clears this accumulation. We analysed plasma proteome profiles before and after ERT to characterise its molecular pathology. METHODS: Two-dimensional electrophoresis and matrix-assisted laser desorption/ionisation-time of flight tandem mass spectrometry (MALDI-TOF MS) and tandem mass spectrometry (MS/MS) were done using plasma samples before and after ERT in eight patients with classical Fabry disease RESULTS: After short-term ERT (4-12 months), the levels of 15 plasma proteins involved in inflammation, oxidative and ischaemic injury, or complement activation were reduced significantly. Among them, ß-actin (ACTB), inactivated complement C3b (iC3b), and C4B were elevated significantly in pre-ERT Fabry disease plasma compared with control plasma. After longer-term ERT (46-96 months), iC3b levels gradually decreased, whereas the levels of other proteins varied. The gradual reduction of iC3b was comparable to that of Gb3 levels. In addition, iC3b increased significantly in pre-ERT Fabry disease mouse plasma, and C3 deposits were notable in renal tissues of pre-enzyme replacement therapy patients. CONCLUSION: These results indicated that C3-mediated complement activation might be altered in Fabry disease and ERT might promote its stabilisation.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Terapia de Reemplazo Enzimático , Enfermedad de Fabry/tratamiento farmacológico , Plasma/química , Adolescente , Adulto , Animales , Biomarcadores/sangre , Niño , Enfermedad de Fabry/enzimología , Enfermedad de Fabry/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Proteómica , Trihexosilceramidas/sangre
11.
Int J Mol Sci ; 19(8)2018 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-30110925

RESUMEN

Charcot-Marie-Tooth disease type 1A (CMT1A) is the most common inherited motor and sensory neuropathy, and is caused by duplication of PMP22, alterations of which are a characteristic feature of demyelination. The clinical phenotype of CMT1A is determined by the degree of axonal loss, and patients suffer from progressive muscle weakness and impaired sensation. Therefore, we investigated the potential of Schwann-like cells differentiated from human tonsil-derived stem cells (T-MSCs) for use in neuromuscular regeneration in trembler-J (Tr-J) mice, a model of CMT1A. After differentiation, we confirmed the increased expression of Schwann cell (SC) markers, including glial fibrillary acidic protein (GFAP), nerve growth factor receptor (NGFR), S100 calcium-binding protein B (S100B), glial cell-derived neurotrophic factor (GDNF), and brain-derived neurotrophic factor (BDNF), which suggests the differentiation of T-MSCs into SCs (T-MSC-SCs). To test their functional efficiency, the T-MSC-SCs were transplanted into the caudal thigh muscle of Tr-J mice. Recipients' improved locomotive activity on a rotarod test, and their sciatic function index, which suggests that transplanted T-MSC-SCs ameliorated demyelination and atrophy of nerve and muscle in Tr-J mice. Histological and molecular analyses showed the possibility of in situ remyelination by T-MSC-SCs transplantation. These findings demonstrate that the transplantation of heterologous T-MSC-SCs induced neuromuscular regeneration in mice and suggest they could be useful for the therapeutic treatment of patients with CMT1A disease.


Asunto(s)
Diferenciación Celular , Enfermedad de Charcot-Marie-Tooth/terapia , Células Madre Mesenquimatosas/metabolismo , Tonsila Palatina/metabolismo , Recuperación de la Función , Células de Schwann/trasplante , Animales , Enfermedad de Charcot-Marie-Tooth/genética , Enfermedad de Charcot-Marie-Tooth/metabolismo , Enfermedad de Charcot-Marie-Tooth/patología , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Masculino , Células Madre Mesenquimatosas/patología , Ratones , Ratones Mutantes , Tonsila Palatina/patología , Células de Schwann/metabolismo , Células de Schwann/patología
12.
Ann Neurol ; 79(2): 231-43, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26506222

RESUMEN

OBJECTIVE: Distal myopathy is a heterogeneous group of muscle diseases characterized by predominant distal muscle weakness. A study was done to identify the underlying cause of autosomal recessive adolescent onset distal myopathy. METHODS: Four patients from 2 unrelated Korean families were evaluated. To isolate the genetic cause, exome sequencing was performed. In vitro and in vivo assays using myoblast cells and zebrafish models were performed to examine the ADSSL1 mutation causing myopathy pathogenesis. RESULTS: Patients had an adolescent onset distal myopathy phenotype that included distal dominant weakness, facial muscle weakness, rimmed vacuoles, and mild elevation of serum creatine kinase. Exome sequencing identified completely cosegregating compound heterozygous mutations (p.D304N and p.I350fs) in ADSSL1, which encodes a muscle-specific adenylosuccinate synthase in both families. None of the controls had both mutations, and the mutation sites were located in well-conserved regions. Both the D304N and I350fs mutations in ADSSL1 led to decreased enzymatic activity. The knockdown of the Adssl1 gene significantly inhibited the proliferation of mouse myoblast cells, and the addition of human wild-type ADSSL1 reversed the reduced viability. In an adssl1 knockdown zebrafish model, muscle fibers were severely disrupted, which was evaluated by myosin expression and birefringence. In these conditions, supplementing wild-type ADSSL1 protein reversed the muscle defect. INTERPRETATION: We suggest that mutations in ADSSL1 are the novel genetic cause of the autosomal recessive adolescent onset distal myopathy. This study broadens the genetic and clinical spectrum of distal myopathy and will be useful for exact molecular diagnostics.


Asunto(s)
Adenilosuccinato Sintasa/genética , Miopatías Distales/genética , Adulto , Edad de Inicio , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Miopatías Distales/enzimología , Miopatías Distales/fisiopatología , Femenino , Humanos , Masculino , Ratones , Mutación , Linaje , Fenotipo , República de Corea , Adulto Joven , Pez Cebra , Proteínas de Pez Cebra
13.
J Peripher Nerv Syst ; 22(3): 200-207, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28544463

RESUMEN

Mutations in the NEFH gene encoding the heavy neurofilament protein are usually associated with neuronal damage and susceptibility to amyotrophic lateral sclerosis (ALS). Recently, frameshift variants in NEFH (p.Asp1004Glnfs*58 and p.Pro1008Alafs*56) have been reported to be the underlying cause of axonal Charcot-Marie-Tooth disease type 2CC (CMT2CC). The frameshift mutation resulted in a stop loss and translation of a cryptic amyloidogenic element (CAE) encoded by the 3' untranslated region (UTR). This study also identified a de novo c.3015_3027dup frameshift mutation predicting p.Lys1010Glnfs*57 in NEFH from a CMT2 family with an atypical clinical symptom of prominent proximal weakness. This mutation is located near the previously reported frameshift mutations, suggesting a mutational hotspot. Lower limb magnetic resonance imaging (MRI) revealed marked hyperintense signal changes in the thigh muscles compared with those in the calf muscles. Therefore, this study suggests that the stop loss and translational elongations by the 3' UTR of the NEFH mutations may be a relatively frequent genetic cause of axonal peripheral neuropathy with the specific characteristics of proximal dominant weakness.


Asunto(s)
Regiones no Traducidas 3'/genética , Axones/patología , Enfermedad de Charcot-Marie-Tooth/genética , Enfermedad de Charcot-Marie-Tooth/fisiopatología , Mutación del Sistema de Lectura/genética , Proteínas de Neurofilamentos/genética , Adulto , Enfermedad de Charcot-Marie-Tooth/patología , Análisis Mutacional de ADN , Femenino , Humanos , Extremidad Inferior/diagnóstico por imagen , Imagen por Resonancia Magnética , Conducción Nerviosa/genética , Linaje
14.
Hum Mutat ; 37(5): 473-80, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26786738

RESUMEN

Charcot-Marie-Tooth disease (CMT) is the most common inherited peripheral neuropathy and is a genetically and clinically heterogeneous disorder. We examined a Korean family in which two individuals had an autosomal-dominant axonal CMT with early-onset, sensory ataxia, tremor, and slow disease progression. Pedigree analysis and exome sequencing identified a de novo missense mutation (p.Y223H) in the diacylglycerol O-acyltransferase 2 (DGAT2) gene. DGAT2 encodes an endoplasmic reticulum-mitochondrial-associated membrane protein, acyl-CoA:diacylglycerol acyltransferase, which catalyzes the final step of the triglyceride (TG) biosynthesis pathway. The patient showed consistently decreased serum TG levels, and overexpression of the mutant DGAT2 significantly inhibited the proliferation of mouse motor neuron cells. Moreover, the variant form of human DGAT2 inhibited the axonal branching in the peripheral nervous system of zebrafish. We suggest that mutation of DGAT2 is the novel underlying cause of an autosomal-dominant axonal CMT2 neuropathy. This study will help provide a better understanding of the pathophysiology of axonal CMT and contribute to the molecular diagnostics of peripheral neuropathies.


Asunto(s)
Axones/patología , Enfermedad de Charcot-Marie-Tooth/genética , Diacilglicerol O-Acetiltransferasa/genética , Diacilglicerol O-Acetiltransferasa/metabolismo , Mutación Missense , Adulto , Edad de Inicio , Animales , Axones/metabolismo , Línea Celular , Proliferación Celular , Enfermedad de Charcot-Marie-Tooth/metabolismo , Enfermedad de Charcot-Marie-Tooth/patología , Niño , Predisposición Genética a la Enfermedad , Humanos , Masculino , Ratones , Neuronas Motoras/citología , Neuronas Motoras/metabolismo , Linaje , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
15.
Differentiation ; 90(1-3): 27-39, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26391447

RESUMEN

Tonsil-derived (T-) mesenchymal stem cells (MSCs) display mutilineage differentiation potential and self-renewal capacity and have potential as a banking source. Diabetes mellitus is a prevalent disease in modern society, and the transplantation of pancreatic progenitor cells or various stem cell-derived insulin-secreting cells has been suggested as a novel therapy for diabetes. The potential of T-MSCs to trans-differentiate into pancreatic progenitor cells or insulin-secreting cells has not yet been investigated. We examined the potential of human T-MSCs to trans-differentiate into pancreatic islet cells using two different methods based on ß-mercaptoethanol and insulin-transferin-selenium, respectively. First, we compared the efficacy of the two methods for inducing differentiation into insulin-producing cells. We demonstrated that the insulin-transferin-selenium method is more efficient for inducing differentiation into insulin-secreting cells regardless of the source of the MSCs. Second, we compared the differentiation potential of two different MSC types: T-MSCs and adipose-derived MSCs (A-MSCs). T-MSCs had a differentiation capacity similar to that of A-MSCs and were capable of secreting insulin in response to glucose concentration. Islet-like clusters differentiated from T-MSCs had lower synaptotagmin-3, -5, -7, and -8 levels, and consequently lower secreted insulin levels than cells differentiated from A-MSCs. These results imply that T-MSCs can differentiate into functional pancreatic islet-like cells and could provide a novel, alternative cell therapy for diabetes mellitus.


Asunto(s)
Transdiferenciación Celular , Técnicas de Reprogramación Celular , Células Secretoras de Insulina/citología , Células Madre Mesenquimatosas/citología , Tonsila Palatina/citología , Tejido Adiposo/citología , Animales , Tratamiento Basado en Trasplante de Células y Tejidos , Células Cultivadas , Diabetes Mellitus Experimental/cirugía , Humanos , Insulina/farmacología , Células Secretoras de Insulina/trasplante , Mercaptoetanol/farmacología , Células Madre Mesenquimatosas/metabolismo , Ratones , Tonsila Palatina/efectos de los fármacos , Selenio/farmacología , Sinaptotagminas/deficiencia , Transferrina/farmacología
16.
Int J Mol Sci ; 17(12)2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27983688

RESUMEN

Maternal malnutrition influences the early development of foetal adaptive changes for survival. We explored the effects of maternal undernutrition during gestation and lactation on hepatic growth and function. Sprague-Dawley rats were fed a normal or a food-restricted (FR) diet during gestation and/or lactation. We performed analyses of covariance (adjusting for the liver weight/body weight ratio) to compare hepatic growth and lipid metabolism among the offspring. Maternal FR during gestation triggered the development of wide spaces between hepatic cells and increased the expression of mammalian target of rapamycin (mTOR) in three-week-old male offspring compared with controls (both p < 0.05). Offspring nursed by FR dams exhibited wider spaces between hepatic cells and a lower liver weight/body weight ratio than control offspring, and increased mTOR expression (p < 0.05). Interestingly, the significant decrease in expression of lipogenic-related genes was dependent on carbohydrate-responsive element-binding protein, despite the increased expression of sterol regulatory element-binding protein 1 (SREBP1) (p < 0.05). This study demonstrated increased expression of key metabolic regulators (mTOR and SREBP1), alterations in lipid metabolism, and deficits in hepatic growth in the offspring of FR-treated dams.


Asunto(s)
Alimentos , Lactancia , Metabolismo de los Lípidos , Hígado/crecimiento & desarrollo , Hígado/metabolismo , Animales , Animales Recién Nacidos , Peso Corporal , Femenino , Metabolismo de los Lípidos/genética , Masculino , Tamaño de los Órganos , Embarazo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
17.
Int J Mol Sci ; 17(11)2016 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-27834852

RESUMEN

Schwann cells (SCs), which produce neurotropic factors and adhesive molecules, have been reported previously to contribute to structural support and guidance during axonal regeneration; therefore, they are potentially a crucial target in the restoration of injured nervous tissues. Autologous SC transplantation has been performed and has shown promising clinical results for treating nerve injuries and donor site morbidity, and insufficient production of the cells have been considered as a major issue. Here, we performed differentiation of tonsil-derived mesenchymal stem cells (T-MSCs) into SC-like cells (T-MSC-SCs), to evaluate T-MSC-SCs as an alternative to SCs. Using SC markers such as CAD19, GFAP, MBP, NGFR, S100B, and KROX20 during quantitative real-time PCR we detected the upregulation of NGFR, S100B, and KROX20 and the downregulation of CAD19 and MBP at the fully differentiated stage. Furthermore, we found myelination of axons when differentiated SCs were cocultured with mouse dorsal root ganglion neurons. The application of T-MSC-SCs to a mouse model of sciatic nerve injury produced marked improvements in gait and promoted regeneration of damaged nerves. Thus, the transplantation of human T-MSCs might be suitable for assisting in peripheral nerve regeneration.


Asunto(s)
Células Madre Mesenquimatosas/citología , Regeneración Nerviosa/fisiología , Traumatismos de los Nervios Periféricos/rehabilitación , Células de Schwann/citología , Nervio Ciático/lesiones , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Niño , Técnicas de Cocultivo , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Expresión Génica , Humanos , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuronas/metabolismo , Tonsila Palatina/citología , Tonsila Palatina/metabolismo , Tonsila Palatina/cirugía , Traumatismos de los Nervios Periféricos/metabolismo , Traumatismos de los Nervios Periféricos/patología , Traumatismos de los Nervios Periféricos/cirugía , Recuperación de la Función , Células de Schwann/metabolismo , Células de Schwann/trasplante , Nervio Ciático/metabolismo , Tonsilectomía , Trasplante Heterólogo
18.
J Cell Physiol ; 230(1): 140-9, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24909560

RESUMEN

CCN1 is highly expressed in cancer cells and has been identified in the secretome of bone marrow-derived mesenchymal stem cells (BM-MSC). Although secreted CCN1 is known to promote angiogenesis, its underlying mechanism remains unclear. Here, we examined whether our recently-established tonsil-derived MSC (T-MSC) secrete CCN1 and, if any, how CCN1 promotes the angiogenesis of human umbilical vein endothelial cells (HUVEC). Compared with untreated control T-MSC, a higher level of CCN1 was secreted by T-MSC treated with activin A and sonic hedgehog, drugs known to induce endodermal differentiation. Expectedly, conditioned medium collected from differentiated T-MSC (DCM) significantly increased HUVEC migration and tube formation compared with that from control T-MSC (CCM), and these stimulatory effects were reversed by neutralization with anti-CCN1 antibody. Treatment with recombinant human CCN1 (rh-CCN1) alone also mimicked the stimulatory effects of DCM. Furthermore, treatment with either DCM or rh-CCN1 increased the phosphorylation of AMP kinase (AMPK), and ectopic expression of siRNA of the AMPK gene inhibited all observed effects of both DCM and rh-CCN1. However, no alteration of intracellular ATP levels or phosphorylation of LKB1, a well-known upstream factor of AMPK activation, was observed under our conditions. Finally, the neutralization of integrin α(v) ß(3) with anti-integrin α(v) ß(3) antibody almost completely reversed the effects of CCN1 on AMPK phosphorylation, and EC migration and tube formation. Taken together, we demonstrated that T-MSC increase the secretion of CCN1 in response to endodermal differentiation and that integrin α(v) ß(3) and AMPK mediate CCN1-induced EC migration and tube formation independent of intracellular ATP levels alteration.


Asunto(s)
Proteína 61 Rica en Cisteína/metabolismo , Células Endoteliales de la Vena Umbilical Humana/citología , Células Madre Mesenquimatosas/metabolismo , Neovascularización Fisiológica , Tonsila Palatina/citología , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Activinas/farmacología , Anticuerpos/inmunología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Proteína 61 Rica en Cisteína/inmunología , Proteína 61 Rica en Cisteína/farmacología , Células Endoteliales/metabolismo , Proteínas Hedgehog/farmacología , Humanos , Integrina alfaVbeta3/inmunología , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Cordón Umbilical/citología
19.
J Biomed Sci ; 22: 43, 2015 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-26141737

RESUMEN

BACKGROUND: Mutations in heat shock 27 kDa protein 1 (HSP27 or HSPB1) cause distal hereditary motor neuropathy (dHMN) or Charcot-Marie-Tooth disease type 2 F (CMT2F) according to unknown factors. Mutant HSP27 proteins affect axonal transport by reducing acetylated tubulin. RESULTS: We generated a transgenic mouse model overexpressing HSP27-S135F mutant protein driven by Cytomegalovirus (CMV) immediate early promoter. The mouse phenotype was similar to dHMN patients in that they exhibit motor neuropathy. To determine the phenotypic aberration of transgenic mice, behavior test, magnetic resonance imaging (MRI), electrophysiological study, and pathology were performed. Rotarod test showed that founder mice exhibited lowered motor performance. MRI also revealed marked fatty infiltration in the anterior and posterior compartments at calf level. Electrophysiologically, compound muscle action potential (CMAP) but not motor nerve conduction velocity (MNCV) was reduced in the transgenic mice. Toluidine staining with semi-thin section of sciatic nerve showed the ratio of large myelinated axon fiber was reduced, which might cause reduced locomotion in the transgenic mice. Electron microscopy also revealed abundant aberrant myelination. Immunohistochemically, neuronal dysfunctions included elevated level of phosphorylated neurofilament and reduced level of acetylated tubulin in the sural nerve of transgenic mice. There was no additional phenotype besides motor neuronal defects. CONCLUSIONS: Overexpression of HSP27-S135F protein causes peripheral neuropathy. The mouse model can be applied to future development of therapeutic strategies for dHMN or CMT2F.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/genética , Proteínas de Choque Térmico/biosíntesis , Atrofia Muscular Espinal/genética , Proteínas de Neoplasias/biosíntesis , Enfermedades del Sistema Nervioso Periférico/genética , Animales , Enfermedad de Charcot-Marie-Tooth/fisiopatología , Modelos Animales de Enfermedad , Proteínas de Choque Térmico/genética , Humanos , Ratones , Ratones Transgénicos , Chaperonas Moleculares , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Atrofia Muscular Espinal/patología , Mutación , Proteínas de Neoplasias/genética , Enfermedades del Sistema Nervioso Periférico/fisiopatología
20.
Arterioscler Thromb Vasc Biol ; 34(1): 81-9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24158513

RESUMEN

OBJECTIVE: Globotriaosylceramide (Gb3) induces KCa3.1 downregulation in Fabry disease (FD). We investigated whether Gb3 induces KCa3.1 endocytosis and degradation. APPROACH AND RESULTS: KCa3.1, especially plasma membrane-localized KCa3.1, was downregulated in both Gb3-treated mouse aortic endothelial cells (MAECs) and human umbilical vein endothelial cells. Gb3-induced KCa3.1 downregulation was prevented by lysosomal inhibitors but not by a proteosomal inhibitor. Endoplasmic reticulum stress-inducing agents did not induce KCa3.1 downregulation. Gb3 upregulated the protein levels of early endosome antigen 1 and lysosomal-associated membrane protein 2 in MAECs. Compared with MAECs from age-matched wild-type mice, those from aged α-galactosidase A (Gla)-knockout mice, an animal model of FD, showed downregulated KCa3.1 expression and upregulated early endosome antigen 1 and lysosomal-associated membrane protein 2 expression. In contrast, no significant difference was found in early endosome antigen 1 and lysosomal-associated membrane protein 2 expression between young Gla-knockout and wild-type MAECs. In aged Gla-knockout MAECs, clathrin was translocated close to the cell border and clathrin knockdown recovered KCa3.1 expression. Rab5, an effector of early endosome antigen 1, was upregulated, and Rab5 knockdown restored KCa3.1 expression, the current, and endothelium-dependent relaxation. CONCLUSIONS: -Gb3 accelerates the endocytosis and lysosomal degradation of endothelial KCa3.1 via a clathrin-dependent process, leading to endothelial dysfunction in FD.


Asunto(s)
Células Endoteliales/enzimología , Endotelio Vascular/enzimología , Enfermedad de Fabry/enzimología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Lisosomas/metabolismo , Trihexosilceramidas/metabolismo , Animales , Células Cultivadas , Clatrina/genética , Clatrina/metabolismo , Modelos Animales de Enfermedad , Endocitosis , Endotelio Vascular/fisiopatología , Enfermedad de Fabry/genética , Enfermedad de Fabry/fisiopatología , Células Endoteliales de la Vena Umbilical Humana/enzimología , Humanos , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Ratones , Ratones Noqueados , Transporte de Proteínas , Proteolisis , Interferencia de ARN , Transfección , Vasodilatación , Proteínas de Transporte Vesicular/metabolismo , alfa-Galactosidasa/genética , alfa-Galactosidasa/metabolismo , Proteínas de Unión al GTP rab5/genética , Proteínas de Unión al GTP rab5/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA