Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Mol Ther ; 30(2): 947-962, 2022 02 02.
Article in English | MEDLINE | ID: mdl-34174444

ABSTRACT

Despite increasing interest in the reversal of age-related processes, there is a paucity of data regarding the effects of post-menopausal-associated estrogen loss on cellular function. We studied human adipose-derived mesenchymal stem cells (hASCs) isolated from women younger than 45 years old (pre-menopause, pre-hASC) or older than 55 years old (post-menopause, post-hASC). In this study, we provide proof of concept that the age-related ineffective functionality of ASCs can be reversed to improve their ability in promoting tissue repair. We found reduced estrogen receptor expression, decreased estrogen receptor activation, and reduced sensitivity to 17ß-estradiol in post-hASCs. This correlated with decreased antioxidants (catalase and superoxide dismutase [SOD] expression) and increased oxidative stress compared with pre-hASCs. Increasing catalase expression in post-hASCs restored estrogen receptor (ER) expression and their functional capacity to promote tissue repair as shown in human skin ex vivo wound healing and in vivo mouse model of lung injury. Our results suggest that the consequences of 17ß-estradiol decline on the function of hASCs may be reversible by changing the oxidative stress/antioxidant composition.


Subject(s)
Adipose Tissue , Mesenchymal Stem Cells , Aging , Animals , Catalase/genetics , Catalase/metabolism , Estrogens/metabolism , Estrogens/pharmacology , Female , Humans , Mesenchymal Stem Cells/metabolism , Mice
2.
Chest ; 160(4): 1271-1281, 2021 10.
Article in English | MEDLINE | ID: mdl-33894254

ABSTRACT

COPD is a chronic inflammatory and destructive disease characterized by progressive decline in lung function that can accelerate with aging. Preclinical studies suggest that mesenchymal stem cells (MSCs) may provide a therapeutic option for this incurable disease because of their antiinflammatory, reparative, and immunomodulatory properties. To date, clinical trials using MSCs demonstrate safety in patients with COPD. However, because of the notable absence of large, multicenter randomized trials, no efficacy or evidence exists to support the possibility that MSCs can restore lung function in patients with COPD. Unfortunately, the investigational status of cell-based interventions for lung diseases has not hindered the propagation of commercial businesses, exploitation of the public, and explosion of medical tourism to promote unproven and potentially harmful cell-based interventions for COPD in the United States and worldwide. Patients with COPD constitute the largest group of patients with lung disease flocking to these unregulated clinics. This review highlights the numerous questions and concerns that remain before the establishment of cell-based interventions as safe and efficacious treatments for patients with COPD.


Subject(s)
Clinical Trials as Topic , Fraud , Mesenchymal Stem Cell Transplantation , Pulmonary Disease, Chronic Obstructive/therapy , Animals , Cell- and Tissue-Based Therapy , Evidence-Based Medicine , Extracellular Vesicles , Humans , Registries
3.
Respirology ; 26(2): 161-170, 2021 02.
Article in English | MEDLINE | ID: mdl-32851725

ABSTRACT

BACKGROUND AND OBJECTIVE: IPF is a fatal and debilitating lung disorder increasing in incidence worldwide. To date, two approved treatments only slow disease progression, have multiple side effects and do not provide a cure. MSC have promising therapeutic potential as a cell-based therapy for many lung disorders based on the anti-fibrotic properties of the MSC. METHODS: Critical questions remain surrounding the optimal source, timing and efficacy of cell-based therapies. The present study examines the most effective sources of MSC. Human MSC were derived from adipose, WJ, chorionic membrane (CSC) and chorionic villi (CVC). MSC were injected into the ageing mouse model of BLM-induced lung fibrosis. RESULTS: All sources decreased Aschroft and hydroxyproline levels when injected into BLM-treated mice at day 10 with the exception of CSC cells that did not change hydroxyproline levels. There were also decreases in mRNA expression of αv -integrin and TNFα in all sources except CSC. Only ASC- and WJ-derived cells reduced AKT and MMP-2 activation, while Cav-1 was increased by ASC treatment as previously reported. BLM-induced miR dysregulation of miR-29 and miR-199 was restored only by ASC treatment. CONCLUSION: Our data suggest that sources of MSC may differ in the pathway(s) involved in repair.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/therapy , Adult , Animals , Biomarkers/metabolism , Bleomycin , Caveolin 1/metabolism , Disease Models, Animal , Gene Expression Regulation , Humans , Inflammation/genetics , Inflammation/pathology , Male , Matrix Metalloproteinase 2/metabolism , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transplantation, Homologous
4.
PLoS One ; 13(9): e0204366, 2018.
Article in English | MEDLINE | ID: mdl-30252878

ABSTRACT

Diabetic kidney disease (DKD) is characterized by progressive glomerulosclerosis (GS). ROP mice have a sclerosis-prone phenotype. However, they develop severe, rapidly progressive GS when rendered diabetic. Since GS also develops in aged C57Bl6 mice, and can be reversed using bone marrow from young mice which have lower oxidative stress and inflammation (OS/Infl), we postulated that this might also apply to DKD. Therefore, this pilot study asked whether reducing OS/Infl in young adult sclerosis-prone (ROP) diabetic mice leads to resolution of existing GS in early DKD using safe, FDA-approved drugs.After 4 weeks of stable streptozotocin-induced hyperglycemia 8-12 week-old female mice were randomized and treated for 22 weeks as follows: 1) enalapril (EN) (n = 8); 2) pyridoxamine (PYR)+EN (n = 8); 3) pentosan polysulfate (PPS)+EN (n = 7) and 4) PPS+PYR+EN (n = 7). Controls were untreated (non-DB, n = 7) and hyperglycemic (DB, n = 8) littermates. PPS+PYR+EN reduced albuminuria and reversed GS in DB. Treatment effects: 1) Anti-OS/Infl defenses: a) PPS+PYR+EN increased the levels of SIRT1, Nrf2, estrogen receptor α (ERα) and advanced glycation endproduct-receptor1 (AGER1) levels; and b) PYR+EN increased ERα and AGER1 levels. 2) Pro-OS/Infl factors: a) PPS+PYR+EN reduced sTNFR1, b) all except EN reduced MCP1, c) RAGE was reduced by all treatments. In summary, PYR+PPS+EN modulated GS in sclerosis-prone hyperglycemic mice. PYR+PPS+EN also decreased albuminuria, OS/Infl and the sclerosis-prone phenotype. Thus, reducing OS/Infl may reverse GS in early diabetes in patients, and albuminuria may allow early detection of the sclerosis-prone phenotype.


Subject(s)
Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/metabolism , Disease Progression , Oxidative Stress/drug effects , Albumins/metabolism , Animals , Creatinine/metabolism , Disease Susceptibility , Female , Gene Expression Regulation/drug effects , Mice , Mice, Inbred C57BL , Pilot Projects
5.
J Cell Physiol ; 233(8): 5503-5512, 2018 08.
Article in English | MEDLINE | ID: mdl-29271488

ABSTRACT

Fibrosis can develop in nearly any tissue leading to a wide range of chronic fibrotic diseases. However, current treatment options are limited. In this study, we utilized an established aged mouse model of bleomycin-induced lung fibrosis (BLM) to test our hypothesis that fibrosis may develop simultaneously in multiple organs by evaluating skin fibrosis and wound healing. Fibrosis was induced in lung in aged (18-22-month-old) C57BL/6 male mice by intratracheal BLM administration. Allogeneic adipose-derived mesenchymal stromal cells (ASCs) or saline were injected intravenously 24 hr after BLM administration. Full thickness 8-mm punch wounds were performed 7 days later to study potential systemic anti-fibrotic and wound healing effects of intravenously delivered ASCs. Mice developed lung and skin fibrosis as well as delayed wound closure. Moreover, we observed similar changes in the expression of known pro-fibrotic factors in both lung and skin wound tissue, including miR-199 and protein expression of its corresponding target, caveolin-1, as well as phosphorylation of protein kinase B. Importantly, ASC-treated mice exhibited attenuation of BLM-induced lung and skin fibrosis and accelerated wound healing, suggesting that ASCs may prime injured tissues and prevent end-organ fibrosis.


Subject(s)
Lung/cytology , Mesenchymal Stem Cells/cytology , Pulmonary Fibrosis/prevention & control , Skin Diseases/prevention & control , Skin/cytology , Wound Healing/physiology , Animals , Bleomycin/pharmacology , Caveolin 1/metabolism , Disease Models, Animal , Lung/drug effects , Lung/metabolism , Male , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Skin/drug effects , Skin/metabolism , Skin Diseases/chemically induced , Skin Diseases/metabolism , Wound Healing/drug effects
6.
Surgery ; 163(1): 143-149, 2018 01.
Article in English | MEDLINE | ID: mdl-29128169

ABSTRACT

BACKGROUND: Estrogen receptors can regulate growth in papillary thyroid cancer and may affect prognosis after menopause. This study examines changes of estrogen receptor subtype ratio expression in papillary thyroid cancer cell lines derived from pre- and postmenopausal women. METHODS: Cells were harvested from papillary thyroid cancer and non-papillary thyroid cancer thyroid tissue (control) from pre- (n = 9) and postmenopausal women (n = 11). Protein expression of estrogen receptor α, estrogen receptor ß, and phosphorylated extracellular signal-regulated kinase and protein kinase B were analyzed. Matrix metalloproteinase-2 activity was determined as a measure of tumor invasiveness. Mitochondrial retrograde signaling was altered with ethidium bromide to determine its effect on estrogen receptor α protein expression. RESULTS: Estrogen receptor α expression was increased in postmenopausal papillary thyroid cancer cells compared with controls but was unchanged in premenopausal papillary thyroid cancer. Estrogen receptor ß expression did not change in either group. Increased matrix metalloproteinase-2 activity was observed only in postmenopausal papillary thyroid cancer. Premenopausal papillary thyroid cancer cells demonstrated increased extracellular signal-regulated kinase and unchanged protein kinase B activation. Conversely, postmenopausal papillary thyroid cancer cells had decreased extracellular signal-regulated kinase and increased protein kinase B activation. Ethidium bromide treatment resulted in increased estrogen receptor α protein expression only in premenopausal papillary thyroid cancer cells. CONCLUSION: Increased estrogen receptor α expression may be involved in papillary thyroid cancer aggressiveness after menopause. This process may be regulated by differential activation of intracellular pathways and differing sensitivities to mitochondrial signaling regulation.


Subject(s)
Carcinoma, Papillary/metabolism , DNA, Mitochondrial/metabolism , Postmenopause/metabolism , Premenopause/metabolism , Receptors, Estrogen/metabolism , Thyroid Neoplasms/metabolism , Adult , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Matrix Metalloproteinase 2/metabolism , Middle Aged , Primary Cell Culture , Proto-Oncogene Proteins c-akt/metabolism , Thyroid Cancer, Papillary , Thyroid Gland/metabolism , Young Adult
7.
Clin Geriatr Med ; 33(4): 473-490, 2017 11.
Article in English | MEDLINE | ID: mdl-28991645

ABSTRACT

Natural lung aging is characterized by molecular and cellular changes in multiple lung cell populations. These changes include shorter telomeres, increased expression of cellular senescence markers, increased DNA damage, oxidative stress, apoptosis, and stem cell exhaustion. Aging, combined with the loss of protective repair processes, correlates with the development and incidence of chronic respiratory diseases, including idiopathic pulmonary fibrosis and chronic obstructive pulmonary disease. Ultimately, it is the interplay of age-related changes in biology and the subsequent responses to environmental exposures that largely define the physiology and clinical course of the aging lung.


Subject(s)
Aging/physiology , Cellular Senescence/physiology , Lung Diseases , Lung , Aged , Environmental Exposure/adverse effects , Humans , Lung/metabolism , Lung/pathology , Lung Diseases/diagnosis , Lung Diseases/etiology , Lung Diseases/physiopathology
8.
Front Med (Lausanne) ; 4: 118, 2017.
Article in English | MEDLINE | ID: mdl-28804709

ABSTRACT

Large multicenter clinical trials have led to two recently approved drugs for patients with idiopathic pulmonary fibrosis (IPF); yet, both of these therapies only slow disease progression and do not provide a definitive cure. Traditionally, preclinical trials have utilized mouse models of bleomycin (BLM)-induced pulmonary fibrosis-though several limitations prevent direct translation to human IPF. Spontaneous pulmonary fibrosis occurs in other animal species, including dogs, horses, donkeys, and cats. While the fibrotic lungs of these animals share many characteristics with lungs of patients with IPF, current veterinary classifications of fibrotic lung disease are not entirely equivalent. Additional studies that profile these examples of spontaneous fibroses in animals for similarities to human IPF should prove useful for both human and animal investigators. In the meantime, studies of BLM-induced fibrosis in aged male mice remain the most clinically relevant model for preclinical study for human IPF. Addressing issues such as time course of treatment, animal size and characteristics, clinically irrelevant treatment endpoints, and reproducibility of therapeutic outcomes will improve the current status of preclinical studies. Elucidating the mechanisms responsible for the development of fibrosis and disrepair associated with aging through a collaborative approach between researchers will promote the development of models that more accurately represent the realm of interstitial lung diseases in humans.

9.
Transl Res ; 178: 107-117, 2016 12.
Article in English | MEDLINE | ID: mdl-27519148

ABSTRACT

Female smokers have a faster decline in lung function with increasing age and overall develop a greater loss of lung function than male smokers. This raises the question of whether estrogen status in women affects susceptibility to cigarette smoke (CS)-induced lung disease. Mouse models suggest that female mice are more susceptible than males to CS-induced lung disease. Moreover, young CS-exposed female mice develop emphysema earlier than male mice. The purpose of this study was to characterize the relationship of estrogen status on the pattern and severity of CS-induced lung disease. In this study, 15-month-old female C57BL/6J mice were ovariectomized and administered either placebo (pla) or 17ß-estradiol (E2, 0.025 mg) 2 weeks after ovariectomy. They were further divided into those that were exposed to CS and no-smoke controls (NSC). Mice were exposed to CS in stainless steel inhalation chambers 3 hours a day, 5 days a week for 6 months, and sacrificed after 24 weeks of CS exposure. Blood and urine were collected at sacrifice to measure estrogen and cotinine levels, a metabolite of nicotine. Uterine weight was recorded as an indicator of estrogen status. Results showed that CS in the absence of E2 induced a decrease in hydroxyproline content, macrophage number, and respiratory chain complex-1 protein. CS without E2 also resulted in an increase in matrix metalloproteinase-2 activity and apoptosis and a change in the ratio of estrogen receptor subtype. These findings were abrogated with administration of E2, suggesting that estrogen deficiency increases susceptibility to CS-induced lung disease.


Subject(s)
Aging/pathology , Estrogens/deficiency , Extracellular Matrix/metabolism , Lung/pathology , Smoking/adverse effects , Animals , Apoptosis , Body Weight , Cotinine/urine , Electron Transport , Female , Hydroxyproline/metabolism , Lung/enzymology , Macrophages/metabolism , Matrix Metalloproteinase 2/metabolism , Mice, Inbred C57BL , Organ Size , Oxidative Stress , Receptors, Estrogen/metabolism , Uterus/pathology
10.
PLoS One ; 11(7): e0159666, 2016.
Article in English | MEDLINE | ID: mdl-27428057

ABSTRACT

Age-related increases in oxidant stress (OS) play a role in regulation of estrogen receptor (ER) expression in the kidneys. In this study, we establish that in vivo 17ß-estradiol (E2) replacement can no longer upregulate glomerular ER expression by 21 months of age in female mice (anestrous). We hypothesized that advanced glycation end product (AGE) accumulation, an important source of oxidant stress, contributes to these glomerular ER expression alterations. We treated 19-month old ovariectomized female mice with pyridoxamine (Pyr), a potent AGE inhibitor, in the presence or absence of E2 replacement. Glomerular ERα mRNA expression was upregulated in mice treated with both Pyr and E2 replacement and TGFß mRNA expression decreased compared to controls. Histological sections of kidneys demonstrated decreased type IV collagen deposition in mice receiving Pyr and E2 compared to placebo control mice. In addition, anti-AGE defenses Sirtuin1 (SIRT1) and advanced glycation receptor 1 (AGER1) were also upregulated in glomeruli following treatment with Pyr and E2. Mesangial cells isolated from all groups of mice demonstrated similar ERα, SIRT1, and AGER1 expression changes to those of whole glomeruli. To demonstrate that AGE accumulation contributes to the observed age-related changes in the glomeruli of aged female mice, we treated mesangial cells from young female mice with AGE-BSA and found similar downregulation of ERα, SIRT1, and AGER1 expression. These results suggest that inhibition of intracellular AGE accumulation with pyridoxamine may protect glomeruli against age-related oxidant stress by preventing an increase of TGFß production and by regulation of the estrogen receptor.


Subject(s)
Aging/metabolism , Antioxidants/pharmacology , Estrogen Receptor alpha/genetics , Glycation End Products, Advanced/antagonists & inhibitors , Kidney Glomerulus/drug effects , Pyridoxamine/pharmacology , Aging/genetics , Animals , Collagen Type IV/genetics , Collagen Type IV/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation , Glycation End Products, Advanced/genetics , Glycation End Products, Advanced/metabolism , Glycation End Products, Advanced/pharmacology , Hormone Replacement Therapy , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Mesangial Cells/pathology , Mice , Mice, Inbred C57BL , Ovariectomy , Oxidative Stress , Primary Cell Culture , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism , Serum Albumin, Bovine/antagonists & inhibitors , Serum Albumin, Bovine/pharmacology , Signal Transduction , Sirtuin 1/genetics , Sirtuin 1/metabolism , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
12.
Transl Res ; 166(6): 554-67, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26432923

ABSTRACT

The observation that pulmonary inflammatory lesions and bleomycin (BLM)-induced pulmonary fibrosis spontaneously resolve in young mice, whereas remaining irreversible in aged mice suggests that impairment of pulmonary regeneration and repair is associated with aging. Because mesenchymal stem cells (MSCs) may promote repair after injury, we postulated that differences in MSCs from aged mice may underlie postinjury fibrosis in aging. The potential for young-donor MSCs to inhibit BLM-induced pulmonary fibrosis in aged male mice (>22 months) has not been studied. Adipose-derived MSCs (ASCs) from young (4 months) and old (22 months) male mice were infused 1 day after intratracheal BLM administration. At 21-day sacrifice, aged BLM mice demonstrated lung fibrosis by Ashcroft score, collagen content, and α(v)-integrin messenger RNA (mRNA) expression. Lung tissue from aged BLM mice receiving young ASCs exhibited decreased fibrosis, matrix metalloproteinase (MMP)-2 activity, oxidative stress, and markers of apoptosis vs BLM controls. Lung mRNA expression of tumor necrosis factor-alpha was also decreased in aged BLM mice receiving young-donor ASCs vs BLM controls. In contrast, old-donor ASC treatment in aged BLM mice did not reduce fibrosis and related markers. On examination of the cells, young-donor ASCs had decreased mRNA expression of MMP-2, insulin-like growth factor (IGF) receptor, and protein kinase B (AKT) activation compared with old-donor ASCs. These results show that the BLM-induced pulmonary fibrosis in aged mice could be blocked by young-donor ASCs and that the mechanisms involve changes in collagen turnover and markers of inflammation.


Subject(s)
Adipose Tissue/cytology , Age Factors , Bleomycin/toxicity , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Pulmonary Fibrosis/therapy , Animals , Biomarkers/metabolism , Disease Models, Animal , Enzyme Activation , In Situ Nick-End Labeling , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Fibrosis/chemically induced
13.
Horm Cancer ; 6(2-3): 100-6, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25752763

ABSTRACT

Papillary thyroid cancer (PTC) is the most prevalent of all endocrine cancers. In recent studies, the presence of receptors for pituitary-type growth hormone-releasing hormone (pGHRH-R) has been demonstrated in various human cancers, including human prostate, brain, and other cancer lines. Thyroid malignancies, however, have not yet been investigated in this regard. In this study, we found that pGHRH-R and its functional splice variant, SV1, are present in normal thyroid and PTC cells. We also treated seven normal and PTC tumor thyroid cells in vitro with a GHRH antagonist, MIA-602, to compare its anti-proliferation and anti-invasion potential against vehicle-treated cells. We found that treatment with GHRH antagonist increases the expression of SV1 and pGHRH-R in tumor cells compared to tumor cells exposed to vehicle only, a response which may alter the sensitivity of signaling kinases within the cells. GHRH antagonist treatment of tumor cells also reduced activity of the tumor invasion marker, matrix metalloproteinase (MMP)-2, compared to tumor cells exposed to vehicle only. The expression of pGHRH-R and SV1, as well as MMP-2 activity, in normal thyroid cells remained unaffected by GHRH antagonist treatment. Similarly, cell proliferation rates for tumor or normal thyroid cells were not affected by GHRH antagonist treatment. Our findings have important implications for the therapeutic use of GHRH antagonist in cases of aggressive PTC refractory to conventional treatment modalities, and in which protein expression and MMP-2 activity in normal thyroid tissue is left unaltered.


Subject(s)
Carcinoma/metabolism , Matrix Metalloproteinase 2/metabolism , Receptors, Neuropeptide/biosynthesis , Receptors, Pituitary Hormone-Regulating Hormone/biosynthesis , Thyroid Neoplasms/metabolism , Blotting, Western , Carcinoma, Papillary , Cell Proliferation , Female , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Humans , Middle Aged , Sermorelin/analogs & derivatives , Sermorelin/pharmacology , Thyroid Cancer, Papillary
14.
Endocrinology ; 155(2): 441-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24274985

ABSTRACT

The role that estrogens play in the aging lung is poorly understood. Remodeling of the aging lung with thickening of the alveolar walls and reduction in the number of peripheral airways is well recognized. The present study was designed to address whether estrogen deficiency would affect age-associated changes in the lungs of female C57BL/6J mice. Lungs isolated from old mice (24 months old, estrogen-deficient) demonstrated decreased lung volume and decreased alveolar surface area. There was no difference in alveolar number in the lungs of old and young mice (6 months old, estrogen-replete). Estrogen replacement restored lung volume, alveolar surface area, and alveolar wall thickness to that of a young mouse. Estrogen receptor-α (ERα) protein expression increased without a change in ERß protein expression in the lung tissue isolated from old mice. In the lungs of old mice, the number of apoptotic cells was increased as well as the activation of matrix metalloproteinase-2 and ERK. Young mice had the highest serum 17ß-estradiol levels that decreased with age. Our data suggest that in the aging female mouse lung, estrogen deficiency and an increase of ERα expression lead to the development of an emphysematous phenotype. Estrogen replacement partially prevents these age-associated changes in the lung architecture by restoration of interalveolar septa. Understanding the role of estrogens in the remodeling of the lung during aging may facilitate interventions and therapies for aging-related lung disease in women.


Subject(s)
Aging/physiology , Estradiol/therapeutic use , Estrogen Replacement Therapy , Lung Diseases/drug therapy , Lung/drug effects , Aging/metabolism , Animals , Apoptosis/drug effects , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Lung/metabolism , Lung/pathology , Lung Diseases/metabolism , Lung Diseases/pathology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Organ Size/drug effects , Ovariectomy , RNA, Messenger/genetics , RNA, Messenger/metabolism
15.
Lab Invest ; 92(2): 236-45, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22042083

ABSTRACT

Pentosan polysulfate (PPS), a heparinoid compound essentially devoid of anticoagulant activity, modulates cell growth and decreases inflammation. We investigated the effect of PPS on the progression of established atherosclerosis in Watanabe heritable hyperlipidemic (WHHL) rabbits. After severe atherosclerosis developed on an atherogenic diet, WHHL rabbits were treated with oral PPS or tap water for 1 month. The aortic intima-to-media ratio and macrophage infiltration were reduced, plaque collagen content was increased, and plaque fibrous caps were preserved by PPS treatment. Plasma lipid levels and post-heparin hepatic lipase activity remained unchanged. However, net collagenolytic activity in aortic extracts was decreased, and the levels of matrix metalloproteinase (MMP)-2 and tissue inhibitor of metalloproteinase (TIMP) activity were increased by PPS. Moreover, PPS treatment decreased tumor necrosis factor α (TNFα)-stimulated proinflammatory responses, in particular activation of nuclear factor-κB and p38, and activation of MMPs in macrophages. In conclusion, oral PPS treatment prevents progression of established atherosclerosis in WHHL rabbits. This effect may be partially mediated by increased MMP-2 and TIMP activities in the aortic wall and reduced TNFα-stimulated inflammation and MMP activation in macrophages. Thus, PPS may be a useful agent in inhibiting the progression of atherosclerosis.


Subject(s)
Atherosclerosis/prevention & control , Hyperlipidemias/complications , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Pentosan Sulfuric Polyester/pharmacology , Animals , Atherosclerosis/complications , Atherosclerosis/enzymology , Cell Line , Enzyme Activation , Female , Humans , Hyperlipidemias/enzymology , Immunohistochemistry , Lipids/blood , Mice , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Rabbits , Tumor Necrosis Factor-alpha/pharmacology
16.
Kidney Int ; 79(4): 404-13, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20962747

ABSTRACT

Podocyte damage and apoptosis are thought to be important if not essential in the development of glomerulosclerosis. Female estrogen receptor knockout mice develop glomerulosclerosis at 9 months of age due to excessive ovarian testosterone production and secretion. Here, we studied the pathogenesis of glomerulosclerosis in this mouse model to determine whether testosterone and/or 17ß-estradiol directly affect the function and survival of podocytes. Glomerulosclerosis in these mice was associated with the expression of desmin and the loss of nephrin, markers of podocyte damage and apoptosis. Ovariectomy preserved the function and survival of podocytes by eliminating the source of endogenous testosterone production. In contrast, testosterone supplementation induced podocyte apoptosis in ovariectomized wild-type mice. Importantly, podocytes express functional androgen and estrogen receptors, which, upon stimulation by their respective ligands, have opposing effects. Testosterone induced podocyte apoptosis in vitro by androgen receptor activation, but independent of the TGF-ß1 signaling pathway. Pretreatment with 17ß-estradiol prevented testosterone-induced podocyte apoptosis, an estrogen receptor-dependent effect mediated by activation of the ERK signaling pathway, and protected podocytes from TGF-ß1- or TNF-α-induced apoptosis. Thus, podocytes are target cells for testosterone and 17ß-estradiol. These hormones modulate podocyte damage and apoptosis.


Subject(s)
Estradiol/pharmacology , Estrogen Receptor alpha/deficiency , Glomerulosclerosis, Focal Segmental/etiology , Podocytes/drug effects , Testosterone/pharmacology , Animals , Apoptosis/drug effects , Cell Dedifferentiation/drug effects , Desmin/metabolism , Estradiol/physiology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/physiology , Estrogen Receptor beta/metabolism , Female , Gene Expression/drug effects , Glomerulosclerosis, Focal Segmental/pathology , Glomerulosclerosis, Focal Segmental/physiopathology , In Vitro Techniques , Membrane Proteins/metabolism , Mice , Mice, Knockout , Ovariectomy , Podocytes/pathology , Podocytes/physiology , Receptors, Androgen/metabolism , Smad7 Protein/genetics , Testosterone/physiology , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
17.
Curr Diabetes Rev ; 7(1): 28-34, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21067504

ABSTRACT

It is generally accepted that estrogens affect and modulate the development and progression of chronic kidney diseases (CKD) not related to diabetes. Clinical studies have indeed demonstrated that the severity and rate of progression of renal damage tends to be greater among men, compared with women. Experimental studies also support the notion that female sex is protective and male sex permissive, for the development of CKD in non-diabetics, through the opposing actions of estrogens and testosterone. However, when we consider diabetes-induced kidney damage, in the setting of either type 1 or type 2 diabetes, the contribution of gender to the progression of renal disease is somewhat uncertain. Previous studies on the effects of estrogens in the pathogenesis of progressive kidney damage have primarily focused on mesangial cells. More recently, data on the effects of estrogens on podocytes, the cell type whose role may include initiation of progressive diabetic renal disease, became available. The aim of this review will be to summarize the main clinical and experimental data on the effects of estrogens on the progression of diabetes-induced kidney injury. In particular, we will highlight the possible biological effects of estrogens on podocytes, especially considering those critical for the pathogenesis of diabetic kidney damage.


Subject(s)
Diabetic Nephropathies/etiology , Estrogens/physiology , Animals , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/physiopathology , Disease Progression , Female , Humans , Kidney Diseases/etiology , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Diseases/physiopathology , Male , Models, Biological , Receptors, Estrogen/agonists , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism
18.
Exp Eye Res ; 90(1): 10-6, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19799898

ABSTRACT

Epidemiological data suggest that estrogen deficiency in postmenopausal women may contribute to the severity of AMD. We discovered that 17beta-estradiol (E2) was a crucial regulator of the severity of extracellular matrix turnover (ECM) dysregulation both in vivo and in vitro. We also found in vitro that the presence of estrogen receptor (ER)beta regulates MMP-2 activity. Therefore in an attempt to delineate the role of the ER subtypes, female estrogen receptor knockout (ERKO) mice were fed a high-fat diet, and the eyes were exposed to seven 5-second doses of nonphototoxic levels of blue-green light over 2 weeks. Three months after cessation of blue light treatment, transmission electron microscopy was performed to assess severity of deposits, Bruchs membrane changes, and choriocapillaris endothelial morphology. We found that changes in the trimolecular complex of pro-MMP-2, MMP-14 and TIMP-2 correlated with increased Bruch's membrane thickening or sub-retinal deposit formation (basal laminar deposits) in ERKObeta mice. In addition RPE isolated from ERKObeta mice had an increase in expression of total collagen and a decrease in MMP-2 activity. Finally we found that ERK an intermediate signaling molecule in the MMP pathway was activated in RPE isolated from ERKObeta mice. These data suggest that mice which lack ERbeta are more susceptible to in vivo injury associated with environmental light and high fat diet.


Subject(s)
Estrogen Receptor beta/physiology , Extracellular Matrix/metabolism , Macular Degeneration/prevention & control , Oxidative Stress , Retinal Pigment Epithelium/metabolism , Animals , Blotting, Western , Bruch Membrane/metabolism , Bruch Membrane/ultrastructure , Cell Culture Techniques , Collagen/metabolism , Dietary Fats/administration & dosage , Extracellular Matrix/ultrastructure , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , In Situ Hybridization , Light , Macular Degeneration/metabolism , Macular Degeneration/pathology , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Oligonucleotide Array Sequence Analysis , Retinal Pigment Epithelium/ultrastructure , Reverse Transcriptase Polymerase Chain Reaction , Tissue Inhibitor of Metalloproteinase-2/metabolism
19.
Wounds ; 22(7): 171-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-25901496

ABSTRACT

UNLABELLED: Postmenopausal women are more susceptible to poor wound healing. This phenomenon can be reversed by estrogen replacement therapy in non-diabetic individuals. Postmenopausal women with type 2 diabetes are more susceptible to wound healing complications, potentially secondary to an estrogen deficiency. Few studies have examined the mechanism of action and effects of estrogens on diabetic wound healing in females. It appears that multiple factors influence delayed wound healing among individuals with diabetes including: an imbalance in cytokines, growth factors, extracellular matrix (ECM) turnover, and oxidant stress (OS). Estrogens have been shown to regulate the expression of genes important for extracellular matrix turnover, including collagen and matrix metalloproteinases (MMP). METHODS: For this reason, the effects of 17ß-estradiol (E2) on MMP-2, MMP-13, and MMP-14 and estrogen receptor alpha and beta (ER-α and -ß) expression in the wound tissue of estrogen-deficient female mice with established type 2 diabetes mellitus (C57BL/6J-m Leprdb/2+) were studied. RESULTS: Topical E2 upregulates ERα in wound tissue thereby improving and accelerating diabetic wound healing in estrogen deficient mice. CONCLUSION: The mechanism appears to decrease MMP-2, MMP-13, and MMP-14 mediated tissue matrix destruction and increasing collagen content. .

20.
Ann Plast Surg ; 63(3): 318-22, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19602952

ABSTRACT

Estrogen deficiency may contribute to extracellular matrix turnover in skin. This has led previous authors to postulate that aged skin heals less efficiently when compared to younger skin. Also, cigarette smokers have been shown to heal less efficiently than nonsmokers. Matrix metalloproteinase (MMP)-13, an enzyme that participates in the degradation of the extracellular matrix, has been implicated in physiologic aging and wound healing. This study investigates the effects of smoke exposure and estrogen deficiency on MMP-13 in young and aged female mouse skin. Young and aged female C57Bl/6J mice were ovariectomized. They were then randomly administered either 17beta-estradiol (E2) or placebo pellets. Half the animals in each age group were further randomized to exposure to cigarette smoke for a period of 6 months. Smoking and estrogen deficiency increased MMP-13 protein and activity in aged skin. The tissue inhibitors of metalloproteinases, which inhibit MMPs, activity was unchanged across all groups. E2 replacement decreased the actual level of MMP-13 protein and activity. We also found an increased collagen content and decreased ER receptor protein level in aged, smoke-exposed female mice. Our experimental data show that tobacco smoke exposure and estrogen deficiency are additive risk factors for promoting increased activity of MMP-13 in aged skin. These findings suggest that MMP-13 functions as a mediator of smoke-induced skin injury in susceptible, aged experimental female mice.


Subject(s)
Estradiol/pharmacology , Estrogens/deficiency , Matrix Metalloproteinase 13/metabolism , Skin Aging/physiology , Smoke/adverse effects , Animals , Disease Models, Animal , Enzyme Activation , Female , Hydroxyproline/metabolism , Mice , Mice, Inbred C57BL , Ovariectomy , Random Allocation , Reference Values , Risk Factors , Sensitivity and Specificity , Skin Aging/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...