Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Heliyon ; 9(4): e14777, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37025807

RESUMEN

The extracellular matrix (ECM) components present within all tissues and organs help to maintain the cytoskeletal architecture and tissue morphology. Although the ECM plays a role in cellular events and signaling pathways, it has not been well studied due its insolubility and complexity. Brain tissue has a higher cell density and weaker mechanical strength than other tissues in the body. When removing cells using a general decellularization method to produce scaffolds and obtain ECM proteins, various problems must be considered because tissues are easily damaged. To retain the brain shape and ECM components, we performed decellularization in combination with polymerization. We immersed mouse brains in oil for polymerization and decellularization via O-CASPER (Oil-based Clinically and Experimentally Applicable Acellular Tissue Scaffold Production for Tissue Engineering and Regenerative Medicine) and then isolated ECM components using sequential matrisome preparation reagents (SMPRs), namely, RIPA, PNGase F, and concanavalin A. Adult mouse brains were preserved with our decellularization method. Western blot and LC-MS/MS analyses revealed that ECM components, including collagen and laminin, were isolated efficiently from decellularized mouse brains using SMPRs. Our method will be useful to obtain matrisomal data and perform functional studies using adult mouse brains and other tissues.

2.
Stem Cell Res Ther ; 14(1): 19, 2023 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-36737811

RESUMEN

BACKGROUND: The generation of liver organoids recapitulating parenchymal and non-parenchymal cell interplay is essential for the precise in vitro modeling of liver diseases. Although different types of multilineage liver organoids (mLOs) have been generated from human pluripotent stem cells (hPSCs), the assembly and concurrent differentiation of multiple cell types in individual mLOs remain a major challenge. Particularly, most studies focused on the vascularization of mLOs in host tissue after transplantation in vivo. However, relatively little information is available on the in vitro formation of luminal vasculature in mLOs themselves. METHODS: The mLOs with luminal blood vessels and bile ducts were generated by assembling hepatic endoderm, hepatic stellate cell-like cells (HscLCs), and endothelial cells derived entirely from hPSCs using 96-well ultra-low attachment plates. We analyzed the effect of HscLC incorporation and Notch signaling modulation on the formation of both bile ducts and vasculature in mLOs using immunofluorescence staining, qRT-PCR, ELISA, and live-perfusion imaging. The potential use of the mLOs in fibrosis modeling was evaluated by histological and gene expression analyses after treatment with pro-fibrotic cytokines. RESULTS: We found that hPSC-derived HscLCs are crucial for generating functional microvasculature in mLOs. HscLC incorporation and subsequent vascularization substantially reduced apoptotic cell death and promoted the survival and growth of mLOs with microvessels. In particular, precise modulation of Notch signaling during a specific time window in organoid differentiation was critical for generating both bile ducts and vasculature. Live-cell imaging, a series of confocal scans, and electron microscopy demonstrated that blood vessels were well distributed inside mLOs and had perfusable lumens in vitro. In addition, exposure of mLOs to pro-fibrotic cytokines induced early fibrosis-associated events, including upregulation of genes associated with fibrotic induction and endothelial cell activation (i.e., collagen I, α-SMA, and ICAM) together with destruction of tissue architecture and organoid shrinkage. CONCLUSION: Our results demonstrate that mLOs can reproduce parenchymal and non-parenchymal cell interactions and suggest that their application can advance the precise modeling of liver diseases in vitro.


Asunto(s)
Hepatopatías , Células Madre Pluripotentes , Humanos , Conductos Biliares , Citocinas/metabolismo , Células Endoteliales , Fibrosis , Hígado , Organoides/metabolismo , Receptores Notch
3.
Sci Rep ; 12(1): 5645, 2022 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-35379891

RESUMEN

Formaldehyde is a colorless, pungent, highly reactive, and toxic environmental pollutant used in various industries and products. Inhaled formaldehyde is a human and animal carcinogen that causes genotoxicity, such as reactive oxygen species formation and DNA damage. This study aimed to identify the toxic effects of inhaled formaldehyde through an integrated toxicogenomic approach utilizing database information. Microarray datasets (GSE7002 and GSE23179) were collected from the Gene Expression Omnibus database, and differentially expressed genes were identified. The network analyses led to the construction of the respiratory system-related biological network associated with formaldehyde exposure, and six upregulated hub genes (AREG, CXCL2, HMOX1, PLAUR, PTGS2, and TIMP1) were identified. The expression levels of these genes were verified via qRT-PCR in 3D reconstructed human airway tissues exposed to aerosolized formaldehyde. Furthermore, NRARP was newly found as a potential gene associated with the respiratory and carcinogenic effects of formaldehyde by comparison with human in vivo and in vitro formaldehyde-exposure data. This study improves the understanding of the toxic mechanism of formaldehyde and suggests a more applicable analytic pipeline for predicting the toxic effects of inhaled toxicants.


Asunto(s)
Formaldehído , Exposición por Inhalación , Animales , Carcinógenos/toxicidad , Formaldehído/efectos adversos , Formaldehído/metabolismo , Formaldehído/toxicidad , Exposición por Inhalación/efectos adversos , Hipersensibilidad Respiratoria , Toxicogenética
4.
J Cancer Prev ; 26(2): 83-97, 2021 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-34258247

RESUMEN

The exponential growth of nanotechnology and the industrial production have raised concerns over its impact on human and environmental health and safety (EHS). Although there has been substantial progress in the assessment of pristine nanoparticle toxicities, their EHS impacts require greater clarification. In this review, we discuss studies that have assessed nanoparticle eco-genotoxicity in different test systems and their fate in the environment as well as the considerable confounding factors that may complicate the results. We highlight key mechanisms of nanoparticle-mediated genotoxicity. Then we discuss the reliability of endpoint assays, such as the comet assay, the most favored assessment technique because of its versatility to measure low levels of DNA strand breakage, and the micronucleus assay, which is complementary to the former because of its greater ability to detect chromosomal DNA fragmentation. We also address the current recommendations on experimental design, including environmentally relevant concentrations and suitable exposure duration to avoid false-positive or -negative results. The genotoxicity of nanoparticles depends on their physicochemical features and the presence of co-pollutants. Thus, the effect of environmental processes (e.g., aggregation and agglomeration, adsorption, and transformation of nanoparticles) would account for when determining the actual genotoxicity relevant to environmental systems, and assay procedures must be standardized. Indeed, the engineered nanoparticles offer potential applications in different fields including biomedicine, environment, agriculture, and industry. Toxicological pathways and the potential risk factors related to genotoxic responses in biological organisms and environments need to be clarified before appropriate and sustainable applications of nanoparticles can be established.

5.
Stem Cell Res ; 26: 95-102, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29272857

RESUMEN

During early development in placental mammals, proper trophoblast lineage development is essential for implantation and placentation. Defects in this lineage can cause early pregnancy failures and other pregnancy disorders. However, transcription factors controlling trophoblast development remain poorly understood. Here, we utilize Fosl1, previously implicated in trophoblast giant cell development as a member of the AP-1 complex, to trans-differentiate embryonic stem (ES) cells to trophoblast lineage-like cells. We first show that the ectopic expression of Fosl1 is sufficient to induce trophoblast-specific gene expression programs in ES cells. Surprisingly, we find that this transcriptional reprogramming occurs independently of changes in levels of ES cell core factors during the cell fate change. This suggests that Fosl1 acts in a novel way to orchestrate the ES to trophoblast cell fate conversion compared to previously known reprogramming factors. Mapping of Fosl1 targets reveals that Fosl1 directly activates TE lineage-specific genes as a pioneer factor. Our work suggests Fosl1 may be used to reprogram ES cells into differentiated cell types in trophoblast lineage, which not only enhances our knowledge of global trophoblast gene regulation but also may provide a future therapeutic tool for generating induced trophoblast cells from patient-derived pluripotent stem cells.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Madre Pluripotentes/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Trofoblastos/metabolismo , Animales , Células Cultivadas , Células Madre Embrionarias/citología , Femenino , Ratones , Células Madre Pluripotentes/citología , Embarazo , Proteínas Proto-Oncogénicas c-fos/genética , Trofoblastos/citología
6.
Mol Med Rep ; 16(2): 1079-1086, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28586027

RESUMEN

Stichopus japonicus has been used as a folk medicine and as an ingredient in traditional food in East Asian countries. In recent years, the bioactive compounds found in S. japonicus have been reported to possess efficacy in wound healing and may be of potential use in the cosmeceutical, pharmaceutical and biomedical industries. Although the components and their functions require further investigation, S. japonicus extracts exhibit anti­inflammatory properties, and may be used for cancer prevention and treatment. Although several reports have examined different aspects of S. japo-nicus, the effects of S. japonicus extract on melanogenesis in the skin has not been reported to date. Therefore the present study aimed to investigate the effects of S. japonicus extract on melanogenesis. Treatment with a mixture of S. japonicus extracts (MSCE) reduced melanin synthesis and tyrosinase (TYR) activity in mouse melanocyte cells lines, B16F10 and Melan­A. In addition, MSCE treatment reduced the protein expression levels of TYR, tyrosinase­related protein­1 and tyrosinase­related protein­2. The reduced protein levels may be the result of decreased microphthalmia­associated transcription factor (MITF) expression, which is an important regulator of melanogenesis. The reduced expression level of MITF was associated with delayed phosphorylation of extracellular signal­regulated kinase (ERK) induced by MSCE treatment. A specific MEK inhibitor, PD98059, significantly blocked MSCE­mediated inhibition of melanin synthesis. In conclusion, these results indicate that MSCE may be useful as a potential skin­whitening compound in the skin medical industry.


Asunto(s)
Mezclas Complejas/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Melaninas/biosíntesis , Stichopus/química , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Melanocitos/efectos de los fármacos , Melanocitos/metabolismo , Melanoma Experimental/enzimología , Melanoma Experimental/patología , Ratones , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
8.
Mol Med Rep ; 15(5): 2681-2688, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28447741

RESUMEN

The present study aimed to investigate the anti­inflammatory effect and mechanism of action of isosecotanapartholide (ISTP), isolated from Artemisia princeps Pampanini extract (APE). The effects of ISTP and APE on the proliferation of human keratinocytes following stimulation by tumor necrosis factor­α/interferon­Î³ were assessed. ISTP and APE downregulated the expression levels of signal transducer and activator of transcription­1 (STAT­1), and reduced interleukin­33 (IL­33) production. ISTP and APE inhibited the mRNA expression levels of thymus and activation­regulated chemokine (TARC/CCL17) in a dose­dependent manner. Western blot analysis demonstrated that ISTP and APE dose­dependently inhibited protein expression levels of intercellular adhesion molecule­1 and phosphorylation of STAT­1. The results of the present study indicate that ISTP may inhibit TARC/CCL17 production in human epidermal keratinocytes via the STAT­1 signaling pathway and may be associated with the inhibition of IL­33 production. The current study indicated that ISTP is an active component in APE and may be a potential therapeutic agent for the treatment of inflammatory skin disorders.


Asunto(s)
Artemisia/química , Interleucina-33/biosíntesis , Queratinocitos/metabolismo , Extractos Vegetales/farmacología , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/efectos de los fármacos , Línea Celular , Humanos , Queratinocitos/citología , Extractos Vegetales/química
9.
Biosci Biotechnol Biochem ; 81(7): 1348-1355, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28372490

RESUMEN

Multiple etiologies of liver injury are associated with fibrosis in which the key event is the activation of hepatic stellate cells (HSCs). Although microRNAs (miRNAs) are reportedly involved in fibrogenesis, the complete array of miRNA signatures associated with the disease has yet to be elucidated. Here, deep sequencing analysis revealed that compared to controls, 80 miRNAs were upregulated and 21 miRNAs were downregulated significantly in the thioacetamide (TAA)-induced mouse fibrotic liver. Interestingly, 58 of the upregulated miRNAs were localized to an oncogenic miRNA megacluster upregulated in liver cancer. Differential expression of some of the TAA-responsive miRNAs was confirmed, and their human orthologs were similarly deregulated in TGF-ß1-activated HSCs. Moreover, a functional analysis of the experimentally validated high-confidence miRNA targets revealed significant enrichment for the GO terms and KEGG pathways involved in HSC activation and liver fibrogenesis. This is the first comprehensive report of miRNAs profiles during TAA-induced mouse liver fibrosis.


Asunto(s)
Regulación de la Expresión Génica , Células Estrelladas Hepáticas/metabolismo , Cirrosis Hepática/genética , Hígado/metabolismo , MicroARNs/genética , Actinas/genética , Actinas/metabolismo , Animales , Línea Celular Transformada , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/patología , Humanos , Hígado/efectos de los fármacos , Hígado/patología , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Anotación de Secuencia Molecular , Transducción de Señal , Tioacetamida , Factor de Crecimiento Transformador beta1/farmacología
10.
BMB Rep ; 50(2): 58-59, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28115038

RESUMEN

The beneficial paracrine roles of mesenchymal stem cells (MSCs) in tissue repair have potential in therapeutic strategies against various diseases. However, the key therapeutic factors secreted from MSCs and their exact molecular mechanisms of action remain unclear. In this study, the cell-free secretome of umbilical cord-derived MSCs showed significant anti-fibrotic activity in the mouse models of liver fibrosis. The involved action mechanism was the regulation of hepatic stellate cell activation by direct inhibition of the TGFß/Smad-signaling. Antagonizing the milk fat globule-EGF factor 8 (MFGE8) activity blocked the anti-fibrotic effects of the MSC secretome in vitro and in vivo. Moreover, MFGE8 was secreted by MSCs from the umbilical cord as well as other tissues, including teeth and bone marrow. Administration of recombinant MFGE8 protein alone had a significant anti-fibrotic effect in two different models of liver fibrosis. Additionally, MFGE8 downregulated TGFß type I receptor expression by binding to αvß3 integrin on HSCs. These findings revealed the potential role of MFGE8 in modulating TGFß-signaling. Thus, MFGE8 could serve as a novel therapeutic agent for liver fibrosis. [BMB Reports 2017; 50(2): 58-59].


Asunto(s)
Antígenos de Superficie/aislamiento & purificación , Antígenos de Superficie/fisiología , Cirrosis Hepática/prevención & control , Células Madre Mesenquimatosas/metabolismo , Proteínas de la Leche/aislamiento & purificación , Proteínas de la Leche/metabolismo , Animales , Células Cultivadas , Humanos , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Trasplante de Células Madre Mesenquimatosas , Metaboloma/fisiología , Ratones
11.
Gastroenterology ; 152(5): 1174-1186, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27956229

RESUMEN

BACKGROUND & AIMS: Mesenchymal stem cells (MSCs) mediate tissue repair and might be used to prevent or reduce liver fibrosis. However, little is known about the anti-fibrotic factors secreted from MSCs or their mechanisms. METHODS: Umbilical cord-derived MSCs (UCMSCs) were differentiated into hepatocyte-like cells (hpUCMSCs), medium was collected, and secretome proteins were identified and quantified using nanochip-liquid chromatography/quadrupole time-of-flight mass spectrometry. Liver fibrosis was induced in mice by intraperitoneal injection of thioacetamide or CCl4; some mice were then given injections of secretomes or proteins. Liver tissues were collected and analyzed by histology or polymerase chain reaction array to analyze changes in gene expression patterns. We analyzed the effects of MSC secretomes and potential anti-fibrotic proteins on transforming growth factor ß 1 (TGFß1)-mediated activation of human hepatic stellate cell (HSC) lines (hTert-HSC and LX2) and human primary HSCs. Liver tissues were collected from 16 patients with liver cirrhosis and 16 individuals without cirrhosis (controls) in Korea and analyzed by immunohistochemistry and immunoblots. RESULTS: In mice with fibrosis, accumulation of extracellular matrix proteins was significantly reduced 3 days after injecting secretomes from UCMSCs, and to a greater extent from hpUCMSCs; numbers of activated HSCs that expressed the myogenic marker α-smooth muscle actin (α-SMA, encoded by ACTA2 [actin, alpha 2, smooth muscle]) were also reduced. Secretomes from UCMSCs, and to a greater extent from hpUCMSCs, reduced liver expression of multiple fibrotic factors, collagens, metalloproteinases, TGFß, and Smad proteins in the TGFß signaling pathways. In HSC cell lines and primary HSCs, TGFß1-stimulated upregulation of α-SMA was significantly inhibited (and SMAD2 phosphorylation reduced) by secretomes from UCMSCs, and to a greater extent from hpUCMSCs. We identified 32 proteins in secretomes of UCMSCs that were more highly concentrated in secretomes from hpUCMSCs and inhibited TGFß-mediated activation of HSCs. One of these, milk fat globule-EGF factor 8 (MFGE8), was a strong inhibitor of activation of human primary HSCs. We found MFGE8 to down-regulate expression of TGFß type I receptor by binding to αvß3 integrin on HSCs and to be secreted by MSCs from umbilical cord, teeth, and bone marrow. In mice, injection of recombinant human MFGE8 had anti-fibrotic effects comparable to those of the hpUCMSC secretome, reducing extracellular matrix deposition and HSC activation. Co-injection of an antibody against MFGE8 reduced the anti-fibrotic effects of the hpUCMSC secretome in mice. Levels of MFGE8 were reduced in cirrhotic liver tissue from patients compared with controls. CONCLUSIONS: MFGE8 is an anti-fibrotic protein in MSC secretomes that strongly inhibits TGFß signaling and reduces extracellular matrix deposition and liver fibrosis in mice.


Asunto(s)
Antígenos de Superficie/metabolismo , Cirrosis Hepática/metabolismo , Proteínas de la Leche/metabolismo , Animales , Tetracloruro de Carbono/toxicidad , Línea Celular , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Células Estrelladas Hepáticas , Hepatocitos , Humanos , Integrina alfaVbeta3/metabolismo , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/patología , Masculino , Células Madre Mesenquimatosas/metabolismo , Metaboloma , Metaloproteasas/metabolismo , Ratones , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteína Smad2/metabolismo , Tioacetamida/toxicidad , Factor de Crecimiento Transformador beta1/metabolismo
12.
Dermatol Ther ; 29(6): 463-465, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27543426

RESUMEN

Filler granuloma is considered to be the result of delayed immune responses; growing evidence suggests that they may be secondary to biofilm formation. Dermal filler is technically a foreign body, and as the development of newer generations of dermal fillers lengthens their duration, it is possible that there is also an increased risk of biofilm formation. Here, we present a case report of a patient with Streptococcus sanguinis isolated from a filler granuloma, suggestive of biofilm formation. This case demonstrates the effective use of antibiotics after incision and drainage on antibiotic resistant biofilm.


Asunto(s)
Antibacterianos/uso terapéutico , Biopelículas/efectos de los fármacos , Técnicas Cosméticas/efectos adversos , Rellenos Dérmicos/efectos adversos , Drenaje , Granuloma de Cuerpo Extraño/terapia , Infecciones Estreptocócicas/terapia , Streptococcus sanguis/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Biopsia , Terapia Combinada , Rellenos Dérmicos/administración & dosificación , Femenino , Granuloma de Cuerpo Extraño/diagnóstico , Granuloma de Cuerpo Extraño/microbiología , Humanos , Imagen por Resonancia Magnética , Persona de Mediana Edad , Infecciones Estreptocócicas/diagnóstico , Infecciones Estreptocócicas/microbiología , Streptococcus sanguis/crecimiento & desarrollo , Streptococcus sanguis/aislamiento & purificación , Resultado del Tratamiento
13.
Lasers Med Sci ; 31(7): 1437-45, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27394442

RESUMEN

Phototherapy with 311-nm narrowband-UVB (NBUVB) is an effective adjuvant treatment modality for atopic dermatitis (AD). In this study, we evaluated the therapeutic effect of the newly developed gain-switched 311-nm Ti:Sapphire laser device using a NC/Nga mouse AD model. A total number of 50 mice were used in this study. Atopic dermatitis (AD) was induced in mice by exposure to Dermatophagoides farina. These, NC/Nga mice were then treated with conventional 311-nm NBUVB or the newly developed gain-switched 311-nm Ti:Sapphire laser. The clinical features, dermatitis severity scores, and scratching behavior were assessed. In addition, serologic analyses including inflammatory cytokines and histological analyses were performed. Gain-switched 311-nm Ti:Sapphire laser improved the AD-like skin lesions, severity, and symptoms of AD in the NC/Nga mouse model. This new laser also modulated the immune response found in the AD model, including hyper-IgE, upregulated Th2 cytokines, and the Th2-mediated allergic inflammatory reaction. Gain-switched 311-nm Ti:Sapphire laser shows therapeutic promise via an immune-modulation mechanism in an AD mouse model. These data suggest that gain-switched 311-nm Ti:Sapphire laser may be useful as a targeted phototherapy modality for AD.


Asunto(s)
Óxido de Aluminio/química , Dermatitis Atópica/radioterapia , Terapia por Láser , Animales , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Hipersensibilidad/inmunología , Hipersensibilidad/patología , Inmunoglobulina E/biosíntesis , Inflamación/inmunología , Inflamación/patología , Mediadores de Inflamación/metabolismo , Ratones , Piel/patología , Piel/efectos de la radiación , Células Th2/inmunología
14.
Lasers Surg Med ; 48(6): 629-37, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27075746

RESUMEN

BACKGROUND AND OBJECTIVE: Atopic dermatitis (AD) is a common inflammatory skin disease that can affect all age groups. It has a relapsing course, which dramatically affects the quality of life of patients. A 308-nm excimer laser has been reported to be a safe and effective treatment for inflammatory skin diseases, although the range of potential application has not been fully explored. The purpose of this study was to evaluate the therapeutic effects of a 308-nm laser on AD-like skin lesions in NC/Nga mice. STUDY DESIGN/MATERIALS AND METHODS: Dermatophagoides farinae-exposed NC/Nga mice with a clinical score of 12 were treated with either a 308-nm excimer laser or narrowband-UVB (NB-UVB). The effects of the 308-nm excimer laser were evaluated by dermatitis scores, skin histology, skin barrier function, and immunological parameters, including IgE and Th2-mediated cytokines. RESULTS: The 308-nm excimer laser significantly reduced the severity of skin lesions and decreased the total serum levels of IgE and Th2-mediated cytokines. The excimer laser also significantly reduced the inflammatory cellular infiltrate into AD-induced skin lesions. Moreover, treatment with the 308-nm excimer laser led to recovery of skin barrier function in AD-induced skin lesions. CONCLUSION: The 308-nm excimer laser can be considered a valid and safe therapeutic option for the treatment of localized AD. Lasers Surg. Med. 48:629-637, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Dermatitis Atópica/cirugía , Láseres de Excímeros/uso terapéutico , Animales , Biomarcadores/sangre , Citocinas/sangre , Dermatitis Atópica/sangre , Dermatitis Atópica/patología , Masculino , Ratones , Resultado del Tratamiento
15.
J Korean Med Sci ; 31(2): 171-7, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26839468

RESUMEN

We investigated the potential of human dental pulp stem cells (hDPSCs) to differentiate into dopaminergic neurons in vitro as an autologous stem cell source for Parkinson's disease treatment. The hDPSCs were expanded in knockout-embryonic stem cell (KO-ES) medium containing leukemia inhibitory factor (LIF) on gelatin-coated plates for 3-4 days. Then, the medium was replaced with KO-ES medium without LIF to allow the formation of the neurosphere for 4 days. The neurosphere was transferred into ITS medium, containing ITS (human insulin-transferrin-sodium) and fibronectin, to select for Nestin-positive cells for 6-8 days. The cells were then cultured in N-2 medium containing basic fibroblast growth factor (FGF), FGF-8b, sonic hedgehog-N, and ascorbic acid on poly-l-ornithine/fibronectin-coated plates to expand the Nestin-positive cells for up to 2 weeks. Finally, the cells were transferred into N-2/ascorbic acid medium to allow for their differentiation into dopaminergic neurons for 10-15 days. The differentiation stages were confirmed by morphological, immunocytochemical, flow cytometric, real-time PCR, and ELISA analyses. The expressions of mesenchymal stem cell markers were observed at the early stages. The expressions of early neuronal markers were maintained throughout the differentiation stages. The mature neural markers showed increased expression from stage 3 onwards. The percentage of cells positive for tyrosine hydroxylase was 14.49%, and the amount was 0.526 ± 0.033 ng/mL at the last stage. hDPSCs can differentiate into dopaminergic neural cells under experimental cell differentiation conditions, showing potential as an autologous cell source for the treatment of Parkinson's disease.


