Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Environ Monit Assess ; 195(2): 298, 2023 Jan 13.
Article in English | MEDLINE | ID: mdl-36637512

ABSTRACT

Polychlorinated dibenzo-p-dioxins and dibenzofurans in the sediments of aquatic systems are a persistent global problem that poses serious health risks. Identifying the sources of dioxins in natural water systems and the extent of their contributions to observed sediment concentrations is important from a health advisory and mitigation perspective. This paper proposes novel distribution-based qualitative and quantitative methods as source apportionment techniques and alternatives to conventional source attribution methods. Using sampled data, air, runoff, industrial effluent, and industrial paper and pulp wastes were identified as four distinct dioxin contributors to concentrations found in the sediments of the test bed region: the Houston Ship Channel-San Jacinto River-Galveston Bay (HSC-SJR-GB) estuarine system that also includes 2 Superfund sites with dioxin contamination. Two qualitative methods, the Kullback-Leibler divergence (K-L divergence) and the Bhattacharya measure (BM), and a quantitative method, the L2 norm, were used to investigate the spatial and temporal sourcing patterns of dioxins in the system sediments. The results indicated a global contribution from air and runoff sources across the estuarine system and over time with more localized impacts of the Superfund sites and the industrial sources. The results using the developed methodologies were compared with the output from the more conventional positive matrix factorization (PMF) method. Statistically significant correlations were observed among source contributions from the proposed methods and the PMF method, with Spearman's ρ ranging between - 0.596 to - 0.963 and 0.652 to 0.719, demonstrating the utility of the sourcing approaches used in the study. Additionally, the proposed methods were found to be rigorous in terms of elucidating spatial and temporal changes in the sourcing of dioxin to the estuary, indicating their suitability for use for other contaminants and other estuarine systems.


Subject(s)
Dioxins , Polychlorinated Biphenyls , Polychlorinated Dibenzodioxins , Water Pollutants, Chemical , Polychlorinated Dibenzodioxins/analysis , Dioxins/analysis , Dibenzofurans , Estuaries , Environmental Monitoring , Geologic Sediments , Water Pollutants, Chemical/analysis , Dibenzofurans, Polychlorinated
2.
Entropy (Basel) ; 24(8)2022 Aug 20.
Article in English | MEDLINE | ID: mdl-36010826

ABSTRACT

The high energy transfer efficiency of photosynthetic complexes has been a topic of research across many disciplines. Several attempts have been made in order to explain this energy transfer enhancement in terms of quantum mechanical resources such as energetic and vibration coherence and constructive effects of environmental noise. The developments in this line of research have inspired various biomimetic works aiming to use the underlying mechanisms in biological light harvesting complexes for the improvement of synthetic systems. In this article, we explore the effect of an auxiliary hierarchically structured environment interacting with a system on the steady-state heat transport across the system. The cold and hot baths are modeled by a series of identically prepared qubits in their respective thermal states, and we use a collision model to simulate the open quantum dynamics of the system. We investigate the effects of system-environment, inter-environment couplings and coherence of the structured environment on the steady state heat flux and find that such a coupling enhances the energy transfer. Our calculations reveal that there exists a non-monotonic and non-trivial relationship between the steady-state heat flux and the mentioned parameters.

3.
FASEB J ; 30(1): 230-40, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26373802

ABSTRACT

Estrogen and estrogen receptor (ER)-α suppress visceral fat development through actions in several organs via unclear mechanisms that we sought to identify. Using mice that express only nuclear ER-α [nuclear-only ER-α (NOER) mice] or plasma membrane ER-α [membrane-only ER-α (MOER) mice], we found that 10-wk-old mice that lacked either receptor pool showed extensive abdominal visceral fat deposition and weight gain compared with wild-type (WT) mice. Differentiation of cultured bone marrow stem cells (BMSCs) into the adipocyte lineage was suppressed by 17-ß-estradiol (E2) in WT female mice but not in NOER or MOER mice. This finding correlated with E2 inhibition of prominent differentiation genes in WT BMSCs. In contrast, triglyceride content in differentiated BMSCs or 3T3-L1 cells was suppressed as a result of membrane ER-α signaling through several kinases to inhibit carbohydrate response element-binding protein-α and -ß. We concluded that extranuclear and nuclear ER-α collaborate to suppress adipocyte development, but inhibition of lipid synthesis in mature cells does not involve nuclear ER-α.


Subject(s)
Adipogenesis , Estrogen Receptor alpha/metabolism , Triglycerides/metabolism , Animals , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Female , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice
4.
Mol Biol Cell ; 17(5): 2125-37, 2006 May.
Article in English | MEDLINE | ID: mdl-16495339

ABSTRACT

Steroid hormones have been reported to indirectly impact mitochondrial functions, attributed to nuclear receptor-induced production of proteins that localize in this cytoplasmic organelle. Here we show high-affinity estrogen receptors in the mitochondria of MCF-7 breast cancer cells and endothelial cells, compatible with classical estrogen receptors ERalpha and ERbeta. We report that in MCF-7, estrogen inhibits UV radiation-induced cytochrome C release, the decrease of the mitochondrial membrane potential, and apoptotic cell death. UV stimulated the formation of mitochondrial reactive oxygen species (mROS), and mROS were essential to inducing mitochondrial events of cell death. mROS mediated the UV activation of c-jun N-terminal kinase (JNK), and protein kinase C (PKC) delta, underlying the subsequent translocation of Bax to the mitochondria where oligomerization was promoted. E2 (estradiol) inhibited all these events, directly acting in mitochondria to inhibit mROS by rapidly up-regulating manganese superoxide dismutase activity. We implicate novel functions of ER in the mitochondria of breast cancer that lead to the survival of the tumor cells.


Subject(s)
Breast Neoplasms/metabolism , Mitochondria/chemistry , Mitochondria/metabolism , Receptors, Estrogen/analysis , Receptors, Estrogen/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Cytochromes c/metabolism , Estradiol/pharmacology , Estrogens/pharmacology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mitochondria/drug effects , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Receptors, Estrogen/antagonists & inhibitors , bcl-2-Associated X Protein/metabolism
5.
Endocrinology ; 149(7): 3361-9, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18372323

ABSTRACT

Estrogen has been reported to prevent development of cardiac hypertrophy in female rodent models and in humans. However, the mechanisms of sex steroid action are incompletely understood. We determined the cellular effects by which 17beta-estradiol (E2) inhibits angiotensin II (AngII)-induced cardiac hypertrophy in vivo. Two weeks of angiotensin infusion in female mice resulted in marked hypertrophy of the left ventricle, exacerbated by the loss of ovarian steroid hormones from oophorectomy. Hypertrophy was 51% reversed by the administration of E2 (insertion of 0.1 mg/21-d-release tablets). The effects of E2 were mainly mediated by the estrogen receptor (ER) beta-isoform, because E2 had little effect in ERbeta-null mice but comparably inhibited AngII-induced hypertrophy in wild-type or ERalpha-null mice. AngII induced a switch of myosin heavy chain production from alpha to beta, but this was inhibited by E2 via ERbeta. AngII-induced ERK activation was also inhibited by E2 through the beta-receptor. E2 stimulated brain natriuretic peptide protein expression and substantially prevented ventricular interstitial cardiac fibrosis (collagen deposition) as induced by AngII. Importantly, E2 inhibited calcineurin activity that was stimulated by AngII, related to E2 stimulating the modulatory calcineurin-interacting protein (MCIP) 1 gene and protein expression. E2 acting mainly through ERbeta mitigates the important signaling by AngII that produces cardiac hypertrophy and fibrosis in female mice.


Subject(s)
Calcineurin/metabolism , Cardiomegaly/drug therapy , Estrogen Receptor beta/physiology , Estrogens/pharmacology , Angiotensin II/pharmacology , Animals , Calcium-Binding Proteins , Cardiomegaly/chemically induced , Collagen/metabolism , Estradiol/pharmacology , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Female , Fibrosis , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myocardium/metabolism , Myocardium/pathology , Myosin Heavy Chains/metabolism , Ovariectomy
6.
Mol Cell Biol ; 23(5): 1633-46, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12588983

ABSTRACT

Estrogen receptors (ER) have been localized to the cell plasma membrane (PM), where signal transduction mediates some estradiol (E2) actions. However, the precise structural features of ER that result in membrane localization have not been determined. We obtained a partial tryptic peptide/mass spectrometry analysis of membrane mouse ERalpha protein. Based on this, we substituted alanine for the determined serine at amino acid 522 within the E domain of wild-type (wt) ERalpha. Upon transfection in CHO cells, the S522A mutant ERalpha resulted in a 62% decrease in membrane receptor number and reduced colocalization with caveolin 1 relative to those with expression of wt ERalpha. E2 was significantly less effective in stimulating multiple rapid signals from the membranes of CHO cells expressing ERalpha S522A than from those of CHO cells expressing wt ERalpha. In contrast, nuclear receptor expression and transcriptional function were very similar. The S522A mutant was also 60% less effective than wt ERalpha in binding caveolin 1, which facilitates ER transport to the PM. All functions of ERalpha mutants with other S-to-A substitutions were comparable to those of wt ER, and deletion of the A/B or C domain had little consequence for membrane localization or function. Transfection of ERalpha S522A into breast cancer cells that express native ER downregulated E2 binding at the membrane, signaling to ERK, and G1/S cell cycle events and progression. However, there was no effect on the E2 transactivation of an ERE-luciferase reporter. In summary, serine 522 is necessary for the efficient translocation and function of ERalpha at the PM. The S522A mutant also serves as a dominant-negative construct, identifying important functions of E2 that originate from activating PM ER.


Subject(s)
Cell Membrane/metabolism , Receptors, Estrogen/metabolism , Animals , Binding, Competitive , Breast Neoplasms/metabolism , CHO Cells , Caveolin 1 , Caveolins/metabolism , Cell Cycle , Cell Nucleus/metabolism , Cricetinae , Cyclin D1/metabolism , Dimerization , Dose-Response Relationship, Drug , Endoplasmic Reticulum/metabolism , Estrogen Receptor alpha , G1 Phase , Genes, Dominant , Kinetics , Mice , Microscopy, Fluorescence , Mitogen-Activated Protein Kinases/metabolism , Mutagenesis, Site-Directed , Mutation , Myristic Acid/metabolism , Palmitic Acid/metabolism , Protein Binding , Protein Structure, Tertiary , S Phase , Signal Transduction , Transcription, Genetic , Transcriptional Activation , Transfection , Tumor Cells, Cultured , Type C Phospholipases/metabolism
7.
Mol Cell Biol ; 24(13): 5900-13, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15199145

ABSTRACT

BRCA1 mutations and estrogen use are risk factors for the development of breast cancer. Recent work has identified estrogen receptors localized at the plasma membrane that signal to cell biology. We examined the impact of BRCA1 on membrane estrogen and growth factor receptor signaling to breast cancer cell proliferation. MCF-7 and ZR-75-1 cells showed a rapid and sustained activation of extracellular signal-related kinase (ERK) in response to estradiol (E2) that was substantially prevented by wild-type (wt) but not mutant BRCA1. The proliferation of MCF-7 cells induced by E2 was significantly inhibited by PD98059, a specific ERK inhibitor, or by dominant negative ERK2 expression and by expression of wt BRCA1 (but not mutant BRCA1). E2 induced the synthesis of cyclins D1 and B1, the activity of cyclin-dependent kinases Cdk4 and CDK1, and G(1)/S and G(2)/M cell cycle progression. The intact tumor suppressor inhibited all of these. wt BRCA1 also inhibited epidermal growth factor and insulin-like growth factor I-induced ERK and cell proliferation. The inhibition of ERK and cell proliferation by BRCA1 was prevented by phosphatase inhibitors and by interfering RNA knockdown of the ERK phosphatase, mitogen-activated kinase phosphatase 1. Our findings support a novel tumor suppressor function of BRCA1 that is relevant to breast cancer and identify a potential interactive risk factor for women with BRCA1 mutations.


Subject(s)
BRCA1 Protein/physiology , Breast Neoplasms/etiology , Receptors, Estrogen/antagonists & inhibitors , Receptors, Growth Factor/antagonists & inhibitors , BRCA1 Protein/genetics , Breast Neoplasms/pathology , Cell Division , Cell Line, Tumor , Estradiol/pharmacology , Humans , Membrane Proteins , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Mutation/physiology , Receptors, Estrogen/physiology , Receptors, Growth Factor/physiology , Signal Transduction , Transfection
8.
Mol Endocrinol ; 20(9): 1996-2009, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16645038

ABSTRACT

Although rapid signaling by estrogen at the plasma membrane is established, it is controversial as to the nature of the receptor protein. Estrogen may bind membrane proteins comparable to classical nuclear estrogen receptors (ERs), but some studies identify nonclassical receptors, such as G protein-coupled receptor (GPR)30. We took several approaches to define membrane-localized estrogen-binding proteins. In endothelial cells (ECs) from ERalpha/ERbeta combined-deleted mice, estradiol (E2) failed to specifically bind, and did not activate cAMP, ERK, or phosphatidyinositol 3-kinase or stimulate DNA synthesis. This is in contrast to wild-type ECs, indicating the lack of any functional estrogen-binding proteins in ERalpha/ERbeta combined-deleted ECs. To directly determine the identity of membrane and nuclear-localized ER, we isolated subcellular receptor pools from MCF7 cells. Putative ER proteins were trypsin digested and subjected to tandem array mass spectrometry. The output analysis identified membrane and nuclear E2-binding proteins as classical human ERalpha. We also determined whether GPR30 plays any role in E2 rapid actions. MCF7 (ER and GPR30 positive) and SKBR-3 (ER negative, GPR30 positive) cells were incubated with E2. Only MCF7 responded with significantly increased signaling. In MCF7, the response to E2 was not different in cells transfected with small interfering RNA to green fluorescent protein or GPR30. In contrast, interfering RNA to ERalpha or ER inhibition prevented rapid signaling and resulting biology in MCF7. In breast cancer and ECs, nuclear and membrane ERs are the same proteins. Furthermore, classical ERs mediate rapid signals induced by E2 in these cells.


Subject(s)
Cell Membrane/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Amino Acid Sequence , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Membrane/drug effects , Cells, Cultured , Estradiol/pharmacology , Estrogen Receptor alpha/deficiency , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/isolation & purification , Estrogen Receptor beta/deficiency , Estrogen Receptor beta/genetics , Estrogen Receptor beta/isolation & purification , Estrogens/pharmacology , Humans , Mass Spectrometry , Mice , Mice, Knockout , Molecular Sequence Data , RNA, Small Interfering/genetics , Signal Transduction/drug effects
9.
PLoS One ; 12(5): e0178723, 2017.
Article in English | MEDLINE | ID: mdl-28542512

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0174951.].

10.
PLoS One ; 12(4): e0174951, 2017.
Article in English | MEDLINE | ID: mdl-28384250

ABSTRACT

Due to the rise in awareness of environmental issues and the depletion of virgin resources, many firms have attempted to increase the sustainability of their activities. One efficient way to elevate sustainability is the consideration of corporate social responsibility (CSR) by designing a closed loop supply chain (CLSC). This paper has developed a mathematical model to increase corporate social responsibility in terms of job creation. Moreover the model, in addition to increasing total CLSC profit, provides a range of strategic decision solutions for decision makers to select a best action plan for a CLSC. A proposed multi-objective mixed-integer linear programming (MILP) model was solved with non-dominated sorting genetic algorithm II (NSGA-II). Fuzzy set theory was employed to select the best compromise solution from the Pareto-optimal solutions. A numerical example was used to validate the potential application of the proposed model. The results highlight the effect of CSR in the design of CLSC.


Subject(s)
Models, Organizational , Social Responsibility , Fuzzy Logic
11.
Waste Manag ; 60: 460-470, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27406308

ABSTRACT

This paper attempts to integrate both a forward and reverse supply chain to design a closed-loop supply chain network (CLSC). The problem in the design of a CLSC network is uncertainty in demand, return products and the quality of return products. Scenario analyses are generated to overcome this uncertainty. In contrast to the existing supply chain network design models, a new application of a CLSC network was studied in this paper to reduce waste. A multi-product, multi-tier mixed integer linear model is developed for a CLSC network design. The main objective is to maximize profit and provide waste management decision support in order to minimize pollution. The result shows applicability of the model in the tire industry. The model determines the number and the locations of facilities and the material flows between these facilities.


Subject(s)
Models, Theoretical , Waste Management , Costs and Cost Analysis , Environment , Environmental Pollution/prevention & control , Industry , Iran , Programming, Linear , Rubber , Waste Products
12.
Mol Cell Endocrinol ; 434: 57-68, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27321970

ABSTRACT

Cardiac fibrosis evolves from the cardiac hypertrophic state. In this respect, estrogen and estrogen receptor beta (ERß) inhibit the effects of cardiac hypertrophic peptides that also stimulate fibrosis. Here we determine details of the anti-fibrotic functions of ERß. In acutely isolated rat cardiac fibroblasts. E2 or a specific ERß agonist (ßLGND2) blocked angiotensin II (AngII) signaling to fibrosis. This resulted from ERß activating protein kinase A and AMP kinase, inhibiting both AngII de-phosphorylation of RhoA and the resulting stimulation of Rho kinase. Inhibition of Rho kinase from ERß signaling resulted in marked decrease of TGFß expression, connective tissue growth factor production and function, matrix metalloproteinases 2 and 9 expression and activity, and the conversion of fibroblasts to myofibroblasts. Production of collagens I and III were also significantly decreased. Several important aspects were corroborated in-vivo from ßLGND2-treated mice that underwent AngII-induced cardiac hypertrophy. Thus, ERß in cardiac fibroblasts prevents key aspects of cardiac fibrosis development.


Subject(s)
Angiotensin II/metabolism , Cardiomegaly/metabolism , Estrogen Receptor beta/metabolism , Myofibroblasts/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Cardiomegaly/pathology , Cells, Cultured , Collagen Type I/metabolism , Collagen Type III/metabolism , Disease Models, Animal , Estrogen Receptor beta/agonists , Fibrosis , Male , Mice , Myofibroblasts/drug effects , Myofibroblasts/pathology , Rats , Signal Transduction/drug effects
13.
Mol Endocrinol ; 16(1): 100-15, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11773442

ABSTRACT

Recent evidence supports the existence of a plasma membrane ER. In many cells, E2 activates signal transduction and cell proliferation, but the steroid inhibits signaling and growth in other cells. These effects may be related to interactions of ER with signal-modulating proteins in the membrane. It is also unclear how ER moves to the membrane. Here, we demonstrate ER in purified vesicles from endothelial cell plasma membranes and colocalization of ERalpha with the caveolae structural coat protein, caveolin-1. In human vascular smooth muscle or MCF-7 (human breast cancer) cell membranes, coimmunoprecipitation shows that ER associates with caveolin-1 and -2. Importantly, E2 rapidly and differentially stimulates ER-caveolin association in vascular smooth muscle cells but inhibits association in MCF-7 cells. E2 also stimulates caveolin-1 and -2 protein synthesis and activates a caveolin-1 promoter/luciferase reporter in smooth muscle cells. However, the steroid inhibits caveolin synthesis in MCF-7 cells. To determine a function for caveolin-ER interaction, we expressed caveolin-1 in MCF-7 cells. This stimulated ER translocation to the plasma membrane and also inhibited E2-induced ERK (MAPK) activation. Both functions required the caveolin-1 scaffolding domain. Depending upon the target cell, membrane ERs differentially associate with caveolin, and E2 differentially modulates the synthesis of this signaling-inhibitory scaffold protein. This may explain the discordant signaling and actions of E2 in various cell types. In addition, caveolin-1 is capable of facilitating ER translocation to the membrane.


Subject(s)
Caveolae/metabolism , Estradiol/metabolism , Receptors, Estrogen/metabolism , Signal Transduction , Animals , Breast Neoplasms/metabolism , Caveolin 1 , Caveolins , Cell Membrane/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha , Humans , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Rats , Receptors, Estrogen/drug effects , Receptors, Estrogen/genetics , Tumor Cells, Cultured
14.
Mol Endocrinol ; 18(12): 2854-65, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15231873

ABSTRACT

A small pool of estrogen receptors (ERalpha and -beta) localize at the plasma membrane and rapidly signal to affect cellular physiology. Although nuclear ERs function mainly as homodimers, it is unknown whether membrane-localized ER exists or functions with similar requirements. We report that the endogenous ER isoforms at the plasma membrane of breast cancer or endothelial cells exist predominantly as homodimers in the presence of 17beta-estradiol (E2). Interestingly, in endothelial cells made from ERalpha /ERbeta homozygous double-knockout mice, membrane ERalpha or ERbeta are absent, indicating that the endogenous membrane receptors derive from the same gene(s) as the nuclear receptors. In ER-negative breast cancer cells or Chinese hamster ovary cells, we expressed and compared wild-type and dimer mutant mouse ERalpha. Only wild-type ERalpha supported the ability of E2 to rapidly activate ERK, cAMP, and phosphatidylinositol 3-kinase signaling. This resulted from E2 activating Gsalpha and Gqalpha at the membrane in cells expressing the wild-type, but not the dimer mutant, ERalpha. Intact, but not dimer mutant, ERalpha also supported E2-induced epidermal growth factor receptor transactivation and cell survival. We also confirmed the requirement of dimerization for membrane ER function using a second, less extensively mutated, human ERalpha. In summary, endogenous membrane ERs exist as dimers, a structural requirement that supports rapid signal transduction and affects cell physiology.


Subject(s)
Cell Membrane/physiology , Estrogen Receptor alpha/physiology , Signal Transduction , Animals , Apoptosis/drug effects , Breast Neoplasms/metabolism , CHO Cells , Cell Physiological Phenomena , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Dimerization , Endothelial Cells/metabolism , ErbB Receptors/metabolism , Estradiol/pharmacology , Estradiol/physiology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Humans , Mice , Mice, Knockout , Mutation/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation
15.
Dev Cell ; 29(4): 482-90, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24871949

ABSTRACT

Steroid receptors are found in discrete cellular locations, but it is unknown whether extranuclear pools are necessary for normal organ development. To assess this, we developed a point mutant estrogen receptor α (ERα) knockin mouse (C451A) that precludes palmitoylation and membrane trafficking of the steroid receptor in all organs. Homozygous knockin female mice (nuclear-only ERα [NOER]) show loss of rapid signaling that occurs from membrane ERα in wild-type mice. Multiple developmental abnormalities were found, including infertility, relatively hypoplastic uteri, abnormal ovaries, stunted mammary gland ductal development, and abnormal pituitary hormone regulation in NOER mice. These abnormalities were rescued in heterozygous NOER mice that were comparable to wild-type mice. mRNAs implicated in organ development were often poorly stimulated by estrogen only in homozygous NOER mice. We conclude that many organs require membrane ERα and resulting signal transduction to collaborate with nuclear ERα for normal development and function.


Subject(s)
Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Mammary Glands, Animal/embryology , Organogenesis , Ovary/embryology , Acylation , Animals , Cell Membrane/genetics , Cell Nucleus/genetics , Female , Lipoylation , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Ovulation/genetics , RNA, Messenger/genetics , Signal Transduction , Transcription, Genetic
16.
Biomaterials ; 34(38): 10228-37, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24055523

ABSTRACT

Drug resistance acquired by cancer cells is a significant challenge in the clinic and requires impairing the responsible pathological pathway. Administering chemotherapeutics along with silencing resistance-basis activity using RNA interference (RNAi) is expected to restore the activity of the chemotherapeutic and generate synergistic cancer eradication. This study attempted to reverse tamoxifen (TAM)-resistance in breast cancer by silencing a mitochondrial enzyme, manganese superoxide dismutase (MnSOD), which dismutates TAM-induced reactive oxygen species (ROS) (i.e., superoxide) to less harmful hydrogen peroxide and hampers therapeutic effects. Breast cancer cells were co-treated with TAM and MnSOD siRNA-delivering nanoparticles (NPs) made of a siRNA/poly(amidoamine) (PAMAM) dendriplex core and an acid-degradable polyketal (PK) shell. The (siRNA/PAMAM)-PK NPs were designed for the PK shell to shield siRNA from nucleases, minimize detrimental aggregation in serum, and facilitate cytosolic release of siRNA from endosomal compartments. This method of forming the PK shell around the siRNA/PAMAM core via surface-initiated photo-polymerization enables ease of tuning NPs' size for readily controlled siRNA release kinetics. The resulting NPs were notably homogenous in size, resistant to aggregation in serum, and invulnerable to heparan sulfate-mediated disassembly, compared to siRNA/PAMAM dendriplexes. Gel electrophoresis and confocal microscopy confirmed efficient siRNA release from the (siRNA/PAMAM)-PK NPs upon stimuli-responsive hydrolysis of the PK shell. Sensitization of TAM-resistant MCF7-BK-TR breast cancer cells with (MnSOD siRNA/PAMAM)-PK NPs restored TAM-induced cellular apoptosis in vitro and significantly suppressed tumor growth in vivo, as confirmed by biochemical assays and histological observations. This study implies that combined gene silencing and chemotherapy is a promising strategy to overcoming a significant challenge in cancer therapy.


Subject(s)
Acids/chemistry , Breast Neoplasms/drug therapy , Nanoparticles/chemistry , Superoxide Dismutase/metabolism , Tamoxifen/chemistry , Tamoxifen/therapeutic use , Animals , Apoptosis/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Gene Silencing/physiology , Humans , Mice , Mice, Nude , Microscopy, Confocal , Microscopy, Electron, Transmission , Superoxide Dismutase/genetics
17.
Sci Signal ; 6(276): ra36, 2013 May 21.
Article in English | MEDLINE | ID: mdl-23695162

ABSTRACT

Estrogen induces signal transduction through estrogen receptor α (ERα), which localizes to both the plasma membrane and nucleus. Using wild-type mice, ERα knockout (ERKO) mice, or transgenic mice expressing only the ligand-binding domain of ERα exclusively at the plasma membrane (MOER), we compared the transcriptional profiles of liver tissue extracts after mice were injected with the ERα agonist propyl-pyrazole-triol (PPT). The expression of many lipid synthesis-related genes was comparably decreased in livers from MOER or wild-type mice but was not suppressed in ERKO mice, indicating that only membrane-localized ERα was necessary for their suppression. Cholesterol, triglyceride, and fatty acid content was decreased only in livers from wild-type and MOER mice exposed to PPT, but not in the livers from the ERKO mice, validating the membrane-driven signaling pathway on a physiological level. PPT-triggered activation of ERα at the membrane induced adenosine monophosphate-activated protein kinase to phosphorylate sterol regulatory element-binding factor 1 (Srebf1), preventing its association with and therefore its proteolytic cleavage by site-1 protease. Consequently, Srebf1 was sequestered in the cytoplasm, preventing the expression of cholesterol synthesis-associated genes. Thus, we showed that inhibition of gene expression mediated by membrane-localized ERα caused a metabolic phenotype that did not require nuclear ERα.


Subject(s)
Estrogens/physiology , Lipids/analysis , Liver/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction , Adenylate Kinase/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Estrogen Receptor alpha/metabolism , Liver/cytology , Liver/enzymology , Mice , Mice, Knockout , Protein Binding , Sterol Regulatory Element Binding Protein 1/metabolism
18.
Endocrinology ; 154(11): 4352-64, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23970786

ABSTRACT

Cardiac hypertrophy in humans can progress to cardiac failure if the underlying impetus is poorly controlled. An important direct stimulator of hypertrophy and its progression is the angiotensin II (AngII) peptide. AngII also causes hypertension that indirectly contributes to cardiac hypertrophy. Others and we have shown that estrogens acting through the estrogen receptor (ER)-ß can inhibit AngII-induced or other forms of cardiac hypertrophy in mice. However, the proliferative effects of estrogen in breast and uterus that promote the development of malignancy preclude using the steroid to prevent cardiac disease progression. We therefore tested whether an ERß selective agonist, ß-LGND2, can prevent hypertension and cardiac pathology in female mice. AngII infusion over 3 weeks significantly stimulated systolic and diastolic hypertension, cardiac hypertrophy, and cardiac fibrosis, all significantly prevented by ß-LGND2 in wild-type but not in ERß genetically deleted mice. AngII stimulated the Akt kinase to phosphorylate and inhibit the glycogen synthase kinase-3ß kinase, leading to GATA4 transcription factor activation and hypertrophic mRNA expression. As a novel mechanism, all these actions were opposed by estradiol and ß-LGND2. Our findings provide additional understanding of the antihypertrophic effects of ERß and serve as an impetus to test specific receptor agonists in humans to prevent the worsening of cardiovascular disease.


Subject(s)
Angiotensins/toxicity , Cardiomegaly/chemically induced , Estrogen Receptor beta/agonists , Isoquinolines/pharmacology , Animals , Cardiomegaly/prevention & control , Estradiol/pharmacology , Female , Heart/drug effects , Mice , Myocardium/pathology , Ovariectomy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
19.
Mol Biol Cell ; 24(24): 3805-18, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24152730

ABSTRACT

The development and progression of cardiac hypertrophy often leads to heart failure and death, and important modulators of hypertrophy include the histone deacetylase proteins (HDACs). Estrogen inhibits cardiac hypertrophy and progression in animal models and humans. We therefore investigated the influence of 17-ß-estradiol on the production, localization, and functions of prohypertrophic (class I) and antihypertrophic (class II) HDACs in cultured neonatal rat cardiomyocytes. 17-ß-Estradiol or estrogen receptor ß agonists dipropylnitrile and ß-LGND2 comparably suppressed angiotensin II-induced HDAC2 (class I) production, HDAC-activating phosphorylation, and the resulting prohypertrophic mRNA expression. In contrast, estrogenic compounds derepressed the opposite effects of angiotensin II on the same parameters for HDAC4 and 5 (class II), resulting in retention of these deacetylases in the nucleus to inhibit hypertrophic gene expression. Key aspects were confirmed in vivo from the hearts of wild-type but not estrogen receptor ß (ERß) gene-deleted mice administered angiotensin II and estrogenic compounds. Our results identify a novel dual regulation of cardiomyocyte HDACs, shown here for the antihypertrophic sex steroid acting at ERß. This mechanism potentially supports using ERß agonists as HDAC modulators to treat cardiac disease.


Subject(s)
Cardiomegaly/prevention & control , Estradiol/agonists , Estrogen Receptor beta/agonists , Estrogens/agonists , Histone Deacetylase 2/biosynthesis , Angiotensin II/pharmacology , Animals , Cells, Cultured , Female , GATA4 Transcription Factor/genetics , Heart/physiology , Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylase 2/genetics , Histone Deacetylases/biosynthesis , Isoquinolines/pharmacology , Mice , Mice, Inbred C57BL , Myocytes, Cardiac , Phosphorylation/drug effects , Protein Kinase C-delta/genetics , RNA, Messenger/biosynthesis , Rats , Repressor Proteins/biosynthesis , Transcriptional Activation
20.
Mol Biol Cell ; 23(1): 188-99, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22031296

ABSTRACT

Classical estrogen, progesterone, and androgen receptors (ERs, PRs, and ARs) localize outside the nucleus at the plasma membrane of target cells. From the membrane, the receptors signal to activate kinase cascades that are essential for the modulation of transcription and nongenomic functions in many target cells. ER, PR, and AR trafficking to the membrane requires receptor palmitoylation by palmitoylacyltransferase (PAT) protein(s). However, the identity of the steroid receptor PAT(s) is unknown. We identified the DHHC-7 and -21 proteins as conserved PATs for the sex steroid receptors. From DHHC-7 and -21 knockdown studies, the PATs are required for endogenous ER, PR, and AR palmitoylation, membrane trafficking, and rapid signal transduction in cancer cells. Thus the DHHC-7 and -21 proteins are novel targets to selectively inhibit membrane sex steroid receptor localization and function.


Subject(s)
Acyltransferases/metabolism , Estrogen Receptor alpha/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Receptors, Androgen/metabolism , Receptors, Progesterone/metabolism , Acetyltransferases , Acyltransferases/genetics , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Epigenesis, Genetic , Estradiol/pharmacology , Estradiol/physiology , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lipoylation , MAP Kinase Signaling System , Presenilin-2/genetics , Presenilin-2/metabolism , Protein Binding , Protein Transport , RNA Interference , Response Elements , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL