Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 199
Filtrar
1.
J Ethnopharmacol ; 299: 115658, 2022 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-36075273

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Artemisia argyi H. Lév. & Vaniot (Asteraceae), also called "Chinese mugwort", is frequently used as a herbal medicine in China, Japan, Korea, and eastern parts of Russia. It is known as "ai ye" in China and "Gaiyou" in Japan. In ancient China, the buds and leaves of A. argyi were commonly consumed before and after Tomb-sweeping Day. It is used to treat malaria, hepatitis, cancer, inflammatory diseases, asthma, irregular menstrual cycle, sinusitis, and pathologic conditions of the kidney and liver. Although A. argyi extract (AAE) has shown anti-tumor activity against various cancers, the therapeutic effect and molecular mechanism of AAE remains to be further studied in lung cancer. AIM OF THE STUDY: This study aimed to demonstrate the anti-tumor effect of AAE and its associated biological mechanisms in CL1-0 parent and gemcitabine-resistant (CL1-0-GR) lung cancer cells. EXPERIMENTAL PROCEDURE: Human lung cancer cells CL1-0 and CL1-0-GR cells were treated with AAE. Cell viability was assessed using the MTT, colony, and spheroid formation assays. Migration, invasion, and immunofluorescence staining were used to determine the extent of epithelial- mesenchymal transition (EMT). JC-1 and MitoSOX fluorescent assays were performed to investigate the effect of AAE on mitochondria. Apoptosis was detected using the TUNEL assay and flow cytometry with Annexin V staining. RESULT: We found that A. argyi significantly decreased cell viability and induced apoptosis, accompanied by mitochondrial membrane depolarization and increased ROS levels in both parent cells (CL1-0) and gemcitabine-resistant lung cancer cells (CL1-0-GR). AAE-induced apoptosis is regulated via the PI3K/AKT and MAPK signaling pathways. It also prevents CL1-0 and CL1-0-GR cancer cell invasion, migration, EMT, colony formation, and spheroid formation. In addition, AAE acts cooperative with commercial chemotherapy drugs to enhance tumor spheroid shrinkage. CONCLUSION: Our study provides the first evidence that A. argyi treatment suppresses both parent and gemcitabine-resistant lung cancer cells by inducing ROS, mitochondrial membrane depolarization, and apoptosis, and reducing EMT. Our finding provides insights into the anti-cancer activity of A. argyi and suggests that A. argyi may serve as a chemotherapy adjuvant that potentiates the efficacy of chemotherapeutic agents.


Asunto(s)
Apoptosis , Artemisia , Neoplasias Pulmonares , Anexina A5/metabolismo , Anexina A5/farmacología , Anexina A5/uso terapéutico , Apoptosis/efectos de los fármacos , Artemisia/química , Línea Celular Tumoral , Desoxicitidina/análogos & derivados , Resistencia a Antineoplásicos , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Gemcitabina
2.
J Diabetes Res ; 2022: 4067812, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35155684

RESUMEN

Systemic vascular impairment is the most common complication of diabetes. Advanced glycation end products (AGEs) can exacerbate diabetes-related vascular damage by affecting the intima and media through a variety of mechanisms. In the study, we demonstrated that AGEs and their membrane receptor RAGE could induce the differentiation of EPCs into osteoblasts under certain circumstances, thereby promoting accelerated atherosclerosis. Differentiation into osteoblasts was confirmed by positive staining for DiI-acetylated fluorescently labeled low-density lipoprotein and FITC-conjugated Ulex europaeus agglutinin. During differentiation, expression of receptor for AGE (RAGE) was significantly upregulated. This upregulation was attenuated by transfection with RAGE-targeting small interfering (si)RNA. siRNA-mediated knockdown of RAGE expression significantly inhibited the upregulation of AGE-induced calcification-related proteins, such as runt-related transcription factor 2 (RUNX2) and osteoprotegerin (OPG). Additional experiments showed that AGE induction of EPCs significantly induced ERK, p38MAPK, and JNK activation. The AGE-induced upregulation of osteoblast proteins (RUNX2 and OPG) was suppressed by treatment with a p38MAPK inhibitor (SB203580) or JNK inhibitor (SP600125), but not by treatment with an ERK inhibitor (PD98059), which indicated that AGE-induced osteoblast differentiation from EPCs may be mediated by p38MAPK and JNK signaling, but not by ERK signaling. These data suggested that AGEs may bind to RAGE on the EPC membrane to trigger differentiation into osteoblasts. The underlying mechanism appears to involve the p38MAPK and JNK1/2 pathways, but not the ERK1/2 pathway.


Asunto(s)
Antígenos de Neoplasias/farmacología , Células Progenitoras Endoteliales/efectos de los fármacos , Productos Finales de Glicación Avanzada/farmacología , Proteínas Quinasas Activadas por Mitógenos/farmacología , Osteogénesis/genética , Animales , Antígenos de Neoplasias/metabolismo , Médula Ósea , Modelos Animales de Enfermedad , Células Progenitoras Endoteliales/fisiología , Productos Finales de Glicación Avanzada/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Osteogénesis/efectos de los fármacos , Osteogénesis/fisiología , Ratas , Ratas Sprague-Dawley/metabolismo
3.
Pharmacol Res Perspect ; 9(5): e00852, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34390539

RESUMEN

Myocardial ischemia (MI) remains the leading cause of mortality worldwide. Therefore, it is urgent to seek the treatment to protect the heart. [8]-Gingerol (8-Gin), one of the most active ingredients in ginger, has antioxidant, cardiotonic, and cardiovascular protective properties. The present study elucidated the cardioprotection effects and underlying mechanisms of 8-Gin in isoproterenol (ISO)-induced MI. ISO (85 mg/kg/d) was subcutaneously injected for 2 consecutive days to induce acute MI model in rats. Electrocardiography, oxidative stress levels, calcium concentrations, and apoptosis degree were observed. The effects of 8-Gin on L-type Ca2+ current (ICa-L ), contraction, and Ca2+ transients were monitored in rat myocytes via patch-clamp and IonOptix detection systems. 8-Gin decreased J-point elevation and heart rate and improved pathological heart damage. Moreover, 8-Gin reduced the levels of CK, LDH, and MDA, ROS production, and calcium concentrations in myocardial tissue, while increased the activities of SOD, CAT, and GSH. In addition, 8-Gin down-regulated Caspase-3 and Bax expressions, while up-regulated Bcl-2 expression. 8-Gin produced a marked decrease in the expression of p38, JNK, and ERK1/2 proteins. 8-Gin inhibited ICa-L , cell contraction, and Ca2+ transients in isolated rat myocytes. The results indicate that 8-Gin could exert anti-myocardial ischemic effects, which may be associated with oxidative stress reduction, cardiomyocytes apoptosis inhibition through MAPK signaling pathway, and Ca2+ homeostasis regulation via ICa-L modulation.


Asunto(s)
Canales de Calcio Tipo L/efectos de los fármacos , Cardiotónicos/farmacología , Catecoles/farmacología , Alcoholes Grasos/farmacología , Corazón/efectos de los fármacos , Isquemia Miocárdica/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Agonistas Adrenérgicos beta/toxicidad , Animales , Canales de Calcio Tipo L/metabolismo , Electrocardiografía/efectos de los fármacos , Isoproterenol/toxicidad , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Isquemia Miocárdica/patología , Miocardio/patología , Miocitos Cardíacos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Transducción de Señal
4.
Expert Rev Gastroenterol Hepatol ; 15(1): 65-79, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32946312

RESUMEN

INTRODUCTION: In colorectal carcinogenesis, genetic alterations in RAS and BRAF oncogenes play an important role for cancer initiation and/or progression and represent a key focus in the search for targeted therapies. Despite many years of research and a great amount of studies, until very recently this pathway was considered extremely hard to downregulate to obtain a significant clinical impact in colorectal cancer patients. But better times are coming with the advent of new promising drugs and combinations strategies. AREAS COVERED: In this review, we go over the biological characteristics of the MAPK pathway in colorectal tumors, while illustrating the clinical correlation of RAS and BRAF mutations, particularly its prognostic and predictive value. We also present newly data about recent improvements in the treatment strategy for patients harboring these types of tumors. EXPERT COMMENTARY: With great advances in the knowledge of molecular basis of RAS and BRAF mutant colorectal cancer in conjunction with biotechnology development and the constant effort for improvement, in the near future many new therapeutic options would be available for the management of this group of patient with dismal prognosis.


Asunto(s)
Neoplasias Colorrectales/genética , Genes ras/genética , Proteínas Proto-Oncogénicas B-raf/genética , Biomarcadores de Tumor/genética , Neoplasias Colorrectales/tratamiento farmacológico , Genes ras/efectos de los fármacos , Humanos , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Mutación , Oncogenes/efectos de los fármacos , Oncogenes/genética , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/efectos de los fármacos
5.
Anticancer Drugs ; 32(5): 517-525, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33290316

RESUMEN

Phosphatidylinositol-4,5-bisphosphate 3-kinases (PI3Ks) regulate several important cellular and subcellular processes including cell proliferation and differentiation. LY294002 was originally reported to be a selective inhibitor of PI3K-Akt. Later, it showed that this compound also inhibits several other molecules. In this study, we investigated the effect of LY294002 on the growth of suspension (MV4-11 and TF-1a) and tissue (Hep-G2) cells. In exponential phase, MV4-11 cells, but not TF-1a and Hep-G2 cells, expressed a low level of PI3Kp85 and addition of LY294002 inhibited the phosphorylation of PI3Kp85. LY294002 also significantly inhibited the proliferation of MV4-11, TF-1a and Hep-G2 cell and caused formation of cell clusters/aggregates measured by MTT and BrdU assays, and observed under an inverted microscope, respectively. Surprisingly, we found that LY294002 markedly repressed the activation of mitogen-activated protein kinase (MAPK) signal molecules, MEK and ERK, in all these cells. The inhibition of MEK and ERK was confirmed by using MEK stimulators, GM-CSF and phorbol 12-myristate 13-acetate, and MEK-specific inhibitor, PD98059. Although transforming growth factor beta (TGFß) also inhibited the growth of Hep-G2 cells, it had no effect on the activity of MEK and ERK. The clusters/aggregates found in LY294002-treated cells were not detectable in TGFß-treated cells. Our data suggest that LY294002 may directly inhibit the activation of MEK and ERK by its ability to bind to the ATP-binding site of the MAPK molecules.


Asunto(s)
Cromonas/farmacología , Inhibidores Enzimáticos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular , Células Hep G2 , Humanos , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Transducción de Señal
6.
Plant Commun ; 1(3): 100040, 2020 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33367237

RESUMEN

Although ABSCISIC ACID INSENSITIVE 4 (ABI4) was initially demonstrated as a key positive regulator in the phytohormone abscisic acid (ABA) signaling cascade, multiple studies have now shown that it is actually involved in the regulation of several other cascades, including diverse phytohormone biogenesis and signaling pathways, various developmental processes (such as seed dormancy and germination, seedling establishment, and root development), disease resistance and lipid metabolism. Consistent with its versatile biological functions, ABI4 either activates or represses transcription of its target genes. The upstream regulators of ABI4 at both the transcription and post-transcription levels have also been documented in recent years. Consequently, a complicated network consisting of the direct target genes and upstream regulators of ABI4, through which ABI4 participates in several phytohormone crosstalk networks, has been generated. In this review, we summarize current understanding of the sophisticated ABI4-mediated molecular networks, mainly focusing on diverse phytohormone (including ABA, gibberellin, cytokinin, ethylene, auxin, and jasmonic acid) crosstalks. We also discuss the potential mechanisms through which ABI4 receives the ABA signal, focusing on protein phosphorylation modification events.


Asunto(s)
Ácido Abscísico/metabolismo , Arabidopsis/efectos de los fármacos , Arabidopsis/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Regulación de la Expresión Génica de las Plantas , Genes de Plantas , Reguladores del Crecimiento de las Plantas/genética , Reguladores del Crecimiento de las Plantas/metabolismo
7.
Nat Commun ; 11(1): 5463, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-33122628

RESUMEN

Metastatic melanoma remains an incurable disease for many patients due to the limited success of targeted and immunotherapies. BRAF and MEK inhibitors reduce metastatic burden for patients with melanomas harboring BRAF mutations; however, most eventually relapse due to acquired resistance. Here, we demonstrate that ABL1/2 kinase activities and/or expression are potentiated in cell lines and patient samples following resistance, and ABL1/2 drive BRAF and BRAF/MEK inhibitor resistance by inducing reactivation of MEK/ERK/MYC signaling. Silencing/inhibiting ABL1/2 blocks pathway reactivation, and resensitizes resistant cells to BRAF/MEK inhibitors, whereas expression of constitutively active ABL1/2 is sufficient to promote resistance. Significantly, nilotinib (2nd generation ABL1/2 inhibitor) reverses resistance, in vivo, causing prolonged regression of resistant tumors, and also, prevents BRAFi/MEKi resistance from developing in the first place. These data indicate that repurposing the FDA-approved leukemia drug, nilotinib, may be effective for prolonging survival for patients harboring BRAF-mutant melanomas.


Asunto(s)
Resistencia a Antineoplásicos/genética , Melanoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Línea Celular Tumoral , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Melanoma/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Mutación/efectos de los fármacos , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-abl/genética , Proteínas Proto-Oncogénicas c-abl/metabolismo
8.
Biomed Pharmacother ; 130: 110522, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32736236

RESUMEN

Myocardial fibrosis is well-known to be the aberrant deposition of extracellular matrix (ECM), which may cause cardiac dysfunction, morbidity, and death. Traditional Chinese medicine formula Si-Miao-Yong-An Decoction (SMYAD), which is used clinically in cardiovascular diseases has been recently reported to able to resist myocardial fibrosis. The anti-fibrosis effects of SMYAD have been evaluated; however, its intricate mechanisms remain to be clarified. Here, we found that SMYAD treatment reduced the fibrosis injury and collagen fiber deposition that could improve cardiac function in isoprenaline (ISO)-induced fibrosis rat models. Combined with our systematic RNA-seq data of SMYAD treatment, we demonstrated that the remarkable up-regulation or down-regulation of several genes were closely related to the functional enrichment of TGF-ß and AMPK pathways that were involved in myocardial fibrosis. Accordingly, we further explored the molecular mechanisms of SMYAD were mainly caused by AMPK activation and thereby suppressing its downstream Akt/mTOR and TGF-ß/SMAD3 pathways. Moreover, we showed that the ECM deposition and secretion process were attenuated, suggesting that the fibrosis pathological features are changed. Interestingly, we found the similar AMPK-driven pathways in NIH-3T3 mouse fibroblasts treated with ISO. Taken together, these results demonstrate that SMYAD may be a new candidate agent by regulating AMPK-driven Akt/mTOR and TGF-ß/SMAD3 pathways for potential therapeutic implications of myocardial fibrosis.


Asunto(s)
Agonistas Adrenérgicos beta , Cardiomiopatías/prevención & control , Medicamentos Herbarios Chinos/uso terapéutico , Isoproterenol , Transducción de Señal/efectos de los fármacos , Animales , Cardiomiopatías/inducido químicamente , Cardiomiopatías/diagnóstico por imagen , Colágeno , Medicamentos Herbarios Chinos/farmacología , Ecocardiografía , Matriz Extracelular/efectos de los fármacos , Fibrosis , Expresión Génica/efectos de los fármacos , Masculino , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Células 3T3 NIH , Proteína Oncogénica v-akt/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Transducción de Señal/genética , Proteína smad3/efectos de los fármacos , Serina-Treonina Quinasas TOR/efectos de los fármacos , Factor de Crecimiento Transformador beta/efectos de los fármacos
9.
Nat Biomed Eng ; 4(7): 686-703, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32661307

RESUMEN

Theranostic agents should ideally be renally cleared and biodegradable. Here, we report the synthesis, characterization and theranostic applications of fluorescent ultrasmall gold quantum clusters that are stabilized by the milk metalloprotein alpha-lactalbumin. We synthesized three types of these nanoprobes that together display fluorescence across the visible and near-infrared spectra when excited at a single wavelength through optical colour coding. In live tumour-bearing mice, the near-infrared nanoprobe generates contrast for fluorescence, X-ray computed tomography and magnetic resonance imaging, and exhibits long circulation times, low accumulation in the reticuloendothelial system, sustained tumour retention, insignificant toxicity and renal clearance. An intravenously administrated near-infrared nanoprobe with a large Stokes shift facilitated the detection and image-guided resection of breast tumours in vivo using a smartphone with modified optics. Moreover, the partially unfolded structure of alpha-lactalbumin in the nanoprobe helps with the formation of an anti-cancer lipoprotein complex with oleic acid that triggers the inhibition of the MAPK and PI3K-AKT pathways, immunogenic cell death and the recruitment of infiltrating macrophages. The biodegradability and safety profile of the nanoprobes make them suitable for the systemic detection and localized treatment of cancer.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/tratamiento farmacológico , Oro/química , Oro/farmacología , Lactalbúmina/química , Lactalbúmina/farmacología , Animales , Apoptosis , Neoplasias de la Mama/patología , Muerte Celular , Femenino , Xenoinjertos , Lipoproteínas , Imagen por Resonancia Magnética/métodos , Ratones , Ratones Endogámicos BALB C , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Nanotecnología/métodos , Imagen Óptica , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Proteómica , Nanomedicina Teranóstica/métodos
10.
J BUON ; 25(2): 1172-1177, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32521923

RESUMEN

PURPOSE: The main purpose of the current research work was to investigate the anticancer activity of Genistein - a plant derived isoflavone - in squamous cell carcinoma SK-MEL-28 (SCC) cells along with studying its effects on cellular apoptosis, DNA damage, cell migration and invasion and MEK/ERK/JNK signalling pathway. METHODS: Cell proliferation was examined by CCK-8 (Cell Counting Kit-8) assay while the effects on apoptosis were evaluated by DAPI staining and Comet assay using fluorescence microscopy. Transwell assay was used for checking the effects on cell migration and invasion while western blot method was used to evaluate the effects on the expression of MEK/ERK/JNK proteins. RESULTS: The results showed that Genistein led to dose-dependent cytotoxic effects in these cells showing an IC50 value of 14.5 µM. It also led to dose-dependent apoptosis and induced DNA damage as shown by fluorescence microscopy. Genistein also inhibited cell migration and invasion dose-dependently, along with inhibiting matrix metalloproteinase (MMP)-9 expression. Genistein also led to inhibition of the expression of p-JNK with no apparent effects on the total JNK expression. It also showed significant and dose-dependent inhibition of the expression of p-MEK and p-ERK proteins. CONCLUSIONS: Genistein has a significant anticancer activity in SK-MEL-28 human SCC cells, inducing apoptosis, DNA damage, cell migration and invasion and inhibiting MEK/ERK and JNK signalling pathway.


Asunto(s)
Anticarcinógenos/uso terapéutico , Carcinoma de Células Escamosas/tratamiento farmacológico , Genisteína/uso terapéutico , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Anticarcinógenos/farmacología , Movimiento Celular , Proliferación Celular , Genisteína/farmacología , Humanos , Invasividad Neoplásica
11.
Cell Death Dis ; 11(5): 380, 2020 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-32424251

RESUMEN

Ovarian cancer represents the first cause of mortality from gynecologic malignancies due to frequent chemoresistance occurrence. Increasing the [BH3-only Bim, Puma, Noxa proapoptotic]/[Bcl-xL, Mcl-1 antiapoptotic] proteins ratio was proven to efficiently kill ovarian carcinoma cells and development of new molecules to imbalance Bcl-2 member equilibrium are strongly required. Drug repurposing constitutes an innovative approach to rapidly develop therapeutic strategies through exploitation of established drugs already approved for the treatment of noncancerous diseases. This strategy allowed a renewed interest for Naftopidil, an α1-adrenergic receptor antagonist commercialized in Japan for benign prostatic hyperplasia. Naftopidil was reported to decrease the incidence of prostate cancer and its derivative was described to increase BH3-only protein expression in some cancer models. Based on these arguments, we evaluated the effects of Naftopidil on ovarian carcinoma and showed that Naftopidil reduced cell growth and increased the expression of the BH3-only proteins Bim, Puma and Noxa. This effect was independent of α1-adrenergic receptors blocking and involved ATF4 or JNK pathway depending on cellular context. Finally, Naftopidil-induced BH3-only members sensitized our models to ABT-737 and Trametinib treatments, in vitro as well as ex vivo, in patient-derived organoid models.


Asunto(s)
Compuestos de Bifenilo/farmacología , Naftalenos/farmacología , Nitrofenoles/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Piperazinas/farmacología , Piridonas/farmacología , Pirimidinonas/farmacología , Sulfonamidas/farmacología , Proteína bcl-X/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Femenino , Humanos , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Proteína bcl-X/metabolismo
12.
Oxid Med Cell Longev ; 2020: 7509612, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32273948

RESUMEN

Glutamic acid (Glu) is a worldwide flavor enhancer with various positive effects. However, Glu-induced neurotoxicity has been reported less. Tetrastigma hemsleyanum (TH), a rare herbal plant in China, possesses high medicinal value. More studies paid attention to tuber of TH whereas vine part (THV) attracts fewer focus. In this study, we extracted and purified flavones from THV (THVF), and UPLC-TOF/MS showed THVF was consisted of 3-caffeoylquinic acid, 5-caffeoylquinic acid, quercetin-3-O-rutinoside, and kaempferol-3-O-rutinoside. In vitro, Glu caused severe cytotoxicity, genotoxicity, mitochondrial dysfunction, and oxidative damage to rat phaeochromocytoma (PC12) cells. Conversely, THVF attenuated Glu-induced toxicity via MAPK pathways. In vivo, the neurotoxicity triggered by Glu restrained the athletic ability in Caenorhabditis elegans (C. elegans). The treatment of THVF reversed the situation induced by Glu. In a word, Glu could cause neurotoxicity and THVF owns potential neuroprotective effects both in vitro and in vivo via MAPK pathways.


Asunto(s)
Antioxidantes/química , Caenorhabditis elegans/efectos de los fármacos , Ácido Glutámico/efectos adversos , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Síndromes de Neurotoxicidad/tratamiento farmacológico , Extractos Vegetales/química , Hojas de la Planta/química , Vitaceae/química , Animales , Flavonas , Humanos , Ratas
13.
Acta Cir Bras ; 34(12): e201901202, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32049183

RESUMEN

PURPOSE: To explore the potential role and unclear molecular mechanisms of vaccarin in wound healing. METHODS: Rats' skin excision model to study the effects of vaccarin on wound healing in vivo . Hematoxylin and eosin staining was performed to evaluate Histopathologic characteristics. Immunohistochemistry was employed to assess the effects of vaccarin in accelerating angiogenesis. Western blot was used to evaluate relative protein expressed levels. RESULTS: Vaccarin could significantly promote wound healing and endothelial cells and fibroblasts proliferation in the wound site. Immunohistochemistry and Western blot studies showed that the nodal proteins and receptor (bFGFR) related to angiogenesis signaling pathway were activated, and the microvascular density in the wound site was markedly higher than that in the control group. CONCLUSIONS: The present study was the first to demonstrate that vaccarin is able to induce angiogenesis and accelerate wound healing in vivo by increasing expressions of p-Akt, p-Erk and p-bFGFR. This process is mediated by MAPK/ERK and PI3K/AKT signaling pathways.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Caryophyllaceae/química , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Extractos Vegetales/farmacología , Cicatrización de Heridas/efectos de los fármacos , Animales , Western Blotting , Proliferación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Inmunohistoquímica , Masculino , Quinasas de Proteína Quinasa Activadas por Mitógenos/análisis , Fosfatidilinositol 3-Quinasas/análisis , Extractos Vegetales/química , Ratas Sprague-Dawley , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/análisis , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/efectos de los fármacos , Reproducibilidad de los Resultados , Transducción de Señal , Factores de Tiempo
14.
J Cell Physiol ; 235(3): 2847-2856, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31535369

RESUMEN

Septic cardiomyopathy is associated with mitochondrial damage and endoplasmic reticulum (ER) dysfunction. However, the upstream mediator of mitochondrial injury and ER stress has not been identified and thus little drug is available to treat septic cardiomyopathy. Here, we explored the role of B-cell receptor-associated protein 31 (BAP31) in septic cardiomyopathy and figure out whether melatonin could attenuate sepsis-mediated myocardial depression via modulating BAP31. Lipopolysaccharide (LPS) was used to establish the septic cardiomyopathy model. Pathway analysis was performed via western blot, quantitative polymerase chain reaction and immunofluorescence. Mitochondrial function and ER stress were detected via enzyme-linked immunosorbent assay, western blot, and immunofluorescence. After exposure to LPS, cardiac function was reduced due to excessive inflammation response and extensive cardiomyocyte death. Mechanistically, melatonin treatment could dose-dependently improve cardiomyocyte viability via preserving mitochondrial function and reducing ER stress. Further, we found that BAP31 transcription was repressed by LPS whereas melatonin could restore BAP31 expression; this effect was dependent on the MAPK-ERK pathway. Inhibition of the ERK pathway and/or knockdown of BAP31 could attenuate the beneficial effects of melatonin on mitochondrial function and ER homeostasis under LPS stress. Altogether, our results indicate that ERK-BAP31 pathway could be used as a critical mediator for mitochondrial function and ER homeostasis in sepsis-related myocardial injury. Melatonin could stabilize BAP31 via the ERK pathway and thus contribute to the preservation of cardiac function in septic cardiomyopathy.


Asunto(s)
Cardiomiopatías/tratamiento farmacológico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Melatonina/farmacología , Proteínas de la Membrana/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Cardiomiopatías/metabolismo , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Lipopolisacáridos/farmacología , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Regulación hacia Arriba/efectos de los fármacos
15.
Biofactors ; 46(2): 283-291, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31721330

RESUMEN

Rheumatoid arthritis (RA) is a chronic and accelerated autoimmune illness with proliferative and damaging synovitis, resulting in joint death and cartilage and bone erosion. This study focused on the potential therapeutic effect of wogonin on complete Freund's adjuvant (CFA) induced RA in rats and the underlying mechanisms. Arthritis was experimentally caused in rats by subcutaneously injecting 0.1 mL of CFA into the subplantar area of the left hind paw under moderate anesthesia on day zero. The regular oral doses of indomethacin/wogonin began on day zero and proceeded after injection to day 35. Wogonin reduced arthritic score considerably, enhanced body weight, and reduced paw thickness. Wogonin also boosted red blood cell considerably along with hemoglobin and reduced white blood cell count and erythrocyte sedimentation rate. Wogonin substantially improved an altered level of oxidative stress markers, antioxidant proteins, and inflammatory cytokines in a dose-dependent way. Wogonin inhibited p38 phosphorylation triggered by CFA and p65 nuclear translocation.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Artritis Reumatoide/tratamiento farmacológico , Flavanonas/uso terapéutico , Adyuvante de Freund , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , FN-kappa B/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos
16.
Neurochem Int ; 134: 104647, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31877349

RESUMEN

BACKGROUND: Neonatal sepsis is defined as a systemic inflammatory response caused by a suspected or proven infection, occurring in the first month of life, and remains one of the main causes of morbidity and mortality in newborn and preterm infants. Frequently, survivors of neonatal sepsis have serious long-term cognitive impairment and adverse neurologic outcomes. There is currently no specific drug treatment for sepsis. Indole-3-guanylhydrazone hydrochloride (LQM01) is an aminoguanidine derivative that has been described as an anti-inflammatory, antihypertensive and antioxidant with potential applicability in inflammatory diseases. METHODS: We used a LPS-challenged neonatal sepsis rodent model to investigate the effect of LQM01 on cognitive impairment and anxiety-like behavior in sepsis mice survivors, and examined the possible molecular mechanisms involved. RESULTS: It was found that LQM01 exposure during the neonatal period reduces anxiety-like behavior and cognitive impairment caused by lipopolysaccharides (LPS) in adult life. Additionally, treatment with LQM01 decreased pro-inflammatory cytokine levels and reduced NFκB, COX-2, MAPK and microglia activation in hippocampus of neonatal mice. Furthermore, LQM01 was also able to prevent oxidative damage in hippocampus of neonatal mice and preserve brain barrier integrity. CONCLUSIONS: LQM01 attenuated inflammatory reactions in an LPS-challenged neonatal sepsis mice model through the MAPK and NFκB signaling pathways and microglia activation suppression. All these findings are associated with mitigated cognitive impairment in 70 days-old LQM01 treated-mice. GENERAL SIGNIFICANCE: We revealed the effect of LQM01 as an anti-septic agent, and the role of crucial molecular pathways in mitigating the potential damage caused by neonatal sepsis.


Asunto(s)
Disfunción Cognitiva/tratamiento farmacológico , Guanidina/análogos & derivados , Indoles/farmacología , Inflamación/tratamiento farmacológico , Sepsis Neonatal/tratamiento farmacológico , Animales , Antiinflamatorios/farmacología , Disfunción Cognitiva/metabolismo , Modelos Animales de Enfermedad , Femenino , Guanidina/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Masculino , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , FN-kappa B/efectos de los fármacos , Sepsis Neonatal/inducido químicamente , Sepsis Neonatal/metabolismo
17.
Neuropharmacology ; 158: 107748, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31465784

RESUMEN

Stroke is a condition with few medical treatments available. Semaglutide, a novel Glucagon-like peptide-1 (GLP-1) analogue, has been brought to the market as a treatment for diabetes. We tested the protective effects of semaglutide against middle cerebral artery occlusion injury in rats. Animals were treated with 10 nmol/kg bw ip. starting 2 h after surgery and every second day for either 1, 7, 14 or 21 days. Semaglutide-treated animals showed significantly reduced scores of neurological impairments in several motor and grip strength tasks. The cerebral infarction size was also reduced, and the loss of neurons in the hippocampal areas CA1, CA3 and the dentate gyrus was much reduced. Chronic inflammation as seen in levels of activated microglia and in the activity of the p38 MAPK - MKK - c-Jun- NF-κB p65 inflammation signaling pathway was reduced. In addition, improved growth factor signaling as shown in levels of activated ERK1 and IRS-1, and a reduction in the apoptosis signaling pathway C-raf, ERK2, Bcl-2/BAX and Caspase-3 was observed. Neurogenesis had also been normalized by the drug treatment as seen in increased neurogenesis (DCX-positive cells) in the dentate gyrus and a normalization of biomarkers for neurogenesis. In conclusion, semaglutide is a promising candidate for re-purposing as a stroke treatment.


Asunto(s)
Apoptosis/efectos de los fármacos , Encéfalo/efectos de los fármacos , Péptidos Similares al Glucagón/farmacología , Hipocampo/efectos de los fármacos , Hipoglucemiantes/farmacología , Infarto de la Arteria Cerebral Media/patología , Neurogénesis/efectos de los fármacos , Animales , Encéfalo/inmunología , Encéfalo/patología , Modelos Animales de Enfermedad , Proteína Doblecortina , Péptido 1 Similar al Glucagón/análogos & derivados , Hipocampo/citología , Infarto de la Arteria Cerebral Media/inmunología , Infarto de la Arteria Cerebral Media/fisiopatología , Inflamación/inmunología , Proteínas Sustrato del Receptor de Insulina/efectos de los fármacos , Proteínas Sustrato del Receptor de Insulina/metabolismo , Microglía/efectos de los fármacos , Microglía/inmunología , Proteína Quinasa 3 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/inmunología , Actividad Motora/efectos de los fármacos , Neuronas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-jun/efectos de los fármacos , Proteínas Proto-Oncogénicas c-jun/inmunología , Ratas , Accidente Cerebrovascular/inmunología , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/fisiopatología , Factor de Transcripción ReIA/efectos de los fármacos , Factor de Transcripción ReIA/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología
18.
Mar Drugs ; 17(2)2019 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-30717280

RESUMEN

The skin, the largest organ in humans, is exposed to major sources of outdoor air pollution, such as fine particulate matter with a diameter ≤ 2.5 µm (PM2.5). Diphlorethohydroxycarmalol (DPHC), a marine-based compound, possesses multiple activities including antioxidant effects. In the present study, we evaluated the protective effect of DPHC on PM2.5-induced skin cell damage and elucidated the underlying mechanisms in vitro and in vivo. The results showed that DPHC blocked PM2.5-induced reactive oxygen species generation in human keratinocytes. In addition, DPHC protected cells against PM2.5-induced DNA damage, endoplasmic reticulum stress, and autophagy. HR-1 hairless mice exposed to PM2.5 showed lipid peroxidation, protein carbonylation, and increased epidermal height, which were inhibited by DPHC. Moreover, PM2.5 induced apoptosis and mitogen-activated protein kinase (MAPK) protein expression; however, these changes were attenuated by DPHC 5. MAPK inhibitors were used to elucidate the molecular mechanisms underlying these actions, and the results demonstrated that MAPK signaling pathway may play a key role in PM2.5-induced skin damage.


Asunto(s)
Compuestos Heterocíclicos con 3 Anillos/farmacología , Material Particulado/farmacología , Piel/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Humanos , Queratinocitos/efectos de los fármacos , Queratinocitos/patología , Peroxidación de Lípido/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Pelados , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mitofagia/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno , Piel/patología
19.
Biomed Res Int ; 2019: 9395146, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31976330

RESUMEN

The phytochemical substances, coumarin derivatives, have demonstrated antiresorptive bone effects by suppressing osteoclast differentiation in vitro and in vivo. Recently, we have identified 5'-hydroxy auraptene (5'-HA), a coumarin derivative isolated from Lotus lalambensis Schweinf, as a novel stimulator for osteoblast differentiation. In this study, we investigated the effect of 5'-HA on osteoclast differentiation of mouse bone marrow (BM) cells. The effect of 5'-HA on BM cell proliferation and osteoclast differentiation was determined by measuring cell viability and tartrate-resistant acid phosphatase (TRAP) enzyme activity, quantification of TRAP+ multinucleated cells (TRAP+MNCs), and quantitative real-time PCR (qPCR) of osteoclastic gene expression. Regulation of NF-κB, c-Fos/NFATc1, and MAPK signaling pathways by 5'-HA during osteoclastogenesis was measured by the NF-κB reporter assay and Western blot analysis. 5'-HA significantly suppresses the receptor activator of NF-κB ligand (RANKL) induced osteoclast differentiation of BM cells in a dose-dependent manner. Consistently, treatment of BM cells with 5'-HA significantly inhibited RANKL-induced activation of NF-κB and c-Fos/NFATc1 pathways in a dose-dependent manner. Furthermore, RANKL-induced phosphorylation of ERK1/2, p-38, and JNK was significantly inhibited by 5'-HA in BM cells. In conclusion, we identified 5'-HA as a novel coumarin derivative that suppresses RANKL-induced osteoclastogenesis via inhibiting c-Fos/NFATc1 and MAPK signaling pathways.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Cumarinas/farmacología , Factores de Transcripción NFATC/metabolismo , Osteoclastos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Expresión Génica , Lotus/química , Masculino , Ratones , Ratones Endogámicos C57BL , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , FN-kappa B/metabolismo , Osteoclastos/metabolismo , Osteogénesis/efectos de los fármacos , Extractos Vegetales/farmacología , Ligando RANK/efectos de los fármacos , Ligando RANK/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos
20.
Acta cir. bras ; 34(12): e201901202, 2019. graf
Artículo en Inglés | LILACS | ID: biblio-1054685

RESUMEN

Abstract Purpose To explore the potential role and unclear molecular mechanisms of vaccarin in wound healing. Methods Rats' skin excision model to study the effects of vaccarin on wound healing in vivo . Hematoxylin and eosin staining was performed to evaluate Histopathologic characteristics. Immunohistochemistry was employed to assess the effects of vaccarin in accelerating angiogenesis. Western blot was used to evaluate relative protein expressed levels. Results Vaccarin could significantly promote wound healing and endothelial cells and fibroblasts proliferation in the wound site. Immunohistochemistry and Western blot studies showed that the nodal proteins and receptor (bFGFR) related to angiogenesis signaling pathway were activated, and the microvascular density in the wound site was markedly higher than that in the control group. Conclusions The present study was the first to demonstrate that vaccarin is able to induce angiogenesis and accelerate wound healing in vivo by increasing expressions of p-Akt, p-Erk and p-bFGFR. This process is mediated by MAPK/ERK and PI3K/AKT signaling pathways.


Asunto(s)
Animales , Masculino , Cicatrización de Heridas/efectos de los fármacos , Extractos Vegetales/farmacología , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Caryophyllaceae/química , Inductores de la Angiogénesis/farmacología , Factores de Tiempo , Inmunohistoquímica , Extractos Vegetales/química , Transducción de Señal , Western Blotting , Reproducibilidad de los Resultados , Ratas Sprague-Dawley , Fosfatidilinositol 3-Quinasas/análisis , Quinasas de Proteína Quinasa Activadas por Mitógenos/análisis , Células Endoteliales/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/análisis , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/efectos de los fármacos , Fibroblastos/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...