Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 167
Filtrar
1.
Int Immunopharmacol ; 137: 112423, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-38861914

RESUMEN

Fibrosis is the excessive deposition of extracellular matrix in an organ or tissue that results from an impaired tissue repair in response to tissue injury or chronic inflammation. The progressive nature of fibrotic diseases and limited treatment options represent significant healthcare challenges. Despite the substantial progress in understanding the mechanisms of fibrosis, a gap persists translating this knowledge into effective therapeutics. Here, we discuss the critical mediators involved in fibrosis and the role of tranilast as a potential antifibrotic drug to treat fibrotic conditions. Tranilast, an antiallergy drug, is a derivative of tryptophan and has been studied for its role in various fibrotic diseases. These include scleroderma, keloid and hypertrophic scars, liver fibrosis, renal fibrosis, cardiac fibrosis, pulmonary fibrosis, and uterine fibroids. Tranilast exerts antifibrotic effects by suppressing fibrotic pathways, including TGF-ß, and MPAK. Because it disrupts fibrotic pathways and has demonstrated beneficial effects against keloid and hypertrophic scars, tranilast could be used to treat other conditions characterized by fibrosis.


Asunto(s)
Fibrosis , Transducción de Señal , ortoaminobenzoatos , Humanos , ortoaminobenzoatos/uso terapéutico , ortoaminobenzoatos/farmacología , Fibrosis/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Animales , Antifibróticos/uso terapéutico , Antifibróticos/farmacología , Queloide/tratamiento farmacológico , Queloide/patología , Queloide/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
2.
Int J Mol Sci ; 24(13)2023 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-37445642

RESUMEN

Tranilast (N-3, 4-dimethoxycinnamoyl anthranilic acid) is an orally administered drug with antiallergic properties and approved in Japan and the Republic of Korea for the treatment of asthma and hypertrophic scars. Previous in vitro studies indicated that tranilast reduced fibroid growth through its inhibitory effects on cell proliferation and induction of apoptosis. The objective of this study was to determine the efficacy of tranilast for treatment of human-derived fibroids in a mouse model. SCID mice (ovariectomized, supplemented with estrogen and progesterone) were implanted with fibroid explants and treated for two months with tranilast (50 m/kg/daily) or the vehicle. After sacrifice, xenografts were excised and analyzed. Tranilast was well tolerated without adverse side effects. There was a 37% reduction in tumor weight along with a significant decrease in staining for Ki67, CCND1, and E2F1; a significant increase in nuclear staining for cleaved caspase 3; and reduced staining for TGF-ß3 and Masson's trichrome in the tranilast treated mice. There was a significant inhibition of mRNA and protein expression of fibronectin, COL3A1, CCND1, E2F1, and TGF-ß3 in the xenografts from the tranilast-treated mice. These promising therapeutic effects of tranilast warrant additional animal studies and human clinical trials to evaluate its efficacy for treatment of fibroids.


Asunto(s)
Leiomioma , Factor de Crecimiento Transformador beta3 , Humanos , Ratones , Animales , Ratones SCID , Leiomioma/metabolismo , ortoaminobenzoatos/farmacología , ortoaminobenzoatos/uso terapéutico , Modelos Animales de Enfermedad
3.
Nat Commun ; 13(1): 7165, 2022 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-36418896

RESUMEN

Nano-immunotherapy improves breast cancer outcomes but not all patients respond and none are cured. To improve efficacy, research focuses on drugs that reprogram cancer-associated fibroblasts (CAFs) to improve therapeutic delivery and immunostimulation. These drugs, however, have a narrow therapeutic window and cause adverse effects. Developing strategies that increase CAF-reprogramming while limiting adverse effects is urgent. Here, taking advantage of the CAF-reprogramming capabilities of tranilast, we developed tranilast-loaded micelles. Strikingly, a 100-fold reduced dose of tranilast-micelles induces superior reprogramming compared to free drug owing to enhanced intratumoral accumulation and cancer-associated fibroblast uptake. Combination of tranilast-micelles and epirubicin-micelles or Doxil with immunotherapy increases T-cell infiltration, resulting in cures and immunological memory in mice bearing immunotherapy-resistant breast cancer. Furthermore, shear wave elastography (SWE) is able to monitor reduced tumor stiffness caused by tranilast-micelles and predict response to nano-immunotherapy. Micellar encapsulation is a promising strategy for TME-reprogramming and SWE is a potential biomarker of response.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Neoplasias , Ratones , Animales , Micelas , Microambiente Tumoral , Inmunoterapia , ortoaminobenzoatos/farmacología , ortoaminobenzoatos/uso terapéutico , Factores Inmunológicos , Polímeros
4.
Acta Med Okayama ; 75(5): 653-657, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34703050

RESUMEN

We present an extremely rare case of deep angiomyxoma (DAM) in the thigh that was misdiagnosed as desmoid-type fibromatosis. A 40-year-old Japanese woman presented with a mass on the left thigh. The histological diagnosis by needle biopsy was desmoid-type fibromatosis; the tumor grew slowly and was resected 4 years later. The histological diagnosis from the resected tumor was DAM. As of 16 months post-surgery, the patient has not noticed any local recurrence. Although DAM in a lower extremity is extremely rare, clinicians must be aware of its possible occurrence in areas relatively close to the pelvis.


Asunto(s)
Mixoma/diagnóstico , Mixoma/cirugía , Adulto , Antiinflamatorios no Esteroideos/uso terapéutico , Femenino , Humanos , Meloxicam/uso terapéutico , Mixoma/patología , ortoaminobenzoatos/uso terapéutico
5.
Biomed Pharmacother ; 141: 111844, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34174504

RESUMEN

Tranilast (TRN) or (N-3,4 -dimethoxy cinnamoyl]-anthranilic acid) is an analog of a tryptophan metabolite and is identified mainly as an anti-allergic agent with limited side effects. The anti-cancer effects of tranilast either alone or in combination with chemotherapeutic drugs have been evidenced in several pre-clinical studies. The main mechanism of action of tranilast includes targeting and modulation of various signaling and immune regulatory pathways including Transforming growth factor-beta (TGF-ß), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), phosphatidylinositol 3-kinase (PI3K), MAP-Kinase (MAPK), Protein kinase B ( Akt/PKB), c-Jun N-terminal kinase, modulation of cancer stem cells, etc. Most of these pathways are involved in tumor proliferation, invasion, and metastasis and it is postulated that tranilast, with its low toxicity profile and high anti-carcinogenic abilities, can serve as a potential anti-tumorigenic agent. The main aim of this review is to provide updated information on the anti-cancer effects of tranilast and its significance as a therapeutic agent.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Neoplasias/tratamiento farmacológico , ortoaminobenzoatos/farmacología , Animales , Antibióticos Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica , Humanos , Neoplasias/genética , Transducción de Señal/efectos de los fármacos , ortoaminobenzoatos/uso terapéutico
6.
J Med Chem ; 64(7): 3976-3996, 2021 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-33784097

RESUMEN

The transient receptor potential melastatin 2 (TRPM2) channel is associated with ischemia/reperfusion injury, inflammation, cancer, and neurodegenerative diseases. However, the limit of specific inhibitors impedes the development of TRPM2-targeted therapeutic agents. To discover more potent and selective TRPM2 inhibitors, 59 N-(p-amylcinnamoyl) anthranilic acid (ACA) derivatives were synthesized and evaluated using calcium imaging and electrophysiology approaches. Systematic structure-activity relationship studies resulted in some potent compounds inhibiting the TRPM2 channel with sub-micromolar half-maximal inhibitory concentration values. Among them, the preferred compound A23 exhibited TRPM2 selectivity over TRPM8 and TRPV1 channels as well as phospholipase A2 and showed neuroprotective activity in vitro. Following pharmacokinetic studies, A23 was further evaluated in a transient middle cerebral artery occlusion model in vivo, which significantly reduced cerebral infarction. These data indicate that A23 might serve as a useful tool for TRPM2-related research as well as a lead compound for the development of therapeutic agents for ischemic injury.


Asunto(s)
Cinamatos/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Canales Catiónicos TRPM/antagonistas & inhibidores , ortoaminobenzoatos/uso terapéutico , Animales , Línea Celular Tumoral , Cinamatos/síntesis química , Cinamatos/farmacocinética , Glucosa/deficiencia , Células HEK293 , Humanos , Masculino , Ratones Endogámicos C57BL , Estructura Molecular , Fármacos Neuroprotectores/síntesis química , Fármacos Neuroprotectores/farmacocinética , Oxígeno/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Relación Estructura-Actividad , ortoaminobenzoatos/síntesis química , ortoaminobenzoatos/farmacocinética
7.
Medicine (Baltimore) ; 99(50): e23633, 2020 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-33327342

RESUMEN

BACKGROUND: Transient receptor potential vanilloid 2 (TRPV2) was previously shown to play an important role in the maintenance of cancer stem cells, and its specific inhibitor, tranilast, also has potential as a targeted therapeutic agent for esophageal squamous cell carcinoma (ESCC). The present study is being conducted to confirm the safety and efficacy of the additional use of tranilast with conventional preoperative adjuvant chemotherapy for patients with advanced ESCC. PATIENTS AND METHODS: Between 56 and 59 patients aged between 20 and 74 years with clinically diagnosed Stage II or Stage III ESCC will be enrolled. Eligible patients will receive preoperative adjuvant chemotherapy, 2 cycles of combination therapy with cisplatin, 5-fluorouracil, and tranilast. Recruitment started in November 2019, with the final follow-up being planned for March 2029. One subject has been enrolled since October 21, 2020. The pathological therapeutic effect is the primary endpoint. The objective response rate, safety of preoperative adjuvant chemotherapy, recurrence-free survival (RFS), and overall survival (OS) are the secondary endpoints. RFS and OS will be calculated as the time from surgery to first recurrence and all-cause death, respectively. ETHICS AND DISSEMINATION: This protocol has been approved by the Institutional Review Boards of Kyoto Prefectural University of Medicine and all participating hospitals in August 30, 2019 (Number: CRB5180001). Written informed consent will be obtained from all patients before their registration, which is in accordance with the Declaration of Helsinki. The results of the present study will be disseminated via publication in peer-reviewed journals. TRIAL REGISTRATION: Trial registration number jRCTs051190076.


Asunto(s)
Quimioterapia Adyuvante/métodos , Neoplasias Esofágicas/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , ortoaminobenzoatos/uso terapéutico , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Quimioterapia Adyuvante/efectos adversos , Cisplatino/uso terapéutico , Relación Dosis-Respuesta a Droga , Femenino , Fluorouracilo/uso terapéutico , Humanos , Masculino , Persona de Mediana Edad , Índice de Severidad de la Enfermedad , Análisis de Supervivencia , Adulto Joven , ortoaminobenzoatos/administración & dosificación , ortoaminobenzoatos/efectos adversos
8.
Anticancer Res ; 40(6): 3287-3296, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32487624

RESUMEN

BACKGROUND/AIM: Transforming growth factor ß1 (TGF-ß1) is an important epithelial-mesenchymal transition (EMT) activator that regulates the expression of E-cadherin and vimentin through Smad signalling. Tranilast is an anti-allergic drug that inhibits TGF-ß1, and is used in the treatment of keloids and hypertrophic scars. We investigated whether tranilast inhibits TGF-ß1-induced EMT and invasiveness in human non-small cell lung cancer cell lines. MATERIALS AND METHODS: We examined the effects of tranilast treatment on EMT markers, TGF-ß1/Smad signalling, and cell invasiveness in A549 and PC14 cells. Tumours from a mouse orthotopic lung cancer model with or without tranilast treatment were also immunohistochemically evaluated. RESULTS: Tranilast increased E-cadherin expression via Smad4 suppression and inhibited cell invasion in TGF-ß1-stimulated cells. Tranilast treatment of the in vivo mouse model reduced the pleural dissemination of cancer cells and suppressed vimentin and Smad4 expression. CONCLUSION: Tranilast inhibited TGF-ß1-induced EMT and cellular invasion/metastasis by suppressing Smad4 expression in cancer cells.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Transición Epitelial-Mesenquimal/efectos de los fármacos , Neoplasias Pulmonares/genética , Proteína Smad4/genética , Factor de Crecimiento Transformador beta1/efectos de los fármacos , ortoaminobenzoatos/uso terapéutico , Animales , Antiinflamatorios no Esteroideos/farmacología , Línea Celular Tumoral , Humanos , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Desnudos , Invasividad Neoplásica , Metástasis de la Neoplasia , ortoaminobenzoatos/farmacología
9.
Dig Dis Sci ; 65(7): 1904-1916, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32279173

RESUMEN

Despite recent advances aimed to treat transmural inflammation in Crohn's disease (CD) patients, the progression to a structuring behavior still represents an issue for clinicians. As inflammation becomes chronic and severe, the attempt to repair damaged tissue can result in an excessive production of extracellular matrix components and deposition of connective tissue, thus favoring the formation of strictures. No specific and accurate clinical predictors or diagnostic tools for intestinal fibrosis exist, and to date, no genetic or serological marker is in routine clinical use. Therefore, intestinal fibrosis is usually diagnosed when it becomes clinically evident and strictures have already occurred. Anti-fibrotic agents such as tranilast, peroxisome proliferator-activated receptor gamma agonists, rho kinase inhibitors, and especially mesenchymal stem cell therapy have provided interesting results, but most of the evidence has been derived from studies performed in vitro. Therefore, current therapy of fibrotic strictures relies mainly on endoscopic and surgical procedures. Although its long-term outcomes may be debated, endoscopic balloon dilation appears to be the safest and most effective approach to treat appropriately selected strictures. The use of endoscopic stricturotomy is currently limited by the expertise needed to perform it and by the few data available in the literature. Some good results have been achieved by the positioning of self-expandable metal stents (SEMS). However, there is no concordance regarding the type of stent to use and for how long it should be left in place. The development of new specific SEMS may lead to better outcomes and to an increased use of this alternative in CD-related strictures.


Asunto(s)
Constricción Patológica/fisiopatología , Constricción Patológica/terapia , Enfermedad de Crohn/fisiopatología , Endoscopía Gastrointestinal , Intestinos/patología , Stents Metálicos Autoexpandibles , Antiinflamatorios no Esteroideos/uso terapéutico , Constricción Patológica/etiología , Constricción Patológica/metabolismo , Enfermedad de Crohn/complicaciones , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/terapia , Dilatación , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Fibrosis/metabolismo , Fibrosis/fisiopatología , Fármacos Gastrointestinales/uso terapéutico , Humanos , Inflamación/metabolismo , Inflamación/fisiopatología , PPAR gamma/agonistas , Inhibidores de Proteínas Quinasas/uso terapéutico , Inhibidores del Factor de Necrosis Tumoral/uso terapéutico , ortoaminobenzoatos/uso terapéutico , Quinasas Asociadas a rho/antagonistas & inhibidores
10.
J Biochem Mol Toxicol ; 34(5): e22466, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32045101

RESUMEN

Drug-induced organ toxicity is a frequently encountered obstacle in the field of medical practice that limits the use of numerous pharmacologically valuable drugs. Methotrexate (MTX)-induced organ toxicity is unfortunately the rate-limiting factor for its clinical application. In the current study, MTX injection induced significant renal and hepatic toxicities manifested on functional, biochemical, and histopathological scales. This was associated with a significant elevation in both renal and hepatic contents of TNF-related apoptosis-inducing ligand (TRAIL) and caspase-8, biomarkers of tissue apoptosis. Inline, immunohistochemical analysis confirmed that tissue increased expression of Ki67 as a biomarker of tissue regeneration in both organs. Tranilast (TRAN) is a small molecular weight anti-inflammatory and antiallergic agent. TRAN's coadministration with MTX in the current study induced a significant tissue recovery via modulation of TRAIL/caspase-8 signaling and modulation of apoptosis-induced tissue proliferation confirmed by quantification of Ki67 expression. In conclusion, TRAN can be proposed as an effective drug to attenuate MTX-induced organ toxicity via modulation of apoptosis-induced tissue proliferation pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Riñón/efectos de los fármacos , Hígado/efectos de los fármacos , Metotrexato/farmacología , ortoaminobenzoatos/farmacología , Animales , Biomarcadores/metabolismo , Caspasa 8/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Antígeno Ki-67/metabolismo , Riñón/metabolismo , Hígado/metabolismo , Regeneración Hepática , Masculino , Metotrexato/efectos adversos , Ratones , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , ortoaminobenzoatos/uso terapéutico
11.
Int J Mol Sci ; 20(18)2019 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-31540249

RESUMEN

Cancer represents one of the leading causes of death worldwide. Progresses in treatment of cancer have continued at a rapid pace. However, undesirable side effects and drug resistance remain major challenges for therapeutic success. Natural products represent a valuable starting point to develop new anticancer strategies. Polyphenols, well-known as antioxidant, exert anticancer effects through the modulation of multiple pathways and mechanisms. Oat (Avena sativa L., Poaceae) is a unique source of avenanthramides (AVAs), a group of polyphenolic alkaloids, considered as its signature compounds. The present review aims to offer a comprehensive and critical perspective on the chemopreventive and chemotherapeutic potential of AVAs. AVAs prevent cancer mainly by blocking reactive species. Moreover, they exhibit potential therapeutic activity through the modulation of different pathways including the activation of apoptosis and senescence, the block of cell proliferation, and the inhibition of epithelial mesenchymal transition and metastatization. AVAs are promising chemopreventive and anticancer phytochemicals, which need further clinical trials and toxicological studies to define their efficacy in preventing and reducing the burden of cancer diseases.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Avena/química , Neoplasias/tratamiento farmacológico , ortoaminobenzoatos/uso terapéutico , Animales , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos , Neoplasias/metabolismo , Extractos Vegetales/química , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Transducción de Señal/efectos de los fármacos , ortoaminobenzoatos/química , ortoaminobenzoatos/farmacología
13.
Toxicol Appl Pharmacol ; 380: 114696, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31381904

RESUMEN

TGFß signaling plays an important role in orchestrating a favorable microenvironment for tumor cell growth and promoting epithelial-mesenchymal transition. As a conventional nonsteroidal anti-inflammation drugs, tolfenamic acid (TA) has been previously reported to exhibit anti-cancer activity. Herein, we investigated the effect of TA on TGFß-mediated pro-metastatic activity and the underlying mechanisms in hepatocellular carcinoma (HCC). As a result, TA suppresses TGFß-induced migration and glycolysis in HCC cells, which is accompanied with reduced Smad phosphorylation and subsequent nuclear transcription activity. Mechanistically, TA promotes lipid raft-caveolar internalization pathway of TGFß receptor, therefore leading to its rapid turnover. Consistently, TA inhibits constitutively active TGFß type I receptor induced Smad phosphorylation and EMT markers, whereas ectopic expression of TGFß type II receptor could partially rescue TGFß-mediated Smad2 phosphorylation and downstream genes expression in the presence of TA. Furthermore, TA inhibited HCC cells invasion in nude mice, associated with the alteration of characteristics related with EMT and glycolysis of cancer cells. Our study suggests TA could activate lipid raft pathway and modulate TGFß mediated metastasis, implicating the potential application of TA as a modulator of tumor microenvironment in HCC.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Antineoplásicos/farmacología , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Microdominios de Membrana/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , ortoaminobenzoatos/farmacología , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Femenino , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Ratones Endogámicos BALB C , Ratones Desnudos , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , ortoaminobenzoatos/uso terapéutico
14.
Brain ; 142(5): 1349-1364, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30938419

RESUMEN

Amyotrophic lateral sclerosis is a deleterious neurodegenerative disease without effective treatment options. Recent studies have indicated the involvement of the dysregulation of RNA metabolism in the pathogenesis of amyotrophic lateral sclerosis. Among the various RNA regulatory machineries, nonsense-mediated mRNA decay (NMD) is a stress responsive cellular surveillance system that degrades selected mRNA substrates to prevent the translation of defective or harmful proteins. Whether this pathway is affected in neurodegenerative diseases is unclear. Here we report the inhibition of NMD by arginine-rich dipeptide repeats derived from C9orf72 hexanucleotide repeat expansion, the most common cause of familial amyotrophic lateral sclerosis. Bioinformatic analysis of multiple transcriptome profiles revealed significant overlap of upregulated genes in NMD-defective cells with those in the brain tissues, micro-dissected motor neurons, or induced pluripotent stem cell-derived motor neurons specifically from amyotrophic lateral sclerosis patients carrying C9orf72 hexanucleotide repeat expansion, suggesting the suppression of NMD pathway in these patients. Using Drosophila as a model, we have validated that the C9orf72 hexanucleotide repeat expansion products could lead to the accumulation of the NMD substrates and identified arginine-rich dipeptide repeats, including poly glycine-arginine and poly proline-arginine, as the main culprits of NMD inhibition. Furthermore, in human SH-SY5Y neuroblastoma cells and in mouse brains, expression of glycine-arginine with 36 repeats (GR36) was sufficient to cause NMD inhibition. In cells expressing GR36, stress granule accumulation was accompanied by decreased processing body formation, which contributed to the inhibition of NMD. Remarkably, expression of UPF1, a core gene in the NMD pathway, efficiently blocked neurotoxicity caused by arginine-rich dipeptide repeats in both cellular and Drosophila models. Although not as effective as UPF1, expression of another NMD gene UPF2 also ameliorated the degenerative phenotypes in dipeptide repeat-expressing flies, indicating that genetically reactivating the NMD pathway could suppress dipeptide repeat toxicity. Finally, after validating tranilast as an NMD-activating drug, we demonstrated the therapeutic potential of this asthma drug in cellular and Drosophila models of C9orf72 dipeptide repeat neurotoxicity. Therefore, our study has revealed a cellular mechanism whereby arginine-rich C9orf72 dipeptide repeats could inhibit NMD activities by reducing the abundance of processing bodies. Furthermore, our results suggested that activation of the NMD pathway could be a potential therapeutic strategy for amyotrophic lateral sclerosis with defective RNA metabolism.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Proteína C9orf72/genética , Proteína C9orf72/metabolismo , Degradación de ARNm Mediada por Codón sin Sentido/fisiología , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Animales , Animales Modificados Genéticamente , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/uso terapéutico , Línea Celular Tumoral , Dipéptidos/genética , Dipéptidos/metabolismo , Drosophila , Femenino , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Degradación de ARNm Mediada por Codón sin Sentido/efectos de los fármacos , ortoaminobenzoatos/farmacología , ortoaminobenzoatos/uso terapéutico
15.
Sci Rep ; 9(1): 1554, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30733502

RESUMEN

Patients treated during leukemia face the risk of complications including pulmonary dysfunction that may result from infiltration of leukemic blast cells (LBCs) into lung parenchyma and interstitium. In LBCs, we demonstrated that transient receptor potential vanilloid type 2 channel (TRPV2), reputed for its role in inflammatory processes, exhibited oncogenic activity associated with alteration of its molecular expression profile. TRPV2 was overexpressed in LBCs compared to normal human peripheral blood mononuclear cells (PBMCs). Additionally, functional full length isoform and nonfunctional short form pore-less variant of TRPV2 protein were up-regulated and down-regulated respectively in LBCs. However, the opposite was found in PBMCs. TRPV2 silencing or pharmacological targeting by Tranilast (TL) or SKF96365 (SKF) triggered caspace-mediated apoptosis and cell cycle arrest. TL and SKF inhibited chemotactic peptide fMLP-induced response linked to TRPV2 Ca2+ activity, and down-regulated expression of surface marker CD38 involved in leukemia and lung airway inflammation. Challenging lung airway epithelial cells (AECs) with LBCs decreased (by more than 50%) transepithelial resistance (TER) denoting barrier function alteration. Importantly, TL prevented such loss in TER. Therefore, TRPV2 merits further exploration as a pharmacodynamic biomarker for leukemia patients (with pulmonary inflammation) who might be suitable for a novel [adjuvant] therapeutic strategy based on TL.


Asunto(s)
Biomarcadores/metabolismo , Leucemia/patología , Neumonía/patología , Canales Catiónicos TRPV/metabolismo , ADP-Ribosil Ciclasa 1/metabolismo , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Humanos , Imidazoles/farmacología , Imidazoles/uso terapéutico , Leucemia/complicaciones , Leucemia/tratamiento farmacológico , Leucocitos Mononucleares/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neumonía/complicaciones , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/genética , Regulación hacia Arriba/efectos de los fármacos , ortoaminobenzoatos/farmacología , ortoaminobenzoatos/uso terapéutico
16.
Oxid Med Cell Longev ; 2019: 5162687, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30755785

RESUMEN

Interest is growing in finding natural sources of effective antitumor agents that generate fewer side effects than conventional chemotherapeutic drugs. Avenanthramides (Avns) are such compounds; these phenolic molecules naturally occur in oats and have antioxidant, anti-inflammatory, and antiproliferative effects making them worthy of further research. The aim of this study is to characterise Avns' curative ability and antineoplastic activity on solid-form Ehrlich tumors. For the study, 75 female mice were randomly and equally allocated to five groups (group 1-control, group 2-DMSO, group 3-positive control receiving Avns, group 4-mice with Ehrlich solid tumor, and group 5-Ehrlich solid tumor treated with Avns). Mice with Ehrlich solid tumors exhibit increased tumor volume; elevated expression of AFP, ALT, AST, Bcl2, CEA, cholesterol, creatinine, urea, MDA, PCNA, potassium, triglycerides, TNF-α, and NF-κB; and a concomitant decline in catalase, GSH, P53, and SOD. In the mice with Ehrlich tumors who received Avns, there appeared to be improvement in NF-κB TNF-α, tumor markers (AFP and CEA), electrolytes, liver and kidney function enzymes, and lipid profiles; reduced MDA level; improved antioxidant parameters; normalised liver protein, P53, and PCNA; and reduced Bcl2 expression. Pathological examination of tumor lesions also indicated improvement. These results suggest that Avns exhibit antineoplastic activity and possess antioxidant properties that enhance the antioxidant defence system, thus reducing the oxidative stress caused by Ehrlich solid tumors.


Asunto(s)
Carcinoma de Ehrlich/tratamiento farmacológico , ortoaminobenzoatos/uso terapéutico , Animales , Femenino , Ratones , ortoaminobenzoatos/farmacología
17.
Oncol Rep ; 41(1): 125-132, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30542720

RESUMEN

Glioblastoma is the most common type of primary brain tumour in adults, and its pathogenesis is particularly complicated. Among the many possible mechanisms underlying its pathogenesis, hyperactivation of the PI3K/Akt pathway is essential to the occurrence and development of glioma through the loss of PTEN or somatic activating mutations in PIK3CA. In the present study, we investigated the effect of the PI3Kß inhibitor AZD6482 on glioma cells. The CCK-8 assay showed dose-dependent cytotoxicity in glioma cell lines treated with AZD6482. Additionally, AZD6482 treatment was found to significantly induce apoptosis and cell cycle arrest as detected using flow cytometry. Moreover, as shown using western blot analysis, the levels of p-AKT, p-GSK-3ß, Bcl-2, and cyclin D1 were decreased after AZD6482 treatment. In addition, we found that AZD6482 inhibited the migration and invasion of glioma cells as detected by wound healing and Transwell invasion assays. Taken together, our findings indicate that AZD6482 exerts an antitumour effect by inhibiting proliferation and inducing apoptosis in human glioma cells. AZD6482 may be applied as an adjuvant therapy to improve the therapeutic efficacy of glioblastoma treatment.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Glioblastoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinonas/farmacología , ortoaminobenzoatos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/genética , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Glioblastoma/genética , Humanos , Mutación , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinonas/uso terapéutico , Transducción de Señal/genética , ortoaminobenzoatos/uso terapéutico
18.
Cell Physiol Biochem ; 51(4): 1894-1907, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30504717

RESUMEN

BACKGROUND/AIMS: Targeting survivin, an anti-apoptotic protein and mitotic regulator, is considered as an effective therapeutic option for pancreatic cancer (PaCa). Tolfenamic acid (TA) showed anti-cancer activity in pre-clinical studies. A recent discovery demonstrated a copper(II) complex of TA (Cu-TA) can result in higher activity. In this study, the ability of Cu-TA to inhibit survivin and its transcription factors, Specificity protein (Sp) 1 and 3 in PaCa cell lines and tumor growth in mouse xenograft model were evaluated. METHODS: Cell growth inhibition was measured in MIA PaCa-2 and Panc1 cells for 2 days using CellTiter-Glo kit. Sp1, Sp3 and survivin expression (by Western blot and qPCR), apoptotic cells and cell cycle phase distribution (by flow cytometry) were evaluated. A pilot study was performed using athymic nude mice [treated with vehicle/Cu-TA (25 or 50 mg/kg) 3 times/week for 4 weeks. RESULTS: The IC50 value for Cu-TA was about half than TA.Both agents repressed the protein expression of Sp1/Sp3/survivin, Cu-TA was more effective than TA. Especially effect on survivin inhibition was 5.2 (MIA PaCa-2) or 6.4 (Panc1) fold higher and mRNA expression of only survivin was decreased. Apoptotic cells increased with Cu-TA treatment in both cell lines, while Panc1 showed both effect on apoptosis and cell cycle (G2/M) arrest. Cu-TA decreased the tumor growth in mouse xenografts (25 mg/kg: 48%; 50 mg/kg: 68%). Additionally, there was no change observed in mice body weights, indicating no overt toxicity was occurring. CONCLUSION: These results show that Cu-TA can serve as an effective survivin inhibitor for inhibiting PaCa cell growth.


Asunto(s)
Antineoplásicos/uso terapéutico , Cobre/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp3/genética , Survivin/antagonistas & inhibidores , ortoaminobenzoatos/uso terapéutico , Animales , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Complejos de Coordinación/química , Complejos de Coordinación/uso terapéutico , Cobre/química , Regulación hacia Abajo/efectos de los fármacos , Femenino , Humanos , Masculino , Ratones , Ratones Desnudos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , ortoaminobenzoatos/química
19.
Int J Nanomedicine ; 13: 5971-5990, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30323586

RESUMEN

BACKGROUND: Abnormal expression of stromal cells and extracellular matrix in tumor stroma creates a tight barrier, leading to insufficient extravasation and penetration of therapeutic agents. Cancer-associated fibroblasts (CAFs) take on pivotal roles encouraging tumor progression. METHOD: To surmount the refractoriness of stroma, we constructed a multi-targeting combined scenario of anti-CAFs agent tranilast and antitumor agent docetaxel micelles (DTX-Ms). Tranilast cut down crosstalk between tumor cells and stromal cells, ameliorated the tumor microenvironment, and enhanced the antiproliferation efficacy of DTX-Ms on cancer cells. RESULTS: Diverse experiments demonstrated that tranilast enhanced DTX-Ms' antitumor effect in a two-stage pattern by CAFs ablation, tumor cell migration blocking, and metastasis inhibition. Along with activated CAFs decreasing in vivo, the two-stage therapy succeeded in reducing interstitial fluid pressure, normalizing microvessels, improving micelles penetration and retention, and inhibiting tumor growth and metastasis. Interestingly, tranilast alone failed to inhibit tumor growth in vivo, and it could only be used as an adjuvant medicine together with an antitumor agent. CONCLUSION: Our proposed two-stage therapy offers a promising strategy to enhance antitumor effects by breaking down CAFs barrier and increasing micellar delivery efficiency.


Asunto(s)
Fibroblastos Asociados al Cáncer/patología , Micelas , Neoplasias/tratamiento farmacológico , Taxoides/uso terapéutico , Células 3T3 , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Peso Corporal , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Docetaxel , Líquido Extracelular/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Microvasos/efectos de los fármacos , Microvasos/patología , Metástasis de la Neoplasia , Neoplasias/irrigación sanguínea , Neoplasias/patología , Especificidad de Órganos , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/patología , Distribución Tisular , Microambiente Tumoral/efectos de los fármacos , ortoaminobenzoatos/farmacología , ortoaminobenzoatos/uso terapéutico
20.
Nutr Cancer ; 70(7): 1159-1165, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30204484

RESUMEN

Moringin (MOR), a glycosyl-isothiocyanate obtained by myrosinase-catalyzed hydrolysis of the precursor 4-(α-l-rhamnosyloxy)-benzyl glucosinolate (glucomoringin), found predominantly in the seeds of Moringa oleifera, shows anticancer effects against several cancer cell lines. Avenanthramide (AVN) 2f is a phytochemical purified from oats with antioxidant and anticancer properties. The aim of this study was to investigate the antiproliferative and proapoptotic effects of MOR and AVN 2f used alone and in combination on Hep3B cancer cells, which are highly resistant to conventional anticancer drugs. We found that a cocktail of MOR and AVN 2f significantly inhibited the Hep3B proliferation rate by markedly increasing the activity of caspases 2, 8, 9, and 3. Extrinsic apoptosis was induced by the AVN 2f-mediated activation of caspase 8, while the intrinsic apoptotic pathway was triggered by MOR-induced increase in the levels of intracellular reactive oxygen species, MOR-mediated activation of caspases 2 and 9 and the MOR-mediated downregulation of the prosurvival gene BIRC5. Our results suggest that the combination MOR + AVN 2f could be an effective chemopreventive cocktail against the development of hepatocarcinoma.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , ortoaminobenzoatos/farmacología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Caspasas/metabolismo , Línea Celular Tumoral , Humanos , Isotiocianatos/administración & dosificación , Isotiocianatos/farmacología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Especies Reactivas de Oxígeno/metabolismo , Survivin/genética , Survivin/metabolismo , ortoaminobenzoatos/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA