Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Ann Rheum Dis ; 81(4): 544-555, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34853001

RESUMEN

OBJECTIVES: Dysregulated chondrocyte metabolism is closely associated with the pathogenesis of osteoarthritis (OA). Suppressing chondrocyte catabolism to restore cartilage homeostasis has been extensively explored, whereas far less effort has been invested toward enhancing chondrocyte anabolism. This study aimed to repurpose clinically approved drugs as potential stimulators of chondrocyte anabolism in treating OA. METHODS: Screening of a Food and Drug Administration-approved drug library; Assays for examining the chondroprotective effects of digoxin in vitro; Assays for defining the therapeutic effects of digoxin using a surgically-induced OA model; A propensity-score matched cohort study using The Health Improvement Network to examine the relationship between digoxin use and the risk of joint OA-associated replacement among patients with atrial fibrillation; identification and characterisation of the binding of digoxin to low-density lipoprotein receptor-related protein 4 (LRP4); various assays, including use of CRISPR-Cas9 genome editing to delete LRP4 in human chondrocytes, for examining the dependence on LRP4 of digoxin regulation of chondrocytes. RESULTS: Serial screenings led to the identification of ouabain and digoxin as stimulators of chondrocyte differentiation and anabolism. Ouabain and digoxin protected against OA and relieved OA-associated pain. The cohort study of 56 794 patients revealed that digoxin use was associated with reduced risk of OA-associated joint replacement. LRP4 was isolated as a novel target of digoxin, and deletion of LRP4 abolished digoxin's regulations of chondrocytes. CONCLUSIONS: These findings not only provide new insights into the understanding of digoxin's chondroprotective action and underlying mechanisms, but also present new evidence for repurposing digoxin for OA.


Asunto(s)
Cartílago Articular , Digoxina , Proteínas Relacionadas con Receptor de LDL , Osteoartritis , Cartílago Articular/metabolismo , Condrocitos/metabolismo , Estudios de Cohortes , Digoxina/farmacología , Reposicionamiento de Medicamentos , Humanos , Proteínas Relacionadas con Receptor de LDL/antagonistas & inhibidores , Osteoartritis/tratamiento farmacológico , Osteoartritis/patología , Ouabaína/farmacología
2.
Int J Cancer ; 144(9): 2227-2238, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30374974

RESUMEN

Vasculogenic mimicry (VM) is a special vascular pattern in malignant tumors, which is composed of highly aggressive tumor cells. This tumor cell-mediated blood supply pattern is closely associated with a poor prognosis in cancer patients. The interaction of axon guidance factor Sema4D and its high affinity receptor plexinB1 could activate small GTPase RhoA and its downstream ROCKs; this process has an active role in the migration of endothelial cells and tumor angiogenesis. Here, we have begun to uncover the role of this pathway in VM formation in non-small cell lung cancer (NSCLC). First, we confirmed this special form of vasculature in NSCLC tissues and found the existence of VM channels in tumor tissues was correlated with Sema4D expression. Further, we found that inhibition of Sema4D in the human NSCLC cells H1299 and HCC827 reduces VM formation both in vitro and in vivo. Moreover, we demonstrated that downregulating the expression of plexinB1 by siRNA expressing vectors and inhibiting the RhoA/ROCK signaling pathway using fasudil can reduce VM formation of H1299 and HCC827 cells. Finally, we found that suppression of Sema4D leads to less stress fibers and depleted the motility of H1299 and HCC827 cells. Collectively, our study implicates Sema4D plays an important role in the process of VM formation in NSCLC through activating the RhoA/ROCK pathway and regulating tumor cell plasticity and migration. Modulation of the Sema4D/plexinB1 and downstream RhoA/ROCK pathway may prevent the tumor blood supply through the VM pattern, which may eventually halt growth and metastasis of NSCLC.


Asunto(s)
Antígenos CD/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/patología , Neovascularización Patológica/patología , Proteínas del Tejido Nervioso/metabolismo , Receptores de Superficie Celular/metabolismo , Semaforinas/metabolismo , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Células A549 , Animales , Línea Celular Tumoral , Movimiento Celular/fisiología , Células Endoteliales/fisiología , Activación Enzimática , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas del Tejido Nervioso/genética , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptores de Superficie Celular/genética , Semaforinas/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
3.
Brain Behav Immun ; 81: 111-121, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31176001

RESUMEN

PURPOSE: Elevated catecholamines in the tumor microenvironment often correlate with tumor development. However, the mechanisms by which catecholamines modulate lung cancer growth are still poorly understood. This study is aimed at examining the functions and mechanisms of catecholamine-induced macrophage polarization in angiogenesis and tumor development. EXPERIMENTAL DESIGN: We established in vitro and in vivo models to investigate the relationship between catecholamines and macrophages in lung cancer. Flow cytometry, cytokine detection, tube formation assay, immunofluorescence, and western blot analysis were performed, and animal models were also used to explore the underlying mechanism of catecholamine-induced macrophage polarization and host immunological response. RESULTS: Catecholamines were shown to be secreted into tumor under the control of the sympathetic nerve system to maintain the pro-tumoral microenvironment. In vivo, the chemical depletion of the natural catecholamine stock with 6OHDA could reduce the release of catecholamines within tumor tissues, restrain the function of alternatively activated M2 macrophage, attenuate tumor neovascularization, and inhibit tumor growth. In vitro, catecholamine treatment triggered the M2 polarization of macrophages, enhanced the expression of VEGF, promoted tumor angiogenesis, and these catecholamine-stimulated effects could be reversed by the adrenergic receptor antagonist propranolol. In addition to regulating tumor-associated macrophages (TAM) recruitment, decreasing catecholamine levels could also shift the immunosuppressive microenvironment by decreasing myeloid-derived suppressor cells' (MDSCs) recruitment and facilitating dendritic cells' (DCs) activation, potentially resulting in a positive antitumor immune response. CONCLUSION: Our study demonstrates the potential of adrenergic stress and catecholamine-driven adrenergic signaling of TAMs to regulate the immune status of a tumor microenvironment and provides promising targets for anticancer therapies.


Asunto(s)
Catecolaminas/metabolismo , Neoplasias Pulmonares/metabolismo , Neovascularización Patológica/fisiopatología , Animales , Catecolaminas/fisiología , Línea Celular Tumoral , Movimiento Celular/inmunología , Citocinas/metabolismo , Humanos , Activación de Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Transducción de Señal , Microambiente Tumoral
4.
Mol Cancer Ther ; 14(7): 1582-90, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25934709

RESUMEN

The aim of this study was to investigate the biologic role of the Rho kinase inhibitor fasudil in the vasculogenic mimicry (VM) of B16 mouse melanoma cells. It was previously reported that RhoA plays a critical role in angiogenesis by coordinating endothelial cell cytoskeleton remodeling and promoting endothelial cell motility. Although RhoA has been implicated in the regulation of angiogenesis, little has been described regarding its control of these tumor cell-lined channels. In this study, we established an in vitro model of VM using 3-dimensional cell culturing of mouse B16 melanoma cells and studied VM in vivo by transplanting B16 cells into C57/BL mice. Next, we explored the effect of RhoA and Rho-associated, coiled-coil containing protein kinase (ROCK) on VM formation using the Rho kinase inhibitor fasudil. We provide direct evidence that fasudil leads to reduced vascular-like channels in Matrigel. Additional experiments suggested that fasudil prevents both initial cellular architecture changes and cell migration in vitro. Finally, we provide in-depth evidence for the underlying mechanisms of fasudil-induced VM destruction using the Rho-GTPase agonist lysophosphatidic acid. In vivo studies revealed that fasudil reduced B16 melanoma cell xenograft tumor growth without causing significant toxicity in mice. Fasudil-treated tumors also displayed fewer VM channels. These results suggest that fasudil may be an emerging therapeutic option for targeting cancer VM.


Asunto(s)
1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , Melanoma Experimental/tratamiento farmacológico , Neovascularización Patológica/prevención & control , Quinasas Asociadas a rho/antagonistas & inhibidores , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Animales , Western Blotting , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Lisofosfolípidos/farmacología , Masculino , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/patología , Ratones Endogámicos C57BL , Microscopía Confocal , Trasplante de Neoplasias/métodos , Inhibidores de Proteínas Quinasas/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Carga Tumoral/efectos de los fármacos , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo
5.
Spine J ; 14(3): 483-90, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23978626

RESUMEN

BACKGROUND CONTEXT: It has been shown that bupivacaine, the most commonly used local anesthetic to relieve or control pain in interventional spine procedures, is cytotoxic to intervertebral disc (IVD) cells in vitro. However, some other common local anesthetics, such as ropivacaine and lidocaine, are also frequently used in the treatment of spine-related pain, and the potential effects of these agents remain unclear. PURPOSE: The purpose of this study was to evaluate the effect of various local anesthetics on rabbit IVD cells in vitro and further compare the cytotoxicity of ropivacaine, bupivacaine, lidocaine, and saline solution control. STUDY DESIGN: Controlled laboratory study. SUBJECTS: Rabbit annulus fibrosus (AF) and nucleus pulposus (NP) cells were isolated from Japanese white rabbits. METHODS: Both AF and NP cells at the second generation maintained in monolayer were exposed to various concentrations of local anesthetics (eg, bupivacaine) or different durations of exposure and evaluated for cell viability by use of cell counting kit-8 (CCK-8). In addition, to compare the cytotoxicity of ropivacaine, bupivacaine, lidocaine, and saline solution control in commercial concentration, the viability was analyzed by flow cytometry after 60-minute exposure, and the morphologic changes were observed by the phase-contrast microscopy. Apoptosis and necrosis of IVD cells were confirmed by using fluorescence microscopy with double staining of Hoechst 33342 and propidium iodide. RESULTS: Rabbit IVD cell death demonstrated a time and dose dependence in response to bupivacaine and lidocaine. However, ropivacaine only exerted a significant time-dependent effect on IVD cells. There was no significant difference in IVD viability after treatment with different doses of ropivacaine. In addition, the results showed that lidocaine was the most toxic of the three local anesthetics and that ropivacaine presented less cytotoxicity than lidocaine and bupivacaine. Fluorescence microscopy also confirmed that the short-term toxic effect of local anesthetics on both AF and NP cells was mainly caused by necrosis rather than apoptosis. CONCLUSIONS: Results show that bupivacaine and lidocaine decrease cell viability in rabbit IVD cells in a dose- and time-dependent manner. All local anesthetics should be avoided if at all possible. Ropivacaine may be a choice if necessary, but it is also toxic. The increase in cell death is more related with cell necrosis rather than cell apoptosis. If these results can be corroborated in tissue explant models or animal studies, caution regarding diagnosing, treating, and controlling spine-related pain with local anesthetics is prompted.


Asunto(s)
Amidas/farmacología , Anestésicos Locales/farmacología , Apoptosis/efectos de los fármacos , Bupivacaína/farmacología , Disco Intervertebral/citología , Disco Intervertebral/efectos de los fármacos , Lidocaína/farmacología , Animales , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Condrocitos/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Técnicas In Vitro , Masculino , Modelos Animales , Conejos , Ropivacaína , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA