Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Prostate ; 78(4): 266-277, 2018 03.
Article in English | MEDLINE | ID: mdl-29243324

ABSTRACT

BACKGROUND: Whereas the androgen receptor (AR) signaling axis remains a therapeutic target in castration-resistant prostate cancer (CRPC), the emergence of AR mutations and splice variants as mechanisms underlying resistance to contemporary inhibitors of this pathway highlights the need for new therapeutic approaches to target this disease. Of significance in this regard is the considerable preclinical data, indicating that histone deacetylase (HDAC) inhibitors may have utility in the treatment of CRPC. However, the results of clinical studies using HDAC inhibitors (directed against HDAC1, 2, 3, and 8) in CRPC are equivocal, a result that some have attributed to their ability to induce an epithelial to mesenchymal transition (EMT) and neuroendocrine differentiation. We posited that it might be possible to uncouple the beneficial effects of HDAC inhibitors on AR signaling from their undesired activities by targeting specific HDACs as opposed to using the pan-inhibitor strategy that has been employed to date. METHODS: The relative abilities of pan- and selective-Class I HDAC inhibitors to attenuate AR-mediated target gene expression and proliferation were assessed in several prostate cancer cell lines. Small interfering RNA (siRNA)-mediated knockdown approaches were used to confirm the importance of of HDAC 1, 2, and 3 expression in these processes. Further, the ability of each HDAC inhibitor to induce the expression of EMT markers (RNA and protein) and EMT-like phenotype(s) (migration) were also assessed. The anti-tumor efficacy of a HDAC3-selective inhibitor, RGFP966, was compared to the pan-HDAC inhibitor Suberoylanilide Hydroxamic Acid (SAHA) in the 22Rv1 xenograft model. RESULTS: Using genetic and pharmacological approaches we demonstrated that a useful inhibition of AR transcriptional activity, absent the induction of EMT, could be achieved by specifically inhibiting HDAC3. Significantly, we also determined that HDAC3 inhibitors blocked the activity of the constitutively active AR V7-splice variant and inhibited the growth of xenograft tumors expressing this protein. CONCLUSIONS: Our studies provide strong rationale for the near-term development of specific HDAC3 inhibitors for the treatment of CRPC.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/metabolism , Acrylamides/pharmacology , Animals , Blotting, Western , Cell Line, Tumor , Cell Migration Assays , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Phenylenediamines/pharmacology , Prostate/pathology , Prostatic Neoplasms, Castration-Resistant/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Androgen/drug effects , Signal Transduction/drug effects , Vorinostat/pharmacology , Xenograft Model Antitumor Assays
2.
J Clin Invest ; 127(6): 2326-2338, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28463227

ABSTRACT

The clinical utility of inhibiting cytochrome P450 17A1 (CYP17), a cytochrome p450 enzyme that is required for the production of androgens, has been exemplified by the approval of abiraterone for the treatment of castration-resistant prostate cancer (CRPC). Recently, however, it has been reported that CYP17 inhibitors can interact directly with the androgen receptor (AR). A phase I study recently reported that seviteronel, a CYP17 lyase-selective inhibitor, ædemonstrated a sustained reduction in prostate-specific antigen in a patient with CRPC, and another study showed seviteronel's direct effects on AR function. This suggested that seviteronel may have therapeutically relevant activities in addition to its ability to inhibit androgen production. Here, we have demonstrated that CYP17 inhibitors, with the exception of orteronel, can function as competitive AR antagonists. Conformational profiling revealed that the CYP17 inhibitor-bound AR adopted a conformation that resembled the unliganded AR (apo-AR), precluding nuclear localization and DNA binding. Further, we observed that seviteronel and abiraterone inhibited the growth of tumor xenografts expressing the clinically relevant mutation AR-F876L and that this activity could be attributed entirely to competitive AR antagonism. The results of this study suggest that the ability of CYP17 inhibitors to directly antagonize the AR may contribute to their clinical efficacy in CRPC.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Antineoplastic Agents, Hormonal/pharmacology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Active Transport, Cell Nucleus , Animals , Benzamides , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Drug Synergism , HEK293 Cells , Humans , Inhibitory Concentration 50 , Male , Metribolone/pharmacology , Mice, Inbred NOD , Mice, SCID , Nitriles , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Protein Binding , Receptors, Androgen/metabolism , Steroid 17-alpha-Hydroxylase/metabolism , Testosterone/pharmacology , Transcriptional Activation/drug effects , Xenograft Model Antitumor Assays
3.
Mol Cancer Res ; 15(6): 660-669, 2017 06.
Article in English | MEDLINE | ID: mdl-28209757

ABSTRACT

Resistance to second-generation androgen receptor (AR) antagonists and CYP17 inhibitors in patients with castration-resistant prostate cancer (CRPC) develops rapidly through reactivation of the androgen signaling axis and has been attributed to AR overexpression, production of constitutively active AR splice variants, or the selection for AR mutants with altered ligand-binding specificity. It has been established that androgens induce cell-cycle progression, in part, through upregulation of cyclin D1 (CCND1) expression and subsequent activation of cyclin-dependent kinases 4 and 6 (CDK4/6). Thus, the efficacy of the newly described CDK4/6 inhibitors (G1T28 and G1T38), docetaxel and enzalutamide, was evaluated as single agents in clinically relevant in vitro and in vivo models of hormone-sensitive and treatment-resistant prostate cancer. CDK4/6 inhibition (CDK4/6i) was as effective as docetaxel in animal models of treatment-resistant CRPC but exhibited significantly less toxicity. The in vivo effects were durable and importantly were observed in prostate cancer cells expressing wild-type AR, AR mutants, and those that have lost AR expression. CDK4/6i was also effective in prostate tumor models expressing the AR-V7 variant or the AR F876L mutation, both of which are associated with treatment resistance. Furthermore, CDK4/6i was effective in prostate cancer models where AR expression was lost. It is concluded that CDK4/6 inhibitors are a viable alternative to taxanes as therapeutic interventions in endocrine therapy-refractory CRPC.Implications: The preclinical efficacy of CDK4/6 monotherapy observed here suggests the need for near-term clinical studies of these agents in advanced prostate cancer. Mol Cancer Res; 15(6); 660-9. ©2017 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Prostatic Neoplasms, Castration-Resistant/drug therapy , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Humans , Male , Mice, Nude , Molecular Targeted Therapy/methods , Prostatic Neoplasms, Castration-Resistant/pathology , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Taxoids/pharmacology , Xenograft Model Antitumor Assays
4.
Nat Chem Biol ; 12(10): 795-801, 2016 10.
Article in English | MEDLINE | ID: mdl-27501397

ABSTRACT

Clinical resistance to the second-generation antiandrogen enzalutamide in castration-resistant prostate cancer (CRPC), despite persistent androgen receptor (AR) activity in tumors, highlights an unmet medical need for next-generation antagonists. We have identified and characterized tetra-aryl cyclobutanes (CBs) as a new class of competitive AR antagonists that exhibit a unique mechanism of action. These CBs are structurally distinct from current antiandrogens (hydroxyflutamide, bicalutamide, and enzalutamide) and inhibit AR-mediated gene expression, cell proliferation, and tumor growth in several models of CRPC. Conformational profiling revealed that CBs stabilize an AR conformation resembling an unliganded receptor. Using a variety of techniques, it was determined that the AR-CB complex was not recruited to AR-regulated promoters and, like apo AR, remains sequestered in the cytoplasm, bound to heat shock proteins. Thus, we have identified third-generation AR antagonists whose unique mechanism of action suggests that they may have therapeutic potential in CRPC.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Antineoplastic Agents/pharmacology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/metabolism , Androgen Receptor Antagonists/chemistry , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Male , Prostatic Neoplasms, Castration-Resistant/pathology , Structure-Activity Relationship
5.
Mol Cancer Res ; 12(12): 1829-39, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25100862

ABSTRACT

UNLABELLED: Tamoxifen, a selective estrogen receptor (ER) modulator (SERM), remains a frontline clinical therapy for patients with ERα-positive breast cancer. However, the relatively rapid development of resistance to this drug in the metastatic setting remains an impediment to a durable response. Although drug resistance likely arises by many different mechanisms, the consensus is that most of the implicated pathways facilitate the outgrowth of a subpopulation of cancer cells that can either recognize tamoxifen as an agonist or bypass the regulatory control of ERα. Notable in this regard is the observation here and in other studies that expression of anterior gradient homology 2 (AGR2), a known proto-oncogene and disulfide isomerase, was induced by both estrogen (17ß-estradiol, E2) and 4-hydroxytamoxifen (4OHT) in breast cancer cells. The importance of AGR2 expression is highlighted here by the observation that (i) its knockdown inhibited the growth of both tamoxifen-sensitive and -resistant breast cancer cells and (ii) its increased expression enhanced the growth of ERα-positive tumors in vivo and increased the migratory capacity of breast cancer cells in vitro. Interestingly, as with most ERα target genes, the expression of AGR2 in all breast cancer cells examined requires the transcription factor FOXA1. However, in tamoxifen-resistant cells, the expression of AGR2 occurs in a constitutive manner, requiring FOXA1, but loses its dependence on ER. Taken together, these data define the importance of AGR2 in breast cancer cell growth and highlight a mechanism where changes in FOXA1 activity obviate the need for ER in the regulation of this gene. IMPLICATIONS: These findings reveal the transcriptional interplay between FOXA1 and ERα in controlling AGR2 during the transition from therapy-sensitive to -resistant breast cancer and implicate AGR2 as a relevant therapeutic target.


Subject(s)
Breast Neoplasms/genetics , Drug Resistance, Neoplasm , Estradiol/pharmacology , Hepatocyte Nuclear Factor 3-alpha/genetics , Proteins/genetics , Tamoxifen/pharmacology , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Estrogen Receptor alpha/metabolism , Female , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , MCF-7 Cells , Mice , Mucoproteins , Oncogene Proteins , Proteins/metabolism , Proto-Oncogene Mas , Xenograft Model Antitumor Assays
6.
Shock ; 38(2): 128-36, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22683727

ABSTRACT

17ß-Estradiol (E2) treatment activates a set of protective response that has been found to protect cells from injury and more importantly to significantly abate the injuries associated with trauma-hemorrhage in vivo. Rapid NF-κB activation has been found to be an important signaling step in E2-mediated protection in cell culture, in vivo ischemia, and trauma-hemorrhage. In the current study, we investigated the signaling cascades linking E2 signaling with NF-κB activation and the protective response and compared them with the effects of two selective estrogen receptor modulators (SERMs), raloxifene and tamoxifen. Two candidate pathways, mitogen-activated protein kinases and phosphatidylinositol-3-kinase (PI3K) were studied. Selective inhibitors were used to identify each pathway's contribution to NF-κB activation. Treatment of human coronary artery endothelial cells with E2 activated PI3K/Akt, p38, and JNK, all of which activated ERK1/2 followed by NF-κB activation. The combined activation of Akt, p38, and JNK was essential to activate NF-κB. The two SERMs activated PI3K and p38, which then phosphorylated ERK1/2 and activated NF-κB independent of the JNK pathway. Nuclear factor κB activation by these compounds protected cells from hypoxia/reoxygenation injury. However, E2, unlike either SERM, led to modest increases in apoptosis through the JNK pathway. Selective estrogen receptor modulator treatment led to increased expression of the protective proteins, Mn superoxide dismutase, and endothelial nitric oxide synthase, which was not seen with E2. These results provide new insight into the pathways activating NF-κB by E2 and SERMs and demonstrate that SERMs may have greater protective benefits than E2 in adult endothelial cells and potentially in vivo, as well.


Subject(s)
Endothelium, Vascular/drug effects , Estradiol/pharmacology , MAP Kinase Signaling System/drug effects , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Adult , Cells, Cultured , Coronary Vessels/drug effects , Endothelium, Vascular/metabolism , Gene Knockdown Techniques , HSP27 Heat-Shock Proteins/metabolism , Humans , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinases/drug effects , Protein Kinase Inhibitors/pharmacology , Superoxide Dismutase/metabolism
7.
Endocrinology ; 152(4): 1589-98, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21303943

ABSTRACT

Heat shock proteins (HSPs) are a cardioprotective class of proteins induced by stress and regulated by the transcription factor, heat shock factor (HSF)-1. 17ß-estradiol (E(2)) indirectly regulates HSP expression through rapid activation of nuclear factor-κB (NF-κB) and HSF-1 and protects against hypoxia. As males experience a loss of protective cellular responses in aging, we hypothesized that aged menopausal (old ovariectomized) rats would have an impaired HSP response, which could be prevented by immediate in vivo E(2) replacement. After measuring cardiac function in vivo, cardiac myocytes were isolated from ovariectomized adult and old rats with and without 9 weeks of E(2) replacement. Myocytes were treated with E(2) in vitro and analyzed for activation of NF-κB, HSF-1, and HSP expression. In addition, we measured inflammatory cytokine expression and susceptibility to hypoxia/reoxygenation injury. Cardiac contractility was reduced in old ovariectomized rats and could prevented by immediate E(2) replacement in vivo. Subsequent investigations in isolated cardiac myocytes found that in vitro E(2) activated NF-κB, HSF-1, and increased HSP 72 expression in adult but not old rats. In response to hypoxia/reoxygenation, myocytes from adult, but not old, rats had increased HSP 72 expression. In addition, expression of the inflammatory cytokines TNF-α and IL-1ß, as well as oxidative stress, were increased in myocytes from old ovariectomized rats; only the change in cytokine expression could be attenuated by in vivo E(2) replacement. This study demonstrates that while aging in female rats led to a loss of the cardioprotective HSP response, E(2) retains its protective cellular properties.


Subject(s)
Aging/physiology , Estradiol/pharmacology , Heart/drug effects , Heart/physiopathology , Inflammation/physiopathology , Animals , Blotting, Western , Cells, Cultured , DNA-Binding Proteins/metabolism , Echocardiography , Female , Heart/physiology , Heat Shock Transcription Factors , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Ovariectomy , Polymerase Chain Reaction , Rats , Reactive Oxygen Species/metabolism , Transcription Factors/metabolism
8.
Mol Endocrinol ; 24(12): 2292-302, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20980435

ABSTRACT

Both pro- and antimitogenic activities have been ascribed to progesterone receptor (PR) agonists and antagonists in breast cancer cells; however, the transcriptional responses that underlie these paradoxical functions are not apparent. Using nontransformed, normal human mammary epithelial cells engineered to express PR and standard microarray technology, we defined 2370 genes that were significantly regulated by the PR agonist R5020. Gene ontology (GO) analysis revealed that GO terms involved in inflammation and nuclear factor-κB (NF-κB) signaling were among the most significantly regulated. Interestingly, on those NF-κB responsive genes that were inhibited by agonist-activated PR, antagonists either 1) mimicked the actions of agonists or 2) reversed the inhibitory actions of agonists. This difference in pharmacological response could be attributed to the fact that although agonist- and antagonist-activated PR is recruited to NF-κB-responsive promoters, the physical presence of PR tethered to the promoter of some genes is sufficient for transcriptional inhibition, whereas on others, an agonist-activated PR conformation is required for inhibition of NF-κB signaling. Importantly, the actions of PR on the latter class of genes were reversed by an activation function-2-inhibiting, LXXLL-containing peptide. Consideration of the relative activities of these distinct antiinflammatory pathways in breast cancer may be instructive with respect to the likely therapeutic activity of PR agonists or antagonists in the treatment of breast cancer.


Subject(s)
Inflammatory Breast Neoplasms/pathology , Receptors, Progesterone/physiology , Amino Acid Sequence , Cell Line , Cell Line, Tumor , Enhancer Elements, Genetic , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gonanes/pharmacology , Humans , Inflammatory Breast Neoplasms/genetics , Inflammatory Breast Neoplasms/metabolism , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Mifepristone/pharmacology , Molecular Sequence Data , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/metabolism , Progesterone/genetics , Progesterone/metabolism , Progestins/metabolism , Promoter Regions, Genetic , Protein Isoforms , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Progesterone/agonists , Receptors, Progesterone/antagonists & inhibitors , Signal Transduction , Transcriptional Activation , Translocation, Genetic
9.
Circ Res ; 105(12): 1186-95, 2009 Dec 04.
Article in English | MEDLINE | ID: mdl-19875724

ABSTRACT

RATIONALE: Previously, we have found that changes in the location of intracellular heat shock protein (HSP)60 are associated with apoptosis. HSP60 has been reported to be a ligand of toll-like receptor (TLR)-4. OBJECTIVE: We hypothesized that extracellular HSP60 (exHSP60) would mediate apoptosis via TLR4. METHODS AND RESULTS: Adult rat cardiac myocytes were treated with HSP60, either recombinant human or with HSP60 purified from the media of injured rat cardiac myocytes. ExHSP60 induced apoptosis in cardiac myocytes, as detected by increased caspase 3 activity and increased DNA fragmentation. Apoptosis could be reduced by blocking antibodies to TLR4 and by nuclear factor kappaB binding decoys, but not completely inhibited, even though similar treatment blocked lipopolysaccharide-induced apoptosis. Three distinct controls showed no evidence for involvement of a ligand other than exHSP60 in the mediation of apoptosis. CONCLUSIONS: This is the first report of HSP60-induced apoptosis via the TLRs. HSP60-mediated activation of TLR4 may be a mechanism of myocyte loss in heart failure, where HSP60 has been detected in the plasma.


Subject(s)
Apoptosis , Chaperonin 60/metabolism , Myocytes, Cardiac/metabolism , Signal Transduction , Toll-Like Receptor 4/metabolism , Animals , Antibodies , Caspase 3/metabolism , DNA Fragmentation , Endotoxins/metabolism , HSP27 Heat-Shock Proteins/metabolism , Humans , Interleukin-1beta/genetics , Interleukin-6/genetics , Ligands , Lipopolysaccharide Receptors/metabolism , Male , Myocytes, Cardiac/pathology , NF-kappa B/metabolism , Phosphorylation , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/immunology , Tumor Necrosis Factor-alpha/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Cardiovasc Res ; 84(1): 91-9, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19493956

ABSTRACT

AIMS: Mitochondrial fusion and fission are essential processes for preservation of normal mitochondrial function. We hypothesized that fusion proteins would be decreased in heart failure (HF), as the mitochondria in HF have been reported to be small and dysfunctional. METHODS AND RESULTS: Expression of optic atrophy 1 (OPA1), a mitochondrial fusion protein, was decreased in both human and rat HF, as observed by western blotting. OPA1 is important for maintaining normal cristae structure and function, for preserving the inner membrane structure and for protecting cells from apoptosis. Confocal and electron microscopy studies demonstrated that the mitochondria in the failing hearts were small and fragmented, consistent with decreased fusion. OPA1 mRNA levels did not differ between failing and normal hearts, suggesting post-transcriptional control. Simulated ischaemia in the cardiac myogenic cell line H9c2 cells reduced OPA protein levels. Reduction of OPA1 expression with shRNA resulted in increased apoptosis and fragmentation of the mitochondria. Overexpression of OPA1 increased mitochondrial tubularity, but did not protect against simulated ischaemia-induced apoptosis. Cytochrome c release from the mitochondria was increased both with reduction in OPA1 and with overexpression of OPA1. CONCLUSION: This is the first report, to our knowledge, of changes in mitochondrial fusion/fission proteins in cardiovascular disease. These changes have implications for mitochondrial function and apoptosis, contributing to the cell loss which is part of the downward progression of the failing heart.


Subject(s)
Apoptosis , GTP Phosphohydrolases/physiology , Heart Failure/metabolism , Mitochondria/physiology , Animals , Cell Line , Cyclosporine/pharmacology , Cytochromes c/metabolism , GTP Phosphohydrolases/analysis , GTP Phosphohydrolases/genetics , Heart Failure/pathology , Humans , Membrane Proteins/analysis , Membrane Transport Proteins/analysis , Mitochondrial Membrane Transport Proteins , Mitochondrial Proteins/analysis , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , RNA, Messenger/analysis , Rats
11.
Future Cardiol ; 5(1): 93-103, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19371207

ABSTRACT

Estrogen is a powerful hormone with pleiotropic effects. Estrogens have potent antioxidant effects and are able to reduce inflammation, induce vasorelaxation and alter gene expression in both the vasculature and the heart. Estrogen treatment of cultured cardiac myocytes and endothelial cells rapidly activates NFkappaB, induces heat-shock protein (HSP)-72, a potent intracellular protective protein, and protects cells from simulated ischemia. In in vivo models, estrogens protect against ischemia and trauma/hemorrhage. Estrogens may decrease the expression of soluble epoxide hydrolase, which has deleterious effects on the cardiovascular system through metabolism of epoxyeicosatrienoic acids. Natural (endogenous) estrogens in premenopausal women appear to protect against cardiovascular disease and yet controlled clinical trials have not indicated a benefit from estrogen replacement postmenopause. Much remains to be understood in regards to the many properties of this powerful hormone and how changes in this hormone interact with aging-associated changes. The unexpected negative results of trials of estrogen replacement postmenopause probably arise from our lack of understanding of the many effects of this hormone.


Subject(s)
Aging/physiology , Cardiovascular Diseases/physiopathology , Cardiovascular System , Estrogens/physiology , Animals , Antioxidants/pharmacology , Antioxidants/physiology , Cardiovascular System/drug effects , Estrogens/pharmacology , Female , Humans , Postmenopause/physiology , Rats
12.
Endocrinology ; 150(1): 212-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18787021

ABSTRACT

Although aging is known to lead to increased vascular stiffness, the role of estrogens in the prevention of age-related changes in the vasculature remains to be elucidated. To address this, we measured vascular function in the thoracic aorta in adult and old ovariectomized (ovx) rats with and without immediate 17beta-estradiol (E2) replacement. In addition, aortic mRNA and protein were analyzed for proteins known to be involved in vasorelaxation. Aging in combination with the loss of estrogens led to decreased vasorelaxation in response to acetylcholine and sodium nitroprusside, indicating either smooth muscle dysfunction and/or increased fibrosis. Loss of estrogens led to increased vascular tension in response to phenylephrine, which could be partially restored by E2 replacement. Levels of endothelial nitric oxide synthase and inducible nitric oxide synthase did not differ among the groups, nor did total nitrite plus nitrate levels. Old ovx exhibited decreased expression of both the alpha and beta-subunits of soluble guanylyl cyclase (sGC) and had impaired nitric oxide signaling in the vascular smooth muscle. Immediate E2 replacement in the aged ovx prevented both the impairment in vasorelaxation, and the decreased sGC receptor expression and abnormal sGC signaling within the vascular smooth muscle.


Subject(s)
Aging/physiology , Blood Vessels/physiology , Estrogens/deficiency , Isometric Contraction/physiology , Animals , Aorta/growth & development , Aorta/physiology , Blood Vessels/growth & development , Female , Muscle, Smooth, Vascular/growth & development , Muscle, Smooth, Vascular/physiology , Nitric Oxide/blood , Nitric Oxide/metabolism , Ovariectomy , Polymerase Chain Reaction , RNA/genetics , Rats , Rats, Inbred BN , Vasoconstriction/physiology , Vasodilation/physiology
13.
J Cardiovasc Pharmacol Ther ; 13(4): 261-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18682551

ABSTRACT

Endothelial dysfunction occurs in heart disease and may reduce functional capacity via attenuations in peripheral blood flow. Dietary decosahexaenoic acid (DHA) may improve this dysfunction, but the mechanism is unknown. This study determined if DHA enhances expression and activity of eNOS in cultured human coronary artery endothelial cells (HCAEC). HCAEC from 4 donors were treated with 5 nM, 50 nM, or 1 microM DHA for 7 days to model chronic DHA exposure. A trend for increased expression of endothelial nitric oxide synthase (eNOS) and phospho-eNOS was observed with 5 and 50 nM DHA. DHA also enhanced expression of 2 proteins instrumental in activation of eNOS: phospho-Akt (5 and 50 nM) and HSP90 (50 nM and 1 microM). Vascular endothelial growth factor-induced activation of Akt increased NOx in treated (50 nM DHA) versus untreated HCAEC (9.2 +/- 1.0 vs 3.3 +/- 1.1 micromol/microg protein/microL). Findings suggest that DHA enhances eNOS and Akt activity, augments HSP90 expression, and increases NO bioavailability in response to Akt kinase activation.


Subject(s)
Docosahexaenoic Acids/pharmacology , Endothelial Cells/drug effects , Nitric Oxide Synthase Type III/metabolism , Analysis of Variance , Blotting, Western , Cells, Cultured , Coronary Vessels/cytology , Cyclic GMP/metabolism , Dietary Supplements , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , HSP90 Heat-Shock Proteins/metabolism , Humans , Male , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Time Factors , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Vascular Endothelial Growth Factors/pharmacology
14.
Mol Med ; 14(7-8): 517-27, 2008.
Article in English | MEDLINE | ID: mdl-18431462

ABSTRACT

Estrogen has pleiotropic actions, among which are its anti-apoptotic, anti-inflammatory, and vasodilatory effects. Recently, an interaction between 17beta-estradiol (E2) and the transcription factor nuclear factor kappaB (NFkappaB) has been identified. NFkappaB has a central role in the control of genes involved in inflammation, proliferation, and apoptosis. Prolonged activation of NFkappaB is associated with numerous inflammatory pathological conditions. An important facet of E2 is its ability to modulate activity of NFkappaB via both genomic and nongenomic actions. E2 can activate NFkappaB rapidly via nongenomic pathways, increase cellular resistance to injury, and induce expression of the protective class of proteins, heat shock proteins (HSPs). HSPs can bind to many of the pro-apoptotic and pro-inflammatory targets of NFkappaB and, thus, indirectly inhibit many of its deleterious effects. In addition, HSPs can block NFkappaB activation and binding directly. Similarly, genomic E2 signaling can inhibit NFkappaB, but does so through alternative mechanisms. This review focuses on the molecular mechanisms of cross-talk between E2, NFkappaB, and HSPs, and the biological relevance of this cross-talk.


Subject(s)
Estrogens/physiology , Heat-Shock Response/physiology , NF-kappa B/physiology , Animals , Cardiovascular Diseases/etiology , Cytoprotection/drug effects , Cytoprotection/physiology , Estrogens/pharmacology , Heat-Shock Proteins/physiology , Humans , Models, Biological , Sex Characteristics , Stress, Physiological/physiopathology
15.
Circ Res ; 100(3): 381-90, 2007 Feb 16.
Article in English | MEDLINE | ID: mdl-17234968

ABSTRACT

High levels of triglyceride-rich lipoproteins (TGRLs) in blood are linked to development of atherosclerosis, yet the mechanisms by which these particles initiate inflammation of endothelium are unknown. TGRL isolated from human plasma during the postprandial state was examined for its capacity to bind to cultured human aortic endothelial cells (HAECs) and alter the acute inflammatory response to tumor necrosis factor-alpha. HAECs were repetitively incubated with dietary levels of freshly isolated TGRL for 2 hours per day for 1 to 3 days to mimic postprandial lipidemia. TGRL induced membrane upregulation of the low-density lipoprotein family receptors LRP and LR11, which was inhibited by the low-density lipoprotein receptor-associated protein-1. TGRLs alone did not elicit inflammation in HAECs but enhanced the inflammatory response via a 10-fold increase in sensitivity to cytokine stimulation. This was reflected by increased mitogen-activated protein kinase activation, nuclear translocation of NF-kappaB, amplified expression of endothelial selectin and VCAM-1, and a subsequent increase in monocyte-specific recruitment under shear flow as quantified in a microfabricated vascular mimetic device.


Subject(s)
Aortic Diseases/etiology , Arteriosclerosis/etiology , Arteritis/etiology , Dietary Fats/adverse effects , Endothelial Cells/drug effects , Hypertriglyceridemia/complications , LDL-Receptor Related Proteins/metabolism , Lipoproteins, HDL/toxicity , Lipoproteins, LDL/toxicity , Lipoproteins, VLDL/toxicity , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Membrane Transport Proteins/metabolism , Receptors, LDL/metabolism , Triglycerides/toxicity , Tumor Necrosis Factor-alpha/pharmacology , Aorta , Apolipoprotein C-III/metabolism , Apolipoprotein C-III/pharmacology , Cell Adhesion/drug effects , Cell Adhesion Molecules/metabolism , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Chylomicrons/blood , Dietary Fats/administration & dosage , E-Selectin/biosynthesis , E-Selectin/genetics , Endocytosis , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Fat Emulsions, Intravenous/pharmacology , Gene Expression Regulation/drug effects , Humans , Hypertriglyceridemia/blood , Hypoglycemia , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , LDL-Receptor Related Protein-Associated Protein/pharmacology , LDL-Receptor Related Proteins/drug effects , Leukocytes/cytology , Leukocytes/drug effects , Lipopolysaccharides/pharmacology , Lipoproteins, HDL/blood , Lipoproteins, LDL/blood , Lipoproteins, VLDL/blood , Low Density Lipoprotein Receptor-Related Protein-1/drug effects , Membrane Transport Proteins/drug effects , Models, Cardiovascular , Monocytes/cytology , Monocytes/drug effects , NF-kappa B/metabolism , Oxidative Stress , Receptors, LDL/drug effects , Rheology , Signal Transduction/drug effects , Triglycerides/blood , Tumor Necrosis Factor-alpha/physiology , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...