Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 141
Filtrar
1.
Respir Res ; 22(1): 250, 2021 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-34548087

RESUMEN

BACKGROUND: Mechanical ventilation can induce or aggravate lung injury, which is termed ventilator-induced lung injury (VILI). Piezo1 is a key element of the mechanotransduction process and can transduce mechanical signals into biological signals by mediating Ca2+ influx, which in turn regulates cytoskeletal remodeling and stress alterations. We hypothesized that it plays an important role in the occurrence of VILI, and investigated the underlying mechanisms. METHODS: High tidal volume mechanical ventilation and high magnitude cyclic stretch were performed on Sprague-Dawley rats, and A549 and human pulmonary microvascular endothelial cells, respectively, to establish VILI models. Immunohistochemical staining, flow cytometry, histological examination, enzyme-linked immunosorbent assay, western blotting, quantitative real-time polymerase chain reaction and survival curves were used to assess the effect of Piezo1 on induction of lung injury, as well as the signaling pathways involved. RESULTS: We observed that Piezo1 expression increased in the lungs after high tidal volume mechanical ventilation and in cyclic stretch-treated cells. Mechanistically, we observed the enhanced expression of RhoA/ROCK1 in both cyclic stretch and Yoda1-treated cells, while the deficiency or inhibition of Piezo1 dramatically antagonized RhoA/ROCK1 expression. Furthermore, blockade of RhoA/ROCK1 signaling using an inhibitor did not affect Piezo1 expression. GSMTx4 was used to inhibit Piezo1, which alleviated VILI-induced pathologic changes, water content and protein leakage in the lungs, and the induction of systemic inflammatory mediators, and improved the 7-day mortality rate in the model rats. CONCLUSIONS: These findings indicate that Piezo1 affects the development and progression of VILI through promotion of RhoA/ROCK1 signaling.


Asunto(s)
Mecanotransducción Celular/fisiología , Proteínas de la Membrana/biosíntesis , Respiración Artificial/efectos adversos , Lesión Pulmonar Inducida por Ventilación Mecánica/metabolismo , Proteínas de Unión al GTP rho/biosíntesis , Quinasas Asociadas a rho/biosíntesis , Células A549 , Animales , Humanos , Masculino , Ratas , Ratas Sprague-Dawley , Volumen de Ventilación Pulmonar/fisiología , Lesión Pulmonar Inducida por Ventilación Mecánica/patología
2.
Sci Rep ; 11(1): 5540, 2021 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-33692394

RESUMEN

Previously, we identified increased maternal circulating DAAM2 mRNA in pregnancies complicated by preterm fetal growth restriction (FGR). Here, we assessed whether circulating DAAM2 mRNA could detect FGR, and whether the DAAM2 gene, known to play roles in the Wnt signalling pathway is expressed in human placenta and associated with dysfunction and FGR. We performed linear regression analysis to calculate area under the ROC curve (AUC) for DAAM2 mRNA expression in the maternal circulation of pregnancies complicated by preterm FGR. DAAM2 mRNA expression was assessed across gestation by qPCR. DAAM2 protein and mRNA expression was assessed in preterm FGR placenta using western blot and qPCR. DAAM2 expression was assessed in term cytotrophoblasts and placental explant tissue cultured under hypoxic and normoxic conditions by qPCR. Small interfering RNAs were used to silence DAAM2 in term primary cytotrophoblasts. Expression of growth, apoptosis and oxidative stress genes were assessed by qPCR. Circulating DAAM2 mRNA was elevated in pregnancies complicated by preterm FGR [p < 0.0001, AUC = 0.83 (0.78-0.89)]. Placental DAAM2 mRNA was detectable across gestation, with highest expression at term. DAAM2 protein was increased in preterm FGR placentas but demonstrated no change in mRNA expression. DAAM2 mRNA expression was increased in cytotrophoblasts and placental explants under hypoxia. Silencing DAAM2 under hypoxia decreased expression of pro-survival gene, BCL2 and oxidative stress marker, NOX4, whilst increasing expression of antioxidant enzyme, HMOX-1. The increased DAAM2 associated with FGR and hypoxia implicates a potential role in placental dysfunction. Decreasing DAAM2 may have cytoprotective effects, but further research is required to elucidate its role in healthy and dysfunctional placentas.


Asunto(s)
Retardo del Crecimiento Fetal/metabolismo , Regulación de la Expresión Génica , Hipoxia/metabolismo , Proteínas de Microfilamentos/biosíntesis , Placenta/metabolismo , ARN Mensajero/biosíntesis , Proteínas de Unión al GTP rho/biosíntesis , Adulto , Femenino , Humanos , Placenta/irrigación sanguínea , Embarazo
3.
Neurotherapeutics ; 17(4): 2028-2040, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32822001

RESUMEN

Rho GTPase family members were identified as critical regulators of cell morphology, actin cytoskeleton organization, cell movement, and cell cycle and also contributed to tumor progression, which have been implicated in various types of cancer metastasis and growth. Here, we firstly reported the dysregulation of Rhoj in glioblastoma multiforme (GBM) and aimed to investigate the role and mechanism of Rhoj in GBM. We analyzed the expression of 21 Rho GTPases family members and validated the expression of Rhoj in GBM by immunohistochemistry. We further investigated the role and mechanism of Rhoj in GBM both in vitro and in vivo. We observed that Rhoj is significantly overexpressed in GBM and associated with patients' survival. However, the role and underlying molecular mechanism of Rhoj in GBM are still unclear. We demonstrated that transcription factor c-Jun regulated the expression of Rhoj, and Rhoj interacted with moesin to promote GBM cell proliferation and migration by potentiating the activation of Rac1/PAK pathway and cytoskeletal dynamics. Rhoj may promote migration and invasion of GBM cells by regulating epithelial-mesenchymal transition (EMT)-like process. In conclusion, the Rhoj/Rac1/PAK signaling mediates invasion and progression of GBM and is a potential therapeutic target for GBM treatment. Rhoj may also be a promising biomarker for GBM diagnosis and prognosis.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Citoesqueleto/metabolismo , Progresión de la Enfermedad , Glioblastoma/metabolismo , Proteínas de Unión al GTP rho/biosíntesis , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Citoesqueleto/genética , Citoesqueleto/patología , Glioblastoma/genética , Glioblastoma/patología , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Proteínas de Unión al GTP rho/genética
4.
J Cell Mol Med ; 24(18): 10573-10588, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32779334

RESUMEN

Epileptogenesis is a potential process. Mossy fibre sprouting (MFS) and synaptic plasticity promote epileptogenesis. Overexpression of repulsive guidance molecule a (RGMa) prevents epileptogenesis by inhibiting MFS. However, other aspects underlying the RGMa regulatory process of epileptogenesis have not been elucidated. We studied whether RGMa could be modulated by microRNAs and regulated RhoA in epileptogenesis. Using microRNA databases, we selected four miRNAs as potential candidates. We further experimentally confirmed miR-20a-5p as a RGMa upstream regulator. Then, in vitro, by manipulating miR-20a-5p and RGMa, we investigated the regulatory relationship between miR-20a-5p, RGMa and RhoA, and the effects of this pathway on neuronal morphology. Finally, in the epilepsy animal model, we determined whether the miR-20a-5p-RGMa-RhoA pathway influenced MFS and synaptic plasticity and then modified epileptogenesis. Our results showed that miR-20a-5p regulated RGMa and that RGMa regulated RhoA in vitro. Furthermore, in primary hippocampal neurons, the miR-20a-5p-RGMa-RhoA pathway regulated axonal growth and neuronal branching; in the PTZ-induced epilepsy model, silencing miR-20a-5p prevented epileptogenesis through RGMa-RhoA-mediated synaptic plasticity but did not change MFS. Overall, we concluded that silencing miR-20a-5p inhibits axonal growth and neuronal branching and prevents epileptogenesis through RGMa-RhoA-mediated synaptic plasticity in the PTZ-induced epilepsy model, thereby providing a possible strategy to prevent epileptogenesis.


Asunto(s)
Proteínas Ligadas a GPI/fisiología , Proteínas de la Membrana/fisiología , MicroARNs/genética , Proteínas del Tejido Nervioso/fisiología , Plasticidad Neuronal/fisiología , Convulsiones/prevención & control , Proteínas de Unión al GTP rho/fisiología , Regiones no Traducidas 3' , Animales , Axones/ultraestructura , Células Cultivadas , Convulsivantes/toxicidad , Dependovirus/genética , Modelos Animales de Enfermedad , Femenino , Proteínas Ligadas a GPI/biosíntesis , Proteínas Ligadas a GPI/genética , Regulación de la Expresión Génica , Silenciador del Gen , Vectores Genéticos , Hipocampo/citología , Masculino , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , MicroARNs/antagonistas & inhibidores , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Plasticidad Neuronal/genética , Neuronas/ultraestructura , Pentilenotetrazol/toxicidad , ARN/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Convulsiones/inducido químicamente , Convulsiones/genética , Convulsiones/fisiopatología , Transducción de Señal , Proteínas de Unión al GTP rho/biosíntesis , Proteínas de Unión al GTP rho/genética
5.
J Neurosurg ; 134(5): 1599-1609, 2020 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-32470939

RESUMEN

OBJECTIVE: Neuropathic pain caused by traumatic neuromas is an extremely intractable clinical problem. Disorderly scar tissue accumulation and irregular and immature axon regeneration around the injury site mainly contribute to traumatic painful neuroma formation. Therefore, successfully preventing traumatic painful neuroma formation requires the effective inhibition of irregular axon regeneration and disorderly accumulation of scar tissue. Considering that chondroitin sulfate proteoglycans (CSPGs) can act on the growth cone and effectively inhibit axon regeneration, the authors designed and manufactured a CSPG-gelatin blocker to regulate the CSPGs' spatial distribution artificially and applied it in a rat model after sciatic nerve neurectomy to evaluate its effects in preventing traumatic painful neuroma formation. METHODS: Sixty female Sprague Dawley rats were randomly divided into three groups (positive group: no covering; blank group: covering with gelatin blocker; and CSPG group: covering with the CSPG-gelatin blocker). Pain-related factors were evaluated 2 and 8 weeks postoperatively (n = 30). Neuroma growth, autotomy behavior, and histological features of the neuromas were assessed 8 weeks postoperatively (n = 30). RESULTS: Eight weeks postoperatively, typical bulb-shaped neuromas did not form in the CSPG group, and autotomy behavior was obviously better in the CSPG group (p < 0.01) than in the other two groups. Also, in the CSPG group the regenerated axons showed a lower density and more regular and improved myelination (p < 0.01). Additionally, the distribution and density of collagenous fibers and the expression of α-smooth muscle actin were significantly lower in the CSPG group than in the positive group (p < 0.01). Regarding pain-related factors, c-fos, substance P, interleukin (IL)-17, and IL-1ß levels were significantly lower in the CSPG group than those in the positive and blank groups 2 weeks postoperatively (p < 0.05), while substance P and IL-17 remained lower in the CSPG group 8 weeks postoperatively (p < 0.05). CONCLUSIONS: The authors found that CSPGs loaded in a gelatin blocker can prevent traumatic neuroma formation and effectively relieve pain symptoms after sciatic nerve neurotomy by blocking irregular axon regeneration and disorderly collagenous fiber accumulation in the proximal nerve stump. These results indicate that covering the proximal nerve stump with CSPGs may be a new and promising strategy to prevent traumatic painful neuroma formation in the clinical setting.


Asunto(s)
Proteoglicanos Tipo Condroitín Sulfato/uso terapéutico , Regeneración Nerviosa/efectos de los fármacos , Neuralgia/prevención & control , Neuroma/prevención & control , Neoplasias del Sistema Nervioso Periférico/prevención & control , Neuropatía Ciática/tratamiento farmacológico , Ciática/prevención & control , Administración Tópica , Animales , Axones/efectos de los fármacos , Conducta Animal , Proteoglicanos Tipo Condroitín Sulfato/administración & dosificación , Cicatriz/etiología , Femenino , Ganglios Espinales/metabolismo , Gelatina , Conos de Crecimiento/efectos de los fármacos , Interleucina-17/sangre , Interleucina-1beta/sangre , Iridoides/administración & dosificación , Neuralgia/etiología , Neuroma/etiología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Ciática/etiología , Método Simple Ciego , Proteínas de Unión al GTP rho/biosíntesis , Proteínas de Unión al GTP rho/genética
6.
Inflammation ; 43(4): 1476-1487, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32240450

RESUMEN

Sanggenon C (SC), a natural flavonoid extracted from Cortex Mori (Sang Bai Pi), is reported to possess anti-inflammatory and antioxidant properties in hypoxia. The present study aimed to investigate the therapeutic potential and the underlying mechanisms of SC in cerebral ischemia-reperfusion (I/R) injury. A rat model of reversible middle cerebral artery occlusion (MCAO) was used to induce cerebral I/R injury in vivo, and SC was administrated intragastrically. Brain injuries were evaluated using Bederson scores, brain water content, and 2, 3, 5-triphenyltetrazolium chloride (TTC) staining. The levels of inflammatory factors and oxidative stress were examined using corresponding kits. Cell apoptosis was evaluated by TUNEL. Moreover, the expressions of apoptosis-related and RhoA/ROCK signaling-related proteins were detected through western blotting. In vitro, RhoA was overexpressed in oxygen-glucose deprivation and reperfusion (OGD/R)-induced PC12 cells to confirm the contribution of RhoA-ROCK signaling inhibition by SC to the neuroprotective effects post OGD/R. Pretreatment with SC significantly ameliorated the neurologic impairment, brain edema, and cerebral infarction post MCAO-reperfusion, associated with reductions of inflammation, oxidative stress, and cell apoptosis in the brain. Furthermore, SC remarkably downregulated the expression of RhoA/ROCK signaling-related proteins post MCAO-reperfusion in rats, while overexpression of RhoA reversed the beneficial effects of SC on protecting against inflammation and oxidative stress in OGD/R-induced PC12 cells. Taken together, these findings demonstrated that SC exerts neuroprotective effects after cerebral I/R injury via inhibiting inflammation and oxidative stress through regulating RhoA-ROCK signaling, suggesting a therapeutic potential of SC in cerebral I/R injury.


Asunto(s)
Antiinflamatorios/uso terapéutico , Benzofuranos/uso terapéutico , Isquemia Encefálica/tratamiento farmacológico , Cromonas/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Daño por Reperfusión/tratamiento farmacológico , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Quinasas Asociadas a rho/antagonistas & inhibidores , Animales , Antiinflamatorios/farmacología , Benzofuranos/farmacología , Isquemia Encefálica/metabolismo , Cromonas/farmacología , Relación Dosis-Respuesta a Droga , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Masculino , Estrés Oxidativo/fisiología , Células PC12 , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/metabolismo , Proteínas de Unión al GTP rho/biosíntesis , Quinasas Asociadas a rho/biosíntesis
7.
Pharmacol Rep ; 72(4): 903-911, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32144744

RESUMEN

BACKGROUND: Chronic heart failure (CHF) is characterized by left ventricular dysfunction and altered autonomic control of cardiac function. This study aimed to investigate the effects of atorvastatin on left ventricular remodeling (LVR) and cardiac function in rats with isoproterenol-induced CHF and the possible mechanism. METHODS: An isoproterenol-induced CHF model was established in rata, which were subsequently treated with atorvastatin. Echocardiography, hemodynamic, and left ventricular mass indexes were assessed. The mRNA expression of RhoA, Rho kinase, and endothelial nitric oxide synthase (eNOS) was determined by RT-qPCR. The protein expression of myosin-binding subunit (MBS), MBS-P, eNOS, phosphorylated-eNOS, RhoA, and Rho kinase was measured by Western blot analysis. The relative activity of NADPH oxidase, ROS, and NO was assessed by ELISA. RESULTS: Isoproterenol-induced CHF rats treated with atorvastatin exhibited decreased left ventricular end-systolic dimension, left ventricular end-diastolic dimension, left ventricular end-diastolic pressure, left ventricular mass index, maximum fall rate of change in left ventricular pressure, heart rate (p < 0.001), expression of RhoA, Rho kinase, MBS and MBS-P (p < 0.01), and relative activity of NADPH oxidase, ROS and NO (p < 0.05) and increased left ventricular short axis fractional shortening, left ventricular end-systolic pressure, maximum rise rate of change in left ventricular pressure (p < 0.001) and expression of eNOS, and phosphorylated-eNOS ser1177 (all p < 0.05) compared with those of rats with isoproterenol-induced CHF. CONCLUSION: We demonstrated that atorvastatin inhibits LVR and improves cardiac function in rats with isoproterenol-induced CHF through inhibition of the RhoA/Rho kinase signaling pathway.


Asunto(s)
Atorvastatina/uso terapéutico , Insuficiencia Cardíaca/tratamiento farmacológico , Isoproterenol/toxicidad , Remodelación Ventricular/efectos de los fármacos , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Quinasas Asociadas a rho/antagonistas & inhibidores , Animales , Atorvastatina/farmacología , Cardiotónicos/toxicidad , Enfermedad Crónica , Ecocardiografía/métodos , Insuficiencia Cardíaca/inducido químicamente , Insuficiencia Cardíaca/diagnóstico por imagen , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Masculino , Óxido Nítrico Sintasa de Tipo III/biosíntesis , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Remodelación Ventricular/fisiología , Proteínas de Unión al GTP rho/biosíntesis , Quinasas Asociadas a rho/biosíntesis
8.
Pathol Res Pract ; 216(3): 152736, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31757662

RESUMEN

Recent studies have reported that dishevelled-associated activator of morphogenesis 1 (DAAM1) is remarkably essential for mediating cell migration and invasion in breast cancer (BrCa). Nonetheless, the definite expression profile of DAAM1 in BrCa patients and the impact on metastasis of BrCa in vivo have not been explored up to now. The differential expression of DAAM1 in BrCa and adjacent tissues was assessed via immunohistochemistry (IHC) staining. The metastatic capacities of BrCa SUM-1315 cells were examined in BALB/c nude mice. Besides, the prognostic values of DAAM1 mRNA in BrCa were explored based on Kaplan-Meier (KM) plotter. The expression of DAAM1 protein was notably overexpressed in BrCa tissues compared with that in paired normal breast tissues. The high expression of DAAM1 in BrCa tissues was significantly associated with lymph-node metastasis. Furthermore, DAAM1 overexpression promoted the invasive capacity of BrCa cells and stimulated lung metastatic extent in vivo. We also found that overexpressed DAAM1 mRNA was significantly associated with poor relapse-free survival (RFS), overall survival (OS), distance-metastasis-free survival (DMFS), and post-progression survival (PPS). Our findings reveal that DAAM1 might be a novel therapeutic target to manage the deteriorated metastasis of BrCa and identified DAAM1 as a promising biomarker for unfavorable prognosis in BrCa patients.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/patología , Metástasis Linfática/patología , Proteínas de Microfilamentos/biosíntesis , Proteínas de Unión al GTP rho/biosíntesis , Animales , Neoplasias de la Mama/mortalidad , Femenino , Xenoinjertos , Humanos , Metástasis Linfática/genética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Pronóstico , Regulación hacia Arriba
9.
Biochem Pharmacol ; 168: 412-428, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31325448

RESUMEN

Although having promising anti-myeloma properties, the pan-histone deacetylase inhibitor (HDACi) panobinostat lacks therapeutic activity as a single agent. The aim of the current study was to elucidate the mechanisms underlying multiple myeloma (MM) resistance to panobinostat monotherapy and to define strategies to overcome it. Sensitivity of MM cell lines and primary CD138+ cells from MM patients to panobinostat correlated with reduced expression of the chemokine receptor CXCR4, whereas overexpression of CXCR4 in MM cell lines increased their resistance to panobinostat. Decreased sensitivity to HDACi was associated with reversible G0/G1 cell growth arrest while response was characterized by apoptotic cell death. Analysis of intra-cellular signaling mediators revealed the pro-survival mTOR pathway to be regulated by CXCR4 overexpression. Combining panobinostat with mTOR inhibitor everolimus abrogated the resistance to HDACi and induced synergistic cell death. The combination of panobinostat/everolimus resulted in sustained DNA damage and irreversible suppression of proliferation accompanied by robust apoptosis. Gene expression analysis revealed distinct genetic profiles of single versus combined agent exposure. Whereas panobinostat increased the expression of the cell cycle inhibitor p21, co-treatment with everolimus abrogated the increase in p21 and synergistically downregulated the expression of DNA repair genes and mitotic checkpoint regulators. Importantly, the combination of panobinostat with everolimus effectively targeted CXCR4-expressing resistant MM cells in vivo in the BM niche. In summary, our results uncover the mechanism responsible for the strong synergistic anti-MM activity of dual HDAC and mTOR inhibition and provide the rationale for a novel potential therapeutic approach to treat MM.


Asunto(s)
Antineoplásicos/administración & dosificación , Everolimus/administración & dosificación , Inhibidores de Histona Desacetilasas/administración & dosificación , Mitosis/efectos de los fármacos , Panobinostat/administración & dosificación , Receptores CXCR4/antagonistas & inhibidores , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Línea Celular Tumoral , Células Cultivadas , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/fisiología , Humanos , Ratones , Mitosis/fisiología , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Receptores CXCR4/biosíntesis , Receptores CXCR4/genética , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/biosíntesis , Serina-Treonina Quinasas TOR/genética , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Proteínas de Unión al GTP rho/biosíntesis , Proteínas de Unión al GTP rho/genética
10.
J Pineal Res ; 67(2): e12586, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31077613

RESUMEN

Disruption of circadian time structure and suppression of circadian nocturnal melatonin (MLT) production by exposure to dim light at night (dLAN), as occurs with night shift work and/or disturbed sleep-wake cycles, is associated with a significantly increased risk of breast cancer and resistance to tamoxifen and doxorubicin. Melatonin inhibition of human breast cancer chemoresistance involves mechanisms including suppression of tumor metabolism and inhibition of kinases and transcription factors which are often activated in drug-resistant breast cancer. Signal transducer and activator of transcription 3 (STAT3), frequently overexpressed and activated in paclitaxel (PTX)-resistant breast cancer, promotes the expression of DNA methyltransferase one (DNMT1) to epigenetically suppress the transcription of tumor suppressor Aplasia Ras homolog one (ARHI) which can sequester STAT3 in the cytoplasm to block PTX resistance. We demonstrate that breast tumor xenografts in rats exposed to dLAN and circadian MLT disrupted express elevated levels of phosphorylated and acetylated STAT3, increased DNMT1, but reduced sirtuin 1 (SIRT1) and ARHI. Furthermore, MLT and/or SIRT1 administration blocked/reversed interleukin 6 (IL-6)-induced acetylation of STAT3 and its methylation of ARH1 to increase ARH1 mRNA expression in MCF-7 breast cancer cells. Finally, analyses of the I-SPY 1 trial demonstrate that elevated MT1 receptor expression is significantly correlated with pathologic complete response following neo-adjuvant therapy in breast cancer patients. This is the first study to demonstrate circadian disruption of MLT by dLAN driving intrinsic resistance to PTX via epigenetic mechanisms increasing STAT3 expression and that MLT administration can reestablish sensitivity of breast tumors to PTX and drive tumor regression.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Resistencia a Antineoplásicos/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Melatonina/farmacología , Paclitaxel/farmacología , Factor de Transcripción STAT3/metabolismo , Proteínas Supresoras de Tumor/biosíntesis , Proteínas de Unión al GTP rho/biosíntesis , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ritmo Circadiano/efectos de los fármacos , Femenino , Humanos , Células MCF-7 , Ratas Desnudas , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Physiol Res ; 68(1): 89-98, 2019 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-30433799

RESUMEN

The contraction of gastrointestinal (GI) smooth muscles is regulated by both Ca(2+)-dependent and Ca(2+) sensitization mechanisms. Proline-rich tyrosine kinase 2 (Pyk2) is involved in the depolarization-induced contraction of vascular smooth muscle via a Ca(2+) sensitization pathway. However, the role of Pyk2 in GI smooth muscle contraction is unclear. The spontaneous contraction of colonic smooth muscle was measured by using isometric force transducers. Protein and phosphorylation levels were determined by using western blotting. Pyk2 protein was expressed in colonic tissue, and spontaneous colonic contractions were inhibited by PF-431396, a Pyk2 inhibitor, in the presence of tetrodotoxin (TTX). In cultured colonic smooth muscle cells (CSMCs), PF-431396 decreased the levels of myosin light chain (MLC20) phosphorylated at Ser19 and ROCK2 protein expression, but myosin light chain kinase (MLCK) expression was not altered. However, Y-27632, a Rho kinase inhibitor, increased phosphorylation of Pyk2 at Tyr402 and concomitantly decreased ROCK2 levels; the expression of MLCK in CSMCs did not change. The expression of P(Tyr402)-Pyk2 and ROCK2 was increased when CSMCs were treated with Ach. Pyk2 is involved in the process of colonic smooth muscle contraction through the RhoA/ROCK pathway. These pathways may provide very important targets for investigating GI motility disorders.


Asunto(s)
Colon/metabolismo , Quinasa 2 de Adhesión Focal/biosíntesis , Contracción Muscular/fisiología , Músculo Liso/metabolismo , Proteínas de Unión al GTP rho/biosíntesis , Quinasas Asociadas a rho/biosíntesis , Animales , Técnicas de Cultivo de Célula , Colon/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Quinasa 2 de Adhesión Focal/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos ICR , Contracción Muscular/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Técnicas de Cultivo de Órganos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Proteína de Unión al GTP rhoA
12.
Methods Mol Biol ; 1821: 247-256, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30062417

RESUMEN

RhoA is a founding member of Rho GTPase family and is a well-recognized regulator of actin cytoskeleton dynamics. Signal pathways transduced by RhoA are involved in cell migration, polarity, morphogenesis, proliferation, survival, and cell fate decision. Conditional gene targeting of Rhoa in murine blood system induces acute hematopoietic failure due to the loss of multilineage hematopoietic progenitor cells (HPC) caused by a cytokinesis defect and necrosis. Here we describe a method to conditionally induce Rhoa gene knockout in murine blood cells and a rescue by exogenous RhoA expression with lentivirus in HPCs, an approach that has general applicability in studying in vivo function of Rho GTPases and their regulators/effectors by gene targeting.


Asunto(s)
Técnicas de Inactivación de Genes/métodos , Células Madre Hematopoyéticas/enzimología , Lentivirus , Transducción Genética/métodos , Proteínas de Unión al GTP rho , Animales , Células HEK293 , Células Madre Hematopoyéticas/citología , Humanos , Ratones , Células 3T3 NIH , Proteínas de Unión al GTP rho/biosíntesis , Proteínas de Unión al GTP rho/genética , Proteína de Unión al GTP rhoA
13.
Methods Mol Biol ; 1821: 359-370, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30062424

RESUMEN

Rho GTPases are Ras-family G proteins that regulate many critical cellular functions. Due to their requirement during early embryonic development, investigations into the function of Rho GTPases at a tissue-specific level require inducible and spatially targeted modulation of Rho GTPase activity. Here, we describe the use of ten novel zebrafish transgenics enabling GAL4-specific expression of Rho GTPases to modulate Rho GTPase activity with spatial and temporal control.


Asunto(s)
Animales Modificados Genéticamente , Proteínas de Unión al ADN , Embrión no Mamífero/embriología , Desarrollo Embrionario , Factores de Transcripción , Proteínas de Pez Cebra , Pez Cebra , Proteínas de Unión al GTP rho , Animales , Animales Modificados Genéticamente/embriología , Animales Modificados Genéticamente/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Proteínas de Unión al GTP rho/biosíntesis , Proteínas de Unión al GTP rho/genética
14.
J Neurochem ; 147(1): 84-98, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30071134

RESUMEN

The nucleus accumbens (NAc) is a critical brain reward region that mediates the rewarding effects of drugs of abuse, including those of morphine and other opiates. Drugs of abuse induce widespread alterations in gene transcription and dendritic spine morphology in medium spiny neurons (MSNs) of the NAc that ultimately influence NAc excitability and hence reward-related behavioral responses. Growing evidence indicates that within the NAc small GTPases are common intracellular targets of drugs of abuse where these molecules regulate drug-mediated transcriptional and spine morphogenic effects. The RhoA small GTPase is among the most well-characterized members of the Ras superfamily of small GTPases, and recent work highlights an important role for hippocampal RhoA in morphine-facilitated reward behavior. Despite this, it remains unclear how RhoA pathway signaling in the NAc is affected by withdrawal from morphine. To investigate this question, using subcellular fractionation and subsequent protein profiling we examined the expression of key components of the RhoA pathway in NAc nuclear, cytoplasmic, and synaptosomal compartments during multiple withdrawal periods from repeated morphine administration. Furthermore, using in vivo viral-mediated gene transfer, we determined the consequences of revealed RhoA pathway alterations on NAc MSN dendritic spine morphology. Our findings reveal an important role for RhoA signaling cascades in mediating the effects of long-term morphine withdrawal on NAc MSN dendritic spine elimination. OPEN PRACTICES: Open Science: This manuscript was awarded with the Open Materials Badge. For more information see: https://cos.io/our-services/open-science-badges/.


Asunto(s)
Morfina , Narcóticos , Núcleo Accumbens/metabolismo , Núcleo Accumbens/patología , Síndrome de Abstinencia a Sustancias/metabolismo , Síndrome de Abstinencia a Sustancias/patología , Sinapsis/metabolismo , Sinapsis/patología , Proteínas de Unión al GTP rho/biosíntesis , Animales , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Núcleo Accumbens/efectos de los fármacos , Recompensa , Transducción de Señal/efectos de los fármacos , Sinapsis/efectos de los fármacos , Sinaptosomas/efectos de los fármacos , Sinaptosomas/metabolismo , Proteínas de Unión al GTP rho/genética , Proteína de Unión al GTP rhoA
15.
Int J Dev Neurosci ; 69: 88-96, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30009881

RESUMEN

Methyl mercury is a teratogenic and neurodevelopmental toxicant in the environment. MeHg affects several biological pathways critical for brain development. The present study validated the effect of Fisetin on developmental MeHg exposure induced alterations in mitochondrial apoptotic pathway and Rho GTPase mRNA expressions in hippocampus of F1 generation rats. Pregnant Wistar rats were grouped as Group I : administered with vehicle control, Group II: MeHg (1.5 mg/kg b.w), Group III: MeHg + Fisetin (10 mg/kg b.w), Group IV: MeHg + Fisetin (30 mg/kg b.w), Group V: MeHg + Fisetin (50 mg/kg b.w), Group VI: MeHg + Fisetin (70 mg/kg b.w), Group VII: Fisetin (30 mg/kg b.w) alone. Fisetin reduced mercury accumulation in offspring brain. In hippocampus, Fisetin preserved mitochondrial total thiol status, glutathione antioxidant system, mitochondrial metabolic integrity and respiratory chain activity. Fisetin ameliorated apoptotic signals by preventing Cytochrome c release, down regulating ERK 1/2 and Caspase 3 gene expression. Fisetin also upregulated mRNA expressions of RhoA/Rac1/Cdc42 in hippocampus. Predominant effect of Fisetin was to reduce mercury accumulation in offspring brain there by diminishing the toxic effect of MeHg. Hence we showed that, gestational intake of Fisetin (30 mg/kg b.w.) impedes developmental MeHg neurotoxicity by regulating mitochondrial apoptotic and Rho GTPase signalling molecules and by reducing the mercury accumulation in hippocampus of F1 generation rats.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteínas Reguladoras de la Apoptosis/efectos de los fármacos , Flavonoides/uso terapéutico , Hipocampo/metabolismo , Intoxicación del Sistema Nervioso por Mercurio/tratamiento farmacológico , Intoxicación del Sistema Nervioso por Mercurio/psicología , Compuestos de Metilmercurio , Fármacos Neuroprotectores/uso terapéutico , Transducción de Señal/efectos de los fármacos , Proteínas de Unión al GTP rho/biosíntesis , Animales , Antioxidantes/metabolismo , Relación Dosis-Respuesta a Droga , Transporte de Electrón/efectos de los fármacos , Femenino , Flavonoles , Hipocampo/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mercurio/metabolismo , Embarazo , Ratas , Ratas Wistar , Proteínas de Unión al GTP rho/genética
16.
Int J Food Microbiol ; 278: 1-10, 2018 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-29689332

RESUMEN

Results from this study explored the inhibitory effect of RHO1 gene (GenBank accession number KY859864) from the antagonistic yeast, Cryptococcus laurentii ZJU10, on the control of Penicillium expansum in pear fruit and its possible mechanism involved. The RHO1 gene was successfully cloned and overexpressed in Saccharomyces cerevisiae. Sequence analysis showed high similarity with Rho family proteins, implying a primary role of Rho1 in the cell wall integrity (CWI) signaling pathway. Gene expression of RHO1 and other five CWI-related genes (including Pkc1, Rlm1, Fks1, Fks2 and Chs3) were significant up-regulated in the treatment of SC/Rho1-induced strain (Saccharomyces cerevisiae transformed with RHO1 and induced by galactose). Meanwhile, SC/Rho1-induced treatment reduced about 61.5% of disease incidence and almost 5-times lower lesion diameter compared to the control. In addition, the growth of transformed strains was slightly lower in contrast to the wild Saccharomyces cerevisiae and the induction of fruit resistance was significantly enhanced, which was tightly linked with triggering stronger host defensive responses by priming activation. This is the first study that Rho1 has a potential function of suppressing fungal disease in harvested fruit by activating CWI signaling pathway and indicates an alternative strategy for postharvest disease management.


Asunto(s)
Agentes de Control Biológico/metabolismo , Cryptococcus/metabolismo , Frutas/microbiología , Penicillium/crecimiento & desarrollo , Pyrus/microbiología , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Unión al GTP rho/genética , Pared Celular/metabolismo , China , Quitina Sintasa/genética , Clonación Molecular , Cryptococcus/genética , Equinocandinas/genética , Glucosiltransferasas/genética , Proteínas de la Membrana/genética , Enfermedades de las Plantas/microbiología , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/biosíntesis , Transducción de Señal , Proteínas de Unión al GTP rho/biosíntesis
17.
Acta Biomater ; 72: 434-446, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29649639

RESUMEN

Histological assessments of synovial tissues from patients with failed CoCr alloy hip prostheses demonstrate extensive infiltration and accumulation of macrophages, often loaded with large quantities of particulate debris. The resulting adverse reaction to metal debris (ARMD) frequently leads to early joint revision. Inflammatory response starts with the recruitment of immune cells and requires the egress of macrophages from the inflamed site for resolution of the reaction. Metal ions (Co2+ and Cr3+) have been shown to stimulate the migration of T lymphocytes but their effects on macrophages motility are still poorly understood. To elucidate this, we studied in vitro and in vivo macrophage migration during exposure to cobalt and chromium ions and nanoparticles. We found that cobalt but not chromium significantly reduces macrophage motility. This involves increase in cell spreading, formation of intracellular podosome-type adhesion structures and enhanced cell adhesion to the extracellular matrix (ECM). The formation of podosomes was also associated with the production and activation of matrix metalloproteinase-9 (MMP9) and enhanced ECM degradation. We showed that these were driven by the down-regulation of RhoA signalling through the generation of reactive oxygen species (ROS). These novel findings reveal the key mechanisms driving the wear/corrosion metallic byproducts-induced inflammatory response at non-toxic concentrations. STATEMENT OF SIGNIFICANCE: Adverse tissue responses to metal wear and corrosion products from CoCr alloy implants remain a great challenge to surgeons and patients. Macrophages are the key regulators of these adverse responses to the ions and debris generated. We demonstrated that cobalt, rather than chromium, causes macrophage retention by restructuring the cytoskeleton and inhibiting cell migration via ROS production that affects Rho Family GTPase. This distinctive effect of cobalt on macrophage behaviour can help us understand the pathogenesis of ARMD and the cellular response to cobalt based alloys, which provide useful information for future implant design and biocompatibility testing.


Asunto(s)
Cobalto , Regulación de la Expresión Génica/efectos de los fármacos , Macrófagos/metabolismo , Nanopartículas del Metal , Especies Reactivas de Oxígeno/metabolismo , Proteínas de Unión al GTP rho/biosíntesis , Proteína de Unión al GTP rhoA/biosíntesis , Animales , Cobalto/efectos adversos , Cobalto/química , Cobalto/farmacología , Reacción a Cuerpo Extraño/inducido químicamente , Reacción a Cuerpo Extraño/metabolismo , Reacción a Cuerpo Extraño/patología , Humanos , Macrófagos/patología , Nanopartículas del Metal/efectos adversos , Nanopartículas del Metal/química , Ratones , Transducción de Señal/efectos de los fármacos , Células U937
18.
Cancer Biomark ; 21(4): 787-795, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29504523

RESUMEN

BACKGROUND: Aplysia ras homology member I/ARHI is known as ovarian cancer suppressor gene and potential inhibitor of signal transducer and activator of transcription 3/STAT3 signaling. Resveratrol suppresses growth and STAT3 activation of ovarian cancer cells, while its influence in ARHI expression remains unknown. OBJECTIVE: The current study aims to elucidate the status of ARHI expression and its relevance with growth suppression and STAT3 inactivation of resveratrol-treated cells. METHODS: ARHI expression patterns of three ovarian cancer cell lines (human CAOV-3, OVCAR-3 and rat NUTU-19) without and with 100 µM resveratrol treatment were checked by immunocytochemical staining, Western blotting and RT-PCR. The involvement of ARHI in the growth inhibition and STAT3 inactivation of resveratrol-treated OVCAR-3 cells was investigated by transfection of ARHI-specific siRNA. RESULTS: ARHI is expressed in low levels in three ovarian cancer cell lines, which is upregulated upon resveratrol treatment accompanied with growth arrest, extensive apoptosis, increased autophagic activity and inactivated STAT3 signaling. Specific siRNA transfection efficiently knocked down ARHI expression in resveratrol-treated CAOV-3 and OVCAR-3 cells and increased the total cell number in limited extents (P> 0.05) in comparison with that of resveratrol-treated ovarian cancer cells without any transfection or transfected with mock oligonucleotides. ARHI knockdown failed to prevent resveratrol-caused STAT3 inactivation and cell crisis. CONCLUSION: ARHI upregulation is another molecular event caused by resveratrol and one of the elements related with resveratrol's anti-ovarian cancer efficacy. Resveratrol may inactivate STAT3 signaling of ovarian cancer cells in ARHI unrelated pattern(s).


Asunto(s)
Antioxidantes/farmacología , Neoplasias Ováricas/patología , Factor de Transcripción STAT3/metabolismo , Estilbenos/farmacología , Proteínas de Unión al GTP rho/biosíntesis , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Neoplasias Ováricas/metabolismo , Resveratrol , Regulación hacia Arriba
19.
J Am Heart Assoc ; 6(5)2017 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-28526701

RESUMEN

BACKGROUND: Netrin-1 (NTN-1) has been established to be a novel intrinsic regulator of blood-brain barrier (BBB) maintenance. This study was carried out to investigate the potential roles of exogenous NTN-1 in preserving BBB integrity after experimental subarachnoid hemorrhage (SAH) as well as the underlying mechanisms of its protective effects. METHODS AND RESULTS: A total of 309 male Sprague-Dawley rats were subjected to an endovascular perforation model of SAH. Recombinant NTN-1 was administered intravenously 1 hour after SAH induction. NTN-1 small interfering RNA or Deleted in Colorectal Cancer small interfering RNA was administered intracerebroventricular at 48 hours before SAH. Focal adhesion kinase inhibitor was administered by intraperitoneal injection at 1 hour prior to SAH. Neurological scores, brain water content, BBB permeability, RhoA activity, Western blot, and immunofluorescence staining were evaluated. The expression of endogenous NTN-1 and its receptor Deleted in Colorectal Cancer were increased after SAH. Administration of exogenous NTN-1 significantly reduced brain water content and BBB permeability and ameliorated neurological deficits at 24 and 72 hours after SAH. Exogenous NTN-1 treatment significantly promoted phosphorylated focal adhesion kinase activation and inhibited RhoA activity, as well as upregulated the expression of ZO-1 and Occludin. Conversely, depletion of endogenous NTN-1 aggravated BBB breakdown and neurological impairments at 24 hours after SAH. The protective effects of NTN-1 at 24 hours after SAH were also abolished by pretreatment with Deleted in Colorectal Cancer small interfering RNA and focal adhesion kinase inhibitor. CONCLUSIONS: NTN-1 treatment preserved BBB integrity and improved neurological functions through a Deleted in Colorectal Cancer/focal adhesion kinase/RhoA signaling pathway after SAH. Thus, NTN-1 may serve as a promising treatment to alleviate early brain injury following SAH.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Experimentales , Netrina-1/genética , ARN Neoplásico/genética , Hemorragia Subaracnoidea/genética , Animales , Western Blotting , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Neoplasias Colorrectales/complicaciones , Neoplasias Colorrectales/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/antagonistas & inhibidores , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Infusiones Intraventriculares , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Masculino , Netrina-1/administración & dosificación , Netrina-1/biosíntesis , Ocludina/biosíntesis , Ocludina/genética , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/administración & dosificación , Transducción de Señal , Hemorragia Subaracnoidea/etiología , Hemorragia Subaracnoidea/prevención & control , Proteína de la Zonula Occludens-1/biosíntesis , Proteína de la Zonula Occludens-1/genética , Proteínas de Unión al GTP rho/biosíntesis , Proteínas de Unión al GTP rho/genética
20.
Calcif Tissue Int ; 101(3): 328-340, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28474171

RESUMEN

Approximately 45% of people of East Asian descent have the inactive aldehyde dehydrogenase 2 (ALDH2) phenotype. The enzyme defect of ALDH2 has been found to adversely influence the risk of osteoporosis. The aim of this study was to clarify the effect of skeletal loading on trabecular bone structure and dynamics in Aldh2-disrupted mice in the absence of alcohol consumption. Four-week-old male Aldh2-/- (KO) and Aldh2+/+ (WT) mice were divided into a ground control (GC) group and a climbing exercise (CE) group in each genotype. The trabecular bone mineral density of the distal femur measured by peripheral quantitative computed tomography in the wild-type CE (WTCE) group was significantly higher than that in the wild-type GC (WTGC) group; however, there was no significant difference between the knockout CE (KOCE) and knockout GC (KOGC) groups. Bone histomorphometry revealed that osteogenic parameters were significantly increased in the WTCE group compared with the WTGC group, but not increased in the KOCE group compared with the KOGC group. Quantitative reverse transcriptase polymerase chain reaction and flow cytometry revealed that mRNA and protein expression levels of p21 were significantly decreased in the WTCE group compared with those in the WTGC group, while these differences were not observed between the KOGC and KOCE groups. This study provides the first in vivo evidence that p21 expression in the bone marrow is not decreased after skeletal loading and osteoblast differentiation is impaired in the absence of Aldh2 gene.


Asunto(s)
Aldehído Deshidrogenasa Mitocondrial/genética , Densidad Ósea/fisiología , Huesos/fisiología , Diferenciación Celular/genética , Proteínas de Unión al GTP rho/biosíntesis , Animales , Células de la Médula Ósea/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoblastos/citología , Condicionamiento Físico Animal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...