Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Craniofac Surg ; 2024 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-39038207

RESUMEN

This study examined the biocompatibility and expansion volume of tissue expanders utilizing rabbits and beagles as experimental models. The self-inflatable expander was provided using a Tissue balloon® (Neobiotech Co., Seoul, Korea). In 9 adult rabbits, a Tissue balloon® was placed under the lateral mandibular periosteum via an extraoral approach. After 2, 3, and 4 weeks (n=3), the expanders were removed, and soft tissue and bone samples were collected for analysis. Histomorphometric analysis and measurements of expander characteristics were performed. In 5 Beagles, all premolars were extracted. Three months after extraction, tissue expanders were placed in the maxilla and mandible, adjacent to dental extraction sites, and removed after 3 weeks. Gingival parameters were measured before and after expansion. Blood perfusion in the oral mucosa was assessed using a laser Doppler flowmeter at different time points. After three weeks, dogs were euthanized, and tissue samples were collected for histologic analysis, focusing on inflammatory response, bone formation, and gingival thickness changes. In the rabbit study, measurements of size, weight, and volume showed significant increases over 4 weeks. Swelling peaked at 2 weeks postimplantation and then gradually stabilized. Histologic examination revealed no signs of inflammation, and expanders were covered by collagen-rich capsules. Some bone resorption was noted due to pressure from the expanders, but the resorption was not significant. In the beagle study, twenty tissue expanders were implanted; 1 expander was lost, and 9 wounds dehisced during soft tissue healing. However, no signs of infection were noted. Histologic evaluation showed bone atrophy, attributed to swelling pressure, at the recipient site. Gingival thickness was not decreased. Perfusion measurements indicated that expansion did not affect microvascular circulation adversely. Overall, both studies suggest that tissue expanders demonstrate biocompatibility and successful expansion volume in vivo, with minimal adverse effects on surrounding tissues and microvascular circulation.

2.
Cereb Cortex ; 31(12): 5598-5612, 2021 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-34171088

RESUMEN

Early-life exposure to bisphenol A (BPA), synthetic compound used in polycarbonate plastic, is associated with altered cognitive and emotional behavior later in life. However, the brain mechanism underlying the behavioral deficits is unknown. Here, we show that maternal BPA exposure disrupted self-renewal and differentiation of neural progenitors during cortical development. The BPA exposure reduced the neuron number, whereas it increased glial cells in the cerebral cortex. Also, synaptic formation and transmission in the cerebral cortex were suppressed after maternal BPA exposure. These changes appeared to be associated with autophagy as a gene ontology analysis of RNA-seq identified an autophagy domain in the BPA condition. Mouse behavioral tests revealed that maternal BPA caused hyperactivity and social deficits in adult offspring. Together, these results suggest that maternal BPA exposure leads to abnormal cortical architecture and function likely by activating autophagy.


Asunto(s)
Compuestos de Bencidrilo , Efectos Tardíos de la Exposición Prenatal , Animales , Compuestos de Bencidrilo/toxicidad , Corteza Cerebral , Ratones , Neurogénesis , Fenoles/toxicidad , Efectos Tardíos de la Exposición Prenatal/inducido químicamente
3.
Toxicol Appl Pharmacol ; 428: 115696, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34419494

RESUMEN

Bisphenol A (BPA) is a well-known endocrine-disrupting chemical that is widely used in a variety of products, including plastics, medical equipment and receipts. Hence, most people are exposed to BPA through the skin, via inhalation and via the digestive system, and such exposure has been linked to cardiovascular diseases including coronary artery disease, hypertension, atherosclerosis, and myocardial infarction. However, the underlying mechanisms of cardiac dysfunction caused by BPA remain poorly understood. In this study, we found that BPA exposure altered cardiac function in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Acute BPA exposure in hiPSC-CMs resulted in reduced field potential, as measured by multielectrode array (MEA). Furthermore, we observed that BPA dose-dependently inhibited ICa, INa or IKr channels. In addition, BPA exposure dose-dependently inhibited calcium transients and contraction in hiPSC-CMs. Our findings suggest that BPA exposure leads to cardiac dysfunction and cardiac risk factors such as arrhythmia.


Asunto(s)
Contaminantes Ocupacionales del Aire/toxicidad , Compuestos de Bencidrilo/toxicidad , Cardiotoxinas/toxicidad , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Fenoles/toxicidad , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología
4.
Int J Mol Sci ; 22(23)2021 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-34884818

RESUMEN

Alzheimer's disease (AD), an age-dependent, progressive neurodegenerative disorder, is the most common type of dementia, accounting for 50-70% of all dementia cases. Due to the increasing incidence and corresponding socioeconomic burden of dementia, it has rapidly emerged as a challenge to public health worldwide. The characteristics of AD include the development of extracellular amyloid-beta plaques and intracellular neurofibrillary tangles, vascular changes, neuronal inflammation, and progressive brain atrophy. However, the complexity of the biology of AD has hindered progress in elucidating the underlying pathophysiological mechanisms of AD, and the development of effective treatments. MicroRNAs (miRNAs, which are endogenous, noncoding RNAs of approximately 22 nucleotides that function as posttranscriptional regulators of various genes) are attracting attention as powerful tools for studying the mechanisms of diseases, as they are involved in several biological processes and diseases, including AD. AD is a multifactorial disease, and several reports have suggested that miRNAs play an important role in the pathological processes of AD. In this review, the basic biology of miRNAs is described, and the function and physiology of miRNAs in the pathological processes of AD are highlighted. In addition, the limitations of current pharmaceutical therapies for the treatment of AD and the development of miRNA-based next-generation therapies are discussed.


Asunto(s)
Enfermedad de Alzheimer/genética , MicroARNs/metabolismo , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Biomarcadores/metabolismo , Inhibidores de la Colinesterasa/uso terapéutico , Humanos , MicroARNs/uso terapéutico , ARN Mensajero/metabolismo , Sinapsis/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
5.
Int J Mol Sci ; 22(15)2021 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-34360711

RESUMEN

The acute demise of stem cells following transplantation significantly compromises the efficacy of stem cell-based cell therapeutics for infarcted hearts. As the stem cells transplanted into the damaged heart are readily exposed to the hostile environment, it can be assumed that the acute death of the transplanted stem cells is also inflicted by the same environmental cues that caused massive death of the host cardiac cells. Pyroptosis, a highly inflammatory form of programmed cell death, has been added to the list of important cell death mechanisms in the damaged heart. However, unlike the well-established cell death mechanisms such as necrosis or apoptosis, the exact role and significance of pyroptosis in the acute death of transplanted stem cells have not been explored in depth. In the present study, we found that M1 macrophages mediate the pyroptosis in the ischemia/reperfusion (I/R) injured hearts and identified miRNA-762 as an important regulator of interleukin 1ß production and subsequent pyroptosis. Delivery of exogenous miRNA-762 prior to transplantation significantly increased the post-transplant survival of stem cells and also significantly ameliorated cardiac fibrosis and heart functions following I/R injury. Our data strongly suggest that suppressing pyroptosis can be an effective adjuvant strategy to enhance the efficacy of stem cell-based therapeutics for diseased hearts.


Asunto(s)
MicroARNs , Daño por Reperfusión Miocárdica , Piroptosis , Trasplante de Células Madre , Células Madre , Animales , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , MicroARNs/genética , MicroARNs/farmacología , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/terapia , Piroptosis/efectos de los fármacos , Piroptosis/genética , Células RAW 264.7 , Ratas , Ratas Sprague-Dawley , Células Madre/metabolismo , Células Madre/patología
6.
Int J Mol Sci ; 21(23)2020 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-33265983

RESUMEN

Depression is a serious medical illness that is one of the most prevalent psychiatric disorders. Corticosterone (CORT) increases depression-like behavior, with some effects on anxiety-like behavior. 2-Phenethylamine (PEA) is a monoamine alkaloid that acts as a central nervous system stimulant in humans. Here, we show that PEA exerts antidepressant effects by modulating the Brain-derived neurotrophic factor (BDNF)/tropomyosin receptor kinase B (TrkB)/cAMP response element binding protein (CREB) signaling pathway in CORT-induced depression. To investigate the potential effects of PEA on CORT-induced depression, we first treated CORT (50 µM)-induced hippocampal neurons with 100 µM PEA for 24 h. We found that treatment with CORT altered dendritic spine architecture; however, treatment with PEA rescued dendritic spine formation via regulation of BDNF/TrkB/CREB signaling. Next, we used a mouse model of CORT-induced depression. Mice were treated with CORT (20 mg/kg) for 21 days, followed by assessments of a battery of depression-like behaviors. During the final four days of CORT exposure, the mice were treated with PEA (50 mg/kg). We found that CORT injection promoted depression-like behavior and significantly decreased BDNF and TrkB expression in the hippocampus. However, treatment with PEA significantly ameliorated the behavioral and biochemical changes induced by CORT. Our findings reveal that PEA exerts antidepressant effects by modulating the BDNF/TrkB/CREB signaling pathway in a mouse model of CORT-induced depression.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Depresión/inducido químicamente , Depresión/metabolismo , Fenetilaminas/farmacología , Receptor trkB/metabolismo , Transducción de Señal , Animales , Conducta Animal/efectos de los fármacos , Corticosterona , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/metabolismo , Espinas Dendríticas/patología , Hipocampo/patología , Ratones Endogámicos C57BL , Modelos Biológicos , Fenotipo , Sinapsis/efectos de los fármacos
7.
Molecules ; 25(22)2020 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-33198253

RESUMEN

Pathological cardiac hypertrophy is characterized by an abnormal increase in cardiac muscle mass in the left ventricle, resulting in cardiac dysfunction. Although various therapeutic approaches are being continuously developed for heart failure, several studies have suggested natural compounds as novel potential strategies. Considering relevant compounds, we investigated a new role for Pterosin B for which the potential life-affecting biological and therapeutic effects on cardiomyocyte hypertrophy are not fully known. Thus, we investigated whether Pterosin B can regulate cardiomyocyte hypertrophy induced by angiotensin II (Ang II) using H9c2 cells. The antihypertrophic effect of Pterosin B was evaluated, and the results showed that it reduced hypertrophy-related gene expression, cell size, and protein synthesis. In addition, upon Ang II stimulation, Pterosin B attenuated the activation and expression of major receptors, Ang II type 1 receptor and a receptor for advanced glycation end products, by inhibiting the phosphorylation of PKC-ERK-NF-κB pathway signaling molecules. In addition, Pterosin B showed the ability to reduce excessive intracellular reactive oxygen species, critical mediators for cardiac hypertrophy upon Ang II exposure, by regulating the expression levels of NAD(P)H oxidase 2/4. Our results demonstrate the protective role of Pterosin B in cardiomyocyte hypertrophy, suggesting it is a potential therapeutic candidate.


Asunto(s)
Angiotensina II/química , Hipertrofia/tratamiento farmacológico , Indanos/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Animales , Línea Celular , Supervivencia Celular , Citosol/metabolismo , Proteína HMGB1/metabolismo , Corazón/efectos de los fármacos , FN-kappa B/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Transducción de Señal
8.
Biochem Biophys Res Commun ; 499(2): 368-373, 2018 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-29577900

RESUMEN

A previous study showed that small G protein signaling modulator 3 (SGSM3) was highly correlated with Cx43 in heart functions and that high levels of SGSM3 may induce Cx43 turnover through lysosomal degradation in infarcted rat hearts. Here, we investigated the protective effects of kenpaullone on cardiomyocytes following H2O2-induced oxidative stress mediated by the interaction of SGSM3 with Cx43. We found that the gap junction protein Cx43 was significantly down-regulated in an H2O2 concentration-dependent manner, whereas expression of SGSM3 was up-regulated upon H2O2 exposure in H9c2 cells. The effect of kenpaullone pretreatment on H2O2-induced cytotoxicity was evaluated in H9c2 cells. H2O2 markedly increased the release of lactate dehydrogenase (LDH), while kenpaullone pretreatment suppressed LDH release in H9c2 cells. Moreover, kenpaullone pretreatment significantly reduced ROS fluorescence intensity and significantly down-regulated the level of apoptosis-activating genes (cleaved caspase-3, cleaved caspase-9 and cytochrome C), autophagy markers (LC3A/B), and the Cx43-interacting partner SGSM3. These results suggest that kenpaullone plays a role in protecting cardiomyocytes from oxidative stress and that the turnover of Cx43 through SGSM3-induced lysosomal degradation underlies the anti-apoptotic effect of kenpaullone.


Asunto(s)
Benzazepinas/farmacología , Cardiotónicos/farmacología , Conexina 43/metabolismo , Peróxido de Hidrógeno/toxicidad , Indoles/farmacología , Proteínas de Unión al GTP Monoméricas/metabolismo , Miocitos Cardíacos/patología , Estrés Oxidativo/efectos de los fármacos , Proteolisis/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Biomarcadores/metabolismo , Línea Celular , Técnicas de Silenciamiento del Gen , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Unión Proteica/efectos de los fármacos , Ratas
9.
Mol Cell Biochem ; 439(1-2): 105-115, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28795305

RESUMEN

During the past decade, microRNAs have continuously been suggested as a promising therapeutic tool due to their beneficial effects, such as their multi-targets and multi-functions in pathologic conditions. As a pathologic phenotype is generally regulated by multiple signaling pathways, in this study we identified a microRNA regulating multiple target genes within cardiac hypertrophic signaling pathways. microRNA-133a is known to play a crucial role in cardiac hypertrophy. However, the role of microRNA-133a, which may regulate several signaling pathways in norepinephrine-induced cardiac hypertrophy via multi-targeting, has not been investigated. In the current study, we showed that microRNA-133a can protect cardiomyocyte hypertrophy against norepinephrine stimulation in neonatal rat ventricular cardiomyocytes via new targets, PKCδ and Gq, all of which are related to downstream signaling pathways of the α1-adrenergic receptor. Taken together, these results suggest the advantages of the therapeutic use of microRNAs as an effective potential drug regulating multiple signaling pathways under pathologic conditions.


Asunto(s)
Cardiomegalia/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , MicroARNs/metabolismo , Miocitos Cardíacos/metabolismo , Proteína Quinasa C-delta/metabolismo , Transducción de Señal , Animales , Cardiomegalia/patología , Miocitos Cardíacos/patología , Ratas , Receptor de Adenosina A1/metabolismo
10.
Biol Pharm Bull ; 41(12): 1748-1756, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30504677

RESUMEN

Alternative medicines attract attention because stroke is rarely expected to make a full recovery with the most advanced medical technology. Angelica gigas (AG) is a well-known herbal medicine as a neuroprotective agent. The present study introduced mesenchymal stem cells (MSCs) to identify for the advanced treatment of the cerebrovascular disease. The objective of this research is validation of the enhanced effects of multiple combined treatment of AG extract with MSCs on stroke through angiogenesis. Our results confirmed that AG extract with MSCs improved the neovascularization increasing expression of angiogenesis-regulated molecules. The changes of brain and the behavioral ability showed the increased effects of AG extract with MSCs. As a result, AG extract and MSCs may synergistically increase the therapeutic potential by enhancing neovascularization. This mixed approach provides a new experimental protocol of herbal medicine therapy for the treatment of a variety of diseases including stroke, trauma, and spinal cord injury.


Asunto(s)
Angelica/química , Medicamentos Herbarios Chinos/uso terapéutico , Trasplante de Células Madre Mesenquimatosas , Fármacos Neuroprotectores/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Conducta Animal/efectos de los fármacos , Terapia Combinada , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/aislamiento & purificación , Masculino , Fármacos Neuroprotectores/aislamiento & purificación , Raíces de Plantas/química , Ratas Sprague-Dawley , Accidente Cerebrovascular/terapia , Resultado del Tratamiento
11.
Biol Res ; 51(1): 41, 2018 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-30384862

RESUMEN

BACKGROUND: Osteoarthritis (OA) can be defined as degradation of articular cartilage of the joint, and is the most common degenerative disease. To regenerate the damaged cartilage, different experimental approaches including stem cell therapy have been tried. One of the major limitations of stem cell therapy is the poor post-transplantation survival of the stem cells. Anoikis, where insufficient matrix support and adhesion to extracellular matrix causes apoptotic cell death, is one of the main causes of the low post-transplantation survival rate of stem cells. Therefore, enhancing the initial interaction of the transplanted stem cells with chondrocytes could improve the therapeutic efficacy of stem cell therapy for OA. Previously, protein kinase C activator phorbol 12-myristate 13-acetate (PMA)-induced increase of mesenchymal stem cell adhesion via activation of focal adhesion kinase (FAK) has been reported. In the present study, we examine the effect PMA on the adipose-derived stem cells (ADSCs) adhesion and spreading to culture substrates, and further on the initial interaction between ADSC and chondrocytes. RESULTS: PMA treatment increased the initial adhesion of ADSC to culture substrate and cellular spreading with increased expression of adhesion molecules, such as FAK, vinculin, talin, and paxillin, at both RNA and protein level. Priming of ADSC with PMA increased the number of ADSCs attached to confluent layer of cultured chondrocytes compared to that of untreated ADSCs at early time point (4 h after seeding). CONCLUSION: Taken together, the results of this study suggest that priming ADSCs with PMA can increase the initial interaction with chondrocytes, and this proof of concept can be used to develop a non-invasive therapeutic approach for treating OA. It may also accelerate the regeneration process so that it can relieve the accompanied pain faster in OA patients. Further in vivo studies examining the therapeutic effect of PMA pretreatment of ADSCs for articular cartilage damage are required.


Asunto(s)
Cartílago Articular/citología , Condrocitos/citología , Proteína Quinasa C/farmacología , Células Madre/efectos de los fármacos , Western Blotting , Adhesión Celular , Comunicación Celular , Técnicas de Cultivo de Célula , Diferenciación Celular , Supervivencia Celular , Condrocitos/efectos de los fármacos , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Cell Physiol Biochem ; 44(1): 53-65, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29131029

RESUMEN

BACKGROUND/AIMS: Stromal vascular fraction (SVF) cells are a mixed cell population, and their regenerative capacity has been validated in various therapeutic models. The purpose of this study was to investigate the regenerative mechanisms utilized by implanted SVF cells. Using an in vitro co-culture system, we sought to determine whether SVF implantation into impaired tissue affects endogenous mesenchymal stem cell (MSC) differentiation; MSCs can differentiate into a variety of cell types, and they have a strong regenerative capacity despite their low numbers in impaired tissue. METHODS: Adipose-derived SVF cells obtained from four donors were co-cultured with bone marrow-derived MSCs, and the differential expression of osteogenic markers and osteogenic differentiation inducers over time was analyzed in mono-cultured MSCs and MSCs co-cultured with SVF cells. RESULTS: The co-cultivation of MSCs with SVF cells significantly and mutually induced the expression of osteogenic-specific markers via paracrine and/or autocrine regulation but did not induce adipocyte, chondrocyte or myoblast marker expression. More surprisingly, subsequent osteogenesis and/or comparable effects were rapidly induced within 48 h. CONCLUSION: To the best of our knowledge, this is the first study in which osteogenesis and/or comparable effects were rapidly induced in bone marrow-derived MSCs and adipose-derived SVF cells through co-cultivation. Our findings suggest that the positive effects of SVF implantation into impaired bone may be attributed to the rapid induction of MSC osteogenesis, and the transplantation of co-cultured and preconditioned SVF cells and/or MSCs may be more effective than the transplantation of untreated cells for the treatment of bone defects.


Asunto(s)
Células Madre Mesenquimatosas/citología , Células del Estroma/citología , Tejido Adiposo/citología , Adulto , Células de la Médula Ósea/citología , Diferenciación Celular , Células Cultivadas , Técnicas de Cocultivo , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Humanos , Masculino , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Células Madre Mesenquimatosas/metabolismo , Persona de Mediana Edad , Osteocalcina/genética , Osteocalcina/metabolismo , Osteogénesis , Osteopontina/genética , Osteopontina/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Células del Estroma/metabolismo
13.
Biochem Biophys Res Commun ; 491(2): 429-435, 2017 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-28716730

RESUMEN

Connexin 43 (Cx43), a ubiquitous connexin expressed in the heart and skin, is associated with a variety of hereditary conditions. Therefore, the characterization of Cx43-interacting proteins and their dynamics is important to understand not only the molecular mechanisms underlying pathological malfunction of gap junction-mediated intercellular communication but also to identify novel and unanticipated biological functions of Cx43. In the present study, we observed potential targets of Cx43 to determine new molecular functions in cardio-protection. MALDI-TOF mass spectrometry analysis of Cx43 co-immunoprecipitated proteins showed that Cx43 interacts with several proteins related to metabolism. In GeneMANIA network analysis, SGSM3, which has not been previously associated with Cx43, was highly correlated with Cx43 in heart functions, and high levels of SGSM3 appeared to induce the turnover of Cx43 through lysosomal degradation in myocardial infarcted rat hearts. Moreover, we confirmed that lysosomal degradation of Cx43 is dependent upon the interaction between SGSM3 and Cx43 in H9c2 cardiomyocytes. The functional importance of the interaction between SGSM3 and Cx43 was confirmed by results showing that Cx43 expression was enhanced by SGSM3 siRNA knockdown in H9c2 cells. In summary, the results of this study elucidate the molecular mechanisms in which Cx43 with SGSM3 is degraded in myocardial infarcted rat hearts, which may contribute to the establishment of new therapeutic targets to modulate cardiac function in physiological and pathological conditions.


Asunto(s)
Conexina 43/metabolismo , Uniones Comunicantes/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Comunicación Celular , Línea Celular , Conexina 43/genética , Vasos Coronarios/patología , Vasos Coronarios/cirugía , Uniones Comunicantes/patología , Uniones Comunicantes/ultraestructura , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ligadura , Lisosomas/metabolismo , Masculino , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Proteínas de Unión al GTP Monoméricas/genética , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Miocardio/patología , Miocitos Cardíacos/patología , Miocitos Cardíacos/ultraestructura , Unión Proteica , Mapeo de Interacción de Proteínas , Proteolisis , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Proteína de la Zonula Occludens-1/genética , Proteína de la Zonula Occludens-1/metabolismo
14.
J Vasc Res ; 54(2): 100-108, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28407626

RESUMEN

Adult stem cells have been studied as a promising therapeutic modality for the functional restoration of the damaged heart. In the present study, a strategy for enhancing the angiogenic efficacy of human mesenchymal stem cells (hMSCs) using micro-RNA was examined. We investigated whether micro-RNA-146a (miR-146a) influences the secretion of vascular endothelial growth factor (VEGF) and angiogenesis of MSCs. Our data indicated that miR-146a-transfected hMSCs (hMSCmiR-146a) decreased the expression of neurofibromin 2, an inhibitor of p21-activated kinase-1 (PAK1). miR-146a also increased the expression of Ras-related C3 botulinum toxin substrate 1 and PAK1, which are known to induce VEGF expression, and the formation of vascular branches was increased in hMSCmiR-146a compared to hMSCs treated with VEGF. VEGF and p-Akt were increased in hMSCmiR-146a. Furthermore, injection of hMSCmiR-146a after ischemia/reperfusion (I/R) injury led to a reduction of fibrosis area and increased VEGF expression, confirming the regenerative capacity such as reparative angiogenesis in the infarcted area. Cardiac functions in I/R injury were improved following injection of hMSCmiR-146a compared to the I/R group. Taken together, these data suggest that miR-146 is a novel microRNA that regulates VEGF expression, and its use may be an effective strategy for enhancing the therapeutic efficacy of hMSC transplantation into the I/R-injured heart.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , MicroARNs/metabolismo , Infarto del Miocardio/cirugía , Daño por Reperfusión Miocárdica/cirugía , Miocardio/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Regiones no Traducidas 3' , Animales , Sitios de Unión , Células Cultivadas , Modelos Animales de Enfermedad , Fibrosis , Humanos , Masculino , MicroARNs/genética , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocardio/patología , Neovascularización Fisiológica , Neurofibromina 2/genética , Neurofibromina 2/metabolismo , Ratas Sprague-Dawley , Recuperación de la Función , Regeneración , Transducción de Señal , Transfección , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/genética , Quinasas p21 Activadas/metabolismo , Proteínas de Unión al GTP rac/metabolismo
15.
Mol Cell Biochem ; 427(1-2): 177-185, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28039611

RESUMEN

Gliomas are the most common type of malignant primary brain tumors. Some treatments of gliomas exist, but they are rarely curative. Mesenchymal stem cells (MSCs) are emerging as potential modes of targeted cancer therapy owing to their capacity for homing toward tumor sites. It has been proposed that MSCs derived from various sources, such as bone marrow, adipose tissue and umbilical cord blood, can be used as cell-based therapy for brain tumors. Here, MSCs obtained from the synovial fluid of osteoarthritis or rheumatoid arthritis patients were investigated as therapeutic candidates. Specifically, we compared migratory and adhesive abilities, as well as expression levels of related genes and microRNA in bone marrow derived-MSCs (BMMSCs), adipose derived-MSCs (ADMSCs), and synovial fluid derived-MSCs (SFMSCs) after treatment with conditioned medium from gliomas. Migration and adhesion of SFMSCs increased through upregulation of the activated lymphocyte cell adhesion molecule (ALCAM) and N-cadherin by microRNA-192 and -218 downregulation, similar to BMMSCs and ADMSCs. Migratory capacities of all types of MSCs were evaluated in vivo, and SFMSCs migrated intensively toward gliomas. These results suggest that SFMSCs have potential for use in cell-based antitumor therapies.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Movimiento Celular , Proteínas Fetales/metabolismo , Células Madre Mesenquimatosas/metabolismo , MicroARNs/biosíntesis , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/terapia , Línea Celular Tumoral , Femenino , Glioma/metabolismo , Glioma/terapia , Humanos , Masculino
16.
Biol Res ; 50(1): 1, 2017 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-28100269

RESUMEN

BACKGROUND: Pathologic vascular smooth muscle cell (VSMC) proliferation and migration after vascular injury promotes the development of occlusive vascular disease. Therefore, an effective chemical agent to suppress aberrant proliferation and migration of VSMCs can be a potential therapeutic modality for occlusive vascular disease such as atherosclerosis and restenosis. To find an anti-proliferative chemical agent for VSMCs, we screened an in-house small molecule library, and the selected small molecule was further validated for its anti-proliferative effect on VSMCs using multiple approaches, such as cell proliferation assays, wound healing assays, transwell migration assays, and ex vivo aortic ring assay. RESULTS: Among 43 initially screened small molecule inhibitors of kinases that have no known anti-proliferative effect on VSMCs, a spleen tyrosine kinase (Syk) inhibitor (BAY61-3606) showed significant anti-proliferative effect on VSMCs. Further experiments indicated that BAY61 attenuated the VSMC proliferation in both concentration- and time-dependent manner, and it also significantly suppressed the migration of VSMCs as assessed by both wound healing assays and transwell assays. Additionally, BAY61 suppressed the sprouting of VSMCs from endothelium-removed aortic rings. CONCLUSION: The present study identified a Syk kinase inhibitor as a potent VSMC proliferation and migration inhibitor and warrants further studies to elucidate its underlying molecular mechanisms, such as its primary target, and to validate its in vivo efficacy as a therapeutic agent for restenosis and atherosclerosis.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Niacinamida/análogos & derivados , Pirimidinas/farmacología , Quinasa Syk/antagonistas & inhibidores , Animales , Aorta Torácica/efectos de los fármacos , Western Blotting , Ensayos de Migración Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Músculo Liso Vascular/citología , Niacinamida/farmacología , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Factores de Tiempo , Cicatrización de Heridas/efectos de los fármacos
17.
Cell Physiol Biochem ; 39(4): 1595-607, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27627433

RESUMEN

BACKGROUND/AIMS: It is known that mesenchymal stem cells (MSCs) can have variable responses to hypoxic conditions and that hypoxia may specifically stimulate differentiation into osteogenic, chondrogenic, or adipogenic cells. Based on our previous study, we hypothesized that hypoxia may also induce MSC differentiation into cardiomyocytes and/or cells with comparable phenotypes. METHODS: The differences in the proteomes were specifically investigated in bone marrow-derived rat MSCs (BM-rMSCs) under normoxic and hypoxic conditions using 2-DE combined with a MALDI-TOF-MS analysis and western blot analysis. In addition, genetic and/or proteomic interactions were assessed using a String network analysis. RESULTS: Among the 35 markedly changed spots from a total of 393 matched spots, 24 were highly up-regulated and 11 were significantly down-regulated in hypoxic rMSCs based on a proteomic analysis. Although hypoxia failed to induce the direct differentiation of rMSCs into cardiomyocytes, several cardiomyocyte differentiation-related genes and proteins were significantly increased by hypoxic stress. CONCLUSION: We found that BM-rMSCs alter their expression of several cardiomyocyte differentiation-related genes and proteins under hypoxic conditions, and we examined the interactions between these genes and/or proteins, providing new insights for the applicability of MSCs preconditioned by hypoxic stimulation for use in cardiac diseases.


Asunto(s)
Células de la Médula Ósea/metabolismo , Diferenciación Celular/genética , Células Madre Mesenquimatosas/metabolismo , Miocitos Cardíacos/metabolismo , Proteoma/genética , Animales , Células de la Médula Ósea/citología , Hipoxia de la Célula , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Masculino , Células Madre Mesenquimatosas/citología , Miocitos Cardíacos/citología , Cultivo Primario de Células , Mapeo de Interacción de Proteínas , Proteoma/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal
18.
Cell Physiol Biochem ; 40(1-2): 400-410, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27866198