Asunto(s)
Diferenciación Celular , Pulpa Dental/citología , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Encéfalo/patología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Medios de Cultivo/química , Medios de Cultivo/farmacología , Neuronas Dopaminérgicas/patología , Ensayo de Inmunoadsorción Enzimática , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Ratones , Ratones Endogámicos ICR , Proteína Básica de Mielina/genética , Proteína Básica de Mielina/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Antígenos Embrionarios Específico de Estadio/genética , Antígenos Embrionarios Específico de Estadio/metabolismo , Células Madre/patología , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Tirosina 3-Monooxigenasa/análisis , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
16.
Arch Plast Surg ; 43(1): 105-7, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26848458
17.
J Immunol Res ; 2016: 5781070, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28074193

RESUMEN

The study investigated the association between disease activity and serum 25-hydroxyvitamin D3 (25(OH)-D3), B cell activation of the tumor necrosis factor family (BAFF), or ß2 microglobulin in patients with primary Sjogren's syndrome (SS). Sixty-nine primary SS patients and 22 sicca control patients were included in the study. Disease activity was measured with EULAR Sjogren's syndrome disease activity index (ESSDAI). Serum levels of 25(OH)-D3 and ß2 microglobulin were measured by radioimmunoassay and BAFF was measured by an enzyme-linked immunosorbent assay. Serum levels of 25(OH)-D3 were significantly lower in SS patients compared to the sicca controls (p = 0.036). Serum levels of BAFF tended to be higher (p = 0.225) and those of ß2 microglobulin were significantly higher in patients with SS than in sicca controls (p = 0.023). In univariate regression analyses, ESSDAI was significantly associated with serum levels of 25(OH)-D3, BAFF, and ß2 microglobulin. After stepwise backward multivariate linear regression analyses including age and acute phase reactants, ESSDAI was associated with 25(OH)-D3 (ß = -0.042, p = 0.015) and BAFF (ß = 0.001, p = 0.015) in SS patients. In SS patients, ESSDAI is negatively associated with serum levels of 25(OH)-D3 and positively associated with BAFF.


Asunto(s)
Factor Activador de Células B/sangre , Síndrome de Sjögren/sangre , Vitamina D/análogos & derivados , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunoensayo , Masculino , Persona de Mediana Edad , Síndrome de Sjögren/etiología , Vitamina D/sangre , Microglobulina beta-2/sangre
18.
Photodermatol Photoimmunol Photomed ; 32(3): 120-8, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26577060

RESUMEN

BACKGROUND: Mesenchymal stem cells (MSCs) are promising therapeutic agents for various diseases. AIMS: To investigate the effects of conditioned medium from human bone marrow-derived mesenchymal stem cells (MSC-CdM) on pro-collagen production and wrinkle formation, we performed in vitro and in vivo experiments. METHODS: We assessed the effects of MSC-CdM on proliferation and photo-aging in human dermal fibroblasts after UVB exposure using enzyme activity assays for collagen type I secretion and MMP-1. To determine the effect of topically applied MSC-CdM on wrinkle formation, MSC-CdM (1% and 10%) and vehicle (propylene glycol: ethanol, 7 : 3) were applied to the dorsal skin of UVB-irradiated hairless mice for 8 weeks. We examined the effects on wrinkle formation by assessing visual skin grading, replica, tape stripping, transepidermal water loss (TEWL), and skin hydration measurement. We also examined histology of the lesions using hematoxylin-eosin, Masson's trichrome, and immunohistochemical staining. RESULTS: MSC-CdM markedly reduced UV-induced matrix metalloproteinase-1 expression and increased pro-collagen synthesis in a dose-dependent manner. Our findings suggest that MSC-CdM induces repair of dermal damage and effacement of wrinkles on UVB-irradiated hairless mice through protective effect of hydration. CONCLUSION: These results support an anti-wrinkle effect of MSC-CdM that involves increased collagen synthesis and suggest that MSC-CdM might be a potential candidate for preventing UV-induced skin damage.


Asunto(s)
Células Madre Mesenquimatosas , Envejecimiento de la Piel/efectos de los fármacos , Piel/efectos de los fármacos , Piel/metabolismo , Administración Cutánea , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Colágeno Tipo I/metabolismo , Medios de Cultivo Condicionados/farmacología , Tejido Elástico/efectos de los fármacos , Tejido Elástico/patología , Tejido Elástico/efectos de la radiación , Femenino , Fibroblastos , Humanos , Masculino , Metaloproteinasa 1 de la Matriz/metabolismo , Ratones , Ratones Pelados , Procolágeno/biosíntesis , Piel/efectos de la radiación , Envejecimiento de la Piel/patología , Envejecimiento de la Piel/efectos de la radiación , Rayos Ultravioleta/efectos adversos
19.
J Korean Med Sci ; 30(12): 1754-63, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26713050

RESUMEN

We conducted this study to investigate the synergistic effect of human urine-derived stem cells (USCs) and surface modified composite scaffold for bladder reconstruction in a rat model. The composite scaffold (Polycaprolactone/Pluronic F127/3 wt% bladder submucosa matrix) was fabricated using an immersion precipitation method, and heparin was immobilized on the surface via covalent conjugation. Basic fibroblast growth factor (bFGF) was loaded onto the heparin-immobilized scaffold by a simple dipping method. In maximal bladder capacity and compliance analysis at 8 weeks post operation, the USCs-scaffold(heparin-bFGF) group showed significant functional improvement (2.34 ± 0.25 mL and 55.09 ± 11.81 µL/cm H2O) compared to the other groups (2.60 ± 0.23 mL and 56.14 ± 9.00 µL/cm H2O for the control group, 1.46 ± 0.18 mL and 34.27 ± 4.42 µL/cm H2O for the partial cystectomy group, 1.76 ± 0.22 mL and 35.62 ± 6.69 µL/cm H2O for the scaffold group, and 1.92 ± 0.29 mL and 40.74 ± 7.88 µL/cm H2O for the scaffold(heparin-bFGF) group, respectively). In histological and immunohistochemical analysis, the USC-scaffold(heparin-bFGF) group showed pronounced, well-differentiated, and organized smooth muscle bundle formation, a multi-layered and pan-cytokeratin-positive urothelium, and high condensation of submucosal area. The USCs seeded scaffold(heparin-bFGF) exhibits significantly increased bladder capacity, compliance, regeneration of smooth muscle tissue, multi-layered urothelium, and condensed submucosa layers at the in vivo study.


Asunto(s)
Células Madre Adultas/trasplante , Ingeniería de Tejidos/métodos , Vejiga Urinaria/cirugía , Orina/citología , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Animales , Materiales Biocompatibles/química , Diferenciación Celular , Factor 2 de Crecimiento de Fibroblastos/administración & dosificación , Heparina/administración & dosificación , Humanos , Ensayo de Materiales , Modelos Animales , Poloxámero , Poliésteres , Ratas , Procedimientos de Cirugía Plástica , Regeneración , Andamios del Tejido/química , Vejiga Urinaria/anatomía & histología , Vejiga Urinaria/fisiología
20.
Lasers Surg Med ; 47(10): 824-32, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26413796

RESUMEN

BACKGROUND AND OBJECTIVES: Ultraviolet light-emitting diodes (UV-LEDs) are a novel light source for phototherapy. This research investigated the in vitro safety and efficacy of UV-LEDs as a phototherapeutic device for atopic dermatitis (AD). STUDY DESIGN/MATERIALS AND METHODS: Human keratinocytes and fibroblasts were irradiated by UV-LEDs with a center wavelength of 310 and 340 nm. We examined the effects of UV-LED irradiation on the suppression of TNF-α/IFN-γ-induced activation of STAT1 and ICAM-1 and on NF-κB expression; we used the following methods: cell viability assay, reverse transcription-polymerase chain reaction, enzyme-linked immunosorbent assay, Western blotting, and immunocytochemistry. RESULTS: We observed anti-inflammatory responses through the suppression of TNF-α/IFN-γ-induced expression of TARC and MCP-1/CCL2, IL-1beta, IL-6, and sICAM-1 via blockage of ICAM-1 activation and subsequent activation of STAT1 and NF-κB. The results suggested that UV-LED irradiation inhibited ICAM expression by suppressing TNF-α/IFN-γ-induced NF-κB activation in vitro. CONCLUSION: We concluded that novel UV-LED (310 and 340 nm) modalities were effective for the treatment of AD and may be promising for the treatment of inflammatory skin diseases.


Asunto(s)
Molécula 1 de Adhesión Intercelular/metabolismo , Interferón gamma/metabolismo , Queratinocitos/efectos de la radiación , Fosforilación/efectos de la radiación , Factor de Transcripción STAT1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Rayos Ultravioleta , Biomarcadores/metabolismo , Western Blotting , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Humanos , Inmunohistoquímica , Queratinocitos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Terapia Ultravioleta/instrumentación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA