Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
2.
Int J Mol Sci ; 24(14)2023 Jul 08.
Article in English | MEDLINE | ID: mdl-37511011

ABSTRACT

As metastasis is responsible for most cancer-related deaths, understanding the cellular and molecular events that lead to cancer cell migration and invasion will certainly provide insights into novel anti-metastatic therapeutic targets. Fascin-1 is an actin-bundling protein fundamental to all physiological or pathological processes that require cell migration. It is responsible for cross-linking actin microfilaments during the formation of actin-rich cellular structures at the leading edge of migrating cells such as filopodia, lamellipodia and invadopodia. While most epithelial tissues express low levels of Fascin-1, it is dramatically elevated in the majority of cancers and its expression has been associated with more aggressive disease and decreased overall survival. Hence, it has been proposed as a potential anti-cancer target. In the present review, we studied recent literature with regard to Fascin-1 expression in different cancers, its role in altering the mechanical properties of cancer cells, promoting cancer cell migration, invasion and metastasis and the effect of its inhibition, via various pharmacological inhibitors, in eliminating metastasis in vitro and/or in vivo. Recent studies corroborate the notion that Fascin-1 is critically involved in metastasis and prove that it is a valuable anti-metastatic target that is worth investigating further.


Subject(s)
Actins , Neoplasms , Humans , Actins/metabolism , Cell Movement , Neoplasms/metabolism , Actin Cytoskeleton/metabolism
3.
Life Sci ; 309: 121047, 2022 Nov 15.
Article in English | MEDLINE | ID: mdl-36208660

ABSTRACT

AIMS: Alzheimer's Disease (AD) is characterized by progressive cognitive impairment, and memory loss. It has been shown that depletion of estrogens renders women vulnerable to AD with menopause women presenting higher risk for AD development than men. However, women under hormone replacement therapy (HRT) with 17ß-estradiol (E2) show lower risk for AD, implying that E2 may be protective. It has been shown that E2 exerts its effects through the estrogen receptor (ER) but also via its biologically active metabolites, 2-hydroxyestradiol (2OH), and 2-methoxyestradiol (2ME). We hypothesized that the neuroprotective effects of E2 are partly attributed to its metabolites. MATERIALS AND METHODS: SH-SY5Y neuronal cells were subjected oxidative stress (OS) cell death by hydrogen peroxide (H2O2), in the presence or absence of E2, 2ME and 2OH. Viability was assessed by trypan blue and thiazolyl blue tetrazolium bromide assays, intracellular OS with the Dichlorodihydrofluorescein Diacetate (DCFDA) assay, and Bax, p53 and PUMA quantified by RT-PCR. Tau hyperphosphorylation was studied by western blot. KEY FINDINGS: E2 and its metabolites 2OH and 2ME protect from cell death as assessed by the viability assays. Their effect was partly attributed to their antioxidant properties evidenced by the reduction of intracellular OS. Treatment with 2ME resulted in a reduction of Bax, but not p53 or PUMA in cells challenged with OS. Finally, 2ME was able to inhibit tau hyperphosphorylation as well. SIGNIFICANCE: E2 protects neuron cells partly through its metabolites. Further studies are needed to fully delineate the mechanism for this protection.


Subject(s)
Neuroblastoma , Neuroprotective Agents , Humans , Female , 2-Methoxyestradiol/pharmacology , Hydrogen Peroxide/toxicity , Neuroprotective Agents/pharmacology , Receptors, Estrogen , bcl-2-Associated X Protein , Antioxidants/pharmacology , Trypan Blue/pharmacology , Apoptosis Regulatory Proteins , Estradiol/pharmacology , Estradiol/metabolism , Estrogens/pharmacology , Cell Death
4.
Nat Prod Res ; 36(16): 4210-4214, 2022 Aug.
Article in English | MEDLINE | ID: mdl-34498955

ABSTRACT

Smoking is a public health concern, and even though smoking cessation methods exist, nicotine replacement therapy (NRT) is often ineffective. Smoking behavior is related to the nicotine metabolizing enzyme (NME) P450 2A6 (mouse 2A5) polymorphisms. Accordingly, fast metabolizers are nicotine dependent, and have low quitting rates compared to slow metabolizers. In this study we examined the ability of Ginkgo biloba L (GB) and its constituents to inhibit the NME, using mouse liver microsomes containing the 2A5 enzyme. Our results indicate that GB can inhibit 2A5 (25% inhibition at 5%v/v), with the flavonoids quercetin, isorhamnetin, and kaempferol being responsible for this inhibition (23.5%, 10.7%, 25.2% inhibition at 60 ng/µL, respectively). Importantly, the flavonoids inhibited 2A5 via mechanism based inhibition (for quercetin 30 ng/µl inhibition increased from 20.8% to 26.9% within 15 minutes). Our results suggest that GB if consumed on a regular basis can help NRT enhancement particularly in fast nicotine metabolizers.


Subject(s)
Ginkgo biloba , Smoking Cessation , Animals , Dietary Supplements , Flavonoids/pharmacology , Mice , Microsomes, Liver , Nicotine/pharmacology , Quercetin/pharmacology , Tobacco Use Cessation Devices
5.
Int J Mol Sci ; 21(3)2020 Jan 21.
Article in English | MEDLINE | ID: mdl-31973122

ABSTRACT

Aging-associated neurodegenerative diseases, which are characterized by progressive neuronal death and synapses loss in human brain, are rapidly growing affecting millions of people globally. Alzheimer's is the most common neurodegenerative disease and it can be caused by genetic and environmental risk factors. This review describes the amyloid-ß and Tau hypotheses leading to amyloid plaques and neurofibrillary tangles, respectively which are the predominant pathways for the development of anti-Alzheimer's small molecule inhibitors. The function and structure of the druggable targets of these two pathways including ß-secretase, γ-secretase, and Tau are discussed in this review article. Computer-Aided Drug Design including computational structure-based design and ligand-based design have been employed successfully to develop inhibitors for biomolecular targets involved in Alzheimer's. The application of computational molecular modeling for the discovery of small molecule inhibitors and modulators for ß-secretase and γ-secretase is summarized. Examples of computational approaches employed for the development of anti-amyloid aggregation and anti-Tau phosphorylation, proteolysis and aggregation inhibitors are also reported.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/chemistry , Amyloid Precursor Protein Secretases/metabolism , Drug Design , Amyloid Precursor Protein Secretases/drug effects , Animals , Aspartic Acid Endopeptidases/chemistry , Brain/metabolism , Cheminformatics , Humans , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Neurodegenerative Diseases , Neurofibrillary Tangles/metabolism , Phosphorylation , Plaque, Amyloid/metabolism , Protein Conformation , tau Proteins/metabolism
6.
Expert Opin Drug Discov ; 15(1): 53-62, 2020 01.
Article in English | MEDLINE | ID: mdl-31744341

ABSTRACT

Introduction: Parkinson's Disease (PD) is a neurodegenerative central nervous system (CNS) disorder characterized by dopaminergic neuron degeneration with consequent reduction in striatal dopamine (DA) levels that leads to motor symptoms. Catechol-O-methyltransferase (COMT, E.C 2.1.1.6) inactivates dopamine and other substrates bearing catechol through the methylation of a hydroxyl group. COMT inhibition can block metabolism of catecholamines including DA. Since the increase in DA bioavailability is dependent on the inhibition of DA metabolism at the periphery, the development of COMT inhibitors as adjuvants to levodopa/aromatic amino acid decarboxylase (AADC) inhibitor treatment improves the clinical benefits of PD symptomatic treatment significantly.Areas covered: This review focuses on the contribution of computational studies to develop novel COMT inhibitors as therapeutics of Parkinson's disease with substantially improved efficacy.Expert opinion: The increasing use of in silico methods and the development of new chemoinformatic tools in combination with the knowledge gained from the development of different inhibitors studied both in silico, in vitro and in vivo, could help solve a number of issues related to the shortcomings of currently marketed treatments. They can also aid to open new avenues for centrally acting COMT inhibitors, and perhaps irreversible inhibitors, to be tested for PD and other neurological diseases.


Subject(s)
Antiparkinson Agents/pharmacology , Catechol O-Methyltransferase Inhibitors , Cheminformatics , Drug Evaluation, Preclinical/methods , Molecular Dynamics Simulation , Parkinson Disease/drug therapy , Animals , Antiparkinson Agents/chemistry
7.
Sci Rep ; 9(1): 10050, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31296919

ABSTRACT

Extracellular matrix (ECM)-adhesion proteins and actin cytoskeleton are pivotal in cancer cell invasion. Ras Suppressor-1 (RSU-1), a cell-ECM adhesion protein that interacts with PINCH-1, thus being connected to Integrin Linked Kinase (ILK), alpha-parvin (PARVA), and actin cytoskeleton, is up-regulated in metastatic breast cancer (BC) samples. Apart from the originally-identified gene (RSU-1L), an alternatively-spliced isoform (RSU-1-X1) has been reported. We used non-invasive MCF-7 cells, expressing only RSU-1L, and highly invasive MDA-MB-231-LM2 expressing both isoforms and generated stable shRNA-transduced cells lacking RSU-1L, while the truncated RSU-1-X1 isoform was depleted by siRNA-mediated silencing. RSU-1L depletion in MCF-7 cells resulted in complete abrogation of tumor spheroid invasion in three-dimensional collagen gels, whereas it promoted MDA-MB-231-LM2 invasion, through a compensatory upregulation of RSU-1-X1. When RSU-1-X1 was also eliminated, RSU-1L-depletion-induced migration and invasion were drastically reduced being accompanied by reduced urokinase plasminogen activator expression. Protein expression analysis in 23 human BC samples corroborated our findings showing RSU-1L to be upregulated and RSU-1-X1 downregulated in metastatic samples. We demonstrate for the first time, that both RSU-1 isoforms promote invasion in vitro while RSU-1L elimination induces RSU-1-X1 upregulation to compensate for the loss. Hence, we propose that both isoforms should be blocked to effectively eliminate metastasis.


Subject(s)
Breast Neoplasms/metabolism , Extracellular Matrix/metabolism , Transcription Factors/metabolism , Actin Cytoskeleton/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Movement , Humans , LIM Domain Proteins/metabolism , MCF-7 Cells , Membrane Proteins/metabolism , Molecular Targeted Therapy , Neoplasm Invasiveness , Neoplasm Metastasis , Protein Isoforms/genetics , RNA, Small Interfering/genetics , Transcription Factors/genetics , Up-Regulation , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism
8.
J R Soc Interface ; 16(154): 20190226, 2019 05 31.
Article in English | MEDLINE | ID: mdl-31113335

ABSTRACT

In many solid tumours a desmoplastic reaction takes place, which results in tumour tissue stiffening due to the extensive production of extracellular matrix (ECM) proteins, such as collagen, by stromal cells, mainly fibroblasts (FBs) and cancer-associated fibroblasts (CAFs). In this study, we investigated the effect of collagen stiffness on pancreatic FBs and CAFs, particularly on specific cytoskeleton properties and gene expression involved in tumour invasion. We found that cells become stiffer when they are cultured on stiff substrates and express higher levels of alpha-smooth muscle actin (α-SMA). Also, it was confirmed that on stiff substrates, CAFs are softer than FBs, while on soft substrates they have comparable Young's moduli. Furthermore, the number of spread FBs and CAFs was higher in stiffer substrates, which was also confirmed by Ras-related C3 botulinum toxin substrate 1 ( RAC1) mRNA expression, which mediates cell spreading. Although stress fibres in FBs become more oriented on stiff substrates, CAFs have oriented stress fibres regardless of substrate stiffness. Subsequently, we demonstrated that cells' invasion has a differential response to stiffness, which was associated with regulation of Ras homologue family member ( RhoA) and Rho-associated, coiled-coil containing protein kinase 1 ( ROCK-1) mRNA expression. Overall, our results demonstrate that collagen stiffness modulates FBs and CAFs cytoskeleton remodelling and alters their invasion properties.


Subject(s)
Collagen/metabolism , Cytoskeleton/metabolism , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Pancreas/metabolism , Actins/metabolism , Cell Line , Fibroblasts/cytology , Gene Expression Regulation , Humans , Pancreas/cytology , rac1 GTP-Binding Protein/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
10.
Trends Cancer ; 3(9): 621-630, 2017 09.
Article in English | MEDLINE | ID: mdl-28867166

ABSTRACT

Inefficient delivery of drugs is a main cause of chemotherapy failure in hypoperfused tumors. To enhance perfusion and drug delivery in these tumors, two strategies have been developed: vascular normalization, aiming at normalizing tumor vasculature and blood vessel leakiness, and stress alleviation, aiming at decompressing tumor vessels. Vascular normalization is based on anti-angiogenic drugs, whereas stress alleviation is based on stroma-depleting agents. We present here an alternative approach to normalize tumor vasculature, taking into account that malignant tumors tend to develop at sites of chronic inflammation. Similarly to tumor vessel leakiness, inflammation is also characterized by vascular hyperpermeability. Therefore, testing the ability of anti-inflammatory agents, such as non-steroidal anti-inflammatory drugs (NSAIDs) or inflammation resolution mediators, as an alternative means to increase tumor drug delivery might prove promising.


Subject(s)
Antineoplastic Agents/therapeutic use , Inflammation/pathology , Neoplasms/drug therapy , Neoplasms/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Capillary Permeability , Extracellular Matrix , Humans , Inflammation/complications , Inflammation/drug therapy , Inflammation/metabolism , Inflammation Mediators/metabolism , Neoplasms/etiology , Neoplasms/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Tissue Distribution , Treatment Outcome , Tumor Microenvironment/drug effects
11.
Toxicol Sci ; 159(2): 279-289, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28666357

ABSTRACT

This report aims to determine the permitted daily exposure (PDE) of flutamide, an androgen receptor blocker, as directed by guideline EMA/CHMP/CVPM/SWP/169430/2012 that came into effect on June 2015. A literature review was conducted to identify toxicity studies of flutamide. Hazards and sensitive endpoints were determined. Based on the no adverse effect levels (NOAELs) and lowest observed adverse effect levels (LOAELs) reported from both reproductive, developmental, and 28-day toxicity studies the PDE was calculated. Most of the toxicity studies converge toward a NOAEL of 1 mg/kg/d that translates to a PDE of 0.1 mg/d. However, taking into consideration the worst case scenarios for additional safety a PDE of 0.025 mg/d (25 µg/d) was calculated based on a reported NOAEL of 0.25 mg/kg/d. A PDE of 0.05 mg/d (50 µg/d) was also calculated from reproductive/developmental toxicity studies, which is in close agreement with the PDE from the 28-day toxicity studies. Considering the lowest PDE of 0.025 mg/d, residual flutamide at this dose is unlikely to pose any risk to humans. Nonmonotonic dose response (NMDR) effects of flutamide were not supported by literature. Oral route of administration was considered.


Subject(s)
Androgen Antagonists/toxicity , Flutamide/toxicity , Genitalia, Male/drug effects , Administration, Oral , Androgen Antagonists/administration & dosage , Animals , Body Weight/drug effects , Dose-Response Relationship, Drug , Female , Flutamide/administration & dosage , Follicle Stimulating Hormone/blood , Genitalia, Male/metabolism , Luteinizing Hormone/blood , Male , No-Observed-Adverse-Effect Level , Organ Size/drug effects , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Reproduction/drug effects , Testosterone/blood , Toxicogenetics
12.
Crit Rev Oncog ; 22(3-4): 249-253, 2017.
Article in English | MEDLINE | ID: mdl-29604901

ABSTRACT

Metastasis to distant organs and not the primary tumor itself is usually the cause of death for cancer patients. Hence, studying the key molecules and molecular pathways involved in metastasis are essential. Metastasis is a complex process in which cancer cells detach from the original tumor, migrate, and invade through surrounding tissues and metastasize to other sites of the body through the circulation. The cell-extracellular matrix (ECM) adhesion proteins play a fundamental role in this process as cancer cells need to weaken their adhesions to dissociate from the ECM as well as the neighboring cells within the tumor and finally form new adhesions and invade surrounding tissues. Ras suppressor-1 (RSU-1) was originally identified as a suppressor of Ras-dependent oncogenic transformation and found to be localized to cell-ECM adhesions where it binds to PINCH-1, a focal adhesion involved in cell survival. Although RSU-1 was connected to cancer early on, little is known about its expression in various cancer types or its role in metastasis. In this article, we review the recent literature regarding the expression of RSU-1 in various cancer types and its potential role in metastasis, discussing interesting findings and issues that still need to be addressed.


Subject(s)
Biomarkers, Tumor/biosynthesis , Neoplasms/metabolism , Transcription Factors/biosynthesis , Animals , Biomarkers, Tumor/genetics , Humans , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasms/genetics , Neoplasms/pathology , Transcription Factors/genetics
13.
Clin Exp Metastasis ; 32(3): 255-65, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25647720

ABSTRACT

Metastasis, responsible for most deaths from breast cancer (BC), is a multistep process leading to cancer cell spread. Extracellular matrix (ECM)-related adhesion and apoptosis resistance play pivotal role in metastasis. Ras suppressor-1 (RSU-1) localizes to cell-ECM adhesions and binds to pro-survival adhesion protein PINCH-1. Little is known about the role of RSU-1 in BC. In the present study, we investigated the role of RSU-1 in BC metastasis using two BC cell lines that differ in terms of their metastatic potential and a set of 32 human BC samples from patients with or without lymph node metastasis. We show that RSU-1 is upregulated in the aggressive MDA-MB-231 cells compared to MCF-7 and that its silencing by siRNA leads to upregulation of PINCH-1, induction of proliferation and reduction of apoptosis through downregulation of the pro-apoptotic gene p53-upregulated-modulator-of-apoptosis (PUMA). Our findings in the cell lines were further validated in the human BC tissues where normal adjacent tissues were used as controls. We demonstrate for the first time, that RSU-1 expression is upregulated in metastatic BC samples and downregulated in non-metastatic while it is negatively correlated with PINCH-1 and positively correlated with PUMA expression, suggesting that a pro-apoptotic mechanism is in place in metastatic BC samples and identifying RSU-1 as a potentially interesting molecule that needs to be evaluated further as a novel BC metastasis biomarker.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenocarcinoma, Mucinous/secondary , Apoptosis Regulatory Proteins/metabolism , Apoptosis , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/secondary , Carcinoma, Lobular/secondary , LIM Domain Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Mucinous/metabolism , Adult , Aged , Aged, 80 and over , Apoptosis Regulatory Proteins/genetics , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Lobular/genetics , Carcinoma, Lobular/metabolism , Cell Proliferation , Female , Humans , LIM Domain Proteins/genetics , Lymphatic Metastasis , Membrane Proteins/genetics , Membrane Proteins/metabolism , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prognosis , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Tumor Cells, Cultured
14.
Anticancer Res ; 33(5): 1977-81, 2013 May.
Article in English | MEDLINE | ID: mdl-23645746

ABSTRACT

BACKGROUND: Cell adhesion proteins that connect each cell to neighboring cells and the extracellular matrix play a fundamental role in metastasis. Mitogen-inducible gene-2 (MIG2), is a cell-matrix adhesion protein, which through migfilin, interacts with filamin-A, being linked to actin cytoskeleton. AIM: Recent studies have implicated both MIG2 and migfilin in cancer, but little is known regarding their expression in breast cancer. In this study, we investigated this topic. MATERIALS AND METHODS: mRNA and protein expression was examined in 30 breast cancer samples and compared to that of normal adjacent tissue using real time-polymerase chain reaction (PCR) and western blotting. RESULTS: Our results showed that expression of MIG2 and migfilin was significantly reduced in the majority of the breast cancer tissues compared to normal tissues regardless of metastatic status and disease stage. CONCLUSION: Both MIG2 and migfilin are down-regulated in breast cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast/metabolism , Cell Adhesion Molecules/metabolism , Cytoskeletal Proteins/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Biomarkers, Tumor/genetics , Blotting, Western , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/secondary , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/metabolism , Carcinoma, Intraductal, Noninfiltrating/secondary , Carcinoma, Lobular/genetics , Carcinoma, Lobular/metabolism , Carcinoma, Lobular/secondary , Cell Adhesion Molecules/genetics , Cytoskeletal Proteins/genetics , Female , Humans , Membrane Proteins/genetics , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
15.
Transplantation ; 94(9): 894-902, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23073466

ABSTRACT

BACKGROUND: Liver ischemia-reperfusion injury (IRI) is a known risk factor for the postoperative outcome of patients undergoing liver surgery/transplantation. Attempts to protect from organ damage require multidisciplinary strategies and are of emerging interest in view of patients with higher age and American Society of Anesthesiology status. Ischemic preconditioning has been successfully applied to prevent from IRI during liver resection/transplantation. Because even short periods of ischemia during preconditioning inevitably lead to hypoxia and formation of anti-inflammatory/cytoprotective acting adenosine, we reasoned that short nonischemic hypoxia also protects against hepatic IRI. METHODS: Mice underwent hypoxic preconditioning (HPC) by breathing 10% oxygen for 10 min followed by 10 min of 21% oxygen before left liver lobe ischemia (45 min) and reperfusion (4 hr). The interactions of hypoxia→adenosine→adenosine receptors were tested by pharmacologic antagonism at adenosine receptor (AR) sites in wild-type mice and in mice with genetic deletions at the A1, A2A, A2B, and A3 ARs. Hepatocellular damage, inflammation, and metabolic effects were quantified by enzyme activities, cytokines, liver myeloperoxidase, blood adenosine, and tissue AMP, respectively. RESULTS: Hepatoprotection by HPC was significant in wild-type and A1, A2A, and A3 AR knockout mice as quantified by lower alanine aminotransferase serum activities, cytokine levels, histologic damage scores, tissue myeloperoxidase concentrations, and preserved AMP concentrations. Protection by HPC was blunted in mice pretreated with the A2B AR antagonist MRS1754 or in A2B AR knockout mice. CONCLUSIONS: Because liver protective effects of HPC are negated when the A2B receptor is nonfunctional, the hypoxia→adenosine→A2B receptor pathway plays a critical role in the prevention of warm IRI in vivo. Hypoxic activation of this pathway warrants use of selective A2B AR agonists or even intermittent hypoxia (e.g., in deceased organ donors) to protect from liver IRI.


Subject(s)
Hypoxia/physiopathology , Ischemic Preconditioning , Liver/blood supply , Receptor, Adenosine A2B/physiology , Reperfusion Injury/prevention & control , Warm Ischemia , Acetamides/pharmacology , Adenosine/physiology , Animals , Hepatocytes/pathology , Hepatocytes/physiology , Liver/pathology , Liver/physiopathology , Liver Transplantation/pathology , Liver Transplantation/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Purines/pharmacology , Receptor, Adenosine A2B/deficiency , Receptor, Adenosine A2B/drug effects , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Signal Transduction/physiology
16.
J Clin Endocrinol Metab ; 95(9): E9-17, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20534756

ABSTRACT

CONTEXT: Antimitogenic effects of estradiol on vascular smooth muscle cells (VSMCs) may be cardioprotective, and these effects are mediated by estrogen receptor-alpha-dependent and -independent mechanisms, with the latter involving the conversion of estradiol to 2-hydroxyestradiol/2-methoxyestradiol by CYP450. Because resveratrol inhibits CYP450 and is an estrogen-receptor-alpha antagonist, resveratrol may abrogate the antimitogenic effects of estradiol. OBJECTIVE: The objective of the study was to examine the interaction of pharmacologically relevant concentrations of resveratrol with estradiol, 2-hydroxyestradiol, and 2-methoxyestradiol in human female coronary artery VSMCs. METHODS AND RESULTS: In human female coronary VSMCs, resveratrol (0.1-10 microm) alone did not influence serum-induced DNA or collagen synthesis or cell proliferation or migration; however, resveratrol abrogated the inhibitory effects of estradiol, but not 2-hydroxyestradiol or 2-methoxyestradiol, on these responses. Resveratrol also abrogated the inhibitory effects of estradiol on positive growth regulators (cyclin A, cyclin D, MAPK phosphorylation) and the stimulatory effects of estradiol on negative growth regulators (p21, p27). In microsomes and cells, dietarily relevant levels of resveratrol (0.001-1 microm) inhibited the metabolism of estradiol to 2-hydroxestradiol/2-methoxyestradiol. Propylpyrazoletriol (estrogen receptor-alpha agonist, 100 nmol/liter), but not diarylpropionitrile (estrogen receptor-beta agonist, 10 nmol/liter), inhibited VSMC mitogenesis, and this effect was blocked by resveratrol (5 micromol/liter). Higher concentrations (>25-50 microm) of resveratrol, never attainable in vivo, inhibited VSMC growth, an effect blocked by GW9662 (peroxisomal proliferator-activated receptor-gamma antagonist). CONCLUSION: In conclusion, dietarily relevant levels of resveratrol abrogate the antimitogenic effects of estradiol by inhibiting CYP450-mediated estradiol metabolism and blocking estrogen receptor-alpha.


Subject(s)
Coronary Vessels/drug effects , Estradiol/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Stilbenes/pharmacology , Antimitotic Agents/antagonists & inhibitors , Cell Proliferation/drug effects , Cells, Cultured , Coronary Vessels/growth & development , Coronary Vessels/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Hormone Antagonists/pharmacology , Humans , Muscle, Smooth, Vascular/growth & development , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/physiology , Osmolar Concentration , Resveratrol , Sex Factors , Vitis/chemistry , Wine
17.
Hypertension ; 51(3): 719-26, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18250367

ABSTRACT

Alpha(2)-adrenoceptors potentiate renal vascular responses to angiotensin II via coincident signaling at phospholipase C. This leads to increased activation of the phospholipase C/protein kinase C/c-src pathway. Studies suggest that c-src activates the reduced nicotinamide-adenine dinucleotide phosphate (NADPH) oxidase/superoxide system, and reactive oxygen species stimulate the RhoA/Rho kinase pathway. Therefore, we hypothesized that NADPH oxidase/superoxide and RhoA/Rho kinase are downstream components of the signal transduction pathway that mediate the interaction between alpha(2)-adrenoceptors and angiotensin II on renal vascular resistance. In rat kidneys, both in vivo and in vitro, intrarenal infusions of angiotensin II increased renal vascular resistance, and UK14,304 (alpha(2)-adrenoceptor agonist) enhanced this response. Intrarenal Tempol (superoxide dismutase mimetic) or Y27632 (Rho kinase inhibitor) abolished the interaction between UK14,304 and angiotensin II both in vivo and in vitro. The interaction was also blocked by inhibitors of NADPH oxidase (in vivo using chronic gp91ds-tat administration and in vitro with diphenyleneiodonium). In cultured preglomerular vascular smooth muscle cells, UK14,304 enhanced angiotensin II-induced intracellular superoxide (2-hydroxyethidium production) and potentiated activation of RhoA (Western blot of activated RhoA bound to the binding domain of rhotekin). The interaction between angiotensin II and UK14,304 on superoxide generation and RhoA activation was blocked by inhibitors of phospholipase C (U73312), protein kinase C (GF109203X), c-src (PP1), NADPH oxidase (diphenyleneiodonium), or superoxide (Tempol). We conclude that NADPH oxidase/superoxide and RhoA/Rho kinase are involved in the interaction between alpha(2)-adrenoceptors and angiotensin II on renal vascular resistance by mediating signaling events downstream of the phospholipase C/protein kinase C/c-src pathway.


Subject(s)
Angiotensin II/pharmacology , Kidney/blood supply , NADPH Oxidases/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Vasoconstriction/drug effects , rhoA GTP-Binding Protein/metabolism , Adrenergic alpha-Agonists/pharmacology , Animals , CSK Tyrosine-Protein Kinase , Cells, Cultured , Enzyme Inhibitors/pharmacology , Hypertension/metabolism , Hypertension/physiopathology , Kidney/metabolism , Male , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/metabolism , Rats , Rats, Inbred SHR , Receptors, Adrenergic, alpha-2/drug effects , Signal Transduction/physiology , Superoxides/metabolism , Type C Phospholipases/metabolism , Vasoconstriction/physiology , rho-Associated Kinases/metabolism , src-Family Kinases
18.
Hypertension ; 51(4): 1197-202, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18259021

ABSTRACT

Sequential conversion of estradiol (E) to 2/4-hydroxyestradiols and 2-/4-methoxyestradiols (MEs) by CYP450s and catechol-O-methyltransferase, respectively, contributes to the inhibitory effects of E on smooth muscle cells (SMCs) via estrogen receptor-independent mechanisms. Because medroxyprogesterone (MPA) is a substrate for CYP450s, we hypothesized that MPA may abrogate the inhibitory effects of E by competing for CYP450s and inhibiting the formation of 2/4-hydroxyestradiols and MEs. To test this hypothesis, we investigated the effects of E on SMC number, DNA and collagen synthesis, and migration in the presence and absence of MPA. The inhibitory effects of E on cell number, DNA synthesis, collagen synthesis, and SMC migration were significantly abrogated by MPA. For example, E (0.1micromol/L) reduced cell number to 51+/-3.6% of control, and this inhibitory effect was attenuated to 87.5+/-2.9% by MPA (10 nmol/L). Treatment with MPA alone did not alter any SMC parameters, and the abrogatory effects of MPA were not blocked by RU486 (progesterone-receptor antagonist), nor did treatment of SMCs with MPA influence the expression of estrogen receptor-alpha or estrogen receptor-beta. In SMCs and microsomal preparations, MPA inhibited the sequential conversion of E to 2-2/4-hydroxyestradiol and 2-ME. Moreover, as compared with microsomes treated with E alone, 2-ME formation was inhibited when SMCs were incubated with microsomal extracts incubated with E plus MPA. Our findings suggest that the inhibitory actions of MPA on the metabolism of E to 2/4-hydroxyestradiols and MEs may negate the cardiovascular protective actions of estradiol in postmenopausal women receiving estradiol therapy combined with administration of MPA.


Subject(s)
Contraceptives, Oral, Synthetic/pharmacology , Estradiol/pharmacology , Estrogens/pharmacokinetics , Medroxyprogesterone/pharmacology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Aorta/cytology , Cell Movement/drug effects , Cells, Cultured , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Estradiol/analogs & derivatives , Estradiol/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Female , Humans , Microsomes/drug effects , Microsomes/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Platelet-Derived Growth Factor/pharmacology
19.
Circulation ; 115(17): 2307-15, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17438146

ABSTRACT

BACKGROUND: It is well known that adenosine levels are increased during ischemia and protect the heart during ischemia/reperfusion. However, less is known about the role of adenosine-adenosine receptor (AR) pathways in hearts with left ventricular dilation and dysfunction. Therefore, we assessed adenosine levels and selective AR expression in transgenic mice with left ventricular systolic dysfunction secondary to overexpression of tumor necrosis factor-alpha (TNF 1.6). METHODS AND RESULTS: Cardiac adenosine levels were reduced by 70% at 3 and 6 weeks of age in TNF 1.6 mice. This change was accompanied by a 4-fold increase in the levels of A1-AR and a 50% reduction in the levels of A2A-AR. That the increase in A1-AR density was of physiological significance was shown by the fact that chronotropic responsiveness to the A1-AR selective agonist 2-chloro-N6-cyclopentanyladenosine was enhanced in the TNF 1.6 mice. Similar changes in adenosine levels were found in 2 other models of heart failure, mice overexpressing calsequestrin and mice after chronic pressure overload, suggesting that the changes in adenosine-AR signaling were secondary to myocardial dysfunction rather than to TNF overexpression. CONCLUSIONS: Cardiac dysfunction secondary to the overexpression of TNF is associated with marked alterations in myocardial levels of adenosine and ARs. Modulation of the myocardial adenosine system and its signaling pathways may be a novel therapeutic target in patients with heart failure.


Subject(s)
Adenosine/metabolism , Myocardium/metabolism , Receptor, Adenosine A1/genetics , Receptor, Adenosine A1/metabolism , Ventricular Dysfunction, Left/physiopathology , Adenosine Diphosphate/metabolism , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Disease Models, Animal , Female , Heart Failure/metabolism , Heart Failure/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/metabolism , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/genetics , Up-Regulation/physiology , Ventricular Dysfunction, Left/metabolism
20.
J Pharmacol Exp Ther ; 321(2): 799-809, 2007 May.
Article in English | MEDLINE | ID: mdl-17314200

ABSTRACT

Stimulation of adenylyl cyclase causes cellular efflux of cAMP, and cAMP (unlike adenosine) is stable in blood. Therefore, it is conceivable that cAMP could function as a circulating adenosine prohormone by local target-organ conversion of distally released cAMP to adenosine via the sequential actions of ectophosphodiesterase and ecto-5'-nucleotidase (cAMP==> AMP==> adenosine; called the cAMP-adenosine pathway). A possible specific representation of this general concept is the pancreatohepatorenal cAMP-adenosine mechanism. The pancreas secretes glucagon into the portal circulation, and glucagon is a stimulant of hepatic adenylyl cyclase. Therefore, we hypothesize that the pancreas, via glucagon, stimulates hepatic cAMP production, which provides circulating cAMP for conversion to adenosine in the kidney via the cAMP-adenosine pathway. In normal rats, intravenous cAMP increased urinary and renal interstitial (assessed by renal microdialysis) cAMP and adenosine. Intraportal infusions of glucagon increased plasma cAMP 10-fold, it did not affect plasma adenosine, and it increased urinary and renal interstitial cAMP and adenosine. Local renal interstitial blockade (by adding inhibitors directly to the microdialysis perfusate) of ectophosphodiesterase (using 3-isobutyl-1-methylxanthine or 1,3-dipropyl-8-p-sulfophenylxanthine) or ecto-5'-nucleotidase (using alpha,beta-methyleneadenosine-5'-diphosphate) prevented the cAMP-induced and glucagon-induced increases in renal interstitial adenosine, but not cAMP. In ZSF1 rats with the metabolic syndrome, an oral glucose load increased plasma glucagon and urinary cAMP and adenosine excretion. We conclude that circulating cAMP is a substrate for local conversion to adenosine via the cAMP-adenosine pathway. A specific manifestation of this is the pancreatohepatorenal cAMP-adenosine mechanism (pancreas==> portal glucagon==> liver==> circulating cAMP==> kidney==> local cAMP-adenosine pathway).


Subject(s)
Adenosine/urine , Cyclic AMP/metabolism , Kidney/metabolism , Liver/metabolism , Pancreas/physiology , Adenosine Monophosphate/urine , Animals , Cyclic AMP/administration & dosage , Glucagon/pharmacology , Glucose/pharmacology , Inosine/urine , Male , Microdialysis , Probenecid/pharmacology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...