RESUMEN

BACKGROUND/AIMS: We previously showed that a hypoxic environment modulates the antiarrhythmic potential of mesenchymal stem cells. METHODS: To investigate the mechanism by which secreted proteins contribute to the pathogenesis of antiarrhythmic potential in mesenchymal stem cells, we used two-dimensional electrophoresis combined with MALDI-TOF-MS to perform a proteomic analysis to compare the paracrine media produced by normoxic and hypoxic cells. RESULTS: The proteomic analysis revealed that 66 protein spots out of a total of 231 matched spots indicated differential expression between the normoxic and hypoxic conditioned media of mesenchymal stem cells. Interestingly, two tropomyosin isoforms were dramatically increased in the hypoxic conditioned medium of mesenchymal stem cells. An increase in tropomyosin was confirmed using Western blot to analyze the conditioned media between normoxic and hypoxic cells. In a network analysis based on gene ontology (GO) Molecular Function by GeneMANIA analysis, most of the identified proteins were found to be involved in the regulation of heart processes. CONCLUSION: Our results show that hypoxia up-regulates tropomyosin and other secreted proteins which suggests that tropomyosin may be involved in regulating proarrhythmic and antiarrhythmic functions.


Asunto(s)
Medios de Cultivo Condicionados/farmacología , Células Madre Mesenquimatosas/metabolismo , Comunicación Paracrina/efectos de los fármacos , Proteómica/métodos , Animales , Hipoxia de la Célula , Electroforesis en Gel Bidimensional , Redes Reguladoras de Genes , Células Madre Mesenquimatosas/efectos de los fármacos , Miocardio/metabolismo , Isoformas de Proteínas/metabolismo , Ratas , Tinción con Nitrato de Plata , Tropomiosina/metabolismo
19.
Int J Mol Sci ; 17(10)2016 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-27775615

RESUMEN

Stem cell therapy using adult stem cells, such as mesenchymal stem cells (MSCs) has produced some promising results in treating the damaged heart. However, the low survival rate of MSCs after transplantation is still one of the crucial factors that limit the therapeutic effect of stem cells. In the damaged heart, oxidative stress due to reactive oxygen species (ROS) production can cause the death of transplanted MSCs. Apoptosis signal-regulating kinase 1 (ASK1) has been implicated in the development of oxidative stress-related pathologic conditions. Thus, we hypothesized that down-regulation of ASK1 in human MSCs (hMSCs) might attenuate the post-transplantation death of MSCs. To test this hypothesis, we screened microRNAs (miRNAs) based on a miRNA-target prediction database and empirical data and investigated the anti-apoptotic effect of selected miRNAs on human adipose-derived stem cells (hASCs) and on rat myocardial infarction (MI) models. Our data indicated that miRNA-301a most significantly suppressed ASK1 expression in hASCs. Apoptosis-related genes were significantly down-regulated in miRNA-301a-enriched hASCs exposed to hypoxic conditions. Taken together, these data show that miRNA-mediated down-regulation of ASK1 protects MSCs during post-transplantation, leading to an increase in the efficacy of MSC-based cell therapy.


Asunto(s)
Apoptosis/genética , MAP Quinasa Quinasa Quinasa 5/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , MicroARNs/genética , Estrés Oxidativo/fisiología , Animales , Línea Celular , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Humanos , Infarto del Miocardio , Interferencia de ARN , ARN Interferente Pequeño/genética , Ratas , Especies Reactivas de Oxígeno/metabolismo
20.
Int J Mol Sci ; 16(7): 14961-78, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26147426

RESUMEN

Osteoarthritis (OA) is a chronic, progressive, and irreversible degenerative joint disease. Conventional OA treatments often result in complications such as pain and limited activity. However, transplantation of mesenchymal stem cells (MSCs) has several beneficial effects such as paracrine effects, anti-inflammatory activity, and immunomodulatory capacity. In addition, MSCs can be differentiated into several cell types, including chondrocytes, osteocytes, endothelia, and adipocytes. Thus, transplantation of MSCs is a suggested therapeutic tool for treatment of OA. However, transplanted naïve MSCs can cause problems such as heterogeneous populations including differentiated MSCs and undifferentiated cells. To overcome this problem, new strategies for inducing differentiation of MSCs are needed. One possibility is the application of microRNA (miRNA) and small molecules, which regulate multiple molecular pathways and cellular processes such as differentiation. Here, we provide insight into possible strategies for cartilage regeneration by transplantation of differentiated MSCs to treat OA patients.


Asunto(s)
Condrogénesis , Trasplante de Células Madre Mesenquimatosas/métodos , Osteoartritis/terapia , Animales , Humanos , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA