Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 426
Filtrar
1.
Int J Mol Sci ; 24(7)2023 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-37047322

RESUMEN

Dental follicle stem cells (DFSCs) have been verified to promote periodontal regeneration in an inflammatory microenvironment. When coping with inflammatory stimulation, DFSCs highly express periostin, a bioactive molecule closely related to periodontal homeostasis. It is worth exploring whether and how periostin plays a role in the promotion of periodontal regeneration by DFSCs. By tracking the fate of DFSCs, it was found that DFSCs significantly contributed to periodontal regeneration in rat periodontal defects while they had a low survival rate. They highly expressed periostin and improved the immune microenvironment in the defect area, especially via the recruitment and reprogramming of macrophages. Silencing periostin attenuated the effects of DFSCs in promoting periodontal regeneration and regulating macrophages. Recombinant human periostin (rhPeriostin) could not only directly promote macrophage reprogramming through the integrin αM/phosphorylated extracellular signal-regulated kinase (p-Erk)/Erk signaling pathway, but it also exhibited the potential to promote periodontal regeneration in rats when loaded in a collagen matrix. These results indicated that periostin is actively involved in the process by which DFSCs promote periodontal regeneration through the regulation of macrophages and is a promising molecular agent to promote periodontal regeneration. This study provides new insight into the mechanism by which DFSCs promote periodontal regeneration and suggests a new approach for periodontal regeneration therapy.


Asunto(s)
Moléculas de Adhesión Celular , Saco Dental , Periodoncio , Regeneración , Trasplante de Células Madre , Células Madre , Saco Dental/citología , Saco Dental/fisiología , Células Madre/metabolismo , Periodoncio/efectos de los fármacos , Periodoncio/inmunología , Periodoncio/fisiología , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Moléculas de Adhesión Celular/farmacología , Humanos , Animales , Ratas , Proteínas Recombinantes/farmacología , Periodontitis/inmunología , Periodontitis/terapia , Masculino , Ratas Sprague-Dawley
2.
Gynecol Endocrinol ; 40(1): 2270621, 2023 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-38301727

RESUMEN

OBJECTIVE: Medroxyprogesterone acetate (MPA) may increase the risk of atherosclerosis during hormone replacement therapy (HRT); therefore, the effect of progestogens other than MPA on atherosclerotic lesions requires evaluation. Adhesion of monocytes to vascular endothelial cells is an important early step in atherosclerosis progression. MCP-1 is a key chemokine that promotes monocyte migration and adhesion to vascular endothelial cells. In this study, we investigated the effects of dienogest (DNG), an alternative progestogen, on monocyte adhesion and cytokine expression in human umbilical vein endothelial cells (HUVECs). STUDY DESIGN: HUVECs were treated with DNG, natural progesterone, or MPA, followed by interleukin (IL)-1ß stimulation. The mRNA expression of adhesion molecules (E-selectin and ICAM-1) and cytokines (MCP-1 and IL-6) was examined using real-time PCR. A flow chamber system was used to examine the effect of DNG on the adhesion of U937 monocytic cells to monolayer HUVECs. RESULTS: Unlike MPA, DNG did not alter the mRNA expression of E-selectin, ICAM-1, MCP-1, and IL-6 in HUVECs. Moreover, it did not increase the number of monocytes adhering to HUVECs in the flow chamber system. However, MPA treatment significantly enhanced monocyte adhesion to HUVECs (p < 0.05). CONCLUSIONS: DNG had no effect on the mRNA expression of adhesion molecules and cytokines in HUVECs, as well as the monocyte adhesion to HUVECs, suggesting that DNG can be explored as an alternative to MPA for HRT.


Asunto(s)
Aterosclerosis , Monocitos , Nandrolona/análogos & derivados , Humanos , Monocitos/metabolismo , Molécula 1 de Adhesión Intercelular , Selectina E/genética , Selectina E/metabolismo , Interleucina-6/metabolismo , Molécula 1 de Adhesión Celular Vascular , Moléculas de Adhesión Celular/metabolismo , Moléculas de Adhesión Celular/farmacología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Citocinas/metabolismo , Aterosclerosis/metabolismo , ARN Mensajero/metabolismo , Expresión Génica , Adhesión Celular , Células Cultivadas
3.
Genes Cells ; 27(6): 451-464, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35430770

RESUMEN

Multilayered proliferation in an adherent culture as well as proliferation in a suspension culture is a characteristic feature of cancer cells. We previously showed using T47D human mammary cancer cells that nectin-4, upregulated in many cancer cells, cis-interacts with ErbB2 and its trastuzumab-resistant splice variants, p95-ErbB2 and ErbB2ΔEx16, and enhances DNA synthesis mainly through the PI3K-AKT pathway in an adherent culture. We showed here that only the combination of nectin-4 and p95-ErbB2, but not that of nectin-4 and ErbB2 or that of nectin-4 and ErbB2ΔEx16, cooperatively enhanced multilayered T47D cell proliferation through the Hippo pathway-mediated SOX2 gene expression in an adherent culture. T47D cells expressed the components of the apical junctional complex (AJC) consisting of adherens junctions (AJs) and tight junctions and cell polarity molecules, but not the AJ component afadin. The AJC and apicobasal polarity were disorganized in T47D cells in a monolayer and T47D cells stably expressing both nectin-4 and p95-ErbB2 in multilayers. These results indicate that nectin-4 and p95-ErbB2 play a stimulatory role in multilayered proliferation in an adherent culture.


Asunto(s)
Neoplasias de la Mama , Cadherinas , Moléculas de Adhesión Celular , Fosfatidilinositol 3-Quinasas , Receptor ErbB-2 , Uniones Adherentes/efectos de los fármacos , Neoplasias de la Mama/patología , Cadherinas/metabolismo , Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular/metabolismo , Moléculas de Adhesión Celular/farmacología , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Nectinas/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Receptor ErbB-2/metabolismo , Células Tumorales Cultivadas
4.
Cancer Gene Ther ; 29(10): 1452-1462, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35388173

RESUMEN

Progestin is one of the main hormone treatment regimens for early-stage estrogen receptor- and progesterone receptor (PR)-positive endometrial cancer (EC). However, the response rate of EC to progestins is unsatisfactory. Investigating the mechanisms related to progestin treatment could help improve treatment efficacy. Studies have demonstrated that normal endometrial stromal cells (ESCs) increase the inhibitory effect of progestin on EC cell proliferation via paracrine signaling, but the mechanisms involved remain unclear. In this study, we found that ESCs had different morphological features between progestin-sensitive and -insensitive EC tissues. ESCs presented typical decidualization changes in progestin-sensitive cases, while they remained slim in progestin-insensitive EC lesions, indicating no response. Furthermore, ESCs enhanced the inhibitory effect of medroxyprogesterone acetate (MPA) on EC cell proliferation by secreting neuron cell adhesion molecule (NrCAM). MPA treatment enhanced NrCAM secretion by ESCs. EC xenografts in BALB/C nude mice demonstrated that MPA combined with NrCAM had an increased tumor inhibitory effect compared with MPA or NrCAM alone. Mechanistically, MPA upregulated NrCAM expression in ESCs through PR. Specifically, NrCAM increased PR expression in EC cells through TET1-induced hydroxymethylation of the PRB gene promoter region. These findings indicate that NrCAM or NrCAM combined with progestins could be a new EC treatment.


Asunto(s)
Neoplasias Endometriales , Progestinas , Animales , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Moléculas de Adhesión Celular/farmacología , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/genética , Endometrio , Epigénesis Genética , Femenino , Humanos , Acetato de Medroxiprogesterona/metabolismo , Acetato de Medroxiprogesterona/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Oxigenasas de Función Mixta/genética , Progestinas/metabolismo , Progestinas/farmacología , Proteínas Proto-Oncogénicas/genética , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Células del Estroma/metabolismo , Células del Estroma/patología
5.
Bioengineered ; 13(3): 6558-6566, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35220882

RESUMEN

Adipose-derived mesenchymal stem cells (ADSCs) are a class of pluripotent stem cells isolated from the adipose tissue; they can differentiate into osteoblasts after induction and play an important role in bone repair. EGFL6 protein is secreted by adipocytes and osteoblasts and can promote endothelial cell migration and angiogenesis. This study aimed to explore the effect of recombinant EGFL6 protein on the osteogenic differentiation of ADSCs. The cells were incubated with fluorescein isothiocyanate-conjugated antibodies and analyzed by flow cytometry. Alizarin red staining and alkaline phosphatase staining were used to detect the osteogenic differentiation ability. mRNA expression was analyzed by real-time quantitative polymerase chain reaction (RT-qPCR). Protein expression was determined using Western blotting. The osteogenic differentiation ability of ADSCs isolated from the adipose tissue was significantly weakened after EGFL6 knockdown; this ability was restored upon the addition of EGFL6 recombinant protein. BMP2 knockdown inhibited the effect of EGFL6 recombinant protein on osteogenic differentiation. EGFL6 recombinant protein promoted osteogenic differentiation of ADSCs through the BMP2/SMAD4 signaling pathway. This may provide a potential target for the osteogenic differentiation of ADSCs.


Asunto(s)
Proteína Morfogenética Ósea 2/genética , Proteínas de Unión al Calcio , Moléculas de Adhesión Celular , Osteogénesis/efectos de los fármacos , Proteína Smad4/genética , Células Madre/efectos de los fármacos , Factor de Crecimiento Transformador beta/genética , Adipocitos/efectos de los fármacos , Proteína Morfogenética Ósea 2/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al Calcio/farmacología , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Moléculas de Adhesión Celular/farmacología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Humanos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Proteína Smad4/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
6.
J Asian Nat Prod Res ; 24(10): 987-999, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35023793

RESUMEN

Aloin is the main anthraquinone glycoside from Aloe species. Here, the anti-inflammatory functions of aloin against lipopolysaccharide (LPS)-induced vascular inflammatory responses were tested in endothelial cells or mice such as permeability, expressions of cell adhesion molecule (CAM), migration of leukocytes and lethality. Aloin was found to inhibit LPS-induced barrier disruption, CAM expression, and neutrophil adhesion/transendothelial migration to endothelial cells. Furthermore, aloin inhibited LPS-induced hyperpermeability, leukocyte migration, lethality in vivo. These results suggest that aloin has anti-inflammatory activities against LPS, thereby supporting its usefulness as a treatment for vascular inflammatory.


Asunto(s)
Emodina , Lipopolisacáridos , Ratones , Animales , Humanos , Lipopolisacáridos/farmacología , Células Endoteliales de la Vena Umbilical Humana , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Ratones Endogámicos C57BL , Estructura Molecular , Emodina/farmacología , Antiinflamatorios/farmacología , Moléculas de Adhesión Celular/metabolismo , Moléculas de Adhesión Celular/farmacología
7.
Toxins (Basel) ; 13(9)2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34564617

RESUMEN

Cysteine-Rich Secretory Proteins (CRiSPs) are typically found in many snake venoms; however, the role that these toxins play in the pathophysiology of snakebites is still unclear. Herein, we compared the effects of snake venom CRiSPs (svCRiSPs) from the most medically important species of North American snakes on endothelial cell permeability and vascular permeability. We used reverse phase protein array (RPPA) to identify key signaling molecules on human dermal lymphatic (HDLECs) and blood (HDBECs) endothelial cells treated with svCRiSPs. The results showed that Css-CRiSP isolated from Crotalus scutulatus scutulatus and App-CRiSP from Agkistrodon piscivorus piscivorus are the most potent causes of increase vascular and endothelial permeability in comparison with other svCRiSPs used in this study. We examined the protein expression levels and their activated phosphorylation states in HDLECs and HDBECs induced by App-CRiSP and Css-CRiSP using RPPA. Interestingly, both App-CRiSP and Css-CRiSP induced caveolin-1 expression in HDBECs. We also found that stimulating HDBECs with Css-CRiSP and App-CRiSP significantly induced the phosphorylation of mTOR and Src, respectively. In HDLECs, Css-CRiSP significantly downregulated the expression of N-Cadherin and phospholipase C-gamma, while App-CRiSP significantly enhanced Akt and JNK phosphorylation. These results suggest that the increased endothelial permeability in HDLECs and HDBECs by Css-CRiSP and App-CRiSP may occur through different pathways.


Asunto(s)
Agkistrodon , Moléculas de Adhesión Celular/farmacología , Venenos de Crotálidos/farmacología , Crotalus , Células Endoteliales/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Células Endoteliales/fisiología , Humanos , Análisis por Matrices de Proteínas
8.
Reprod Sci ; 28(10): 2906-2915, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33959891

RESUMEN

The self-renewal of spermatogonial cells (SCs) provides the foundation for life-long spermatogenesis. To date, only a few growth factors have been used for the culture of SCs in vitro, and how to enhance proliferation capacity of SCs in vitro needs further research. This study aimed to explore the effects of periostin (POSTN) on the proliferation of human SCs. GC-1 spg cells were cultured in a medium with POSTN, cell proliferation was evaluated by MTS analysis and EdU assay, and the Wnt/ß-catenin signaling pathway was examined. Thereafter, the proliferations of human SC were detected using immunofluorescence and RT-PCR. In this study, we found that CM secreted by human amniotic mesenchymal stem cells (hAMSCs) could enhance the proliferation capacity of mouse GC-1 spg cells. Label-free mass spectrometry and ELISA analysis demonstrated that high level of POSTN was secreted by hAMSCs. MTS and EdU staining showed that POSTN increased GC-1 spg cell proliferation, whereas CM from POSTN-silenced hAMSCs suppressed cell proliferation capacity. Then POSTN was found to activate the Wnt/ß-catenin signaling pathway to regulate the proliferation of GC-1 spg cells. XAV-939, a Wnt/ß-catenin inhibitor, partially reversed the effects of POSTN on GC-1 spg cell proliferation. We then analyzed human SCs and found that POSTN promoted human SC proliferation in vitro. These findings provide insights regarding the role of POSTN in regulating SC proliferation via the Wnt/ß-catenin signaling pathway and suggest that POSTN may serve as a cytokine for male infertility therapy.


Asunto(s)
Moléculas de Adhesión Celular/farmacología , Proliferación Celular/efectos de los fármacos , Espermatogonias/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , Animales , Línea Celular , Proliferación Celular/fisiología , Relación Dosis-Respuesta a Droga , Humanos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Espermatogonias/metabolismo , Vía de Señalización Wnt/fisiología
9.
Chem Commun (Camb) ; 57(42): 5111-5126, 2021 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-33977972

RESUMEN

DC-SIGN, a lectin discovered two decades ago, plays a relevant role in innate immunity. Since its discovery, it has turned out to be a target for developing antiviral drugs based on carbohydrates due to its participation in the infection process of several pathogens. A plethora of carbohydrate multivalent systems using different scaffolds have been described to achieve this goal. Our group has made significant contributions to this field, which are revised herein.


Asunto(s)
Antracenos/química , Moléculas de Adhesión Celular/química , Glicósidos/química , Lectinas Tipo C/química , Receptores de Superficie Celular/química , Antracenos/farmacología , Antivirales/química , Antivirales/farmacología , Moléculas de Adhesión Celular/metabolismo , Moléculas de Adhesión Celular/farmacología , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Virus del Dengue/efectos de los fármacos , Fulerenos/química , Lectinas Tipo C/metabolismo , Nanopartículas del Metal/química , Conformación Molecular , Receptores de Superficie Celular/metabolismo , Virus Zika/efectos de los fármacos
10.
Am J Physiol Heart Circ Physiol ; 320(6): H2222-H2239, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33834866

RESUMEN

Extracellular matrix (ECM) exerts a series of biological functions and contributes to almost 30% of the osteogenic process. Periostin is a secreted protein that can alter ECM remodeling in response to vascular injury. However, the role of periostin in vascular calcification has yet to be fully investigated. As found in this study, recombinant periostin accelerated the thoracic aortas calcification, increased the expression of glycolysis key enzymes, and disturbed the normal oxidative phosphorylation (OXPHOS) ex vivo, which could be alleviated by the peroxisome proliferation-activated receptor γ (PPARγ) agonist pioglitazone. In vascular smooth muscle cells (VSMCs), periostin promoted VSMC-osteoblastic phenotype transition and calcium deposition and suppressed PPARγ expression. Mechanistically, periostin caused overactivation of glycolysis and mitochondrial dysfunction in VSMCs as assessed by extracellular acidification rate, oxygen consumption rate, and mitochondrial respiratory chain complex activities. Targeted glycolysis inhibitors reduced mitochondrial calcium overload, apoptosis, and periostin-induced VSMCs calcification. PPARγ agonists preserved glycolysis and OXPHOS in the stimulated microenvironment and reversed periostin-promoted VSMC calcification. Furthermore, plasma periostin, lactate, and matrix Gla protein levels were measured in 274 patients undergoing computed tomography to determine coronary artery calcium score (Agatston score). Plasma periostin and lactate levels were both linked to an Agatston score in patients with coronary artery calcification (CAC). There was also a positive correlation between plasma periostin and lactate levels. This study suggests that downregulation of PPARγ is involved in the mechanism by which periostin accelerates arterial calcification partly through excessive glycolysis activation and unbalanced mitochondrial homeostasis.NEW & NOTEWORTHY Periostin caused arterial calcification, overactivated glycolysis, and damaged OXPHOS. PPARγ agonists alleviated periostin-promoted arterial calcification and corrected abnormal glycolysis and unbalanced mitochondrial homeostasis. There exists a relationship between periostin and lactate in patients with CAC.


Asunto(s)
Aorta Torácica/metabolismo , Moléculas de Adhesión Celular/metabolismo , Enfermedad de la Arteria Coronaria/metabolismo , Glucosa/metabolismo , Mitocondrias/metabolismo , Miocitos del Músculo Liso/metabolismo , PPAR gamma/metabolismo , Calcificación Vascular/metabolismo , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/patología , Apoptosis/efectos de los fármacos , Moléculas de Adhesión Celular/farmacología , Angiografía por Tomografía Computarizada , Regulación hacia Abajo , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Glucólisis/efectos de los fármacos , Humanos , Técnicas In Vitro , Ácido Láctico/metabolismo , Masculino , Mitocondrias/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Fosforilación Oxidativa/efectos de los fármacos , Consumo de Oxígeno , PPAR gamma/agonistas , Pioglitazona/farmacología , Ratas
11.
J Pharmacol Toxicol Methods ; 106: 106936, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33191187

RESUMEN

INTRODUCTION: Subcutaneous (SC) formulations of therapeutics with recombinant human hyaluronidase PH20 (rHuPH20) are currently approved across various disease indications. The rHuPH20-mediated enzymatic degradation of SC hyaluronan (HA) facilitates bulk fluid flow and dispersion of co-administered therapeutics. However, current methods of quantifying dispersion in the SC space are limited. Here, a novel method is outlined to quantify and follow rapid SC volumetric dispersion of a representative therapeutic fluid in the presence of rHuPH20 using computed tomography (CT). METHODS: Ten Yucatan miniature swine were randomized to three groups. Animals received simultaneous infusions of contrast agent (CA) alone (left side of the animal) or in combination with rHuPH20 (right side) at infusion rates of 2.5, 5, or 10 mL/min. Spiral CT scans (1.5 mm thickness) were conducted before and after the infusion and at regular time intervals throughout. Scans were used to create three-dimensional (3D) reconstructions of the fluid pockets and analyze surface area, volume, and sphericity. RESULTS: 3D reconstruction showed increased dispersion of CA with rHuPH20 compared with CA alone, with fenestration and increased dispersion in the craniocaudal and lateromedial directions. The CA with rHuPH20 fluid pockets showed an average increase of 46% in surface area (p = 0.001), a 35% increase in volume (p = 0.001) and a 17% decrease in sphericity post-infusion compared with CA alone at 30 min post-infusion. DISCUSSION: This exploratory study confirms the value of CT imaging as a non-invasive method of assessing real-time spatial and temporal behavior of SC-administered fluids. This technique could help to assess the dispersion pattern of novel rHuPH20 SC co-formulations.


Asunto(s)
Moléculas de Adhesión Celular/farmacología , Tomografía Computarizada de Haz Cónico , Portadores de Fármacos/farmacología , Hialuronoglucosaminidasa/farmacología , Tejido Subcutáneo/diagnóstico por imagen , Distribución Tisular/efectos de los fármacos , Animales , Medios de Contraste/administración & dosificación , Medios de Contraste/farmacocinética , Estudios de Factibilidad , Femenino , Humanos , Infusiones Subcutáneas , Modelos Animales , Proteínas Recombinantes/farmacología , Análisis Espacio-Temporal , Tejido Subcutáneo/metabolismo , Porcinos , Porcinos Enanos
12.
Mol Cell Endocrinol ; 518: 111026, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32919022

RESUMEN

Secreted PDZD2 (sPDZD2) is a signaling molecule generated upon proteolytic processing of the multi-PDZ-containing protein PDZD2. Previous analysis of gene-trap mice deficient in the synthesis of full-length PDZD2, but not the secreted form, revealed a role of PDZD2 in the regulation of glucose-stimulated insulin secretion. Here, using the pancreatic INS-1E ß cells as in vitro model, we showed that depletion of PDZD2/sPDZD2 by RNA interference suppressed the expression of ß-cell genes Ins1, Glut2 and MafA whereas treatment with recombinant sPDZD2 rescued the suppressive effect. Similar to GLP-1, sPDZD2 stimulated intracellular cAMP levels, activated ß-cell gene expression in a PKA-dependent manner and induced the phosphorylation and nuclear localization of PDX1. Depletion of PDX1 inhibited the sPDZD2 insulinotropic effect, which could also be demonstrated in mouse islets. In summary, our findings are consistent with sPDZD2 serving a signaling function in regulating ß-cell gene expression.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/farmacología , Moléculas de Adhesión Celular/farmacología , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 2/metabolismo , Insulina/genética , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Ratones , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Ratas , Vías Secretoras/efectos de los fármacos , Vías Secretoras/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
13.
Folia Histochem Cytobiol ; 58(3): 227-234, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32936927

RESUMEN

INTRODUCTION: Intervertebral disc degeneration (IDD) is one of the major causes of low back pain (LBP) which seriously affects health and normal physical activity. Recombinant human tumor necrosis factor-a (TNF-a) induced protein 6 (rhTSG-6) has been reported to have therapeutic effects on a variety of inflammatory diseases, but the effect and mechanism of rhTSG-6 action in IDD are not fully understood. The present study was aimed to explore the functional role of rhTSG-6 in interleukin (IL)-1b-induced nucleus pulposus (NP) cell model. MATERIALS AND METHODS: Experimental human NP cells were isolated from the patients with idiopathic scoliosis and treated with culture medium containing IL-1b (10 ng/mL) for 24 hours to induce extracellular matrix degradation and apoptosis, simulating an IDD model in vitro. The viability of NP cells was analyzed by the CCK-8 assay. The relevant mRNA and protein levels were measured by RT-qPCR and western blot. The apoptosis of NP cells was determined by flow cytometry analysis and western blot. RESULTS: Compared with the NP cells without IL-1b treatment, IL-1b caused approximately 70% reduction in the viability of NP cells, while RhTSG-6 partly increased the decrease of IL-1b on cell viabilities. Moreover, treatment with rhTSG-6 considerably attenuated the upregulation of extracellular matrix (ECM)-catabolic factors (MMP-3, MMP-13, ADAMTS-4, and ADAMTS-5), and increased the downregulation of ECM-anabolic factor (collagen II) in NP cells induced by IL-1b, indicating that ECM degradation was suppressed. Furthermore, rhTSG-6 also protected NP cells from IL-1b-induced apoptosis. Mechanically, rhTSG-6 inhibited the activation of members of mitogen-activated protein kinase (MAPK) pathway by blocking the phosphorylation of p38, c-Jun N-terminal kinase (JNK) and ERK in IL-1b-induced NP cells. CONCLUSIONS: RhTSG-6 can attenuate ECM degradation and apoptosis in IL-1b-induced NP cells by inhibiting the p38, JNK and ERK pathways, which may contribute to its potential application in the therapy of IDD.


Asunto(s)
Apoptosis/efectos de los fármacos , Moléculas de Adhesión Celular/farmacología , Matriz Extracelular/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Núcleo Pulposo/metabolismo , Proteínas Recombinantes/farmacología , Adolescente , Adulto , Supervivencia Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Interleucina-1beta/metabolismo , Degeneración del Disco Intervertebral/tratamiento farmacológico , MAP Quinasa Quinasa 4/metabolismo , Persona de Mediana Edad , Núcleo Pulposo/citología , Escoliosis/patología , Adulto Joven , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Int J Mol Sci ; 21(13)2020 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-32629816

RESUMEN

The formation of 3D multicellular spheroids in the ascites fluid of ovarian cancer patients is an understudied component of the disease progression. Spheroids are less sensitive to chemotherapy, in part due to the protection afforded by their structure, but also due to their slower proliferation rate. Previous studies suggest that the cell adhesion molecule Nectin-4 plays a key role in the formation of ovarian cancer spheroids. In this study, we further examined the role of Nectin-4 at early time points in spheroid formation using real-time digital photography. Human NIH:OVCAR5 ovarian cancer cells formed aggregates within 8 h, which further contracted into compact spheroids over 24 h. In contrast, Nectin-4 knockdown cells did not form tightly compacted spheroids. Synthetic peptides derived from Nectin-4 were tested for their ability to alter spheroid formation in two ovarian cancer cell lines. Nectin-4 peptide 10 (N4-P10) had an immediate effect on disrupting ovarian cancer spheroid formation, which continued for over 24 h, while a scrambled version of the peptide had no effect. N4-P10 inhibited spheroid formation in a concentration-dependent manner and was not cytotoxic; suggesting that N4-P10 treatment could maintain the cancer cells as single cells which may be more sensitive to chemotherapy.


Asunto(s)
Moléculas de Adhesión Celular/farmacología , Péptidos/farmacología , Esferoides Celulares/efectos de los fármacos , Ascitis , Líquido Ascítico , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Carcinoma Epitelial de Ovario/metabolismo , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Moléculas de Adhesión Celular/metabolismo , Agregación Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Femenino , Humanos , Nectinas/metabolismo , Nectinas/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Ovario/metabolismo , Péptidos/síntesis química , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas/efectos de los fármacos
15.
J Cardiovasc Pharmacol ; 76(3): 329-336, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32569018

RESUMEN

Atherosclerosis (AS) is the most common and serious complication in type 2 diabetes mellitus (T2DM). Recent studies have emphasized that inflammation is the main cause of atherosclerosis. Studies have shown that carcinoembryonic antigen-related cellular adhesion molecule 1 (CEACAM1) regulates the expression of matrix metallopeptidase 9 (MMP-9) after ischemic stroke to reduce inflammation. The aim of this study was to elucidate potential molecular mechanism of CEACAM1 on the inflammatory response in atherosclerosis. The serum levels of CEACAM1, MMP-9, and tissue inhibitors of metalloproteinase 1 (TIMP-1) in T2DM patients and healthy control was detected. The results showed that the levels of CEACAM1 and TIMP-1 were significantly decreased, and the levels of MMP-9 were significantly higher than those in the control group. Moreover, we also observed the effect of CEACAM1 on atherosclerosis in T2DM rats. Hematoxylin & eosin (HE) staining and oil red staining showed that CEACAM1 recombinant protein reduced intima-media thickness and the area of atherosclerotic plaques. To further explore the molecular mechanism of CEACAM1 regulating MMP-9/TIMP-1, we conducted experiments in rat aorta vascular endothelial cells and rat aorta smooth muscle cells. The result showed that CEACAM1 inhibits inflammatory response via MMP-9/TIMP-1 axis. Taken together, CEACAM1 attenuates diabetic atherosclerosis by inhibition of IκB/NF-κB signal pathway via MMP-9/TIMP-1 axis, which indicate that CEACAM1 is potentially amenable to therapeutic manipulation for clinical application in atherosclerosis in T2DM.


Asunto(s)
Antiinflamatorios/farmacología , Antígenos CD/farmacología , Arterias/efectos de los fármacos , Aterosclerosis/prevención & control , Moléculas de Adhesión Celular/farmacología , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Angiopatías Diabéticas/prevención & control , Proteínas I-kappa B/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , FN-kappa B/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Animales , Antígenos CD/metabolismo , Arterias/enzimología , Arterias/patología , Aterosclerosis/enzimología , Aterosclerosis/etiología , Aterosclerosis/patología , Estudios de Casos y Controles , Moléculas de Adhesión Celular/metabolismo , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/enzimología , Angiopatías Diabéticas/enzimología , Angiopatías Diabéticas/etiología , Angiopatías Diabéticas/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Placa Aterosclerótica , Ratas Wistar , Transducción de Señal
16.
J Neuroinflammation ; 17(1): 154, 2020 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-32393298

RESUMEN

BACKGROUND: Neuroinflammation plays a vital role in the development and maintenance of neuropathic pain. Recent evidence has proved that bone marrow mesenchymal stem cells (BMSCs) can inhibit neuropathic pain and possess potent immunomodulatory and immunosuppressive properties via secreting a variety of bioactive molecules, such as TNF-α-stimulated gene 6 protein (TSG-6). However, it is unknown whether BMSCs exert their analgesic effect against neuropathic pain by secreting TSG-6. Therefore, the present study aimed to evaluate the analgesic effects of TSG-6 released from BMSCs on neuropathic pain induced by chronic constriction injury (CCI) in rats and explored the possible underlying mechanisms in vitro and in vivo. METHODS: BMSCs were isolated from rat bone marrow and characterized by flow cytometry and functional differentiation. One day after CCI surgery, about 5 × 106 BMSCs were intrathecally injected into spinal cerebrospinal fluid. Behavioral tests, including mechanical allodynia, thermal hyperalgesia, and motor function, were carried out at 1, 3, 5, 7, 14 days after CCI surgery. Spinal cords were processed for immunohistochemical analysis of the microglial marker Iba-1. The mRNA and protein levels of pro-inflammatory cytokines (IL-1ß, TNFα, IL-6) were detected by real-time RT-PCR and ELISA. The activation of the TLR2/MyD88/NF-κB signaling pathway was evaluated by Western blot and immunofluorescence staining. The analgesic effect of exogenous recombinant TSG-6 on CCI-induced mechanical allodynia and heat hyperalgesia was observed by behavioral tests. In the in vitro experiments, primary cultured microglia were stimulated with the TLR2 agonist Pam3CSK4, and then co-cultured with BMSCs or recombinant TSG-6. The protein expression of TLR2, MyD88, p-p65 was evaluated by Western blot. The mRNA and protein levels of IL-1ß, TNFα, IL-6 were detected by real-time RT-PCR and ELISA. BMSCs were transfected with the TSG-6-specific shRNA and then intrathecally injected into spinal cerebrospinal fluid in vivo or co-cultured with Pam3CSK4-treated primary microglia in vitro to investigate whether TSG-6 participated in the therapeutic effect of BMSCs on CCI-induced neuropathic pain and neuroinflammation. RESULTS: We found that CCI-induced mechanical allodynia and heat hyperalgesia were ameliorated by intrathecal injection of BMSCs. Moreover, intrathecal administration of BMSCs inhibited CCI-induced neuroinflammation in spinal cord tissues. The analgesic effect and anti-inflammatory property of BMSCs were attenuated when TSG-6 expression was silenced. We also found that BMSCs inhibited the activation of the TLR2/MyD88/NF-κB pathway in the ipsilateral spinal cord dorsal horn by secreting TSG-6. Meanwhile, we proved that intrathecal injection of exogenous recombinant TSG-6 effectively attenuated CCI-induced neuropathic pain. Furthermore, in vitro experiments showed that BMSCs and TSG-6 downregulated the TLR2/MyD88/NF-κB signaling and reduced the production of pro-inflammatory cytokines, such as IL-1ß, IL-6, and TNF-α, in primary microglia treated with the specific TLR2 agonist Pam3CSK4. CONCLUSIONS: The present study demonstrated a paracrine mechanism by which intrathecal injection of BMSCs targets the TLR2/MyD88/NF-κB pathway in spinal cord dorsal horn microglia to elicit neuroprotection and sustained neuropathic pain relief via TSG-6 secretion.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Células Madre Mesenquimatosas/metabolismo , Microglía/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Neuralgia/metabolismo , Transducción de Señal/fisiología , Receptor Toll-Like 2/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Moléculas de Adhesión Celular/farmacología , Masculino , Trasplante de Células Madre Mesenquimatosas , Microglía/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo
17.
Methods Mol Biol ; 2132: 379-389, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32306345

RESUMEN

Tachyzoites, which are infective forms of Toxoplasma gondii, use their actinomyosin system to move over surfaces and invade host cells. Central to this process is the regulated release of micronemes organelles contents. The microneme protein 4 (MIC4) has the property to recognize galactosides residues linked to glycoproteins on the host cell surface. This property allows that MIC4 binds to TLR2- and TLR4 N-linked glycans and promote the activation of cell innate immune cells and secretion of inflammatory cytokines, acting on resistance against the parasite. Obtention of MIC4 from T. gondii requires several purification steps, is time-consuming and provides low yield. Therefore, this section details the protocol for prokaryotic expression, production, and purification of recombinant MIC4 (rMIC4) and for experimental assays to confirm its biological activity.


Asunto(s)
Moléculas de Adhesión Celular/farmacología , Galactósidos/metabolismo , Proteínas Protozoarias/farmacología , Receptores Toll-Like/agonistas , Toxoplasma/metabolismo , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Membrana Celular/metabolismo , Galactósidos/química , Glicoproteínas/química , Células HEK293 , Humanos , Inmunidad Innata , Ingeniería de Proteínas , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Toxoplasma/genética
18.
Methods Mol Biol ; 2132: 391-400, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32306346

RESUMEN

Some lectins of pathogens interact with host cells through the recognition of specific carbohydrates displayed on the mammals' cell surface. The microneme protein 1 (MIC1) from Toxoplasma gondii has a lectin domain that specifically binds sialic acid residues, often found in the terminal positions of N-glycans of mammalian cells. The necessary studies on the MIC1 biological roles have been limited initially by the laborious purification of the protein from T. gondii tachyzoites and the low yields verified. Then Escherichia coli has been transformed with a construct containing the MIC1 gene, and the obtained recombinant MIC1 (rMIC1) has been purified from the inclusion bodies. Herein, we detail the methodology of heterologous production and purification of rMIC1 and protocols to assay the rMIC1 lectin ability.


Asunto(s)
Moléculas de Adhesión Celular/farmacología , Polisacáridos/metabolismo , Proteínas Protozoarias/farmacología , Toxoplasma/metabolismo , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Cuerpos de Inclusión/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Polisacáridos/química , Ingeniería de Proteínas , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Toxoplasma/genética
19.
PLoS One ; 15(2): e0220756, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32040478

RESUMEN

Adipose tissue derived mesenchymal stem/stromal cell (ASC)-derived extracellular vesicles (EV) have been reported to be beneficial against dextran sulfate sodium (DSS)-induced colitis in mice. However, the underlying mechanisms have not been fully elucidated. We hypothesize that the tumor necrosis factor-α-stimulated gene/protein 6 (TSG-6) in EVs is a key factor influencing the alleviation of colitis symptoms. DSS-induced colitis mice (C57BL/6, male, Naïve = 6, Sham = 8, PBS = 8 EV = 8, CTL-EV = 8, TSG-6 depleted EV = 8) were intraperitoneally administered EVs (100 ug/mice) on day 1, 3, and 5; colon tissues were collected on day 10 for histopathological, RT-qPCR, western blot and immunofluorescence analyses. In mice injected with EV, inflammation was alleviated. Indeed, EVs regulated the levels of pro- and anti-inflammatory cytokines, such as TNF-α, IL-1ß, IFN-γ, IL-6, and IL-10 in inflamed colons. However, when injected with TSG-6 depleted EV, the degree of inflammatory relief was reduced. Furthermore, TSG-6 in EVs plays a key role in increasing regulatory T cells (Tregs) and polarizing macrophage from M1 to M2 in the colon. In conclusion, this study shows that TSG-6 in EVs is a major factor in the relief of DSS-induced colitis, by increasing the number of Tregs and macrophage polarization from M1 to M2 in the colon.


Asunto(s)
Moléculas de Adhesión Celular/farmacología , Colitis/prevención & control , Vesículas Extracelulares/química , Células Madre Mesenquimatosas/química , Animales , Recuento de Células , Colitis/inducido químicamente , Colitis/terapia , Citocinas/metabolismo , Sulfato de Dextran/efectos adversos , Perros , Vesículas Extracelulares/trasplante , Inflamación/terapia , Macrófagos/citología , Células Madre Mesenquimatosas/ultraestructura , Ratones , Linfocitos T Reguladores/citología
20.
Theranostics ; 10(1): 36-49, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31903104

RESUMEN

Mesenchymal stem cells (MSCs) transplantation is a promising antifibrotic strategy but facing clinical controversies. Inspired by advances in nanomedicine, we aimed to bypass these clinical barriers of MSCs by identifying the key antifibrotic molecule of MSCs and developing a specific liver-targeting nanocarrier. Methods: Cytokines secreted by MSCs were examined with serum stimulation of cirrhotic patients. Immunohistochemistry, microarray, immunoblotting, and quantitative real-time PCR (qRT-PCR) were applied to identify the critical antifibrotic cytokine and to discover its role in modulating antifibrotic effects. Biomineralization method was used to prepare calcium phosphate nanoparticles (NPs). The targeting and therapeutic efficiency of NPs were evaluated by in vivo imaging and biochemical studies on fibrotic mice induced by CCl4. Results: The stimulated MSCs exhibited high-level expression of Tumor necrosis factor (TNF)-stimulated gene 6 (TSG-6). On animal study, exogenous administration of TSG-6 alone can ameliorate liver fibrosis while TSG-6 knocked MSCs (Lv-TSG-6 MSCs) lost antifibrotic effects. Further studies verified the importance of TSG-6 and identified its antifibrotic mechanism by modulating M2 macrophages and increasing matrix metalloproteinase 12 (MMP12) expression. Additionally, we found a feedback loop between TSG-6, MMP12 and pro-inflammatory cytokines (TNF-α, IL-6, and IL-1ß), which may improve our understanding of the aggravating process of cirrhosis and antifibrotic mechanisms of TSG-6 and MSCs. Based on these findings, we developed calcium phosphate nanoparticles (CaP@BSA NPs) by biomineralization method using bovine serum albumin (BSA) as the biotemplate. Imaging tracking and drug loading studies showed specific liver targeting and high TSG-6 loading efficacy of as-prepared CaP@BSA NPs. In vivo therapeutic study further demonstrated the improved therapeutic effects of TSG-6 loaded CaP@BSA. Conclusions: TSG-6 was a major antifibrotic cytokine of MSCs, TSG-6 loaded CaP@BSA NPs showed specific liver accumulation and improved therapeutic effects, which indicated translational potentials of CaP@BSA as a promising drug carrier for the liver disease management.


Asunto(s)
Moléculas de Adhesión Celular , Portadores de Fármacos/química , Cirrosis Hepática/tratamiento farmacológico , Nanopartículas/química , Animales , Fosfatos de Calcio/química , Moléculas de Adhesión Celular/administración & dosificación , Moléculas de Adhesión Celular/farmacología , Citocinas/metabolismo , Humanos , Hígado/efectos de los fármacos , Hígado/patología , Macrófagos/metabolismo , Masculino , Metaloproteinasa 12 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Albúmina Sérica Bovina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA