Abstract
Estrogen actions are largely dependent on the intracellular estrogen receptor (ER) levels. During aging the decline of estrogens or ER leads to a loss in antiinflammatory protection and an increase in oxidant stress due to changes in mitochondrial function. Estrogens/ER may also coordinate signaling between the nucleus and mitochondria through ERK activation, which paradoxically decreases ER expression. The changes in ER expression and transcriptional activation that occur with aging as well as the mitochondria-to-nuclear signaling pathways have not been studied in the glomerulus. We found that ER expression and transcriptional activation decreased with age. Whereas ER levels decreased by greater than 90%, serum 17β-estradiol levels decreased by less than 30%, suggesting alternative mechanisms for ER decrease. Because we postulated that this was due in part to age-related oxidant stress, we treated mesangial cells (MCs) with ethidium bromide (EtBr) to deplete mitochondria. EtBr treatment resulted in decreased ERK activation and reactive oxygen species, which were associated with increased ERα expression and transcriptional activation in old MCs. EtBr treatment also decreased apoptosis and caspase-9 protein expression in old MCs. These data suggest that loss of several of the functions of 17β-estradiol during aging could be mainly due to decreased ERα expression, that the ER loss is reversible by reducing reactive oxygen species, and that mitochondrial retrograde signaling plays a role in this regulation.
The glomerulus is an estrogen target tissue, and estrogen action is particularly important for maintaining glomerular structure and function and extracellular matrix turnover (1, 2). Glomerular cells express estrogen receptors (ER), predominantly the subtype ERα, which mediates most estrogen/ER actions in mesangial cells (MCs) (1). The effects of estrogens in the kidney are largely dependent on the intracellular ER levels, which are determined and regulated by genetic traits, the hormonal milieu, and environmental factors. For instance, female ROP Os/+ mice, which have an increased susceptibility to glomerulosclerosis, have lower glomerular and mesangial cell ER levels than C57BL/6 mice, a strain that is more resistant to glomerulosclerosis. In addition, estrogen deficiency at a young age accelerates the progression of glomerulosclerosis in female ROP Os/+ (3).
It is well established that aging and estrogen deficiency are associated with increased oxidant stress, which promotes age-related diseases in the renal vasculature (4, 5). Oxidant stress occurs when free radicals, single reactive oxygen species (ROS), and other reactive intermediates, such as advanced glycation endproducts of lipid peroxidation products, overwhelm antioxidant systems (6, 7). Mitochondria, a primary source of ROS in the cell, play an important role in cellular adaptation to oxidant stress (8). The consequences of increased age-related mitochondrial ROS production coupled with a decline in estrogens on the expression of ER, have not been well studied. We found that female C57BL/6 mice develop glomerular lesions as they age (9). We also showed that aged, ovariectomized C57BL/6 mice develop glomerular lesions after exposure to cigarette smoke (a source of oxidant injury and advanced glycation endproducts) (10), which can be ameliorated by 17β-estradiol (E2) replacement therapy (11). Based on these data, we postulated that the level of ER expression and function in the glomerulus decreases with age and may be mediated by age-associated oxidant stress.
In addition, because E2 regulation of mitochondrial function is most likely mediated through ER-dependent mechanisms (12, 13), a decline in ER expression and function in glomerular cells could lead to altered mitochondrial responses such as apoptosis. We found that aging decreases ERα mRNA and protein expression in glomeruli and MCs isolated from female C57BL6 mice. Transcriptional activation was decreased in parallel. Manipulation of oxidant stress in vitro also regulated ER expression. Surprisingly, in aging MCs, decreased ER expression and function were reversed by preventing signaling from the mitochondria through ERK. Finally, we show that increased apoptosis present in aging MCs was also reversed by reducing mitochondrial ROS production.
Materials and Methods
Isolation of glomeruli and glomerular cells
Glomeruli were isolated as previously described (14) for use in mRNA experiments. MCs were isolated from two separate glomerular isolates from each experimental group 7 and 24 months C57BL/6 mice (15). Glomeruli were plated in Nunc dishes (Rochester, NY) in the presence of 10% fetal bovine serum (FBS). MCs were identified by morphology, immunochemical profile (end to end F-actin filaments, smooth muscle actin, and the absence of nephrin and WT-1). Cell lines isolated from at least two individual mice per group were used for all experiments. All cell lines were studied between passages 5 and 15. Serum E2 levels were measured by ELISA as previously described (11).
Transfection studies
MCs from young and old mice and those treated with ethidium bromide (EtBr) were plated in basal medium with 20% charcoal/dextran-treated fetal bovine serum (<5 pg/ml estrogens) in 24-well plates. Twenty-four hours before transfections, MCs were grown in phenol red-free medium containing 0.1% charcoal stripped serum. MCs were transfected using GenePorter (Promega, Madison, WI) with a 4ERE-TATA-Luc reporter gene construct and the β-galactosidase gene pRSV-βgal (0.4 μg/well) to control for transfection efficacy. MCs were subsequently treated with vehicle or E2 (10 nm). After 48 h, MCs were harvested and luciferase and β-galactosidase assays were performed as previously described (1). Briefly, cells were washed two times in PBS and lysed with 100 μl of reporter lysis buffer (Promega) at room temperature for 15 min. Wells were scraped and the lysate transferred to a Microfuge tube, vortexed, and microcentrifuged for 2 min at 4 C. The supernatant was collected and frozen at −70 C until assayed. In some experiments, cells were transfected with a dominant-negative ERK1 or ERK2 plasmid (kind gift of Melanie Cobb, University of Texas Southwestern Medical Center, Dallas, TX).
Polymerase chain reaction
Amplification and measurement of target RNA was performed on the on the Step 1 real time PCR system as previously described (16). The mRNA sequence was obtained from the National Center for Biotechnology Information (Bethesda, MD) to acquire the copy number for each ER subtype. The number of occurrences of each of the four nucleobases was counted and multiplied by its respective molecular weight. These four numbers were then summed together to obtain the mass of 1 mol of each subtype of the ER. The mass of the purified plasmid of each subtype and the unknown samples was calculated by the A260 method on a Molecular Devices SpectraMax PLUS (Ramsey, MI) (17).
Western blot analysis
For protein analysis, cell lysates were extracted and protein assessed using the Pierce BCA protein assay kit (Rockford, IL). Equal amounts of protein were applied to precast sodium dodecyl sulfate polyacrylamide gels (Life Technologies, Grand Island, NY) and analyzed as previously described (2) for ERα (H184), ERK and phospho-ERK (Santa Cruz Biotechnology, Santa Cruz, CA) and caspase-9 (Cell Signaling Technology, Beverly MA). Blots were analyzed as described (17). In some experiments, cells were treated overnight with 40 μm PD98059. Western blots were also exposed to β-actin (Sigma Chemical, St. Louis, MO) to control for protein loading. Human recombinant ERα was used as a control (PanVera, Madison, WI).
Modulation of ROS
In some experiments, MCs were treated overnight with 25 μm N-acetyl-l-cysteine (NAC), an antioxidant, followed by 50 μm hydrogen peroxide (H2O2) for 4 h. Cells were collected as described for Western analysis and ERα protein expression determined.
ROS measurement
ROS was measured using 5-(and-6)-carboxy-2′,7′-dichlorodihydrofluorescein diacetate according to the manufacturer's directions (carboxy-H2DCFDA; Molecular Probes, Eugene, OR).
EtBr treatment
MCs were treated with 50 ng/ml EtBr to block mitochondrial DNA replication (18). Two cell lines of both young and old MCs were passaged and maintained in medium supplemented with 0.1% uridine and 1% sodium pyruvate and EtBr for 3 wk. These conditions allowed the cells to continue exponential growth while reducing mitochondrial DNA by 50% at each doubling. Sister cell lines were grown in parallel in exactly the same medium without EtBr as controls. Depletion of mitochondria was assessed by measurement of ROS and respiration. Cell viability was assessed by trypan blue.
Cell respiration and OxPhos activity
Oxygen consumption was measured polarographically in 0.3 m mannitol, 10 mm KCl, 5 mm MgCl2, 1 mg/ml BSA, 10 mm KH2PO4 (pH 7.4), with a Clark oxygen electrode (Hansatech Instruments, Norfolk, UK). After measuring intact cell endogenous respiration, antimycin A was added to block respiration at complex III. Ascorbate-N, N, N′, N′-tetramethyl-p-phenylenediamine was then be added to drive complex IV respiration and subsequently inhibited with 0.7 mm KCN.
Mito Tracker staining
Control and EtBr-treated MCs were plated on two-well chamber slides for 24 h (Lab-Tek; Nunc). The medium was replaced to remove FBS, and 25 nm of Mito Tracker Green FM and Mito Tracker Red CMXRos (Invitrogen, Carlsbad, CA) was added for 15 min followed by 4′,6′-diamino-2-phenylindole (DAPI; Vector Laboratories, Inc., Burlingame, CA) for 5 min. The slides were washed, mounted, and visualized on a LSM700 confocal using a ×63 Plan Apochromat 63 × 1.4 oil NA objective.
DNA fragmentation by in situ cell staining and flow cytometry
Staining was performed using Apoptag fluorescence (ApopTag terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling fluorescein in situ apoptosis detection kit; Mllipore, Billerica, MA) according to manufacturer's directions. Then 1 × 106 control and EtBr-treated cells were seeded onto a fibronectin-coated flask. Forty-eight hours later, cells were washed with 0.5 ml PBS, resuspended in equilibration buffer, and centrifuged to remove the supernatant. Cells were incubated with terminal deoxynucleotidyl transferase for 30 min at 37 C. After a stop reaction was added, cells were washed and stained with propidium iodide for 15 min at room temperature. For flow cytometry analysis, the violet annexin V/dead cell apoptosis kit (Invitrogen) was used according to the manufacturer's directions. Briefly, MCs were exposed to the pacific blue and SYTOX stains for 30 min. Analysis was performed by two-color flow cytometry assays using flow cytometer (FACs Calibur) and CellQuest software (BD Biosciences, San Jose, CA).
Statistics
In vitro assays were performed in triplicate. One-way ANOVA and the Dunnett multiple-comparison post hoc test or Student's t test were performed for the statistical analysis. (GraphPad Prism; GraphPad Software Inc., San Diego, CA). Statistical significance was set at P < 0.05.
Results
ERα expression
Glomeruli were isolated from female C57BL/6 mice at different stages of reproductive life: young (7 months old) at which time estrous cycles are regular and approximately 4 d long (reproductive period); during the period of persistent vaginal cornification, 14 months of age (perimenopause); and between 20 and 30 months of age anestrous or rodent menopause (19). The glomerular ERα copy number was decreased at 27 months of age compared with 7 months of age (Fig. 1A, *, P < 0.05). This correlated with 30% decline in serum E2 levels at 27 months of age (Fig. 1B). In addition, ERα mRNA (young, 79 copies; old, 43 copies) and protein expression in MCs isolated from old mice decreased approximately 50% compared with young mice (Fig. 2A, **, P < 0.005).
ER transcriptional activation
To determine whether ER function declined in parallel with ER protein expression, MCs from old and young were transfected in phenol red-free medium containing 10% charcoal-stripped serum with 0.3 μg/well of a luciferase-based reporter construct containing four estrogen response elements. Stimulation with E2 increased transcriptional activation 2-fold in young cells (Fig. 2B, *, P < 0.05), whereas the there was no significant change in MCs from old mice.
ROS modulation of ERα expression
Because we hypothesized that changes in ERα expression and function were possible due to age-associated ROS, we induced ROS by treating cells with H2O2. We found a 45% decrease in ERα expression in young and 65% in old MCs after treatment. Overnight treatment with NAC, an antioxidant, prevented the H2O2 decrease in ERα expression of both young and old MCs (Fig. 3A).
ERK phosphorylation, ERα expression, and ER transcriptional activation
Increased MAPK activation has been reported to regulate ERα expression (20, 21). To determine whether this was one of the signaling pathways responsible for age-associated changes in ER expression, we treated MCs from young and old mice with PD98059. Preventing ERK phosphorylation either by PD98059 (Fig. 3B, *, P < 0.05) or DN ERK (Supplement Fig. 1, **, P < 0.005, published on The Endocrine Society's Journals Online web site at http://endo.endojournals.org) increased ERα protein expression in old MCs.
Altering retrograde signaling between mitochondria and the nucleus could disrupt pathways important for regulation of ER expression; therefore, MCs were depleted of mitochondrial DNA by treatment with EtBr for 3 wk. Cells were viable as assessed by trypan blue. As expected, mitochondrial depletion resulted in a decrease in respiration (Fig. 4A) and a decrease in production of ROS (Fig. 4B). These data were also confirmed using Mito Tracker Green and Red (Invitrogen) to visualize both mitochondrial number and activity respectively (Fig. 4C) of young and old MCs. At baseline, young cells had more mitochondria and less activity (Fig. 4, B and C) compared with old cells (Fig. 4, J and K). EtBr treatment reduced mitochondrial activity in both young and old cells (Fig. 4, H and P).
Blocking mitochondrial transcription/replication by EtBr treatment had no effect on ERK phosphorylation in MCs from young mice, whereas it decreased phosphorylation in MCs isolated from old mice (Fig. 5, *, P < 0.05). Importantly, it increased MC ERα protein expression 2.7-fold (Fig. 6A, *, P < 0.05) and mRNA expression almost 6-fold compared with MCs from young mice (young, 11 copies; old, 63 copies). In addition, transcriptional activation was approximately 50% higher (1 and 10 nm) in old MCs compared with that found in young MCs (Fig. 6B).
DNA fragmentation and apoptosis
Because mitochondria play a significant role in apoptosis, the effect of removal of mitochondria by EtBr treatment was examined. The increase in DNA fragmentation was partially abolished in MCs from old mice (Fig. 7A, panel G). There was no change in young MCs. We also performed flow cytometry (Fig. 7B) and confirmed that there were fewer apoptotic cells at baseline in young vs. old MCs (less than one vs. three cells), which was reduced by at least 80 ± 10% in old MCs (less than one cell, n = 3 individual experiments). In parallel, we found that EtBr treatment decreased caspase-9 protein expression approximately 30% in old (Fig. 7C) but not young MCs.
Discussion
The effect of aging on glomeruli and glomerular cell ERα expression has not been previously described, although there have been studies in the kidney, brain. and inner ear of mice (22–24). In this report, we show for the first time that glomerular ERα mRNA expression decreases with increasing age in glomeruli isolated from female C57BL/6 mice. In addition, ERα protein expression of MCs isolated from young and old mice follow the same pattern. This is in contrast to a study on total kidney isolated from female AK mice, which reported an increase in ERα mRNA and protein expression with age (23). These conflicting results are very likely due to differences in mouse strains and/or the fact that the glomerulus represents a small fraction of total kidney and therefore could have a different expression pattern. We have previously shown that ERα expression levels correlate with the degree of glomerulosclerosis (GS), and we postulated that decreased ERα expression may promote age-related GS in mice as previously described (9). In fact, we show in the current study that transcriptional activation of ER is also decreased in an age-related fashion correlating with the decrease of ERα expression.
Several mechanisms have been reported for regulation of the ER including posttranscriptional destabilization of ERα mRNA (25) and destruction of ERα protein through the ubiquitin-proteasome pathway (26). Because the ERα mRNA levels in our study decreased in parallel with the protein expression, we believe that other mechanisms are more likely. For example, an age-related increase in the hypermethylation of CpG islands in the ER promoter region, which blocks the expression of the ERα gene, has been shown in human vascular smooth muscle cells and aortic endothelial cells (27–30). In addition, oxidant stress activates several transcription factors, including nuclear factor-κB, and MAPK (31, 32), which can regulate ER expression levels. We have previously shown that MCs isolated from mice that develop GS such as db/db and ROP Os/+ mice (33) have decreased ERα expression induced by hyperactivation of MAPK, which is also true for breast cancer cells (21, 34). Our current study suggests that ERK signaling regulates ER expression in old MCs. In addition, it is probable that ERK signaling to and from the mitochondria in old MCs led to the down-regulation of ERα, although this is likely not the sole source of glomerular/mesangial cell ER down-regulation associated with aging.
It is well known that the function of ERs can be modulated by the redox state of the cell (35). Variations in gene expression by both direct and indirect pathways occur due to changes in cellular redox state especially during aging (36). Aging increases oxidant stress as measured by increased levels of endogenous lipid peroxides [8-isoprostane levels in blood and urine (37, 38)]. Young men have higher levels of markers of oxidative stress compared with premenopausal age-matched women (39, 40), but parameters of oxidative stress increase in women after menopause (4). We have evidence that carboxy-methyl-lysine, a marker of oxidant injury (41), is increased with aging in the serum of female C57BL6 mice (Elliot, S. E., and H. Vlassara unpublished data); therefore, we investigated whether ER expression could be manipulated by changing the oxidant stress status of the cell. In fact, treatment of the cells with H2O2 decreased ER, whereas treatment with NAC prevented the decrease. Alternatively, to reduce ROS, we treated young and old MCs with EtBr to block mitochondrial transcription/replication. Confirmation of the effectiveness of the EtBr treatment was confirmed by a decrease in cellular respiration. Reducing mitochondrial ROS decreased ERK phosphorylation in old cells with a corresponding increase of ERα expression and transcriptional activation in old cells. We also found a parallel increase in ERα mRNA expression, suggesting a direct effect of ROS/mitochondrial signaling on the promoter of the ERα gene.
Mitochondria play an important role in cellular adaptation via a mitochondria-to-nucleus signaling pathway called retrograde regulation (36, 42). The retrograde response reacts in a continuous manner to the changing metabolic needs of the cell (43). Low ROS concentrations can act to oxidize redox-sensitive proteins phosphatases or kinases and modify the phosphorylation state of receptors and transcription factors, which in turn could influence the mitochondrial response. In addition, mitochondrial calcium release leads to multiple signaling cascades including activation of MAPK, c-Jun terminal kinase, and p38 MAPK (44). To our knowledge, the mitochondria-to-nuclear signaling pathways involved in the glomerulus have not been determined before our studies. Interestingly, in an effect opposite to that of the old MCs, ERK activation remained unchanged in the young MCs, suggesting different regulation of signaling patterns between old and young MCs.
Because mitochondria are central to the initiation of apoptosis, we investigated whether regulation of mitochondrial signaling might play a role in protection against apoptosis. We used apoptosis as a marker of aging because it is well established that apoptosis increases with age in the kidney and can be prevented by reduction in oxidant stress (45, 46). In fact, EtBr treatment effectively reduced both caspase-9 expression and apoptotic cell number in old mesangial cells.
In summary, ERα expression and transcriptional activation in glomeruli and MCs were decreased in aging. This reduction is reversible by manipulation of ROS and/or ERK activation through changes in mitochondrial signaling. These data suggest a role not only for oxidant stress per se in the regulation of ERα but also for retrograde signaling. In addition, it appears that ERα expression levels have the potential to be modulated, and it is possible that changes in mitochondrial signaling alter repressors of transcription and/or translation. Future studies will extend these findings to determine whether ERK and other relevant signaling pathways increase antioxidant enzymes or other protective mechanisms and whether ERα action is important for regulation of these pathways in the glomerulus.
Supplementary Material
Acknowledgments
We acknowledge the skilled assistance of the Flow Cytometry Core Facility of the Sylvester Comprehensive Cancer Center (University of Miami, Miller School of Medicine) for the provision of sophisticated fluorescence analysis and cell sorting services. The authors also thank Gabriel Gaidosh (Department of Ophthalmology, University of Miami, Miller School of Medicine) for help with the confocal microscopy.
This work was supported by National Institutes of Health Grant RO1 AG17170-11 (to S.E.).
Disclosure Summary: The authors have nothing to disclose.
Footnotes
- DAPI
- 4′,6′-Diamino-2-phenylindole
- E2
- 17β-estradiol
- ER
- estrogen receptor
- EtBr
- ethidium bromide
- FBS
- fetal bovine serum
- GS
- glomerulosclerosis
- MC
- mesangial cell
- NAC
- N-acetyl-l-cysteine
- ROS
- reactive oxygen species.
References
- 1. Potier M, Elliot SJ, Tack I, Lenz O, Striker GE, Striker LJ, Karl M. 2001. Expression and regulation of estrogen receptors in mesangial cells: influence on matrix metalloproteinase-9. J Am Soc Nephrol 12:241–251 [DOI] [PubMed] [Google Scholar]
- 2. Potier M, Karl M, Zheng F, Elliot SJ, Striker GE, Striker LJ. 2002. Estrogen-related abnormalities in glomerulosclerosis-prone mice: reduced mesangial cell estrogen receptor expression and prosclerotic response to estrogens. Am J Pathol 160:1877–1885 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Elliot SJ, Karl M, Berho M, Potier M, Zheng F, Leclercq B, Striker GE, Striker LJ. 2003. Estrogen deficiency accelerates progression of glomerulosclerosis in susceptible mice. Am J Pathol 162:1441–1448 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Helmersson J, Mattsson P, Basu S. 2002. Prostaglandin F(2α) metabolite and F(2)-isoprostane excretion rates in migraine. Clin Sci (Lond) 102:39–43 [PubMed] [Google Scholar]
- 5. Vlassara H, Torreggiani M, Post JB, Zheng F, Uribarri J, Striker GE. 2009. Role of oxidants/inflammation in declining renal function in chronic kidney disease and normal aging. Kidney Int Suppl S3–S11 [DOI] [PubMed] [Google Scholar]
- 6. Lee HC, Wei YH. 2007. Oxidative stress, mitochondrial DNA mutation, and apoptosis in aging. Exp Biol Med (Maywood) 232:592–606 [PubMed] [Google Scholar]
- 7. Cadenas E, Davies KJ. 2000. Mitochondrial free radical generation, oxidative stress, and aging. Free Radic Biol Med 29:222–230 [DOI] [PubMed] [Google Scholar]
- 8. Vendelbo MH, Nair KS. 2011. Mitochondrial longevity pathways. Biochim Biophys Acta 1813:634–644 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Zheng F, Plati AR, Potier M, Schulman Y, Berho M, Banerjee A, Leclercq B, Zisman A, Striker LJ, Striker GE. 2003. Resistance to glomerulosclerosis in B6 mice disappears after menopause. Am J Pathol 162:1339–1348 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Cerami C, Founds H, Nicholl I, Mitsuhashi T, Giordano D, Vanpatten S, Lee A, Al-Abed Y, Vlassara H, Bucala R, Cerami A. 1997. Tobacco smoke is a source of toxic reactive glycation products. Proc Natl Acad Sci USA 94:13915–13920 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Elliot SJ, Karl M, Berho M, Xia X, Pereria-Simon S, Espinosa-Heidmann D, Striker GE. 2006. Smoking induces glomerulosclerosis in aging estrogen-deficient mice through cross-talk between TGF-β1 and IGF-I signaling pathways. J Am Soc Nephrol 17:3315–3324 [DOI] [PubMed] [Google Scholar]
- 12. Pedram A, Razandi M, Wallace DC, Levin ER. 2006. Functional estrogen receptors in the mitochondria of breast cancer cells. Mol Biol Cell 17:2125–2137 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Chen JQ, Russo PA, Cooke C, Russo IH, Russo J. 2007. ERβ shifts from mitochondria to nucleus during estrogen-induced neoplastic transformation of human breast epithelial cells and is involved in estrogen-induced synthesis of mitochondrial respiratory chain proteins. Biochim Biophys Acta 1773:1732–1746 [DOI] [PubMed] [Google Scholar]
- 14. Peten EP, Garcia-Perez A, Terada Y, Woodrow D, Martin BM, Striker GE, Striker LJ. 1992. Age-related changes in α1- and α2-chain type IV collagen mRNAs in adult mouse glomeruli: competitive PCR. Am J Physiol 263:F951–F957 [DOI] [PubMed] [Google Scholar]
- 15. Elliot SJ, Striker LJ, Hattori M, Yang CW, He CJ, Peten EP, Striker GE. 1993. Mesangial cells from diabetic NOD mice constitutively secrete increased amounts of insulin-like growth factor-I. Endocrinology 133:1783–1788 [DOI] [PubMed] [Google Scholar]
- 16. Karl M, Berho M, Pignac-Kobinger J, Striker GE, Elliot SJ. 2006. Differential effects of continuous and intermittent 17β-estradiol replacement and tamoxifen therapy on the prevention of glomerulosclerosis: modulation of the mesangial cell phenotype in vivo. Am J Pathol 169:351–361 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Catanuto P, Doublier S, Lupia E, Fornoni A, Berho M, Karl M, Striker GE, Xia X, Elliot S. 2009. 17-Estradiol and tamoxifen upregulate estrogen receptor α and regulate podocyte signaling pathways in a model of type 2 diabetes. Kidney Int 75:1194–1201 [DOI] [PubMed] [Google Scholar]
- 18. Bacman SR, Moraes CT. 2007. Transmitochondrial technology in animal cells. Methods Cell Biol 80:503–524 [DOI] [PubMed] [Google Scholar]
- 19. vom Saal FS, Finch CE, Nelson JF. 1994. Natural history and mechanisms of reproductive aging in humans, laboratory rodents, and other selected vertebrates. In: Knobil E, Neill JD, eds. The physiology of reproduction. 2nd ed New York: Raven Press; 1213–1314 [Google Scholar]
- 20. Bayliss J, Hilger A, Vishnu P, Diehl K, El-Ashry D. 2007. Reversal of the estrogen receptor negative phenotype in breast cancer and restoration of antiestrogen response. Clin Cancer Res 13:7029–7036 [DOI] [PubMed] [Google Scholar]
- 21. Oh AS, Lorant LA, Holloway JN, Miller DL, Kern FG, El-Ashry D. 2001. Hyperactivation of MAPK induces loss of ERα expression in breast cancer cells. Mol Endocrinol 15:1344–1359 [DOI] [PubMed] [Google Scholar]
- 22. Motohashi R, Takumida M, Shimizu A, Konomi U, Fujita K, Hirakawa K, Suzuki M, Anniko M. 2009. Effects of age and sex on the expression of estrogen receptor α and β in the mouse inner ear. Acta Otolaryngol l1–11 [DOI] [PubMed] [Google Scholar]
- 23. Sharma PK, Thakur MK. 2004. Estrogen receptor α expression in mice kidney shows sex differences during aging. Biogerontology 5:375–381 [DOI] [PubMed] [Google Scholar]
- 24. Sharma PK, Thakur MK. 2006. Expression of estrogen receptor (ER) α and β in mouse cerebral cortex: effect of age, sex and gonadal steroids. Neurobiol Aging 27:880–887 [DOI] [PubMed] [Google Scholar]
- 25. Saceda M, Lippman ME, Chambon P, Lindsey RL, Ponglikitmongkol M, Puente M, Martin MB. 1988. Regulation of the estrogen receptor in MCF-7 cells by estradiol. Mol Endocrinol 2:1157–1162 [DOI] [PubMed] [Google Scholar]
- 26. Nawaz Z, Lonard DM, Dennis AP, Smith CL, O'Malley BW. 1999. Proteasome-dependent degradation of the human estrogen receptor. Proc Natl Acad Sci USA 96:1858–1862 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Cerda S, Weitzman SA. 1997. Influence of oxygen radical injury on DNA methylation. Mutat Res 386:141–152 [DOI] [PubMed] [Google Scholar]
- 28. Kim J, Kim JY, Song KS, Lee YH, Seo JS, Jelinek J, Goldschmidt-Clermont PJ, Issa JP. 2007. Epigenetic changes in estrogen receptor β gene in atherosclerotic cardiovascular tissues and in vitro vascular senescence. Biochimt Biophys Acta 1772:72–80 [DOI] [PubMed] [Google Scholar]
- 29. Post WS, Goldschmidt-Clermont PJ, Wilhide CC, Heldman AW, Sussman MS, Ouyang P, Milliken EE, Issa JP. 1999. Methylation of the estrogen receptor gene is associated with aging and atherosclerosis in the cardiovascular system. Cardiovasc Res 43:985–991 [DOI] [PubMed] [Google Scholar]
- 30. Ying AK, Hassanain HH, Roos CM, Smiraglia DJ, Issa JJ, Michler RE, Caligiuri M, Plass C, Goldschmidt-Clermont PJ. 2000. Methylation of the estrogen receptor-α gene promoter is selectively increased in proliferating human aortic smooth muscle cells. Cardiovasc Res 46:172–179 [DOI] [PubMed] [Google Scholar]
- 31. Kirillova I, Chaisson M, Fausto N. 1999. Tumor necrosis factor induces DNA replication in hepatic cells through nuclear factor κB activation. Cell Growth Differ 10:819–828 [PubMed] [Google Scholar]
- 32. Liang X, Lu B, Scott GK, Chang CH, Baldwin MA, Benz CC. 1998. Oxidant stress impaired DNA-binding of estrogen receptor from human breast cancer. Mol Cell Endocrinol 146:151–161 [DOI] [PubMed] [Google Scholar]
- 33. Karl M, Potier M, Schulman IH, Rivera A, Werner H, Fornoni A, Elliot SJ. 2005. Autocrine activation of the local insulin-like growth factor I system is up-regulated by estrogen receptor (ER)-independent estrogen actions and accounts for decreased ER expression in type 2 diabetic mesangial cells. Endocrinology 146:889–900 [DOI] [PubMed] [Google Scholar]
- 34. Holloway JN, Murthy S, El-Ashry D. 2004. A cytoplasmic substrate of mitogen-activated protein kinase is responsible for estrogen receptor-α down-regulation in breast cancer cells: the role of nuclear factor-κB. Mol Endocrinol 18:1396–1410 [DOI] [PubMed] [Google Scholar]
- 35. Tanaka H, Makino Y, Okamoto K, Iida T, Yoshikawa N, Miura T. 2000. Redox regulation of the nuclear receptor. Oncology 59(Suppl 1):13–18 [DOI] [PubMed] [Google Scholar]
- 36. Finkel T, Holbrook NJ. 2000. Oxidants, oxidative stress and the biology of ageing. Nature 408:239–247 [DOI] [PubMed] [Google Scholar]
- 37. Vlassara H, Uribarri J, Ferrucci L, Cai W, Torreggiani M, Post JB, Zheng F, Striker GE. 2009. Indentifying advanced glycation end products as a major source of oxidants in aging: implications for the management and/or prevention of reduced renal function in elderly persons. Semin Nephrol 29:594–603 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Cai W, He JC, Zhu L, Chen X, Wallenstein S, Striker GE, Vlassara H. 2007. Reduced oxidant stress and extended lifespan in mice exposed to a low glycotoxin diet: association with increased AGER1 expression. Am J Pathol 170:1893–1902 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Loft S, Vistisen K, Ewertz M, Tjønneland A, Overvad K, Poulsen HE. 1992. Oxidative DNA damage estimated by 8-hydroxydeoxyguanosine excretion in humans: influence of smoking, gender and body mass index. Carcinogenesis 13:2241–2247 [DOI] [PubMed] [Google Scholar]
- 40. Uribarri J, Cai W, Peppa M, Goodman S, Ferrucci L, Striker G, Vlassara H. 2007. Circulating glycotoxins and dietary advanced glycation endproducts: two links to inflammatory response, oxidative stress, and aging. J Gerontol A Biol Sci Med Sci 62:427–433 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Vlassara H. 2005. Advanced glycation in health and disease: role of the modern environment. Ann NY Acad Sci 1043:452–460 [DOI] [PubMed] [Google Scholar]
- 42. Butow RA, Avadhani NG. 2004. Mitochondrial signaling: the retrograde response. Mol Cell 14:1–15 [DOI] [PubMed] [Google Scholar]
- 43. Passos JF, Von Zglinicki T. 2006. Oxygen free radicals in cell senescence: are they signal transducers? Free Radic Res 40:1277–1283 [DOI] [PubMed] [Google Scholar]
- 44. Camello-Almaraz C, Gomez-Pinilla PJ, Pozo MJ, Camello PJ. 2006. Mitochondrial reactive oxygen species and Ca2+ signaling. Am J Physiol Cell Physiol 291:C1082–C1088 [DOI] [PubMed] [Google Scholar]
- 45. Kujoth GC, Bradshaw PC, Haroon S, Prolla TA. 2007. The role of mitochondrial DNA mutations in mammalian aging. PLoS Genet 3:e24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Martin JE, Sheaff MT. 2007. Renal ageing. J Pathol 211:198–205 [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.