Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Biosci Rep ; 40(5)2020 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-32342982

RESUMEN

Cutaneous wound is a soft tissue injury that is difficult to heal during aging. It has been demonstrated that adipose-derived stem cells (ADSCs) and its secreted exosomes exert crucial functions in cutaneous wound healing. The present study aimed to elucidate the mechanism of exosomes derived from ADSCs (ADSC-Exos) containing MALAT1 in wound healing. ADSCs were isolated from human normal subcutaneous adipose tissues and identified by flow cytometry analysis. Exosomes were extracted from ADSC supernatants and MALAT1 expression was determined using qRT-PCR analysis. HaCaT and HDF cells were exposed to hydrogen peroxide (H2O2) for simulating the skin lesion model. Subsequently, CCK-8, flow cytometry, wound healing and transwell assays were employed to validate the role of ADSC-Exos containing MALAT1 in the skin lesion model. Besides, cells were transfected with sh-MALAT1 to verify the protective role of MALAT1 in wound healing. The binding relationship between MALAT1 and miR-124 were measured by dual-luciferase reporter assay. ADSC-Exos promoted cell proliferation, migration, and inhibited cell apoptosis of HaCaT and HDF cells impaired by H2O2. However, the depletion of MALAT1 in ADSC-Exos lose these protective effects on HaCaT and HDF cells. Moreover, miR-124 was identified to be a target of MALAT1. Furthermore, ADSC-Exos containing MALAT1 could mediate H2O2-induced wound healing by targeting miR-124 and activating Wnt/ß-catenin pathway. ADSC-Exos containing MALAT1 play a positive role in cutaneous wound healing possibly via targeting miR-124 through activating the Wnt/ß-catenin pathway, which may provide novel insights into the therapeutic target for cutaneous wound healing.


Asunto(s)
Exosomas/metabolismo , Fibroblastos/metabolismo , Queratinocitos/metabolismo , MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Traumatismos de los Tejidos Blandos/metabolismo , Células Madre/metabolismo , Grasa Subcutánea/citología , Vía de Señalización Wnt , Cicatrización de Heridas , Apoptosis , Movimiento Celular , Proliferación Celular , Exosomas/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Células HaCaT , Humanos , Peróxido de Hidrógeno/toxicidad , Queratinocitos/efectos de los fármacos , Queratinocitos/patología , MicroARNs/genética , ARN Largo no Codificante/genética , Traumatismos de los Tejidos Blandos/genética , Traumatismos de los Tejidos Blandos/patología
2.
Biomed Res Int ; 2018: 6165192, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29780826

RESUMEN

Silibinin, also known as silybin, is the major flavonolignan isolated from Silybum marianum. Although previous reports demonstrated that silibinin exhibits significant tumor suppressor activities in various cancers by promoting cell apoptosis, it was also shown to trigger autophagy to counteract apoptosis induced by exogenous stresses in several types of cells. However, there is no report to address the role of silibinin induced autophagy in human A172 and SR glioblastoma cells. Our study showed that silibinin treatment not only inhibited the metabolic activities of glioblastoma cells but also promoted their apoptosis through the regulation of caspase 3 and PARP-1 in concentration- and time-dependent manners. Meanwhile, silibinin induced autophagy through upregulation of microtubule-associated protein a light chain 3- (LC3-) II. And autophagy inhibition with chloroquine, a lysosomotropic agent, significantly enhanced silibinin induced glioblastoma cell apoptosis. Moreover, silibinin dose-dependently downregulated the phosphorylation levels of mTOR at Ser-2448, p70S6K at Thr-389, and 4E-BP1 at Thr-37/46. Furthermore, the expression of YAP, the downstream effector of Hippo signal pathway, was also suppressed by silibinin. These results suggested that silibinin induced glioblastoma cell apoptosis concomitant with autophagy which might be due to simultaneous inhibition of mTOR and YAP and silibinin induced autophagy exerted a protective role against cell apoptosis in both A172 and SR cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis/efectos de los fármacos , Glioblastoma , Fosfoproteínas/metabolismo , Silimarina/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Silibina , Factores de Transcripción , Proteínas Señalizadoras YAP
3.
Biomed Res Int ; 2017: 2578017, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29018809

RESUMEN

MSC treatment can promote cutaneous wound repair through multiple mechanisms, and paracrine mediators secreted by MSC are responsible for most of its therapeutic benefits. Recently, MSC sheet composed of live MSCs and their secreted ECMs was reported to promote wound healing; however, whether its ECM alone could accelerate wound closure remained unknown. In this study, Nc-ECM and Cc-ECM were prepared from nonconditioned and CoCl2-conditioned MSC sheets, respectively, and their wound healing properties were evaluated in a mouse model of full-thickness skin defect. Our results showed that Nc-ECM can significantly promote wound repair through early adipocyte recruitment, rapid reepithelialization, enhanced granulation tissue growth, and augmented angiogenesis. Moreover, conditioning of MSC sheet with CoCl2 dramatically enriched its ECM with collagen I, collagen III, TGF-ß1, VEGF, and bFGF via activation of HIF-1α and hence remarkably improved its ECM's in vivo wound healing potency. All the Cc-ECM-treated wounds completely healed on day 7, while Nc-ECM-treated wounds healed about 85.0% ± 8.6%, and no-treatment wounds only healed 69.8% ± 9.6% (p < 0.05). Therefore, we believe that such growth factor-reinforced ECM fabricated from chemically hypoxic MSC sheet has the potential for clinical translation and will lead to a MSC-derived, cost-effective, bankable biomaterial for wound management.


Asunto(s)
Matriz Extracelular/metabolismo , Matriz Extracelular/fisiología , Hipoxia/fisiopatología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/fisiología , Cicatrización de Heridas/fisiología , Adipocitos/metabolismo , Adipocitos/fisiología , Animales , Células Cultivadas , Medios de Cultivo Condicionados/metabolismo , Femenino , Hipoxia/metabolismo , Ratones , Ratones Endogámicos BALB C , Conejos , Piel/metabolismo , Piel/fisiopatología
4.
J Biomater Appl ; 32(4): 472-483, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28927334

RESUMEN

The extracellular matrix has drawn considerable interest in tissue engineering not only acts as a bioactive three-dimensional scaffold but also regulates cell behaviors through providing biochemical signals. Extracellular matrix-based biomaterials, mainly derived from xenogeneic tissues, have shown positive outcomes in promoting cutaneous wound healing. However, such extracellular matrices only contain low doses of growth factors, which limit their therapeutic efficiency. Recent reports demonstrated that cell sheets made from mesenchymal stem cell can accelerate wound repair through enhanced re-epithelialization and angiogenesis, but its clinical translation is hindered by several limitations, such as the risk of aberrant immune responses and cost implications. In this study, acellular extracellular matrices were prepared from CuCl2-conditioned mesenchymal stem cell sheets and their in vivo wound healing properties were evaluated in a mouse model of full-thickness skin defect. We found that extracellular matrices derived from CuCl2-conditioned mesenchymal stem cell sheets have a compact surface with thick solid-like cross-sectional structure. Moreover, CuCl2 dramatically enriched the extracellular matrices with collagen I, collagen III, transforming growth factor-ß1, vascular endothelial growth factor, and basic fibroblast growth factor via hypoxia-inducible factor-1α activation. And as a consequence, the resulting extracellular matrices showed markedly improved in vivo wound healing potency through early adipocyte mobilization, enhanced granulation tissues formation, rapid re-epithelialization, and augmented angiogenesis. Therefore, we consider that the extracellular matrix derived from CuCl2-conditioned mesenchymal stem cell sheets has the potential for clinical translation and may lead to a novel strategy for wound management.


Asunto(s)
Vendajes , Cobre/química , Matriz Extracelular/química , Células Madre Mesenquimatosas/citología , Cicatrización de Heridas , Adipocitos/fisiología , Animales , Materiales Biocompatibles , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Colágeno/metabolismo , Cobre/metabolismo , Matriz Extracelular/metabolismo , Femenino , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Tejido de Granulación/fisiología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Fenómenos Mecánicos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos BALB C , Neovascularización Fisiológica , Conejos , Ingeniería de Tejidos , Factor A de Crecimiento Endotelial Vascular/metabolismo
5.
Plast Reconstr Surg ; 132(1): 61e-69e, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23806955

RESUMEN

BACKGROUND: Hypertrophic scars result from excessive collagen deposition at sites of healing dermal wounds and could be functionally and cosmetically problematic. The authors tested the ability of the histone deacetylase inhibitor trichostatin A to reduce hypertrophic scar formation in a rabbit ear model. METHODS: The authors have developed a reliable rabbit model that results in hypertrophic scarring. Four 1-cm, full-thickness, circular wounds were made on each ear. After the wounds reepithelialized, 0.02% trichostatin A was injected intradermally into the wounds in the treatment group. Expression of collagen I and fibronectin was detected by reverse transcription polymerase chain reaction and Western blot analysis at postoperative day 23. Scar hypertrophy was quantified by measurement of the scar elevation index at postoperative day 45. RESULTS: Compared with the control group, injection of trichostatin A led to much more normal-appearing scars in the rabbit ear. The scar elevation index at postoperative day 45 was significantly decreased after injection of trichostatin A compared with untreated scars. Furthermore, the authors confirmed the decreased expression of collagen I and fibronectin at postoperative day 23 (after the rabbits had been treated with trichostatin A for 1 week) in the treated scars compared with the control scars according to reverse transcription polymerase chain reaction and Western blot analysis. CONCLUSIONS: The introduction of trichostatin A can result in the decreased formation of hypertrophic scars in a rabbit ear model, which is corroborated by evidence of decreased collagen I and fibronectin synthesis.


Asunto(s)
Cicatriz Hipertrófica/prevención & control , Enfermedades del Oído/prevención & control , Oído Externo/lesiones , Inhibidores de Histona Desacetilasas/uso terapéutico , Ácidos Hidroxámicos/uso terapéutico , Heridas y Lesiones/tratamiento farmacológico , Animales , Western Blotting , Cicatriz Hipertrófica/etiología , Cicatriz Hipertrófica/patología , Colágeno Tipo I/biosíntesis , Colágeno Tipo I/genética , Modelos Animales de Enfermedad , Enfermedades del Oído/etiología , Enfermedades del Oído/patología , Oído Externo/metabolismo , Femenino , Fibronectinas/biosíntesis , Fibronectinas/genética , Estudios de Seguimiento , Regulación de la Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/administración & dosificación , Ácidos Hidroxámicos/administración & dosificación , Inyecciones Intradérmicas , Masculino , ARN Mensajero/análisis , Conejos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Resultado del Tratamiento , Cicatrización de Heridas/efectos de los fármacos , Heridas y Lesiones/complicaciones , Heridas y Lesiones/patología
6.
Med Hypotheses ; 74(1): 85-6, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19682804

RESUMEN

Coronary and peripheral artery bypass grafting are widely being used to deal with vascular deficiencies currently, and a man-made synthetic tube or autogenous arteries or veins are needed a lot. But one's autogenous arteries or veins are limited, and artificial graft substitute isn't yet available in clinical applications because of many disadvantages. Various polymeric materials have been used as scaffolds, but without satisfying results due to intimal hyperplasia and the rate of degradation. Autogenetic dermis, which has the advantages of resistance to immunogenicity, good biocompatibility, and appropriate mechanical and physiological properties, has gained our attention to use it as a scaffold for tissue-engineered blood vessels. What is more, autogenetic dermis can be harvested easily. So we postulate that autogenetic dermis rolled up to form a tube may be an ideal scaffold for tissue-engineered blood vessels.


Asunto(s)
Dermis/fisiología , Ingeniería de Tejidos/métodos , Andamios del Tejido , Animales , Arterias/patología , Materiales Biocompatibles , Prótesis Vascular , Implantación de Prótesis Vascular/métodos , Dermis/patología , Humanos , Modelos Biológicos , Modelos Teóricos , Porcinos , Ingeniería de Tejidos/instrumentación , Venas/patología
7.
Aesthetic Plast Surg ; 33(3): 457-60, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19387723

RESUMEN

BACKGROUND: A successful deep multilayered wound suture should provide a firm tension-relieving closure, good wound-edge eversion, hemostasis, and minimal intradermal extraneous materials. However, this is not always achieved with a single standard technique. The authors describe their modification of a wound closure method that can rapidly and reliably achieve these results. METHODS: A wedge-shaped excision was adopted to obtain a trapezoid pattern transect, after which a modified fully buried vertical mattress suture technique was used to close the wound. These techniques were compared with the standard excision and suture techniques used for the same patient at different times after surgery. RESULTS: The wedge-shaped excision can facilitate good wound-edge eversion, and the modified fully buried vertical mattress suture can provide firm tension relief and optimal apposition. Compared with conventional excision and suture techniques, the described techniques brought about a better outcome in terms of hypertrophic scar prevention. CONCLUSION: The described modified technique seems to be more efficient than conventional procedures used to prevent hypertrophic scar formation.


Asunto(s)
Técnicas de Sutura , Heridas y Lesiones/cirugía , Cicatriz Hipertrófica/prevención & control , Hemostasis Quirúrgica , Humanos , Grasa Subcutánea/cirugía
8.
Med Hypotheses ; 70(2): 343-5, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-17630223

RESUMEN

High mobility group box 1 protein (HMGB1) is a highly conserved, ubiquitous non-histone nuclear protein, which participates in maintaining nucleosome structure, regulation of gene transcription, and modulating the activity of steroid hormone receptors. Substantial evidence demonstrated that HMGB1 could be secreted into the extracellular milieu, acts as a proinflammatory cytokine and mediates the downstream inflammatory responses in endotoxemia, arthritis and sepsis. Recently, several reports suggested that HMGB1 plays a key role in tumor angiogenesis through multiple mechanisms, including up-regulation of proangiogenic factors, promoting endothelial progenitor cells homing to ischemic tumor tissues and induction of endothelial cell migration and sprouting. And blockade of HMGB1 binding to the receptor for advanced glycation end products (RAGE) with anti-HMGB1 antibody, soluble RAGE or anti-RAGE neutralizing antibody has been proved to inhibit angiogenesis efficiently. Since HMGB1 A box peptide could antagonize the HMGB1 whole length protein by competitively binding to RAGE and has been considered as a HMGB1 specific antagonist, we postulate that the HMGB1 A box peptide could function as an anti-angiogenic agent to inhibit tumor angiogenesis. In our opinion, if the hypothesis proved to be practical, HMGB1 A box peptide could be widely used in clinical settings to treat malignant tumors.


Asunto(s)
Proteína HMGB1/farmacología , Neoplasias/irrigación sanguínea , Neovascularización Patológica/prevención & control , Animales , Proteína HMGB1/antagonistas & inhibidores , Proteína HMGB1/fisiología , Humanos , Modelos Biológicos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Neovascularización Patológica/etiología , Neovascularización Patológica/fisiopatología , Fragmentos de Péptidos/farmacología
9.
Med Hypotheses ; 69(4): 888-91, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17376603

RESUMEN

Skin grafts, including skin flaps, are widely used in plastic and reconstructive surgery to cover wounds and tissue defects resulting from mechanic or burn injury. Ischemic necrosis is the main complication in skin graft surgery due to inefficient revascularization. Though the surgical delay procedure has been proved to be the only effective technique to prevent skin flap ischemic necrosis by mechanism of inducing adaption to hypoxia, but it is time consuming, costly, and having high risk of infection due to repeated surgery. Recent research demonstrated that, in addition to protecting cells against apoptosis, the expression of survivin correlates with intratumoral microvessel density in several different types of tumors and survivin could upregulate several proangiogenic factors, including VEGF, Egr-1 and Siah-1. Moreover, Survivin DeltaEx3, one of the survivin alternative splice variants, is necessary for activating the small GTPase Rac1 during endothelial tube formation and required for in vivo endothelial cell invasion. Therefore, we postulate that intracellular delivery of survivin or Survivin DeltaEx3 by fusion with protein transduction domain would enhance skin flap survival through accelerating revascularization by both inhibiting the apoptosis of microvascular endothelial cells and promoting skin flap angiogenesis. If the hypothesis was proved to be practical, the fusion proteins would be widely used in plastic and reconstructive surgery to prevent skin flap from ischemic necrosis in the future.


Asunto(s)
Permeabilidad de la Membrana Celular/fisiología , Supervivencia de Injerto , Proteínas Asociadas a Microtúbulos/fisiología , Proteínas de Neoplasias/fisiología , Trasplante de Piel/fisiología , Colgajos Quirúrgicos , Humanos , Proteínas Inhibidoras de la Apoptosis , Modelos Biológicos , Neovascularización Fisiológica , Piel/irrigación sanguínea , Survivin
10.
Med Hypotheses ; 69(1): 33-5, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17280792

RESUMEN

Hypoxia is a common phenomenon in human solid tumors and has been considered as an important, independent negative prognostic factor for response to treatment and survival of tumor patients. Hypoxia-inducible factor-1 (HIF-1) is the central transcription factor which is activated by hypoxia and modulates the expression of many genes involved in cell metabolism, proliferation, apoptosis, angiogenesis. Recently, it has been reported that HIF-1 contributes to tumor radioresistance by upregulating survivin expression under hypoxic conditions. Moreover, in hypoxic tumor cells, HIF-1 dependent signal transduction pathway is activated and could be further enhanced by radiation, thereby providing survival signals to adjacent vascular endothelial cells by upregulation of VEGF and bFGF and resulting in tumor radioresistance through vascular radioprotection. Recent research revealed that the stability of HIF-1alpha, one of the two subunits of HIF-1, determines the whole HIF-1 activity and the C-terminal transactivation domain of HIF-1alpha could reduce HIF-1 activity when overexpressed in tumor cells by disruption of the assembly of HIF-1 transcription complex. Therefore, we postulate that fusion with protein transduction domains would overcome the inability of C-terminal transactivation domain of HIF-1alpha to cross cellular membrane. Thus the recombinant fusion proteins could serve as cell-permeable HIF-1 antagonists, function as both inhibitors of tumor angiogenesis and tumor radiosensitizers, and would be widely used in clinical settings to improve tumor response to radiotherapy.


Asunto(s)
Antineoplásicos/administración & dosificación , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias/metabolismo , Neoplasias/radioterapia , Fármacos Sensibilizantes a Radiaciones/administración & dosificación , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/efectos de la radiación , Permeabilidad de la Membrana Celular , Humanos , Modelos Biológicos , Neoplasias/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA