Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Brain Topogr ; 36(3): 305-318, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37061591

RESUMEN

In the field of medical imaging, the classification of brain tumors based on histopathological analysis is a laborious and traditional approach. To address this issue, the use of deep learning techniques, specifically Convolutional Neural Networks (CNNs), has become a popular trend in research and development. Our proposed solution is a novel Convolutional Neural Network that leverages transfer learning to classify brain tumors in MRI images as benign or malignant with high accuracy. We evaluated the performance of our proposed model against several existing pre-trained networks, including Res-Net, Alex-Net, U-Net, and VGG-16. Our results showed a significant improvement in prediction accuracy, precision, recall, and F1-score, respectively, compared to the existing methods. Our proposed method achieved a benign and malignant classification accuracy of 99.30 and 98.40% using improved Res-Net 50. Our proposed system enhances image fusion quality and has the potential to aid in more accurate diagnoses.


Asunto(s)
Neoplasias Encefálicas , Redes Neurales de la Computación , Humanos , Neoplasias Encefálicas/diagnóstico por imagen , Imagen por Resonancia Magnética , Recuerdo Mental , Aprendizaje Automático
4.
J Biol Chem ; 291(8): 3882-94, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26703472

RESUMEN

Continuous parathyroid hormone (PTH) blocks its own osteogenic actions in marrow stromal cell cultures by inducing Cox2 and receptor activator of nuclear factor κB ligand (RANKL) in the osteoblastic lineage cells, which then cause the hematopoietic lineage cells to secrete an inhibitor of PTH-stimulated osteoblast differentiation. To identify this inhibitor, we used bone marrow macrophages (BMMs) and primary osteoblasts (POBs) from WT and Cox2 knock-out (KO) mice. Conditioned medium (CM) from RANKL-treated WT, but not KO, BMMs blocked PTH-stimulated cAMP production in POBs. Inhibition was reversed by pertussis toxin (PTX), which blocks Gαi/o activation. Saa3 was the most highly differentially expressed gene in a microarray comparison of RANKL-treated WT versus Cox2 KO BMMs, and RANKL induced Saa3 protein secretion only from WT BMMs. CM from RANKL-stimulated BMMs with Saa3 knockdown did not inhibit PTH-stimulated responses in POBs. SAA added to POBs inhibited PTH-stimulated cAMP responses, which was reversed by PTX. Selective agonists and antagonists of formyl peptide receptor 2 (Fpr2) suggested that Fpr2 mediated the inhibitory actions of Saa3 on osteoblasts. In BMMs committed to become osteoclasts by RANKL treatment, Saa3 expression peaked prior to appearance of multinucleated cells. Flow sorting of WT marrow revealed that Saa3 was secreted only from the RANKL-stimulated B220(-) CD3(-)CD11b(-/low) CD115(+) preosteoclast population. We conclude that Saa3 secretion from preosteoclasts, induced by RANKL in a Cox2-dependent manner, inhibits PTH-stimulated cAMP signaling and osteoblast differentiation via Gαi/o signaling. The induction of Saa3 by PTH may explain the suppression of bone formation when PTH is applied continuously and may be a new therapeutic target for osteoporosis.


Asunto(s)
AMP Cíclico/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Hormona Paratiroidea/farmacología , Sistemas de Mensajero Secundario/efectos de los fármacos , Proteína Amiloide A Sérica/metabolismo , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , AMP Cíclico/genética , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Ratones , Ratones Noqueados , Osteoblastos/citología , Osteoclastos/citología , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Hormona Paratiroidea/metabolismo , Ligando RANK/genética , Ligando RANK/metabolismo , Receptores de Formil Péptido/genética , Receptores de Formil Péptido/metabolismo , Sistemas de Mensajero Secundario/genética , Proteína Amiloide A Sérica/genética
5.
SLAS Technol ; 29(4): 100159, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38909655

RESUMEN

In today's digital world, with growing population and increasing pollution, unhealthy lifestyle habits like irregular eating, junk food consumption, and lack of exercise are becoming more common, leading to various health problems, including kidney issues. These factors directly affect human kidney health. To address this, we require early detection techniques that rely on text data. Text data contains detailed information about a patient's medical history, symptoms, test results, and treatment plans, giving a complete picture of kidney health and enabling timely intervention. In this research paper, we proposed a range of sophisticated models, such as Gradient Boosting Classifier, Light GBM, CatBoost, Support Vector Classifier (SVC), Random Boost, Logistic Regression, XGBoost, Deep Neural Network (DNN), and an Improved DNN. The Improved DNN demonstrated exceptional performance, with an accuracy of 90 %, precision of 89 %, recall of 90 %, and an F1-Score of 89.5 %. By combining traditional machine learning and deep neural networks, this integrative approach enables the identification of intricate patterns in datasets. The model's data-driven processes consistently update internal parameters, guaranteeing flexibility in response to evolving healthcare settings. This research represents a notable advancement in the progress of creating a more detailed and individualised ability to diagnose kidney stones, which could potentially lead to better clinical results and patient treatment.


Asunto(s)
Cálculos Renales , Redes Neurales de la Computación , Cálculos Renales/diagnóstico , Humanos , Aprendizaje Automático
6.
Carcinogenesis ; 34(12): 2891-9, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23825153

RESUMEN

Macrophage migratory inhibitory factor (MIF) is a proinflammatory cytokine shown to promote tumorigenesis. Using the N-butyl-N-(4-hydroxybutyl)-nitrosamine (BBN) model of bladder cancer, we previously showed that MIF knockout mice display decreased angiogenesis and invasion compared with wild-type. This study examines the role of MIF in bladder cancer via use of oral inhibitors of MIF. In vitro, high-grade bladder cancer cells were treated with recombinant human MIF +/- (rhMIF+/-) inhibitor. Measurements included cell counts, proliferation by (3)H-thymidine incorporation (TdR), extracellular signal-regulated kinase (ERK) phosphorylation by western blot analysis, messenger RNA (mRNA) expression by quantitative PCR and protein secretion by enzyme-linked immunosorbent assay. Treatment with rhMIF increased ERK phosphorylation, cell counts, TdR and mRNA expression and protein secretion of vascular endothelial growth factor, which were blocked by specific inhibitors of ERK and MIF. In vivo, 3-month-old male C57Bl/6 mice were given BBN for 22 and 16 weeks in study 1 and study 2, respectively. Mice (n = 8-10 per group) were gavaged with vehicle or doses of MIF inhibitors daily from weeks 16-22 in both studies. Average bladder weights, reflecting tumor mass, tumor stage/burden, mitotic rate and proliferation indices, and microvessel densities were reduced in inhibitor groups versus controls. In summary, MIF promotes bladder cancer via increasing cell proliferation and angiogenesis and oral inhibitors of MIF may prove useful in treatment of this disease.


Asunto(s)
Factores Inhibidores de la Migración de Macrófagos/genética , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Neovascularización Patológica/patología , Neoplasias de la Vejiga Urinaria/patología , Animales , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Células Hep G2 , Humanos , Sistema de Señalización de MAP Quinasas/genética , Factores Inhibidores de la Migración de Macrófagos/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/genética , Fosforilación/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero , Vejiga Urinaria/metabolismo , Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
7.
Bioact Mater ; 25: 42-60, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36733930

RESUMEN

Tendon and ligament injuries are the most common musculoskeletal injuries, which not only impact the quality of life but result in a massive economic burden. Surgical interventions for tendon/ligament injuries utilize biological and/or engineered grafts to reconstruct damaged tissue, but these have limitations. Engineered matrices confer superior physicochemical properties over biological grafts but lack desirable bioactivity to promote tissue healing. While incorporating drugs can enhance bioactivity, large matrix surface areas and hydrophobicity can lead to uncontrolled burst release and/or incomplete release due to binding. To overcome these limitations, we evaluated the delivery of a peptide growth factor (exendin-4; Ex-4) using an enhanced nanofiber matrix in a tendon injury model. To overcome drug surface binding due to matrix hydrophobicity of poly(caprolactone) (PCL)-which would be expected to enhance cell-material interactions-we blended PCL and cellulose acetate (CA) and electrospun nanofiber matrices with fiber diameters ranging from 600 to 1000 nm. To avoid burst release and protect the drug, we encapsulated Ex-4 in the open lumen of halloysite nanotubes (HNTs), sealed the HNT tube endings with a polymer blend, and mixed Ex-4-loaded HNTs into the polymer mixture before electrospinning. This reduced burst release from ∼75% to ∼40%, but did not alter matrix morphology, fiber diameter, or tensile properties. We evaluated the bioactivity of the Ex-4 nanofiber formulation by culturing human mesenchymal stem cells (hMSCs) on matrix surfaces for 21 days and measuring tenogenic differentiation, compared with nanofiber matrices in basal media alone. Strikingly, we observed that Ex-4 nanofiber matrices accelerated the hMSC proliferation rate and elevated levels of sulfated glycosaminoglycan, tendon-related genes (Scx, Mkx, and Tnmd), and ECM-related genes (Col-I, Col-III, and Dcn), compared to control. We then assessed the safety and efficacy of Ex-4 nanofiber matrices in a full-thickness rat Achilles tendon defect with histology, marker expression, functional walking track analysis, and mechanical testing. Our analysis confirmed that Ex-4 nanofiber matrices enhanced tendon healing and reduced fibrocartilage formation versus nanofiber matrices alone. These findings implicate Ex-4 as a potentially valuable tool for tendon tissue engineering.

8.
Environ Sci Pollut Res Int ; 29(42): 63577-63587, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35460489

RESUMEN

Pregnancy and feto-gestational toxicities on exposure to fluoride and its possible amelioration on co-administration with aloe vera were studied in pregnant Swiss albino mice. Once the confirmed pregnancy was tested, animals were equally divided into four groups as follows: group I was given no treatment and served as control, and groups II and III were administered with 100 and 300 ppm sodium fluoride, respectively, while group IV was co- administered aloe vera (300 mg/kg bw) along with sodium fluoride (300 ppm) daily for 14 days prior to gestation and continued till the 18th day of gestation. Animals were sacrificed on the 19th day of gestation for prenatal observations. Maternal body weight, the gravid uterine weight, number of corpora lutea in both the ovaries, number of implantations and resorptions, number of live (mature and immature) fetuses, and number of dead fetuses were examined in each dam. The treatment continued in another set of animals till the completion of the weaning period to observe postnatal changes due to test substances on the mother and pups. Sodium fluoride-treated animals showed morphometric and skeletal changes which were more pronounced in the high-dose group showing significantly decreased body weight gain in pregnant mothers and dead/immature fetuses. Morphometric changes included open eyelids, limb defects, wrinkles on the whole body, anophthalmia, pulmonary edema, enlarged esophagus, and decreased body weight of fetuses and pups. Alizarin-prepared skeletal structures of fetuses of such female mice showed delayed ossification or bending in the number of bones of skull, thoracic, and limb regions. However, concomitant exposure to sodium fluoride and aloe vera in treated animals led to a marked improvement in all the prenatal and postnatal variables. The study suggests that sodium fluoride at high concentrations may be teratogenic while co-administration of aloe vera during fluoride exposure might be beneficial in reducing these toxic effects. The use of aloe vera as a preventive agent or as a complimentary agent is thus recommended following fluoride exposure through the oral route.


Asunto(s)
Aloe , Aloe/química , Animales , Peso Corporal , Femenino , Fluoruros/toxicidad , Ratones , Embarazo , Fluoruro de Sodio/toxicidad , Teratógenos
9.
Wirel Pers Commun ; 123(4): 3497-3509, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34744315

RESUMEN

The COVID-19 lockdown has led all the citizens (mobile subscribers) of India to stay at home and rather work from home. The people have started consuming more channel utilization (in mobile communication) through a continuous long duration conversations and more internet data through more streaming content as well as logging on to work from home. It was also reflected in how data demand from residential areas rose as compared to commercial areas. Consequently the bandwidth and channel saturation has evolved out to be a severe problem thereby affecting the work performance of all online offices and multi-national companies. This research paper proposes the simulation based experimental study of DITMC technique for mitigating this effect with a special concern in North Western Rajasthan part of India. The simulation results show that significant enhancement of 60.52% in channel utilization and bandwidth optimization is possible with negligible overhead of 0.23%. This technique also enables the telecom operators to ponder research in this field that will promisingly lead to manage augmented number of mobile subscribers (independent of any lockdown period) in limited bandwidth thereby using the spectrum efficiently.

10.
Gene ; 773: 145385, 2021 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-33383117

RESUMEN

Tephrosia purpurea (T. purpurea), a plant belonging to Fabaceae (pea) family, is a well-known Ayurvedic herb and commonly known as Sarapunkha in traditional Indian medicinal system. Described as "Sarwa wranvishapaka", i.e. having a capability to heal all types of wounds, it is particularly recognized for its usage in splenomegaly. Towards exploring the comprehensive effects of T. purpurea against polycystic ovarian syndrome (PCOS) and three comorbid neuropsychiatric diseases (anxiety, depression, and bipolar disorder), its constituent phytochemicals (PCs) were extensively reviewed and their network pharmacology evaluation was carried out in this study. The complex regulatory potential of its 76 PCs against PCOS is enquired by developing and analyzing high confidence tripartite networks of protein targets of each phytochemical at both pathway and disease association scales. We also developed a high-confidence human Protein-Protein Interaction (PPI) sub-network specific to PCOS, explored its modular architecture, and probed 30 drug-like phytochemicals (DPCs) having multi-module regulatory potential. The phytochemicals showing good binding affinity towards their protein targets were also evaluated for similarity against currently available approved drugs present in DrugBank. Multi-targeting and synergistic capacities of 12 DPCs against 10 protein targets were identified and evaluated using molecular docking and interaction analyses. Eight DPCs as a potential source of PCOS and its comorbidity regulators are reported in T. purpurea. The results of network-pharmacology study highlight the therapeutic relevance of T. purpurea as PCOS-regulator and demonstrate the effectiveness of the approach in revealing action-mechanism of Ayurvedic herbs from holistic perspective.


Asunto(s)
Medicina Ayurvédica , Fitoquímicos/uso terapéutico , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Tephrosia/química , Ansiedad/tratamiento farmacológico , Ansiedad/psicología , Trastorno Bipolar/tratamiento farmacológico , Depresión/tratamiento farmacológico , Depresión/psicología , Femenino , Humanos , Trastornos Mentales/tratamiento farmacológico , Simulación del Acoplamiento Molecular , Fitoquímicos/química , Fitoquímicos/genética , Síndrome del Ovario Poliquístico/genética , Síndrome del Ovario Poliquístico/patología , Síndrome del Ovario Poliquístico/psicología , Mapas de Interacción de Proteínas/efectos de los fármacos , Mapas de Interacción de Proteínas/genética , Esplenomegalia/tratamiento farmacológico , Esplenomegalia/psicología
11.
Prostaglandins Other Lipid Mediat ; 89(1-2): 20-5, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19464663

RESUMEN

We have examined the effects of varying doses, schedules and routes of administration of prostaglandin E(2) (PGE(2)) on bone in mice. Male C57BL/6 mice treated with a high dose of PGE(2) (6 mg/kg/d) showed decreased trabecular bone volume (BV/TV) by 14 d, indicating increased bone resorption. However, there was also stimulation of bone formation at 14 d after 3 d treatment with PGE(2,) since mineral apposition rate (MAR) and bone formation rate (BFR/BS) were increased. In CD-1 male and female mice, PGE(2) (3mg/kg, 2/wk for 4 wk) increased MAR by 50% and BFR/BS by 100%, but there was no significant change in BV/TV. Tibial mRNA showed an increase in BMP-2 and RUNX-2 expression with PGE(2). Additional experiments using a higher dose or longer exposure did not increase bone mass. We conclude that exposure to high doses of PGE(2) in mice may be anabolic but is balanced by catabolic effects. Studies of PGE(2) in combination with an inhibitor of resorption could lead to development of a true anabolic model and permit assessment of the roles of specific PGE(2) receptors and signal transduction pathways.


Asunto(s)
Huesos/efectos de los fármacos , Huesos/metabolismo , Dinoprostona/farmacología , Animales , Densidad Ósea/efectos de los fármacos , Huesos/fisiología , Dinoprostona/administración & dosificación , Relación Dosis-Respuesta a Droga , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Proteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Tiempo
12.
Prostaglandins Other Lipid Mediat ; 90(3-4): 76-80, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19744575

RESUMEN

Bone morphogenetic protein 2 (BMP-2) is used clinically to stimulate bone formation and accelerate fracture repair. Adding prostaglandin (PG) E(2) or PGE(2) receptor agonists to BMP-2 has been proposed to improve BMP-2 efficacy. However, this may enhance bone resorption, since PGE(2) can increase receptor activator of NF-kappaB ligand (RANKL) expression and decrease osteoprotegerin (OPG) expression in osteoblasts, and the RANKL:OPG ratio is critical for osteoclast formation. We used bone marrow (BM) cultures and BM macrophage (BMM) cultures from outbred CD1 mice to examine effects on osteoclast formation of BMP-2 and PGE(2). In BM cultures, which contain both osteoblastic and osteoclastic lineage cells, BMP-2 (100 ng/ml) alone did not increase osteoclast formation but enhanced the peak response to PGE(2) by 1.6-9.6-fold. In BMM cultures, which must be treated with RANKL because they do not contain osteoblastic cells, BMP-2 did not increase osteoclast formation, with or without PGE(2). Our results suggest that BMP-2 can increase osteoclast formation in response to PGE(2) by increasing the RANKL:OPG ratio in osteoblasts, which may have therapeutic implications for the use of BMP-2.


Asunto(s)
Proteína Morfogenética Ósea 2/farmacología , Macrófagos/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Animales , Recuento de Células , Células Cultivadas , Dinoprostona/farmacología , Humanos , Macrófagos/citología , Ratones , Osteoclastos/citología , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Ligando RANK/genética , Ligando RANK/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Biochem Biophys Res Commun ; 372(4): 536-41, 2008 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-18501188

RESUMEN

PTH is a potent bone anabolic agent in vivo but anabolic effects on osteoblast differentiation in vitro are difficult to demonstrate. This study examined the role of cyclooxygenase (COX)-2 and prostaglandin (PG) production in the effects of PTH on osteoblast differentiation in vitro using marrow stromal cell (MSC) and calvarial osteoblast (COB) cultures from COX-2 knockout (KO) and wild type (WT) mice. Cells were treated with PTH (10 nM) or vehicle throughout culture. Alkaline phosphatase (ALP) and osteocalcin (OCN) mRNA levels were measured at days 14 and 21, respectively, and mineralization at day 21. cAMP concentrations were measured in the presence of a phosphodiesterase inhibitor. PTH did not stimulate differentiation in cultures from WT mice but significantly increased ALP and OCN mRNA expression 6- to 7-fold in KO MSC cultures and 2- to 4-fold in KO COB cultures. PTH also increased mineralization in both KO MSC and COB cultures. Effects in KO cells were mimicked in WT MSC cultures treated with NS-398, an inhibitor of COX-2 activity. PTH increased cAMP concentrations similarly in WT and KO COBs. Differential gene responses to PTH in COX-2 KO COBs relative to WT COBs included greater fold-increases in the cAMP-mediated early response genes, c-fos and Nr4a2; increased IGF-1 mRNA expression; and decreased mRNA expression of MAP kinase phosphatase-1. PTH inhibited SOST mRNA expression 91% in COX-2 KO MSC cultures compared to 67% in WT cultures. We conclude that endogenous PGs inhibit the anabolic responses to PTH in vitro, possibly by desensitizing cAMP pathways.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Osteoblastos/citología , Hormona Paratiroidea/farmacología , Proteínas Adaptadoras Transductoras de Señales , Fosfatasa Alcalina/biosíntesis , Fosfatasa Alcalina/genética , Animales , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Calcificación Fisiológica/efectos de los fármacos , Calcificación Fisiológica/genética , Células Cultivadas , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , AMP Cíclico/biosíntesis , Ciclooxigenasa 2/genética , Expresión Génica/efectos de los fármacos , Marcadores Genéticos/genética , Glicoproteínas , Péptidos y Proteínas de Señalización Intercelular , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/enzimología , Ratones , Ratones Noqueados , Osteoblastos/enzimología , Osteocalcina/biosíntesis , Osteocalcina/genética , ARN Mensajero/análisis , Cráneo/citología
14.
Prostaglandins Other Lipid Mediat ; 86(1-4): 35-40, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18406186

RESUMEN

Studies using prostaglandin E receptor (EP) agonists indicate that prostaglandin (PG) E(2) can have anabolic effects through both EP4 and EP2 receptors. We previously found that the anabolic response to a selective EP4 receptor agonist (EP4A, Ono Pharmaceutical) was substantially greater than to a selective EP2 receptor agonist (EP2A) in cultured murine calvarial osteoblastic cells. To further define the role of the EP2 receptor in PG-mediated effects on bone cells, we examined the effects of EP2A and PGE(2) on both calvarial primary osteoblasts (POB) and marrow stromal cells (MSC) cultured from mice with deletion of one (Het) or both (KO) alleles of the EP2 receptor compared to their wild-type (WT) littermates. Deletion of EP2 receptor was confirmed by quantitative real-time PCR, Western blot and immunohistochemistry. The 1 month-old mice used to provide cells in these studies did not show any significant differences in their femurs by static histomorphometry. EP2A was found to enhance osteoblastic differentiation as measured by alkaline phosphatase mRNA expression and activity as well as osteocalcin mRNA expression and mineralization in the WT cell cultures from both marrow and calvariae. The effects were somewhat diminished in cultures from Het mice and abrogated in cultures from KO mice. PGE(2) effects were greater than those of EP2A, particularly in POB cultures and were only moderately diminished in Het and KO cell cultures. We conclude that activation of the EP2 receptor is able to enhance differentiation of osteoblasts, that EP2A is a true selective agonist for this receptor and that PGE(2) has an additional anabolic effect likely mediated by the EP4 receptor.


Asunto(s)
Dinoprostona/farmacología , Osteoblastos/efectos de los fármacos , Receptores de Prostaglandina E/genética , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Western Blotting , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Osteoblastos/citología , Osteoblastos/metabolismo , Osteocalcina/genética , Osteocalcina/metabolismo , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/fisiología , Subtipo EP2 de Receptores de Prostaglandina E , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo
15.
Cancer Res ; 66(13): 6657-64, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16818639

RESUMEN

Overexpression of cyclooxygenase-2 (COX-2) is generally considered to promote tumorigenesis. To investigate a potential role of COX-2 in osteosarcoma, we overexpressed COX-2 in human osteosarcoma cells. Saos-2 cells deficient in COX-2 expression were retrovirally transduced or stably transfected with murine COX-2 cDNA. Functional expression of COX-2 was confirmed by Northern and Western analyses and prostaglandin production. Overexpression of COX-2 reduced cell numbers by 50% to 70% compared with controls. Decreased proliferation in COX-2-overexpressing cells was associated with cell cycle prolongation in G(2)-M. Apoptosis, measured by both Annexin V binding assay and terminal deoxyribonucleotidyl transferase-mediated dUTP nick end labeling staining, was increased in cells overexpressing COX-2, and the increase was not reversed by treatment with NS-398, indicating that the effects were not mediated by prostaglandins. Retroviral COX-2 overexpression in two other human osteosarcoma cell lines, U2OS and TE85, also decreased cell viability. However, in the human colon carcinoma HCT-116 cell line, which is deficient in COX-2, retroviral overexpression of COX-2, at similar efficiency as in Saos-2 cells, increased resistance to apoptosis. Reactive oxygen species (ROS), measured by flow cytometry, were increased by COX-2 overexpression in Saos-2 cells but not in HCT-116 cells. Inhibition of peroxidase activity, but not of COX activity, blocked the ROS increase. Antioxidants blocked the increase in ROS and the increase in apoptosis due to COX-2 overexpression in Saos-2 cells. Our results suggest that (a) COX-2 overexpression in osteosarcoma cells may increase resistance to tumorigenesis by increasing ROS to levels that decrease cell viability and (b) the effects of COX-2 overexpression are cell type/tissue dependent.


Asunto(s)
Neoplasias Óseas/enzimología , Neoplasias Óseas/patología , Ciclooxigenasa 2/biosíntesis , Osteosarcoma/enzimología , Osteosarcoma/patología , Antioxidantes/farmacología , Apoptosis/fisiología , Neoplasias Óseas/genética , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Ciclooxigenasa 2/genética , Dinoprostona/farmacología , Células HCT116 , Humanos , Osteosarcoma/genética , Especies Reactivas de Oxígeno/metabolismo , Retroviridae/genética , Transducción Genética , Transfección
16.
Urol Oncol ; 36(3): 93.e13-93.e21, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29079132

RESUMEN

PURPOSE: SATB1, a global genome organizer, has been shown to play a role in the development and progression of some solid tumors, but its role in bladder cancer is undetermined. Moreover, there is conflicting data about the role of SATB1 in other tumors. This study was initiated to assess a potential role for SATB1 with the hypothesis that SATB1 acts as a tumor promoter in bladder cancer. MATERIALS AND METHODS: We evaluated SATB1 expression in bladder cancer cell lines (HTB-5, HTB-9) and compared them to a benign urothelial cell line (UROtsa). Short-hairpin RNA was used to silence SATB1 in multiple cell lines, and cell death and cell proliferation were assessed using multiple assays. RESULTS: SATB1 expression was increased significantly in all cancer cell lines compared to benign urothelial cells. SATB1 expression was knocked down by short-hairpin RNA and functional outcomes, including cell number, cell-cycle arrest, cell viability, and apoptosis after cisplatin treatment, were measured. Surprisingly, knockdown of SATB1 in 2 high-grade cancer cell lines showed opposing functional roles. Compared to the non-silencing control, HTB-5 cells, showed decreased cellular proliferation and increased sensitivity to cisplatin, whereas HTB-9 cells, showed increased cell numbers and increased resistance to cisplatin. CONCLUSION: We conclude that our results in bladder cancer are consistent with the conflicting data reported in other cancers, and that SATB1 might have different roles in cancer dependent on genetic background and stage of the cancer.


Asunto(s)
Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Cisplatino/farmacología , Resistencia a Antineoplásicos/genética , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Proteínas de Unión a la Región de Fijación a la Matriz/genética , ARN Interferente Pequeño/metabolismo , Regulación hacia Arriba , Neoplasias de la Vejiga Urinaria/genética
17.
Endocrinology ; 159(7): 2759-2776, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29757436

RESUMEN

Increased bone resorption is considered to explain why intermittent PTH is anabolic for bone but continuous PTH is catabolic. However, when cyclooxygenase-2 (COX2) is absent in mice, continuous PTH becomes anabolic without decreased resorption. In murine bone marrow stromal cells (BMSCs), serum amyloid A (SAA)3, induced in the hematopoietic lineage by the combination of COX2-produced prostaglandin and receptor activator of nuclear factor κB ligand (RANKL), suppresses PTH-stimulated osteoblast differentiation. To determine whether SAA3 inhibits the anabolic effects of PTH in vivo, wild-type (WT) and SAA3 knockout (KO) mice were infused with PTH. In WT mice, continuous PTH induced SAA3 and was catabolic for bone. In KO mice, PTH was anabolic, increasing trabecular bone, serum markers of bone formation, and osteogenic gene expression. In contrast, PTH increased all measurements associated with bone resorption, as well as COX2 gene expression, similarly in KO and WT mice. SAA1 and SAA2 in humans are likely to have analogous functions to SAA3 in mice. RANKL induced both SAA1 and SAA2 in human bone marrow macrophages in a COX2-dependent manner. PTH stimulated osteogenesis in human BMSCs only when COX2 or RANKL was inhibited. Addition of recombinant SAA1 or SAA2 blocked PTH-stimulated osteogenesis. In summary, SAA3 suppresses the bone formation responses but not the bone resorption responses to PTH in mice, and in the absence of SAA3, continuous PTH is anabolic. In vitro studies in human bone marrow suggest that SAA may be a target for enhancing the therapeutic effects of PTH in treating osteoporosis.


Asunto(s)
Resorción Ósea/etiología , Resorción Ósea/metabolismo , Hormona Paratiroidea/metabolismo , Hormona Paratiroidea/farmacología , Proteína Amiloide A Sérica/metabolismo , Animales , Resorción Ósea/sangre , Masculino , Células Madre Mesenquimatosas , Ratones , Ratones Noqueados , Osteogénesis/efectos de los fármacos , Osteogénesis/fisiología , Hormona Paratiroidea/sangre , Ligando RANK/metabolismo
18.
J Bone Miner Res ; 22(7): 1002-10, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17371157

RESUMEN

UNLABELLED: Strontium ranelate is a new anti-osteoporosis treatment. This study showed that strontium ranelate stimulated PGE(2) production and osteoblastic differentiation in murine marrow stromal cells, which was markedly reduced by inhibition of COX-2 activity or disruption of COX-2 gene expression. Hence, some anabolic effects of strontium ranelate may be mediated by the induction of COX-2 and PGE(2) production. INTRODUCTION: Strontium ranelate is an orally active drug that reduces vertebral and hip fracture risk by increasing bone formation and reducing bone resorption. Strontium ranelate effects on bone formation are the result of increased osteoblastic differentiation and activity, but the mechanisms governing these effects are unknown. Based on previous work, we hypothesized that strontium ranelate increases cyclooxygenase (COX)-2 expression and that, consequently, the prostaglandin E(2) (PGE(2)) produced could mediate some effects of strontium ranelate on osteoblasts. MATERIALS AND METHODS: Marrow stromal cells (MSCs) from COX-2 wildtype (WT) and knockout (KO) mice were cultured with and without low-dose dexamethasone. Osteoblastic differentiation was characterized by alkaline phosphatase (ALP) activity, real-time PCR for ALP and osteocalcin (OCN) mRNA expression, and alizarin red staining for mineralization. Medium PGE(2) was measured by radioimmunoassay or enzyme immunoassay. RESULTS AND CONCLUSIONS: In MSCs from COX-2 WT mice, strontium ranelate significantly increased ALP activity, ALP and OCN mRNA expression, and mineralization after 14 or 21 days of culture. A short treatment at the beginning of the culture (0-7 days) with strontium ranelate was as effective as continuous treatment. Strontium ranelate (1 and 3 mM Sr(+2)) dose-dependently increased PGE(2) production, with maximum PGE(2) production occurring during the first week of culture. NS-398, a selective COX-2 inhibitor, blocked the strontium ranelate stimulation of PGE(2) production and significantly inhibited the strontium ranelate stimulation of ALP activity. In MSCs from COX-2 KO mice, the strontium ranelate stimulation of ALP and OCN mRNA expression and mineralization were markedly reduced compared with COX-2 WT cultures. Similar effects of strontium ranelate on osteoblastic markers and on PGE(2) production were seen when MSCs were cultured with or without low-dose dexamethasone (10 nM). We conclude that PGE(2) produced by the strontium ranelate induction of COX-2 expression plays a role in strontium ranelate-induced osteoblastic differentiation in MSCs in vitro.


Asunto(s)
Células de la Médula Ósea/citología , Calcificación Fisiológica/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Compuestos Organometálicos/farmacología , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Tiofenos/farmacología , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Biomarcadores/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Células Cultivadas , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Dexametasona/farmacología , Dinoprostona/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Nitrobencenos/farmacología , Células del Estroma/citología , Sulfonamidas/farmacología , Factores de Tiempo
19.
Bone ; 41(1): 68-76, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17467356

RESUMEN

Cyclooxygenase-2 (COX-2) is highly expressed in osteoblasts, and COX-2 produced prostaglandins (PGs) can increase osteoblastic differentiation in vitro. The goal of this study was to examine effects of COX-2 expression on calvarial osteoblastic proliferation and apoptosis. Primary osteoblasts (POBs) were cultured from calvariae of COX-2 wild-type (WT) and knockout (KO) mice. POB proliferation was evaluated by (3)H-thymidine incorporation and analysis of cell replication and cell cycle distribution by flow cytometry. POB apoptosis was evaluated by annexin and PI staining on flow cytometry. As expected, PGE(2) production and alkaline phosphatase (ALP) activity were increased in WT cultures compared to KO cultures. In contrast, cell numbers were decreased in WT compared to KO cells by day 4 of culture. Proliferation, measured on days 3-7 of culture, was 2-fold greater in KO than in WT POBs and associated with decreased Go/G1 and increased S cell cycle distribution. There was no significant effect of COX-2 genotype on apoptosis under basal culture conditions on day 5 of culture. Cell growth was decreased in KO POBs by the addition of PGE(2) or a protein kinase A agonist and increased in WT POBs by the addition of NS398, a selective COX-2 inhibitor. In contrast, differentiation and cell growth in marrow stromal cell (MSC) cultures, evaluated by ALP and crystal violet staining respectively, were increased in MSCs from WT mice compared to MSCs from KO mice, and exogenous PGE(2) increased cell growth in KO MSC cultures. We conclude that PGs secondary to COX-2 expression decrease osteoblastic proliferation in cultured calvarial cells but increase growth of osteoblastic precursors in MSC cultures.


Asunto(s)
Ciclooxigenasa 2/deficiencia , Ciclooxigenasa 2/genética , Osteoblastos/citología , Osteoblastos/enzimología , Fosfatasa Alcalina/metabolismo , Animales , Apoptosis , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Proliferación Celular , Células Cultivadas , Dinoprostona/biosíntesis , Dinoprostona/farmacología , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoblastos/efectos de los fármacos , Cráneo/citología , Células del Estroma/citología , Células del Estroma/metabolismo
20.
Nat Commun ; 8: 15831, 2017 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-28635959

RESUMEN

During bone remodelling, osteoclasts induce chemotaxis of osteoblasts and yet maintain spatial segregation. We show that osteoclasts express the repulsive guidance factor Semaphorin 4D and induce contact inhibition of locomotion (CIL) in osteoblasts through its receptor Plexin-B1. To examine causality and elucidate how localized Plexin-B1 stimulation may spatiotemporally coordinate its downstream targets in guiding cell migration, we develop an optogenetic tool for Plexin-B1 designated optoPlexin. Precise optoPlexin activation at the leading edge of migrating osteoblasts readily induces local retraction and, unexpectedly, distal protrusions to steer cells away. These morphological changes are accompanied by reorganization of Myosin II, PIP3, adhesion and active Cdc42. We attribute the resultant repolarization to RhoA/ROCK-mediated redistribution of ß-Pix, which activates Cdc42 and promotes protrusion. Thus, our data demonstrate a causal role of Plexin-B1 for CIL in osteoblasts and reveals a previously unknown effect of Semaphorin signalling on spatial distribution of an activator of cell migration.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Movimiento Celular/efectos de la radiación , Polaridad Celular/efectos de la radiación , Luz , Masculino , Ratones , Ratones Endogámicos C57BL , Miosina Tipo II/genética , Miosina Tipo II/metabolismo , Proteínas del Tejido Nervioso/genética , Optogenética , Osteoblastos/citología , Osteoblastos/efectos de la radiación , Osteoclastos/citología , Osteoclastos/efectos de la radiación , Receptores de Superficie Celular/genética , Semaforinas/metabolismo , Transducción de Señal/efectos de la radiación , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA