Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Wound Care ; 32(Sup3): S17-S21, 2023 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-36930279

RESUMEN

OBJECTIVE: To report the efficacy and long-term outcomes of treating the skin defects of aplasia cutis congenita (ACC) with cryopreserved amniotic membrane (AM). METHOD: Human amnion was obtained from the caesarean delivery of a full-term healthy pregnancy and processed in a sterile laminar flow hood, and cryopreserved in liquid nitrogen. The structure of the AM was investigated histologically and the viability of the epithelial cells was assessed after cryopreservation and compared with fresh AM and with AM preserved in phosphate-buffered saline (PBS) at 4°C. The cryopreserved AM was applied onto the lower limb skin defects of a one-month old baby with ACC. Timely AM changes were performed as necessary until the wounds healed. RESULTS: The structure of the cryopreserved AM was intact, with little visible difference compared with fresh AM. The viability of the epithelial cells was partially lost but still much better retained than in those preserved in PBS at 4°C. The limb skin defects were gradually re-epithelialised upon application of the AM and were completely healed after one month. The 4-month and 2-year follow-ups presented good skin texture and colour, without hypertrophic scar formation. CONCLUSION: In this case study, cryopreservation of AM presented a well preserved stromal compartment and viable epithelial layer. It also offered features such as pain relief, good attachment and adhesiveness, improved wound healing and suppressed scar formation in the treatment of ACC skin defects.


Asunto(s)
Amnios , Displasia Ectodérmica , Embarazo , Femenino , Humanos , Lactante , Criopreservación , Piel , Células Epiteliales , Displasia Ectodérmica/terapia
2.
J Nanobiotechnology ; 18(1): 163, 2020 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-33167997

RESUMEN

BACKGROUND: Umbilical cord mesenchymal stem cell (HUCMSC)-based therapies were previously utilised for cartilage regeneration because of the chondrogenic potential of MSCs. However, chondrogenic differentiation of HUCMSCs is limited by the administration of growth factors like TGF-ß that may cause cartilage hypertrophy. It has been reported that extracellular vesicles (EVs) could modulate the phenotypic expression of stem cells. However, the role of human chondrogenic-derived EVs (C-EVs) in chondrogenic differentiation of HUCMSCs has not been reported. RESULTS: We successfully isolated C-EVs from human multi-finger cartilage and found that C-EVs efficiently promoted the proliferation and chondrogenic differentiation of HUCMSCs, evidenced by highly expressed aggrecan (ACAN), COL2A, and SOX-9. Moreover, the expression of the fibrotic marker COL1A and hypertrophic marker COL10 was significantly lower than that induced by TGF-ß. In vivo, C-EVs induced HUCMSCs accelerated the repair of the rabbit model of knee cartilage defect. Furthermore, C-EVs led to an increase in autophagosomes during the process of chondrogenic differentiation, indicating that C-EVs promote cartilage regeneration through the activation of autophagy. CONCLUSIONS: C-EVs play an essential role in fostering chondrogenic differentiation and proliferation of HUCMSCs, which may be beneficial for articular cartilage repair.


Asunto(s)
Autofagia/fisiología , Cartílago/metabolismo , Condrocitos/metabolismo , Vesículas Extracelulares/metabolismo , Células Madre Mesenquimatosas/metabolismo , Cordón Umbilical/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Condrocitos/citología , Condrogénesis , Femenino , Humanos , Masculino , Células Madre Mesenquimatosas/citología , Conejos , Cordón Umbilical/citología
3.
Skin Pharmacol Physiol ; 31(5): 229-237, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29847822

RESUMEN

AIMS: To elucidate the possible mechanisms of how basic fibroblast growth factor (bFGF) influences epidermal homeostasis in a living skin equivalent (LSE) model. METHODS: Several wound healing-related growth factors were analyzed at protein and mRNA levels for dermal fibroblasts of induced alpha-smooth muscle actin (α-SMA)-positive or α-SMA-negative phenotypes. During culturing an LSE model by seeding normal human keratinocytes on a fibroblast-populated type I collagen gel, bFGF or neutralizing antibody for keratinocyte growth factor (KGF) was added to investigate its effects on fibroblast phenotypes and, subsequently, epidermal homeostasis by histology and immunohistochemistry. RESULTS: The α-SMA-positive phenotype of fibroblasts induced by transforming growth factor beta-1 (TGF-ß1) markedly suppressed the expression of KGF and hepatocyte growth factor (HGF), and slightly upregulated vascular endothelial growth factor (VEGF) and TGF-ß1 at mRNA and protein levels, compared with α-SMA-negative fibroblasts treated with bFGF. α-SMA expression of fibroblasts at the epidermal-mesenchymal junction of the LSEs was suppressed by the addition of bFGF, and a better-differentiated epidermis was presented. The abrogation of KGF from fibroblasts by the addition of the KGF neutralizing antibody disenabled the LSE culturing system to develop an epidermis. CONCLUSIONS: bFGF, through affecting the phenotypes and functions of fibroblasts, especially KGF expression, influenced epidermal homeostasis in an LSE model.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/farmacología , Factor 7 de Crecimiento de Fibroblastos/metabolismo , Fibroblastos/efectos de los fármacos , Actinas/metabolismo , Células Cultivadas , Factor 7 de Crecimiento de Fibroblastos/genética , Fibroblastos/metabolismo , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/metabolismo , Homeostasis/efectos de los fármacos , Humanos , Queratinocitos/metabolismo , Fenotipo , Piel/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
4.
Cell Commun Signal ; 15(1): 43, 2017 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-29041943

RESUMEN

BACKGROUND: We evaluated the therapeutic effect and fate of high doses of human umbilical cord Wharton jelly cells (hUCWJCs) after IP administration to streptozotocin (STZ)-induced diabetic mice. METHODS: Type 1 diabetes (T1D) was induced in Kunming mice via IP injection of STZ. hUCWJCs were labeled with 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (DiI). Diabetic animals with sustained hyperglycemia for at least 2 weeks were administered 1 × 107 Dil-hUCWJCs via intraperitoneal injection. Insulin, glucagon and PDX-1 were detected by immunofluorescence with confocal microscopy. Serum mouse and human C-peptide was assayed in blood collected via intracardiac puncture. Specific ß-cell differentiation markers and human DNA were assessed using qPCR performed with 200 ng of target DNA. RESULTS: hUCWJCs migrated to the STZ-damaged organs and contributed to lower blood glucose levels in 30% of the treated mice. Confocal microscopy revealed the presence of resident insulin-positive cells in the liver and kidneys. hUCWJC-treated mice with restored hyperglycemia also showed increased serum mouse C-peptide levels. The qPCR results, particularly in the liver, revealed that after transplantation hUCWJCs upregulated genes of endocrine precursors but failed to express endocrine stage markers. Mice with restored hyperglycemia had reduced urinary volume and lacked glomerular hypertrophy, exhibiting a morphology resembling that of normal glomeruli. Moreover, we also verified that one of the possible mechanisms by which hUCWJCs exert immunosuppressive effects is through down-regulation of the cell surface receptor HLA-1. CONCLUSIONS: We confirmed the potential of IP administration of hUCWJCs and the capability of these cells to migrate to damaged tissues and promote insulin secretion from non-pancreatic local cells and to improve renal damage. These findings confer unique therapeutic properties to hUCWJCs, suggesting a promising future in the treatment of diabetes mellitus.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/terapia , Insulina/biosíntesis , Riñón/lesiones , Trasplante de Células Madre Mesenquimatosas , Páncreas/metabolismo , Cordón Umbilical/citología , Animales , Glucemia/metabolismo , Péptido C/biosíntesis , Diferenciación Celular , Diabetes Mellitus Experimental/patología , Humanos , Masculino , Ratones
5.
Exp Eye Res ; 135: 109-17, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25682729

RESUMEN

Interactions between stromal and epithelial cells play important roles in the development, homeostasis, and pathological conditions of the cornea. Soluble cytokines are critical factors in stromal-epithelial interactions, and growth factors secreted from corneal stromal cells contribute to the regulation of proliferation and differentiation of corneal epithelial cells (CECs). However, the manner in which the expression of growth factors is regulated in stromal cells has not been completely determined. To study stromal-epithelial cell interactions, we used an organotypic culture model. Human or rabbit CECs (HCECs or RCECs) were cultured on amniotic membranes placed on human corneal fibroblasts (HCFs) embedded in a collagen gel. The properties of the organotypic culture were examined by hematoxylin-eosin staining and immunofluorescence. In the organotypic culture, HCECs or RCECs were stratified into two-three layers after five days and five-seven layers after nine days. However, stratification was not observed when the HCECs were seeded on a collagen gel without fibroblasts. K3/K12 were expressed on day 9. The HCF-embedded collagen gels were collected on days 3, 5, or 9 after seeding the RCECs, and mRNA expression of growth factors FGF7, HGF, NGF, EGF, TGF-α, SCF, TGF-ß1, TGF-ß2, and TGF-ß3 were quantified by real-time PCR. mRNA expression of the growth factors in HCFs cultured with RCECs were compared with those cultured without RCECs, as well as in monolayer cultures. mRNA expression of TGF-α was markedly increased in HCFs cultured with RCECs. However, mRNA expression of the TGF-ß family was suppressed in HCFs cultured with RCECs. Principal component analysis revealed that mRNA expression of the growth factors in HCFs were generally similar when they were cultured with RCECs. In organotypic cultures, the morphological changes in the CECs and the expression patterns of the growth factors in the stromal cells clearly demonstrated stromal-epithelial cell interactions, and the results suggest that stromal cells and epithelial cells may act in concert in the cornea.


Asunto(s)
Comunicación Celular/fisiología , Córnea/citología , Células Epiteliales/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Células del Estroma/metabolismo , Análisis de Varianza , Animales , Células Cultivadas , Factor de Crecimiento Epidérmico/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Fibroblastos/metabolismo , Humanos , Modelos Animales , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , ARN Mensajero/metabolismo , Conejos , Factores de Crecimiento Transformadores/genética , Factores de Crecimiento Transformadores/metabolismo
6.
Cell Tissue Res ; 357(3): 571-82, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24906288

RESUMEN

Amyotrophic lateral sclerosis (ALS) is an adult-onset progressive neurodegenerative disease involving degeneration of motor neurons in the central nervous system. Stem cell treatment is a potential therapy for this fatal disorder. The human amniotic membrane (HAM), an extremely rich and easily accessible tissue, has been proposed as an attractive material in cellular therapy and regenerative medicine because of its advantageous characteristics. In the present study, we evaluate the long-term effects of a cellular treatment by intravenous administration of human amniotic mesenchymal stem cells (hAMSCs) derived from HAM into a hSOD1(G93A) mouse model. The mice received systemic administration of hAMSCs or phosphate-buffered saline (PBS) at the onset, progression and symptomatic stages of the disease. hAMSCs were detected in the spinal cord at the final stage of the disease, in the form of isolates or clusters and were negative for ß-tubulin III and GFAP. Compared with the treatment with PBS, multiple hAMSC transplantations significantly retarded disease progression, extended survival, improved motor function, prevented motor neuron loss and decreased neuroinflammation in mice. These findings demonstrate that hAMSC transplantation is a promising cellular treatment for ALS.


Asunto(s)
Amnios/citología , Esclerosis Amiotrófica Lateral/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Conducta Animal , Movimiento Celular , Supervivencia Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inflamación/patología , Inyecciones Intravenosas , Masculino , Ratones , Actividad Motora , Neuronas Motoras/patología
7.
Eur J Med Res ; 29(1): 270, 2024 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-38704575

RESUMEN

BACKGROUND: This study aims to investigate the effects of a conditioned medium (CM) from human umbilical cord mesenchymal stem cells (HuMSCs) cultivated in gelatin sponge (GS-HuMSCs-CM) on hair growth in a mouse model. METHODS: CM was collected from the HuMSCs cultivated in a monolayer or in a gelatin sponge. Vascular endothelial growth factor (VEGF), insulin-like growth factor-1 (IGF-1), keratinocyte growth factor (KGF), and hepatocyte growth factor (HGF) levels in CMs were measured by enzyme-linked immunosorbent assays (ELISAs). A hair loss model by a C57 BL/6J mouse was prepared. The effects of GS-HuMSCs-CM and HuMSCs on hair regrowth in mice were investigated by intradermal injection in the depilated back skin with normal saline (NS) as the control. The time for hair regrowth and full covering in depilated areas was observed, and the hair growth was evaluated histologically and by grossly measuring hair length and diameter. RESULTS: Compared with monolayer cultured cells, the three-dimensional (3D) culture of HuMSCs in gelatin sponge drastically increased VEGF, IGF-1, KGF, and HGF production. GS-HuMSCs-CM and HuMSCs injection both promoted hair regeneration in mice, while GS-HuMSCs-CM presented more enhanced effects in hair length, hair diameter, and growth rate. GS-HuMSCs-CM significantly promoted angiogenesis in injected skin areas, which might also contribute to faster hair regrowth. CONCLUSION: GS-HuMSCs-CM exerted significant effects on inducing hair growth and promoted skin angiogenesis in C57BL/6J mice.


Asunto(s)
Cabello , Factor I del Crecimiento Similar a la Insulina , Células Madre Mesenquimatosas , Cordón Umbilical , Animales , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Humanos , Medios de Cultivo Condicionados/farmacología , Ratones , Cordón Umbilical/citología , Cabello/crecimiento & desarrollo , Cabello/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Gelatina/química , Andamios del Tejido/química , Ratones Endogámicos C57BL , Células Cultivadas , Factor 7 de Crecimiento de Fibroblastos/metabolismo
8.
J Med Chem ; 67(10): 8043-8059, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38730324

RESUMEN

Discoidin domain receptor 1 (DDR1) is a potential target for cancer drug discovery. Although several DDR1 kinase inhibitors have been developed, recent studies have revealed the critical roles of the noncatalytic functions of DDR1 in tumor progression, metastasis, and immune exclusion. Degradation of DDR1 presents an opportunity to block its noncatalytic functions. Here, we report the discovery of the DDR1 degrader LLC355 by employing autophagosome-tethering compound technology. Compound LLC355 efficiently degraded DDR1 protein with a DC50 value of 150.8 nM in non-small cell lung cancer NCI-H23 cells. Mechanistic studies revealed compound LLC355 to induce DDR1 degradation via lysosome-mediated autophagy. Importantly, compound LLC355 potently suppressed cancer cell tumorigenicity, migration, and invasion and significantly outperformed the corresponding inhibitor 1. These results underline the therapeutic advantage of targeting the noncatalytic function of DDR1 over inhibition of its kinase activity.


Asunto(s)
Autofagia , Receptor con Dominio Discoidina 1 , Humanos , Receptor con Dominio Discoidina 1/metabolismo , Receptor con Dominio Discoidina 1/antagonistas & inhibidores , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Animales , Descubrimiento de Drogas , Movimiento Celular/efectos de los fármacos , Proteolisis/efectos de los fármacos , Relación Estructura-Actividad , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/síntesis química , Proliferación Celular/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo
9.
J Appl Biomater Funct Mater ; 21: 22808000221130168, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36633288

RESUMEN

BACKGROUND: To maintain and enhance the wound healing effects of mesenchymal stem cells (MSCs), a scaffold for hosting MSCs is needed, which ought to be completely biocompatible, durable, producible, and of human source. OBJECTIVE: To build a cell-extracellular matrix (ECM) complex assembled by human umbilical cord mesenchymal stem cells (HuMSCs) and to investigate its clinical potentials in promoting wound healing. METHOD: HuMSCs were isolated and expanded. When the cells of third passage reached confluency, ascorbic acid was added to stimulate the cells to deposit ECM where the cells grew in. Four weeks later, a cells-loaded ECM sheet was formed. The cell-ECM complex was observed under the scanning electron microscopy (SEM) and subjected to histological studies. The supernatants were collected and the cell-ECM complex was harvested at different time points and processed for enzyme-linked immune sorbent assay (ELISA) and mRNA analysis. The in vivo experiments were performed by means of implanting the cell-ECM complex on the mice back for up to 6 months and the specimens were collected for histological studies. RESULTS: After 4 weeks of cultivation with ascorbic stimulation, a sheet was formed which is mainly composed with HuMSCs, collagen and hyaluronic acid. The cell-ECM complex can sustain to certain tensile force. The mRNA and protein levels of vascular endothelial growth factor-α (VEGF-α), hepatocyte growth factor (HGF), keratinocyte growth factor (KGF), and transforming growth factor-ß1 (TGF-ß1) were remarkably increased compared to monolayer-cultured cells. The implanted cell-ECM complex on mice was still noticeable with host cells infiltration and vascularization on 6 months. CONCLUSION: Our studies suggested that HuMSCs can be multi-cultivated through adding ascorbic stimulation and ECM containing collagen and hyaluronic acid were enriched around the cells which self-assembly formed a cell-ECM complex. Cell-ECM complex can improve growth factors secretion remarkably which means it may promote wound healing by paracrine.


Asunto(s)
Ácido Hialurónico , Factor A de Crecimiento Endotelial Vascular , Ratones , Humanos , Animales , Factor A de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas/fisiología , Colágeno , Matriz Extracelular
10.
Biochem Biophys Res Commun ; 429(3-4): 180-5, 2012 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-23131552

RESUMEN

Lung cancer is one of the most common causes of cancer-related mortality worldwide. Effective early diagnosis and targeted therapies for lung cancer to reduce incidence and mortality would benefit from a better understanding of the key molecular changes that occur from normal to malignant tumor cells during lung cancer initiation and development, but these are largely unknown. Previous studies have shown that DNA methylation, an important mechanism for the regulation of gene expression, plays a key role in lung carcinogenesis. In this study, we screened a novel methylation gene, ANKRD18A, encoding ankyrin repeat domain 18A, to determine whether it is regulated by DNA methylation in lung cancer. Methylation-specific PCR and bisulfite sequencing PCR were used to analyze gene methylation status, and real-time quantitative reverse transcription-polymerase chain reaction (qRT-PCR) examined mRNA levels. Promoter hypermethylation of ANKRD18A was detected in 68.4% (26/38) of lung cancer tissues but not (0/20) in normal lung tissues (P<0.01), whereas ANKRD18A mRNA expression was significantly decreased in lung cancer tissues compared with adjacent normal tissues. In addition, we found that ANKRD18A expression was significantly decreased in 9 of 10 lung cancer cell lines. This was associated with hypermethylation of the ANKRD18A promoter region. Moreover, weak expression of ANKRD18A in methylated lung cancer cell lines increased markedly after treatment with the DNA methylation inhibitor 5-aza-2'-deoxycytidine. These results suggest that ANKRD18A hypermethylation and consequent mRNA alterations might be a vital molecular mechanism in lung cancer.


Asunto(s)
Repetición de Anquirina , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Azacitidina/análogos & derivados , Azacitidina/farmacología , Metilación de ADN , Metilasas de Modificación del ADN/antagonistas & inhibidores , Decitabina , Regulación hacia Abajo , Humanos , Regiones Promotoras Genéticas
11.
J Allergy Clin Immunol ; 127(3): 806-14.e1-4, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21272927

RESUMEN

BACKGROUND: Atopic dermatitis (AD) is a chronic inflammatory skin disorder caused by multiple factors. Among them, house dust mite (HDM) allergens are important in the development of AD. In airway allergy, HDM allergens activate innate immunity. However, information regarding the activation of innate immunity by HDM allergens in the skin is limited. OBJECTIVES: The inflammasome is a key regulator of pathogen recognition and inflammation. We investigated whether HDM allergens activate the inflammasome in epidermal keratinocytes. METHODS: Keratinocytes were stimulated with Dermatophagoides pteronyssinus, and the activation of caspase-1 and secretion of IL-1ß and IL-18 were examined. Formation of the inflammasome was studied by analyzing the subcellular distributions of inflammasome proteins. The importance of specific inflammasome proteins was studied by knocking down their expression through transfection of keratinocytes with lentiviral particles carrying short hairpin RNAs (shRNAs). RESULTS: D pteronyssinus activated caspase-1 and induced caspase-1-dependent release of IL-1ß and IL-18 from keratinocytes. Moreover, D pteronyssinus stimulated assembly of the inflammasome by recruiting apoptosis-associated specklike protein containing a caspase-recruitment domain (ASC), caspase-1, and nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin-domain containing 3 (NLRP3) to the perinuclear region. Finally, infection with lentiviral particles carrying ASC, caspase-1, or NLRP3 shRNAs suppressed the release of IL-1ß and IL-18 from the keratinocytes. Activation of the NLRP3 inflammasome by D pteronyssinus was dependent on cysteine protease activity. CONCLUSION: House dust mite allergens are danger signals for the skin. In addition, HDM-induced activation of the NLRP3 inflammasome may play a pivotal role in the pathogenesis of AD.


Asunto(s)
Antígenos Dermatofagoides/farmacología , Dermatophagoides pteronyssinus , Inflamasomas/efectos de los fármacos , Queratinocitos/efectos de los fármacos , Piel/efectos de los fármacos , Animales , Antígenos Dermatofagoides/metabolismo , Células Cultivadas , Dermatophagoides pteronyssinus/inmunología , Técnica del Anticuerpo Fluorescente , Humanos , Interleucina-1beta/inmunología , Interleucina-8/inmunología , Queratinocitos/inmunología , Piel/inmunología
12.
Aging (Albany NY) ; 14(6): 2695-2719, 2022 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-35347086

RESUMEN

The beneficial effects of caloric restriction (CR) against cardiac aging and for prevention of cardiovascular diseases are numerous. However, to our knowledge, there is no scientific evidence about how a high-calorie diet (HCD) background influences the mechanisms underlying CR in whole heart tissue (WHT) in experimental murine models. In the current study, CR-treated mice with different alimentary backgrounds were subjected to transthoracic echocardiographic measurements. WHT was then analyzed to determine cardiac energetics, telomerase activity, the expression of energy-sensing networks, tissue-specific adiponectin, and cardiac precursor/cardiac stem cell markers. Animals with a balanced diet consumption before CR presented marked cardiac remodeling with improved ejection fraction (EF) and fractional shortening (FS), enhanced OXPHOS complex I, III, and IV, and CKMT2 enzymatic activity. Mice fed an HCD before CR presented moderate changes in cardiac geometry with diminished EF and FS values, but improved OXPHOS complex IV and CKMT2 activity. Differences in cardiac remodeling, left ventricular systolic/diastolic performance, and mitochondrial energetics, found in the CR-treated mice with contrasting alimentary backgrounds, were corroborated by inconsistencies in the expression of mitochondrial-biogenesis-related markers and associated regulatory networks. In particular, disruption of eNOS and AMPK -PGC-1α-mTOR-related axes. The impact of a past habit of caloric overload on the effects of CR in the WHT is a scarcely explored subject that requires deeper study in combination with analyses of other tissues and organs at higher levels of organization within the organ system. Such research will eventually lead to the development of preventative and therapeutic strategies to promote health and longevity.


Asunto(s)
Restricción Calórica , Promoción de la Salud , Envejecimiento/metabolismo , Animales , Ratones , Mitocondrias/metabolismo , Biogénesis de Organelos
13.
J Appl Biomater Funct Mater ; 19: 22808000211035061, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34519565

RESUMEN

BACKGROUND: Human umbilical cord mesenchymal stem cells (HuMSCs) injected directly have been proven effective for improving chronic wounds. However, HuMSCs largely die within 14 days. The aim of study is to establish a cellularly modified gelatin sponge and investigate its characteristics and clinical potential. METHODS: HuMSCs were isolated, expanded and seeded in a poly-L-lysine (PLL)-coated gelatin sponge. Fabricated gelatin sponges were estimated through observation of morphological surface and ultrastructure, following confirmed by histology method. Supernatants were collected at different times for enzyme-linked immunosorbent assays (ELISAs) to measure growth factors. The cell embedded gelatin sponges were implanted subcutaneously on the backs of mice and the samples were harvested and studied histologically. RESULTS: HuMSCs gradually modified the gelatin sponge by depositing collagen and hyaluronic acid, and degrading the structure of gelatin, resulting in a dense, and elastic structure. Compared with cells cultured in monolayer, the levels of growth factors increased remarkably when HuMSCs were cultivated in the gelatin sponge. Upon subcutaneous implantation in the backs of mice, the cellularized gelatin sponges persisted for up to 2 months and eventually integrated into the host tissue, while blank gelatin sponges degraded completely by the end of the second month. CONCLUSION: Gelatin sponge is a clinically accessible scaffold for HuMSCs implantation to maintain short-term survival of the cells and high-level production of growth factors, which demonstrates good clinical potential for enhancing wound healing.


Asunto(s)
Colágeno , Gelatina , Animales , Ácido Hialurónico , Ratones , Cicatrización de Heridas
14.
Eur J Immunol ; 39(10): 2779-88, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19731362

RESUMEN

IL-20 cytokine subfamily members, including IL-19, IL-20, and IL-24, are highly expressed in psoriatic skin lesions. Here, we demonstrate that psoriasis mediators IL-17 and IL-22 synergistically induce the production of IL-20 subfamily proteins in cultured human keratinocytes. Interestingly, expression of the IL-22 receptor (IL-22R) also increased in epidermal lesions versus normal skin. IL-22R over-expression using an adenoviral vector to mimic psoriatic conditions in cultured keratinocytes significantly enhanced IL-17- and IL-22-induced production of IL-20 subfamily cytokines. Furthermore, IL-17 and IL-22 coordinately enhanced MIP-3alpha, IL-8, and heparin-binding EGF-like growth factor (HB-EGF) production, depending on the amount of IL-22R expression. Additionally, because IL-20 and IL-24 share the IL-22R with IL-22, the function of IL-20 and IL-24 was also increased. IL-20 and IL-24 have effects similar to that of IL-22; IL-24 showed more potent expression than IL-20. A combination of IL-24 and IL-17 increased the production of MIP-3alpha, IL-8, and HB-EGF, as did a combination of IL-22 and IL-17. These data indicate that increased IL-22R expression in epidermal keratinocytes contributes to the pathogenesis of psoriasis through enhancing the coordinated effects of IL-22 and IL-17, inducing the production of the IL-20 subfamily, chemokines, and growth factors.


Asunto(s)
Interleucina-17/farmacología , Interleucinas/metabolismo , Interleucinas/farmacología , Queratinocitos/metabolismo , Psoriasis/metabolismo , Receptores de Interleucina/metabolismo , Células Cultivadas , Quimiocina CCL20/genética , Epidermis/metabolismo , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Factor de Crecimiento Similar a EGF de Unión a Heparina , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Interferón gamma/farmacología , Subunidad beta del Receptor de Interleucina-10/genética , Interleucina-1alfa/farmacología , Interleucina-8/genética , Interleucinas/genética , Queratinocitos/efectos de los fármacos , Modelos Biológicos , Fosforilación/efectos de los fármacos , Receptores de Interleucina/genética , Factor de Transcripción STAT3/metabolismo , Transducción Genética , Factor de Crecimiento Transformador alfa/genética , Factor de Necrosis Tumoral alfa/farmacología , beta-Defensinas/genética , Interleucina-22
15.
Stem Cell Res Ther ; 11(1): 225, 2020 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-32513263

RESUMEN

BACKGROUND: Acetaminophen (APAP) overdose is the common cause of acute liver failure (ALF) due to the oxidative damage of multiple cellular components. This study aimed to investigate whether plasma membrane vesicles (PMVs) from human umbilical cord mesenchymal stem cells (hUCMSCs) could be exploited as a novel stem cell therapy for APAP-induced liver injury. METHODS: PMVs from hUCMSCs were prepared with an improved procedure including a chemical enucleation step followed by a mechanical extrusion. PMVs of hUCMSCs were characterized and supplemented to hepatocyte cultures. Rescue of APAP-induced hepatocyte damage was evaluated. RESULTS: The hUCMSCs displayed typical fibroblastic morphology and multipotency when cultivated under adipogenic, osteogenic, or chondrogenic conditions. PMVs of hUCMSCs maintained the stem cell phenotype, including the presence of CD13, CD29, CD44, CD73, and HLA-ABC, but the absence of CD45, CD117, CD31, CD34, and HLA-DR on the plasma membrane surface. RT-PCR and transcriptomic analyses showed that PMVs were similar to hUCMSCs in terms of mRNA profile, including the expression of stemness genes GATA4/5/6, Nanog, and Oct1/2/4. GO term analysis showed that the most prominent reduced transcripts in PMVs belong to integral membrane components, extracellular vesicular exosome, and extracellular matrix. Immunofluorescence labeling/staining and confocal microscopy assays showed that PMVs enclosed cellular organelles, including mitochondria, lysosomes, proteasomes, and endoplasmic reticula. Incorporation of the fusogenic VSV-G viral membrane glycoprotein stimulated the endosomal release of PMV contents into the cytoplasm. Further, the addition of PMVs and a mitochondrial-targeted antioxidant Mito-Tempo into cultures of APAP-treated HepG2 cells resulted in reduced cell death, enhanced viability, and increased mitochondrial membrane potential. Lastly, this study demonstrated that the redox state and activities of aminotransferases were restored in APAP-treated HepG2 cells. CONCLUSIONS: The results suggest that PMVs from hUCMSCs could be used as a novel stem cell therapy for the treatment of APAP-induced liver injury.


Asunto(s)
Acetaminofén , Células Madre Mesenquimatosas , Acetaminofén/toxicidad , Diferenciación Celular , Membrana Celular , Tratamiento Basado en Trasplante de Células y Tejidos , Células Hep G2 , Humanos , Cordón Umbilical
16.
Int Immunol ; 20(7): 901-9, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18492658

RESUMEN

The epidermis is the primary boundary between the body and the environment, and it serves as the first line of defense against microbial pathogens. Production of chemokines and cytokines is an important step in the initiation of innate immune responses to viral infections. Epidermal keratinocytes produce IFN-alpha, -beta and macrophage inflammatory protein (MIP)-1alpha in response to double-stranded RNA (dsRNA) or viral infections. We showed that human keratinocytes produced cytokines [tumor necrosis factor (TNF)-alpha, IL-1beta and IL-15] and chemokines [MIP-1beta, RANTES and liver and activation-regulated chemokine (LARC)] in response to dsRNA, with activation of the nuclear factor kappaB (NF-kappaB), p38 mitogen-activated protein kinase (MAPK) and signal transducers and activators of transcription 1 (STAT1) pathways. To study the roles of these pathways in their production, we transfected keratinocytes with adenoviral vectors (Ax) carrying a dominant-negative form of inhibitor kappaB alpha (IkappaBalpha) (IkappaBalphaM), a dominant-negative mutant form of STAT1 (STAT1F) or suppressors of cytokine signaling 1 (SOCS1). Transfection with AxIkappaBalphaM or addition of a p38 inhibitor (SB203580) significantly decreased the dsRNA-mediated production of TNF-alpha, IL-1beta and MIP-1alpha, but not of IFN-beta, IL-15, MIP-1beta, RANTES or LARC. Transfection with AxSTAT1F or AxSOCS1 inhibited the dsRNA-mediated production of TNF-alpha, IL-15, MIP-1alpha, MIP-1beta, RANTES and LARC, but not IFN-beta or IL-1beta. In conclusion, the NF-kappaB, p38 MAPK and STAT1 pathways differentially regulate dsRNA-mediated innate immune responses in epidermal keratinocytes.


Asunto(s)
Queratinocitos/inmunología , FN-kappa B/inmunología , ARN Bicatenario/inmunología , Factor de Transcripción STAT1/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología , Células Cultivadas , Citocinas/inmunología , Citocinas/metabolismo , Inhibidores Enzimáticos/farmacología , Células Epidérmicas , Epidermis/inmunología , Regulación de la Expresión Génica , Humanos , Proteínas I-kappa B/genética , Proteínas I-kappa B/inmunología , Proteínas I-kappa B/metabolismo , Imidazoles/farmacología , Inhibidor NF-kappaB alfa , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Piridinas/farmacología , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/inmunología , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Transgenes/genética , Transgenes/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
J Dermatol Sci ; 50(1): 53-60, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18077140

RESUMEN

BACKGROUND: 1 alpha,25-Dihydroxyvitamin D3 (1 alpha,25(OH)2D3), the active form of vitamin D, suppresses keratinocyte proliferation, promotes keratinocyte differentiation, and induces involucrin expression. Peroxisome proliferation-activated receptors (PPARs) are ligand-activated transcription factors. It has been reported that PPARs stimulate keratinocyte differentiation and regulate the expression of differentiation molecules. OBJECTIVE: Keratinocytes treated with 1 alpha,25(OH)2D3 induced PPAR gamma, which was followed by increased involucrin expression. In this study, we investigated whether PPAR gamma is involved in the 1 alpha,25(OH)2D3-induced involucrin expression in human keratinocytes. METHODS: Subconfluent keratinocytes were treated with 10(-7)M 1 alpha,25(OH)2D3 for the indicated times, and PPAR and involucrin mRNA expression were determined by real-time RT-PCR. The levels of PPARs, involucrin, p38, and phospho-p38 proteins were assayed by Western blotting, and the DNA binding activities of PPAR gamma and AP-1 were investigated by electrophoretic mobility shift assays (EMSA). To examine the role of PPAR gamma in 1 alpha,25(OH)2D3 responses, recombinant adenovirus carrying a dominant-negative form of PPAR gamma (Axdn-PPAR gamma) was constructed and transfected into keratinocytes. The p38 inhibitor SB203580 was added to the cultures to evaluate the involvement of p38 in involucrin expression. RESULTS: 1 alpha,25(OH)2D3 induced PPAR gamma expression and stimulated PPAR gamma activity. The introduction of dn-PPAR gamma inhibited the expression of involucrin mRNA and protein induced by 1 alpha,25(OH)2D3, and suppressed AP-1 DNA binding activity. 1 alpha,25(OH)2D3 also triggered the phosphorylation of p38, which contributes to involucrin induction. Moreover, dn-PPAR gamma prevented the 1 alpha,25(OH)2D3-induced phosphorylation of p38. CONCLUSIONS: These results suggest that PPAR gamma regulates involucrin expression by controlling the AP-1 signal and p38 activation in 1 alpha,25(OH)2D3-induced keratinocyte differentiation.


Asunto(s)
Calcitriol/farmacología , Regulación de la Expresión Génica , Queratinocitos/metabolismo , PPAR gamma/fisiología , Precursores de Proteínas/genética , Células Cultivadas , Humanos , Factor de Transcripción AP-1/genética , Activación Transcripcional , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
Mol Med Rep ; 18(2): 1571-1581, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29901205

RESUMEN

In the present study, the effects and underlying mechanism of RbAp48 on the radiosensitivity of AGS gastric cancer cells was investigated. Cell proliferation was determined with an MTT assay. Flow cytometry was performed to evaluate the cell cycle and apoptosis. Reverse transcription­quantitative polymerase chain reaction and western blot analysis were performed to detect mRNA and protein expression, respectively, including RbAp48, phosphoinositide 3­kinase (PI3K) and protein kinase B (Akt). The results revealed that radiation enhanced the expression level of RbAp48 in AGS cells, and that RbAp48 combined with radiation reduced AGS cell proliferation. In addition, RbAp48 combined with radiation resulted in G2 phase arrest and induced apoptosis via regulation of the PI3K/Akt pathway. In conclusion, it was demonstrated that overexpression of RbAp48 enhanced the radiosensitivity of AGS gastric cancer cells via suppression of PI3K/Akt pathway activity, suggesting that RbAp48 may hold potential as a gene therapeutic strategy in the future, aiding in the treatment of gastric cancer.


Asunto(s)
Mucosa Gástrica/efectos de la radiación , Regulación Neoplásica de la Expresión Génica , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Tolerancia a Radiación/genética , Proteína 4 de Unión a Retinoblastoma/genética , Apoptosis/efectos de la radiación , Línea Celular Tumoral , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de la radiación , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patología , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína 4 de Unión a Retinoblastoma/metabolismo , Transducción de Señal
19.
DNA Cell Biol ; 37(6): 560-573, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29608334

RESUMEN

Epithelial mesenchyme transformation (EMT) of the medial edge epithelium (MEE) is the crucial process during palatal fusion. This work is aimed to elucidate the enhancer regulatory mechanism by genome-wide DNA methylation analysis of EMT during palatal fusion. Over 800 million clean reads, 325 million enzyme reads, and 234 million mapping reads were generated. The mapping rate was 68.85-74.32%, which included differentially methylated 17299 CCGG sites and 2363 CCWGG sites (p < 0.05, log2FC >1). Methylated sites in intron and intergenic regions were more compared to other regions of all DNA elements. GO and KEGG analysis indicated that differential methylation sites related to embryonic palatal fusion genes (HDAC4, TCF7L2, and PDGFRB) at the enhancer were located on CCWGG region of non-CpG islands. In addition, the results showed that the enhancer for HDAC4 was hypermethylated, whereas the enhancers for TCF7L2 and PDGFRB were hypomethylated. The methylation status of enhancer regions of HDAC4, PDGFRB, and TCF7L2, involved in the regulation of the EMT during palatal fusion, may enlighten the development of novel epigenetic biomarkers in the treatment of cleft palate.


Asunto(s)
Metilación de ADN , Transición Epitelial-Mesenquimal/genética , Genoma/genética , Hueso Paladar/metabolismo , Animales , Sitios de Unión/genética , Islas de CpG/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Histona Desacetilasas/genética , Masculino , Ratones Endogámicos C57BL , Hueso Paladar/embriología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Proteína 2 Similar al Factor de Transcripción 7/genética
20.
J Dermatol ; 34(8): 523-30, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17683382

RESUMEN

Platelet-derived growth factor (PDGF) is involved in wound healing, but PDGF-induced fibroblast migration and the intracellular signaling mechanisms of fibroblast migration are poorly understood. Signal transducer and activator of transcription 3 (STAT3) is involved in migration and is negatively regulated by the suppressor of cytokine signaling 3 (SOCS3). We studied the PDGF induction of fibroblast migration in vitro and the involvement of STAT3 and SOCS3. We found that PDGF activated STAT3 and strongly induced fibroblast migration. Transfection with a dominant-negative mutant of STAT3 almost completely abolished PDGF-induced fibroblast migration and STAT3 phosphorylation. Next, we studied the mechanisms that regulate fibroblast migration. PDGF enhanced the expression of SOCS3 by 2.8-fold at 1 h. Transfection with SOCS3 almost completely abolished PDGF-induced STAT3 phosphorylation and reduced fibroblast migration to 47% of control, indicating that SOCS3 acts as a negative regulator of PDGF-induced fibroblast migration. In conclusion, PDGF induces fibroblast migration under the control of STAT3-SOCS3.


Asunto(s)
Movimiento Celular/fisiología , Fibroblastos/fisiología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/fisiología , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Adenoviridae/genética , Western Blotting , Células Cultivadas , Fibroblastos/enzimología , Vectores Genéticos , Humanos , Mutación , Fosforilación , Reacción en Cadena de la Polimerasa , Transducción de Señal , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Transfección , Cicatrización de Heridas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA