Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Inflammopharmacology ; 31(6): 2827-2842, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37924473

RESUMEN

Acetylsalicylic acid (ASA), also known as aspirin, was discovered in 1897 as an acetylated form of salicylate. It has been widely used for its anti-inflammatory and antiplatelet effects. It is commonly used for its cardiovascular benefits and is prescribed as secondary prophylaxis after a heart attack. Furthermore, low-dose, long-term ASA is used to reduce the risk of heart attack and stroke in individuals without prior cardiovascular disease. Acetylsalicylic acid acts as a non-selective inhibitor of cyclooxygenase (COX), which inhibits the synthesis of prostaglandins and prevents pro-inflammatory cytokines. Findings suggest that targeting cytokines and growth factors could be a potential therapeutic strategy for reducing neuroinflammation and slowing down the progression of dementia. Additionally, prostaglandins contribute to synaptic plasticity and can act as retrograde messengers in synapses. Research has implicated COX-1, one of the isoforms of the enzyme, in neuroinflammation and neurodegenerative disorders. The inhibition of COX-1 might potentially prevent impairments in working memory and reduce neuroinflammation caused by beta-amyloid proteins in some conditions, such as Alzheimer's disease (AD). Cyclooxygenase-2, an inducible form of the enzyme, is expressed in cortical and hippocampal neurons and is associated with long-term synaptic plasticity. The inhibition or knockout of COX-2 has been shown to decrease long-term potentiation, a process involved in memory formation. Studies have also demonstrated that the administration of COX-2 inhibitors impairs cognitive function and memory acquisition and recall in animal models. There remains a debate regarding the effects of aspirin on dementia and cognitive decline. Although some studies suggest a possible protective effect of non-steroidal anti-inflammatory drugs, including aspirin, against the development of AD, others have shown inconsistent evidence. This review provides an overview of the effects of ASA or its active metabolite salicylate on learning, memory, and synaptic plasticity.


Asunto(s)
Enfermedad de Alzheimer , Infarto del Miocardio , Animales , Humanos , Aspirina/farmacología , Aspirina/uso terapéutico , Enfermedades Neuroinflamatorias , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/prevención & control , Trastornos de la Memoria/tratamiento farmacológico , Antiinflamatorios/uso terapéutico , Prostaglandinas , Ciclooxigenasa 2/metabolismo , Infarto del Miocardio/tratamiento farmacológico , Citocinas
2.
PLoS Pathog ; 18(8): e1010703, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35930608

RESUMEN

Influenza A virus (IAV) infection during pregnancy initiates significant aortic endothelial and vascular smooth muscle dysfunction, with inflammation and T cell activation, but the details of the mechanism are yet to be clearly defined. Here we demonstrate that IAV disseminates preferentially into the perivascular adipose tissue (PVAT) of the aorta in mice. IAV mRNA levels in the PVAT increased at 1-3 days post infection (d.p.i) with the levels being ~4-8 fold higher compared with the vessel wall. IAV infection also increased Ly6Clow patrolling monocytes and Ly6Chigh pro-inflammatory monocytes in the vessel wall at 3 d.p.i., which was then followed by a greater homing of these monocytes into the PVAT at 6 d.p.i. The vascular immune phenotype was characteristic of a "vascular storm"- like response, with increases in neutrophils, pro-inflammatory cytokines and oxidative stress markers in the PVAT and arterial wall, which was associated with an impairment in endothelium-dependent relaxation to acetylcholine. IAV also triggered a PVAT compartmentalised elevation in CD4+ and CD8+ activated T cells. In conclusion, the PVAT of the aorta is a niche that supports IAV dissemination and a site for perpetuating a profound innate inflammatory and adaptive T cell response. The manifestation of this inflammatory response in the PVAT following IAV infection may be central to the genesis of cardiovascular complications arising during pregnancy.


Asunto(s)
Virus de la Influenza A , Tejido Adiposo , Animales , Aorta , Endotelio Vascular , Femenino , Inflamación/genética , Ratones , Embarazo
3.
Microorganisms ; 9(6)2021 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-34205044

RESUMEN

The current outbreak caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), termed coronavirus disease 2019 (COVID-19), has generated a notable challenge for diabetic patients. Overall, people with diabetes have a higher risk of developing different infectious diseases and demonstrate increased mortality. Type 2 diabetes mellitus (T2DM) is a significant risk factor for COVID-19 progression and its severity, poor prognosis, and increased mortality. How diabetes contributes to COVID-19 severity is unclear; however, it may be correlated with the effects of hyperglycemia on systemic inflammatory responses and immune system dysfunction. Using the envelope spike glycoprotein SARS-CoV-2, COVID-19 binds to angiotensin-converting enzyme 2 (ACE2) receptors, a key protein expressed in metabolic organs and tissues such as pancreatic islets. Therefore, it has been suggested that diabetic patients are more susceptible to severe SARS-CoV-2 infections, as glucose metabolism impairments complicate the pathophysiology of COVID-19 disease in these patients. In this review, we provide insight into the COVID-19 disease complications relevant to diabetes and try to focus on the present data and growing concepts surrounding SARS-CoV-2 infections in T2DM patients.

4.
Mol Neurobiol ; 58(7): 3561-3574, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33765290

RESUMEN

The current pandemic of the new human coronavirus (CoV), i.e., severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has created an urgent global condition. The disease, termed coronavirus disease 2019 (COVID-19), is primarily known as a respiratory tract infection. Although SARS-CoV-2 directly invades the lungs, COVID-19 is a complex multi-system disease with varying degrees of severity and affects several human systems including the cardiovascular, respiratory, gastrointestinal, neurological, hematopoietic, and immune systems. From the existing data, most COVID-19 cases develop a mild disease typically presented with fever and respiratory illness. However, in some patients, clinical evidence suggests that COVID-19 might progress to acute respiratory distress syndrome (ARDS), multi-organ dysfunction, and septic shock resulting in a critical condition. Likewise, specific organ dysfunction seems to be related to the disease complication, worsens the condition, and increases the lethality of COVID-19. The neurological manifestations in association with disease severity and mortality have been reported in COVID-19 patients. Despite the continuously increasing reports of the neurological symptoms of SARS-CoV-2, our knowledge about the possible routes of nervous system involvement associated with COVID-19 is limited. Herein, we will primarily describe the critical aspects and clinical features of SARS-CoV-2 related to nervous system impairment and then discuss possible routes of SARS-CoV-2 nervous system involvement based on the current evidence.


Asunto(s)
COVID-19/complicaciones , Síndrome de Liberación de Citoquinas/virología , Enfermedades del Sistema Nervioso/virología , Humanos
5.
Cells ; 8(7)2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31266232

RESUMEN

BACKGROUND: The zinc transporter Zip7 modulates zinc flux and controls cell signaling molecules associated with glucose metabolism in skeletal muscle. The present study evaluated the role of Zip7 in cell signaling pathways involved in insulin-resistant skeletal muscle and mice fed a high-fat diet. METHODS: Insulin-resistant skeletal muscle cells were prepared by treatment with an inhibitor of the insulin receptor, HNMPA-(AM)3 or palmitate, and Zip7 was analyzed along with pAkt, pTyrosine and Glut4. Similarly, mice fed normal chow (NC) or a high-fat diet (HFD) were also analyzed for protein expression of Glut4 and Zip7. An overexpression system for Zip7 was utilized to determine the action of this zinc transporter on several genes implicated in insulin signaling and glucose control. RESULTS: We identified that Zip7 is upregulated by glucose in normal skeletal muscle cells and downregulated in insulin-resistant skeletal muscle. We also observed (as expected) a decrease in pAkt and Glut4 in the insulin-resistant skeletal muscle cells. The overexpression of Zip7 in skeletal muscle cells led to the modulation of key genes involved in the insulin signaling axis and glucose metabolism including Akt3, Dok2, Fos, Hras, Kras, Nos2, Pck2, and Pparg. In an in vivo mouse model, we identified a reduction in Glut4 and Zip7 in the skeletal muscle of mice fed a HFD compared to NC controls. CONCLUSIONS: These data suggest that Zip7 plays a role in skeletal muscle insulin signaling and is downregulated in an insulin-resistant, and HFD state. Understanding the molecular mechanisms of Zip7 action will provide novel opportunities to target this transporter therapeutically for the treatment of insulin resistance and type 2 diabetes.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Resistencia a la Insulina , Fibras Musculares Esqueléticas/patología , Animales , Línea Celular , Diabetes Mellitus Experimental/etiología , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/patología , Dieta Alta en Grasa/efectos adversos , Regulación hacia Abajo , Glucosa/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Insulina/metabolismo , Ratones , Transducción de Señal
6.
Nutrients ; 11(2)2019 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-30781350

RESUMEN

Type 2 diabetes mellitus (T2DM) is a disease associated with dysfunctional metabolic processes that lead to abnormally high levels of blood glucose. Preceding the development of T2DM is insulin resistance (IR), a disorder associated with suppressed or delayed responses to insulin. The effects of this response are predominately mediated through aberrant cell signalling processes and compromised glucose uptake into peripheral tissue including adipose, liver and skeletal muscle. Moreover, a major factor considered to be the cause of IR is endoplasmic reticulum (ER) stress. This subcellular organelle plays a pivotal role in protein folding and processes that increase ER stress, leads to maladaptive responses that result in cell death. Recently, zinc and the proteins that transport this metal ion have been implicated in the ER stress response. Specifically, the ER-specific zinc transporter ZIP7, coined the "gate-keeper" of zinc release from the ER into the cytosol, was shown to be essential for maintaining ER homeostasis in intestinal epithelium and myeloid leukaemia cells. Moreover, ZIP7 controls essential cell signalling pathways similar to insulin and activates glucose uptake in skeletal muscle. Accordingly, ZIP7 may be essential for the control of ER localized zinc and mechanisms that disrupt this process may lead to ER-stress and contribute to IR. Accordingly, understanding the mechanisms of ZIP7 action in the context of IR may provide opportunities to develop novel therapeutic options to target this transporter in the treatment of IR and subsequent T2DM.


Asunto(s)
Proteínas de Transporte de Catión/fisiología , Diabetes Mellitus Tipo 2/metabolismo , Resistencia a la Insulina/fisiología , Zinc/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Estrés del Retículo Endoplásmico/fisiología , Humanos , Transducción de Señal/fisiología
7.
Cells ; 7(10)2018 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-30326660

RESUMEN

Sustained endoplasmic reticular stress (ERS) is implicated in aggressive metastasis of cancer cells and increased tumor cell proliferation. Cancer cells activate the unfolded protein response (UPR), which aids in cellular survival and adaptation to harsh conditions. Inhibition of apoptosis, in contrast, is a mechanism adopted by cancer cells with the help of the inhibitor of an apoptosis (IAP) class of proteins such as Survivin to evade cell death and gain a proliferative advantage. In this study, we aimed to reveal the interrelation between ERS and Survivin. We initially verified the expression of Survivin in Winnie (a mouse model of chronic ERS) colon tissues by using immunohistochemistry (IHC) and immunofluorescence (IF) in comparison with wild type Blk6 mice. Additionally, we isolated the goblet cells and determined the expression of Survivin by IF and protein validation. Tunicamycin was utilized at a concentration of 10 µg/mL to induce ERS in the LS174T cell line and the gene expression of the ERS markers was measured. This was followed by determination of inflammatory cytokines. Inhibition of ERS was carried out by 4Phenyl Butyric acid (4PBA) at a concentration of 10 mM to assess whether there was a reciprocation effect. The downstream cell death assays including caspase 3/7, Annexin V, and poly(ADP-ribose) polymerase (PARP) cleavage were evaluated in the presence of ERS and absence of ERS, which was followed by a proliferative assay (EdU click) with and without ERS. Correspondingly, we inhibited Survivin by YM155 at a concentration of 100 nM and observed the succeeding ERS markers and inflammatory markers. We also verified the caspase 3/7 assay. Our results demonstrate that ERS inhibition not only significantly reduced the UPR genes (Grp78, ATF6, PERKandXBP1) along with Survivin but also downregulated the inflammatory markers such as IL8, IL4, and IL6, which suggests a positive correlation between ERS and the inhibition of apoptosis. Furthermore, we provided evidence that ERS inhibition promoted apoptosis in LS174T cells and shortened the proliferation rate. Moreover, Survivin inhibition by YM155 led to a comparable effect as that of ERS inhibition, which includes attenuation of ERS genes and inflammatory markers as well as the promotion of programmed cell death via the caspase 3/7 pathway. Together, our results propose the interrelation between ERS and inhibition of apoptosis assigning a molecular and therapeutic target for cancer treatment.

8.
Comput Biol Med ; 100: 196-202, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30029049

RESUMEN

The zinc transporter SLC39A7 (ZIP7)1 is a resident endoplasmic reticulum (ER) protein that is involved in controlling the release of zinc from this organelle into the cytosol. Subsequently, zinc plays a major role in processes that preserve cellular homeostasis. The ER contains a high concentration of zinc, and under normal physiological responses, maintains ER function. Disturbances in the concentration and distribution of zinc in the ER leads to abnormal processes that typify many disease states. ZIP7 is protective against ER stress and is a critical 'gate-keeper' of zinc release from the ER during processes that require cellular maintenance. However, it is not known how ZIP7 achieves this protective activity while maintaining cellular function. Bioinformatics analysis was utilised to determine the relationship between ZIP7 and other zinc transporters across humans and the animal and plant kingdom to determine the structure of this transporter in binding zinc in the ER. Analysis of the amino acid sequence of ZIP7 revealed several potential histidine binding sites for zinc in the N-terminal region that were significantly different in comparison to the other members of this family. Moreover, this histidine-rich region in the N-terminal of ZIP7 was highly conserved across the animal and plant kingdom. Accordingly, the highly conserved histidine-rich region in the N-termini of ZIP7 across the animal and plant kingdom suggests that this domain has critical function(s). We hypothesise that ER-localized ZIP7 can potentially sequester zinc to these histidine-rich regions and therefore provides a mechanism that is protective of this cellular structure.


Asunto(s)
Proteínas de Transporte de Catión , Estrés del Retículo Endoplásmico/fisiología , Retículo Endoplásmico , Homeostasis , Zinc , Animales , Proteínas de Transporte de Catión/química , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Humanos , Transporte Iónico/fisiología , Dominios Proteicos , Zinc/química , Zinc/metabolismo
9.
PLoS One ; 13(1): e0191727, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29373583

RESUMEN

Zinc is a metal ion that is an essential cell signaling molecule. Highlighting this, zinc is an insulin mimetic, activating cellular pathways that regulate cellular homeostasis and physiological responses. Previous studies have linked dysfunctional zinc signaling with several disease states including cancer, obesity, cardiovascular disease and type 2 diabetes. The present study evaluated the insulin-like effects of zinc on cell signaling molecules including tyrosine, PRSA40, Akt, ERK1/2, SHP-2, GSK-3ß and p38, and glucose oxidation in human and mouse skeletal muscle cells. Insulin and zinc independently led to the phosphorylation of these proteins over a 60-minute time course in both mouse and human skeletal muscle cells. Similarly, utilizing a protein array we identified that zinc could active the phosphorylation of p38, ERK1/2 and GSK-3B in human and ERK1/2 and GSK-3B in mouse skeletal muscle cells. Glucose oxidation assays were performed on skeletal muscle cells treated with insulin, zinc, or a combination of both and resulted in a significant induction of glucose consumption in mouse (p<0.01) and human (p<0.05) skeletal muscle cells when treated with zinc alone. Insulin, as expected, increased glucose oxidation in mouse (p<0.001) and human (0.001) skeletal muscle cells, however the combination of zinc and insulin did not augment glucose consumption in these cells. Zinc acts as an insulin mimetic, activating key molecules implicated in cell signaling to maintain glucose homeostasis in mouse and human skeletal muscle cells. Zinc is an important metal ion implicated in several biological processes. The role of zinc as an insulin memetic in activating key signaling molecules involved in glucose homeostasis could provide opportunities to utilize this ion therapeutically in treating disorders associated with dysfunctional zinc signaling.


Asunto(s)
Glucemia/metabolismo , Glucosa/metabolismo , Insulina/metabolismo , Músculo Esquelético/metabolismo , Zinc/farmacología , Animales , Línea Celular , Humanos , Ratones , Oxidación-Reducción , Transducción de Señal
10.
J Biomed Sci ; 24(1): 87, 2017 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-29157234

RESUMEN

BACKGROUND: Zinc is a metal ion that is essential for growth and development, immunity, and metabolism, and therefore vital for life. Recent studies have highlighted zinc's dynamic role as an insulin mimetic and a cellular second messenger that controls many processes associated with insulin signaling and other downstream pathways that are amendable to glycemic control. MAIN BODY: Mechanisms that contribute to the decompartmentalization of zinc and dysfunctional zinc transporter mechanisms, including zinc signaling are associated with metabolic disease, including type 2 diabetes. The actions of the proteins involved in the uptake, storage, compartmentalization and distribution of zinc in cells is under intense investigation. Of these, emerging research has highlighted a role for several zinc transporters in the initiation of zinc signaling events in cells that lead to metabolic processes associated with maintaining insulin sensitivity and thus glycemic homeostasis. CONCLUSION: This raises the possibility that zinc transporters could provide novel utility to be targeted experimentally and in a clinical setting to treat patients with insulin resistance and thus introduce a new class of drug target with utility for diabetes pharmacotherapy.


Asunto(s)
Glucemia/metabolismo , Proteínas Portadoras/genética , Diabetes Mellitus Tipo 2/terapia , Resistencia a la Insulina/genética , Zinc/metabolismo , Animales , Transporte Biológico , Proteínas Portadoras/metabolismo , Diabetes Mellitus Tipo 2/genética , Homeostasis , Humanos , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/terapia , Ratones , Ratas
11.
Curr Pharm Des ; 23(16): 2328-2332, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28120719

RESUMEN

BACKGROUND: Zinc is a critical metal ion essential for life. The biological significance of zinc is highlighted by the enormous number of proteins (approximately 10% of the human proteome) that have zinc-binding capacity. Accordingly, zinc concentrations in cells are tightly regulated by two families of zinc transporter proteins: Slc30a (ZnT) and Slc39a (Zip). ZnT and Zip are known to decrease and increase cytosolic zinc concentrations respectively. Both zinc transporters are suggested to be implicated in a number of disorders and disease states through dysfunctional zinc transport including cancer, diabetes and gastrointestinal (GI) disease. METHOD: The goal of this work was to identify the role of zinc transporters in GI disease/disorders. Where possible, reference will be made in the context of the function of zinc transporters and their potential role in GI disorders/ diseases with a view towards their possible therapeutic utility in the treatment of these ailments. PubMed was utilized to search for articles with the terms "zinc and GI disease", "zinc transporters and GI disease", "zinc and gut", zinc transporters and gut", and "zinc transporters and intestinal disorders". RESULTS: We identified a number of reviews on GI disorders/disease states associated with zinc deficiencies, but the very proteins that transport this metal ion in these systems are not well-defined. From a systematic review, we identified the following zinc transporters (Zip1, 2, 4-7, 10, 11 and 14; and ZnT1, 8 and 10) as having some functional role in the GI system and potentially could have a therapeutic role in the treatment of GI disease/disorders. CONCLUSION: An increasingly common health issue in our communities is disorder/disease of the GI system. Although therapies targeting these disorders are somewhat beneficial, there is a need to develop better, more efficient treatments. Despite that many gut-related disorders/disease states have been analyzed in the context of zinc deficiencies, the transport proteins that move zinc across cells are not well-defined. Zinc transporters are expressed in a range of GI tissue and cells, and their roles in the maintenance, integrity and disease processes of the GI system need to be addressed. This might open a whole new avenue of opportunities for the development of novel therapies targeting these receptors in GI disease states.


Asunto(s)
Proteínas Portadoras/metabolismo , Enfermedades Gastrointestinales/metabolismo , Zinc/metabolismo , Humanos
12.
Daru ; 24: 1, 2016 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-26739353

RESUMEN

BACKGROUND: Leukemia is distinguished by abnormal proliferation of leukocytes. Although there has been some progress in developing novel cancer therapies, no significant improvement was observed in the overall survival rate over the last decade. Selective cyclooxygenase-2 (COX-2) inhibitors are known to inhibit tumor growth by exerting antimetastatic and antiangiogenic effects through inhibition of COX -dependent and independent pathways. The ability of two new triaryl-oxadiazole derivatives, compounds A (3-(4-chlorophenyl) -5-(4-flurophenyl)-4-Phenyl-4,5-dihydro-1,2,4-oxadiazole) and B (3,5-bis(4-chlorophenyl)-4-Phenyl-4,5-dihydro-1,2,4-oxadiazole), to induce apoptosis in human erythroleukemia K562 cells was evaluated and the upstream mechanism was investigated. METHODS: K562 cells were treated with compounds A and B at their IC50 concentrations and analyzed by DAPI staining and Annexin-V-FLUOS labelling solution. Nuclear factor kappa-B (NF-κB) activation was evaluated by TransAM kit. Cyclooxygenase-2 (COX-2), Caspase-3, Bax, Bcl-2, ferritin heavy chain (FHC), extra cellular signal-regulated kinase (ERK), p-ERK and early growth response protein-1 (Egr1) levels were determined using Western blotting, while c-Myc mRNA level was investigated by RT-PCR. RESULTS: Changes in nuclear morphology and the increased annexin-V/PI staining revealed the apoptotic cell death in compounds A- and B-treated K562 cells. A significant reduction in NF-κB activity as well as FHC and p-ERK levels were detected in these cells. No change was observed in the levels of Bax, Bcl-2, Caspase-3, COX-2, c-Myc and Egr1, following treatment with the two compounds. Collectively, compounds A and B potentiate apoptosis as shown by DAPI staining, flowcytometry, FHC and p-ERK downregulation and NF-κB inactivation. CONCLUSION: Two compounds induce apoptosis in a COX-2-independent manner which also appears to be independent from mitochondria, caspase and c-Myc/Egr1 pathways.


Asunto(s)
Celecoxib/análogos & derivados , Inhibidores de la Ciclooxigenasa 2/farmacología , Leucemia Eritroblástica Aguda/metabolismo , FN-kappa B/metabolismo , Oxadiazoles/farmacología , Apoptosis , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inhibidores de la Ciclooxigenasa 2/química , Regulación Neoplásica de la Expresión Génica , Humanos , Oxadiazoles/química , Proteínas Proto-Oncogénicas c-myc/genética , Transducción de Señal/efectos de los fármacos
13.
J Cell Biochem ; 116(1): 81-90, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25142612

RESUMEN

Recently, much effort has been directed toward the search for compounds that influence apoptosis and to understand their mechanisms of action. Cyclooxygenase (COX)-2 inhibitors may induce apoptosis through the COX-2-independent mechanism via a mitochondrial pathway. In view of the reported antiproliferative activities of two COX-2 inhibitor derivatives (1, 2) in breast cancer cells (MCF-7), the present study was undertaken to evaluate the potential of these compounds to induce apoptosis and unravel the associated mechanisms. The apoptotic activities of the two compounds were assessed using flow cytometry, fluorescence microscope, and Western blot analysis. Compounds 1 and 2-treated MCF-7 cells revealed the apoptotic cell death, as confirmed by the changes in nuclear morphology and the increased annexin-V/PI staining. Elevation of Bax to Bcl-2 ratio and activation of caspase-3 were found to be associated with the initiation of apoptosis induced by compound 1. Further investigation showed that compounds 1 and 2 inhibited NF-κB, FHC, and ERK activation, while no dramatic change was revealed in c-Myc and EGR-1 levels. Our data suggest that induction of apoptosis by compounds 1 and 2 is not associated with COX-2 expression and occurs through the NF-κB pathway, which sequentially inhibits P-ERK and FHC expression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , Apoferritinas/genética , Apoferritinas/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Femenino , Humanos , Células MCF-7 , FN-kappa B/genética , FN-kappa B/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...