Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Gynecol Oncol ; 186: 17-25, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38554625

ABSTRACT

OBJECTIVE: Histopathologic characteristics after neoadjuvant chemotherapy (NACT) may correlate with outcome. This study evaluates histopathologic features after immunotherapy and NACT/bevacizumab, and associated clinical outcomes. METHODS: Evaluable tissue from IMagyn050/GOG3015/ENGOT-ov39 patients from prespecified anatomic sites from interval cytoreductive surgery (ICS) after NACT/bevacizumab plus atezolizumab/placebo underwent central histopathologic scoring and analyzed with clinical outcomes. RESULTS: The predefined population had 243 evaluable NACT patients, with 48.1% tumors being PD-L1-positive. No statistically significant differences in PFS (16.9 months vs. 19.2 months, p = 0.21) or OS (41.5 months vs. 45.1 months, p = 0.67) between treatment arms were seen. Substantial residual tumor (RT) (3+) was identified in 26% atezolizumab vs. 24% placebo arms (p = 0.94). Most showed no (1+) necrosis (82% vs. 96%, respectively, p = 0.69), moderate (2+) to severe (3+) fibrosis (71% vs. 75%, respectively, p = 0.82), and extensive (2+) inflammation (53% vs. 47% respectively, p = 0.48). No significant histopathologic differences were identified by tissue site or by arm. Multivariate analyses showed increased risk for progression with moderate and substantial RT (13.6 mon vs. 21.1 mon, hazard ratio 2.0, p < 0.01; 13.6 mon vs. 21.1 mon, HR 1.9, p < 0.01, respectively); but decreased risk for death with extensive inflammation (46.9 mon vs. 36.3 mon, HR 0.65, p = 0.02). Inflammation also correlated with greater likelihood of response to NACT/bevacizumab plus immunotherapy (odds ratio 2.9, p < 0.01). Modeling showed inflammation as a consistent but modest predictor for OS. CONCLUSIONS: Detailed histologic assessment of ICS specimens appear to identify characteristics, such as inflammation and residual tumor, that may provide insight to certain clinical outcomes. Future work potentially leveraging emerging tools may provide further insight into outcomes.

2.
Gynecol Oncol ; 177: 20-31, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37625235

ABSTRACT

OBJECTIVE: To determine the impact on overall survival (OS) and patient-reported outcomes (PROs) of combining atezolizumab with standard therapy for newly diagnosed stage III/IV ovarian cancer. METHODS: The placebo-controlled double-blind randomized phase III IMagyn050/GOG 3015/ENGOT-OV39 trial (NCT03038100) assigned eligible patients to 3-weekly atezolizumab 1200 mg or placebo for 22 cycles with platinum-based chemotherapy and bevacizumab. Coprimary endpoints were progression-free survival (already reported) and OS in the PD-L1-positive and intent-to-treat (ITT) populations, tested hierarchically. Prespecified PRO analyses focused on disease-related abdominal pain and bloating symptoms (European Organisation for Research and Treatment of Cancer QLQ-OV28), functioning, and health-related quality of life (HRQoL) (QLQ-C30). RESULTS: After 38 months' median follow-up, the OS hazard ratio in the PD-L1-positive population was 0.83 (95% CI, 0.66-1.06; p = 0.13); median OS was not estimable with atezolizumab versus 49.2 months with placebo. The hazard ratio for OS in the ITT population was 0.92 (95% CI, 0.78-1.09; median 50.5 versus 46.6 months, respectively). At week 9, similar proportions of patients in both arms of the neoadjuvant cohort showed ≥10-point improvement from baseline in abdominal pain and bloating, functioning, and HRQoL. In the primary surgery cohort, similar proportions of patients in each arm had improved, stable, or worsened physical and role function and HRQoL from baseline over time. Neither cohort showed differences between arms in treatment-related symptoms or overall side-effect bother. CONCLUSIONS: Incorporation of atezolizumab into standard therapy for newly diagnosed ovarian cancer does not significantly improve efficacy or impose additional treatment burden for patients. CLINICALTRIALS: gov registration: NCT03038100.


Subject(s)
Ovarian Neoplasms , Quality of Life , Humans , Female , B7-H1 Antigen , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/etiology , Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/etiology , Patient Reported Outcome Measures , Abdominal Pain/etiology , Antineoplastic Combined Chemotherapy Protocols/adverse effects
3.
Clin Cancer Res ; 29(9): 1698-1707, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36595569

ABSTRACT

PURPOSE: To explore whether patients with BRCA1/2-mutated or homologous recombination deficient (HRD) ovarian cancers benefitted from atezolizumab in the phase III IMagyn050 (NCT03038100) trial. PATIENTS AND METHODS: Patients with newly diagnosed ovarian cancer were randomized to either atezolizumab or placebo with standard chemotherapy and bevacizumab. Programmed death-ligand 1 (PD-L1) status of tumor-infiltrating immune cells (IC) was determined centrally (VENTANA SP142 assay). Genomic alterations, including deleterious BRCA1/2 alterations, genomic loss of heterozygosity (gLOH), tumor mutation burden (TMB), and microsatellite instability (MSI), were evaluated using the FoundationOne assay. HRD was defined as gLOH ≥ 16%, regardless of BRCA1/2 mutation status. Potential associations between progression-free survival (PFS) and genomic biomarkers were evaluated using standard correlation analyses and log-rank of Kaplan-Meier estimates. RESULTS: Among biomarker-evaluable samples, 22% (234/1,050) harbored BRCA1/2 mutations and 46% (446/980) were HRD. Median TMB was low irrespective of BRCA1/2 or HRD. Only 3% (29/1,024) had TMB ≥10 mut/Mb, and 0.3% (3/1,022) were MSI-high. PFS was better in BRCA2-mutated versus BRCA2-non-mutated tumors and in HRD versus proficient tumors. PD-L1 positivity (≥1% expression on ICs) was associated with HRD but not BRCA1/2 mutations. PFS was not improved by adding atezolizumab in BRCA2-mutated or HRD tumors; there was a trend toward enhanced PFS with atezolizumab in BRCA1-mutated tumors. CONCLUSIONS: Most ovarian tumors have low TMB despite BRCA1/2 mutations or HRD. Neither BRCA1/2 mutation nor HRD predicted enhanced benefit from atezolizumab. This is the first randomized double-blind trial in ovarian cancer demonstrating that genomic instability triggered by BRCA1/2 mutation or HRD is not associated with improved sensitivity to immune checkpoint inhibitors. See related commentary by Al-Rawi et al., p. 1645.


Subject(s)
B7-H1 Antigen , Ovarian Neoplasms , Humans , Female , B7-H1 Antigen/genetics , Mutation , Double-Blind Method , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/therapeutic use , Genomics , Immunotherapy
4.
J Clin Oncol ; 39(17): 1842-1855, 2021 06 10.
Article in English | MEDLINE | ID: mdl-33891472

ABSTRACT

PURPOSE: To evaluate the addition of the humanized monoclonal antiprogrammed death ligand-1 (PD-L1) antibody, atezolizumab, to platinum-based chemotherapy and bevacizumab in newly diagnosed stage III or IV ovarian cancer (OC). METHODS: This multicenter placebo-controlled double-blind randomized phase III trial (ClinicalTrials.gov identifier: NCT03038100) enrolled patients with newly diagnosed untreated International Federation of Gynecology and Obstetrics (FIGO) stage III or IV OC who either had undergone primary cytoreductive surgery with macroscopic residual disease or were planned to receive neoadjuvant chemotherapy and interval surgery. Patients were stratified by FIGO stage, Eastern Cooperative Oncology Group performance status, tumor immune cell PD-L1 staining, and treatment strategy and randomly assigned 1:1 to receive 3-weekly cycles of atezolizumab 1,200 mg or placebo (day 1, cycles 1-22), with paclitaxel plus carboplatin (day 1, cycles 1-6) plus bevacizumab 15 mg/kg (day 1, cycles 2-22), omitting perioperative bevacizumab in neoadjuvant patients. The co-primary end points were investigator-assessed progression-free survival and overall survival in the intention-to-treat and PD-L1-positive populations. RESULTS: Between March 8, 2017, and March 26, 2019, 1,301 patients were enrolled. The median progression-free survival was 19.5 versus 18.4 months with atezolizumab versus placebo, respectively (hazard ratio, 0.92; 95% CI, 0.79 to 1.07; stratified log-rank P = .28), in the intention-to-treat population and 20.8 versus 18.5 months, respectively (hazard ratio, 0.80; 95% CI, 0.65 to 0.99; P = .038), in the PD-L1-positive population. The interim (immature) overall survival results showed no significant benefit from atezolizumab. The most common grade 3 or 4 adverse events were neutropenia (21% with atezolizumab v 21% with placebo), hypertension (18% v 20%, respectively), and anemia (12% v 12%). CONCLUSION: Current evidence does not support the use of immune checkpoint inhibitors in newly diagnosed OC. Insight from this trial should inform further evaluation of immunotherapy in OC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Ovarian Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , Adolescent , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/administration & dosage , B7-H1 Antigen/metabolism , Bevacizumab/administration & dosage , Carboplatin/administration & dosage , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , Carcinoma, Ovarian Epithelial/surgery , Cytoreduction Surgical Procedures , Double-Blind Method , Female , Humans , Middle Aged , Neoadjuvant Therapy , Neoplasm Staging , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/surgery , Paclitaxel/administration & dosage , Young Adult
5.
Mol Endocrinol ; 30(2): 234-47, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26771535

ABSTRACT

Vesicular transport involving soluble N-ethylmaleimide sensitive factor attachment protein receptor (SNARE) proteins is known to be responsible for many major cellular activities. In steroidogenic tissues, chronic hormone stimulation results in increased expression of proteins involved in the steroidogenic pathway, whereas acute hormone stimulation prompts the rapid transfer of cholesterol to the inner mitochondrial membrane to be utilized as substrate for steroid hormone production. Several different pathways are involved in supplying cholesterol to mitochondria, but mobilization of stored cholesteryl esters appears to initially constitute the preferred source; however, the mechanisms mediating this cholesterol transfer are not fully understood. To study the potential contribution of SNARE proteins in steroidogenesis, we examined the expression levels of various SNARE proteins in response to hormone stimulation in steroidogenic tissues and cells and established an in vitro mitochondria reconstitution assay system to assess the contribution of various SNARE proteins on cholesterol delivery for steroidogenesis. Our results from reconstitution experiments along with knockdown studies in rat primary granulosa cells and in a Leydig cell line show that soluble N-ethylmaleimide sensitive factor attachment protein-α, synaptosomal-associated protein of 25 kDa, syntaxin-5, and syntaxin-17 facilitate the transport of cholesterol to mitochondria. Thus, although StAR is required for efficient cholesterol movement into mitochondria for steroidogenesis, specific SNAREs participate and are necessary to mediate cholesterol movement to mitochondria.


Subject(s)
Cholesterol/metabolism , Mitochondria/metabolism , SNARE Proteins/metabolism , Steroids/biosynthesis , Animals , Biological Transport/drug effects , Cell Line , Female , Gene Knockdown Techniques , Mice, Inbred C57BL , Mitochondria/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , SNARE Proteins/genetics , Steroids/metabolism
6.
PLoS One ; 9(8): e105047, 2014.
Article in English | MEDLINE | ID: mdl-25111084

ABSTRACT

Within cells, lipids are stored in the form of lipid droplets (LDs), consisting of a neutral lipid core, surrounded by a phospholipid monolayer and an outer layer of protein. LDs typically accumulate either triacylglycerol (TAG) and diacylglycerol or cholesteryl ester (CE), depending on the type of tissue. Recently, there has been an increased interest in the proteins that surround LDs. LD proteins have been found to be quite diverse, from structural proteins to metabolic enzymes, proteins involved in vesicular transport, and proteins that may play a role in LD formation. Previous proteomics analyses have focused on TAG-enriched LDs, whereas CE-enriched LDs have been largely ignored. Our study has compared the LD proteins from CE-enriched LDs to TAG-enriched LDs in steroidogenic cells. In primary rat granulosa cells loaded with either HDL to produce CE-enriched LDs or fatty acids to produce TAG-enriched LDs, 61 proteins were found to be elevated in CE-enriched LDs and 40 proteins elevated in TAG-enriched LDs with 278 proteins in similar amounts. Protein expression was further validated by selected reaction monitoring (SRM) mass spectrometry (MS). SRM verified expression of 25 of 27 peptides that were previously detected by tandem mass tagging MS. Several proteins were confirmed to be elevated in CE-enriched LDs by SRM including the intermediate filament vimentin. This study is the first to compare the proteins found on CE-enriched LDs with TAG-enriched LDs and constitutes the first step in creating a better understanding of the proteins found on CE-enriched LDs in steroidogenic cells.


Subject(s)
Cholesterol Esters/metabolism , Lipid Droplets/metabolism , Proteins/analysis , Proteome/analysis , Triglycerides/metabolism , Animals , Cells, Cultured , Drosophila , Female , Gene Expression Profiling , Granulosa Cells/metabolism , Lipid Metabolism , Proteome/metabolism , Proteomics/methods , Rats , Rats, Sprague-Dawley , Tandem Mass Spectrometry
7.
Curr Opin Clin Nutr Metab Care ; 16(6): 632-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24100667

ABSTRACT

PURPOSE OF REVIEW: With the realization that lipid droplets are not merely inert fat storage organelles, but highly dynamic and actively involved in cellular lipid homeostasis, there has been an increased interest in lipid droplet biology. Recent studies have begun to unravel the roles that lipid dropletss play in cellular physiology and provide insights into the mechanisms by which lipid droplets contribute to cellular homeostasis. This review provides a summary of these recent publications on lipid droplet metabolism. RECENT FINDINGS: Perilipins have different preferences for associating with triacylglycerol (TAG) or cholesteryl esters, different tissue distributions, and each contributes to lipid metabolism in its unique way. Cell death-inducing DFF45-like effector proteins are not only involved in lipid droplet expansion, but also in the cellular response to stress and lipid secretion. Lipid droplets undergo an active cycle of lipolysis and re-esterification to form microlipid droplets. TAG synthesis for lipid droplet formation and expansion occurs in the endoplasmic reticulum and on lipid droplets, and TAG transfers between lipid droplets during lipid droplet fusion. Lipid droplets interact with the endoplasmic reticulum and mitochondria to facilitate lipid transfer, lipid droplet expansion, and metabolism. SUMMARY: Lipid droplets are dynamically active, responding to changes in cellular physiology, as well as interacting with cytosolic proteins and other organelles to control lipid homeostasis.


Subject(s)
Lipid Metabolism/physiology , Apoptosis Regulatory Proteins , Carrier Proteins/metabolism , Cell Death , Cholesterol Esters/metabolism , Endoplasmic Reticulum/metabolism , Homeostasis , Humans , Lipolysis , Mitochondria/metabolism , Organelles/metabolism , Perilipin-1 , Phosphoproteins/metabolism , Proteins/metabolism , Triglycerides/metabolism
8.
Mol Cell Endocrinol ; 371(1-2): 15-9, 2013 May 22.
Article in English | MEDLINE | ID: mdl-23089211

ABSTRACT

Intracellular lipid droplets (LDs) are dynamic organelles that contain a number of associated proteins including perilipin (Plin) and vimentin. Cholesteryl ester (CE)-rich LDs normally accumulate in steroidogenic cells and their mobilization is the preferred initial source of cholesterol for steroidogenesis. Plin1a, 1b and 5 were found to preferentially associate with triacylglycerol-rich LDs and Plin1c and Plin4 to associate with CE-rich LDs, but the biological significance of this remains unanswered. Vimentin null mice were found to have decreased ACTH-stimulated corticosterone levels, and decreased progesterone levels in females, but normal hCG-stimulated testosterone levels in males. Smaller LDs were seen in null cells. Lipoprotein cholesterol delivery to adrenals and ovary was normal, as was the expression of steroidogenic genes; however, the movement of cholesterol to mitochondria was reduced in vimentin null mice. These results suggest that vimentin is important in the maintenance of CE-rich LDs and in the movement of cholesterol for steroidogenesis.


Subject(s)
Adrenal Glands/metabolism , Cholesterol Esters/chemistry , Mitochondria/metabolism , Steroids/biosynthesis , Vimentin/deficiency , Adrenocorticotropic Hormone/metabolism , Animals , Carrier Proteins/metabolism , Corticosterone/blood , Humans , Mice , Perilipin-1 , Perilipin-4 , Perilipin-5 , Phosphoproteins/metabolism , Progesterone/blood , Proteins/metabolism , Testosterone/blood , Vimentin/genetics
9.
J Clin Endocrinol Metab ; 96(7): E1153-8, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21543429

ABSTRACT

CONTEXT AND OBJECTIVE: Estrogen sulfotransferase (EST) catalyzes the inactivation of estrone and estradiol in numerous tissues. Animal studies suggest that EST modulates glucose and lipid metabolism in adipose tissue, but it is unknown whether EST is expressed in human adipose tissue and, if so, how its expression relates to features of the metabolic syndrome. DESIGN AND PARTICIPANTS: Cross-sectional data from 16 obese men and women with metabolic dysregulation were collected as part of a larger randomized trial at an academic medical center. OUTCOME MEASURES: Participants underwent assessment of body composition, oral glucose tolerance testing, measurement of serum hormones and inflammatory markers, and sc fat biopsy to assess adipose expression of TNF-α, suppressor of cytokine signaling 3 (SOCS3), leptin, adiponectin, and EST. RESULTS: EST expression was detectable in sc adipose tissue from both men and women. Log(10) EST mRNA was not significantly associated with age, race, sex or menopausal status, or circulating levels of estrogen or testosterone. In univariate analysis, log(10) EST mRNA was significantly associated with visceral adipose tissue area (r = 0.57, P = 0.02) as well as adipose tissue expression of TNF-α (r = 0.94, P < 0.0001) and SOCS3 mRNA (r = 0.93, P < 0.0001). The associations between EST expression and TNF-α and SOCS3 held in multivariate modeling controlling for age, race, sex and menopausal status, and visceral adiposity. EST expression was not significantly associated with the adipose tissue levels of leptin or adiponectin expression. CONCLUSIONS: EST is expressed in abdominal sc adipose tissue of both obese males and females in association with expression of TNF-α and SOCS3, suggesting potential roles in inflammation. Further studies are needed to determine the specific metabolic roles of EST expression in human adipose tissue.


Subject(s)
Metabolic Syndrome/metabolism , Obesity/metabolism , Subcutaneous Fat/metabolism , Sulfotransferases/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adiponectin/blood , Adult , Body Composition , Cross-Sectional Studies , Female , Glucose Tolerance Test , Humans , Insulin Resistance/physiology , Leptin/blood , Male , Metabolic Syndrome/genetics , Middle Aged , Obesity/genetics , Sulfotransferases/genetics , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Tumor Necrosis Factor-alpha/genetics
10.
J Clin Endocrinol Metab ; 96(1): E146-50, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21047923

ABSTRACT

CONTEXT AND OBJECTIVE: Obesity is associated with activation of the TNF-α system, increased inflammatory markers, and insulin resistance. Although studies in rodents suggest that attenuation of TNF activity improves glucose homeostasis, the effect of prolonged inhibition of TNF-α with etanercept on inflammation and glucose homeostasis in a human model of obesity is not known. DESIGN AND PARTICIPANTS: Forty obese subjects with features of metabolic syndrome were randomized to etanercept or placebo, 50 mg twice weekly for 3 months, followed by 50 mg once weekly for 3 months. OUTCOME MEASURES: Subjects underwent oral glucose tolerance testing and measurement of serum inflammatory biomarkers and adipokines. Subcutaneous fat biopsy was performed in a subset for measurement of adipokine and TNF-α mRNA expression. RESULTS: Visceral adiposity was significantly associated with serum concentrations of TNF receptor 1 (TNFR1), TNFR2, and vascular cell adhesion molecule-1 and adipose tissue expression of TNF-α and SOCS-3 (all P < 0.05). Insulin resistance as assessed by homeostasis model assessment was significantly associated with TNFR1, C-reactive protein, IL-6, and soluble intracellular adhesion molecule-1 (sICAM-1) (all P < 0.05). Etanercept significantly improved fasting glucose (treatment effect vs. placebo over 6 months, -10.8 ± 4.4%, P = 0.02). Etanercept also increased the ratio of high molecular weight adiponectin to total adiponectin (+22.1 ± 9.2% vs. placebo, P = 0.02), and decreased levels of sICAM-1 (-11 ± 2% vs. placebo, P < 0.0001). In contrast, body composition, lipids, C-reactive protein, and IL-6 were unchanged after 6 months. CONCLUSIONS: Prolonged therapy with etanercept improved fasting glucose, increased the ratio of high molecular weight to total adiponectin, and decreased sICAM-1 in obese subjects with abnormal glucose homeostasis and significant subclinical inflammation.


Subject(s)
Adipokines/blood , Immunoglobulin G/therapeutic use , Metabolic Syndrome/therapy , Obesity/therapy , Receptors, Tumor Necrosis Factor/therapeutic use , Subcutaneous Fat/metabolism , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Biomarkers/blood , Blood Glucose , Body Composition , Etanercept , Female , Glucose Tolerance Test , Humans , Male , Metabolic Syndrome/blood , Metabolic Syndrome/complications , Obesity/blood , Obesity/complications , Treatment Outcome
11.
Am J Physiol Endocrinol Metab ; 299(4): E657-64, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20682840

ABSTRACT

Estrogen regulates fat mass and distribution and glucose metabolism. We have previously found that estrogen sulfotransferase (EST), which inactivates estrogen through sulfoconjugation, was highly expressed in adipose tissue of male mice and induced by testosterone in female mice. To determine whether inhibition of estrogen in female adipose tissue affects adipose mass and metabolism, we generated transgenic mice expressing EST via the aP2 promoter. As expected, EST expression was increased in adipose tissue as well as macrophages. Parametrial and subcutaneous inguinal adipose mass and adipocyte size were significantly reduced in EST transgenic mice, but there was no change in retroperitoneal or brown adipose tissue. EST overexpression decreased the differentiation of primary adipocytes, and this was associated with reductions in the expression of peroxisome proliferator-activated receptor-γ, fatty acid synthase, hormone-sensitive lipase, lipoprotein lipase, and leptin. Serum leptin levels were significantly lower in EST transgenic mice, whereas total and high-molecular-weight adiponectin levels were not different in transgenic and wild-type mice. Glucose uptake was blunted in parametrial adipose tissue during hyperinsulinemic-euglycemic clamp in EST transgenic mice. In contrast, hepatic insulin sensitivity was improved but muscle insulin sensitivity did not change in EST transgenic mice. These results reveal novel effects of EST on adipose tissue and glucose homeostasis in female mice.


Subject(s)
Adipose Tissue/metabolism , Body Composition/physiology , Glucose/metabolism , Insulin Resistance/physiology , Sulfotransferases/metabolism , Adipocytes, White/enzymology , Adipocytes, White/metabolism , Adipose Tissue/cytology , Adipose Tissue/enzymology , Animals , Cell Differentiation/physiology , Estrogens/metabolism , Fatty Acid Synthases/blood , Female , Glucose Clamp Technique , Leptin/blood , Lipoprotein Lipase/blood , Mice , Mice, Inbred C57BL , Mice, Transgenic , PPAR gamma/blood , Random Allocation , Sterol Esterase/blood
12.
Endocrinology ; 149(11): 5440-8, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18669602

ABSTRACT

Although primarily regarded as a sex steroid, estrogen plays an important role in many other physiological processes including adipose development and disposition. Estrogen sulfotransferase (EST) regulates estrogen activity by catalyzing the sulfoconjugation and inactivation of estrogens. In the present study, we report the gender-specific expression of EST in adipose tissues of the mouse and describe contrasting mechanisms of EST regulation in the fat and liver. EST is expressed in the white adipose tissues of the male but not female mouse. Within the various fat depots of male mice, it is most abundantly expressed in the epididymal fat pad, with variable levels in other white fats and no expression in the brown fat. Fractionation of epididymal fat cells showed EST to be predominantly associated with stromal vascular cells (preadipocyte). EST expression in male mouse adipose tissues is dependent on testosterone as castration ablated, and administration of exogenous testosterone restored, EST expression. Furthermore, testosterone treatment induced abnormal EST expression in the parametrial fat of female mice. EST induction by testosterone in female mice is tissue specific because testosterone treatment had no effect on liver EST expression. Conversely, the liver X receptor agonist TO-901317 induced EST expression in female mouse liver but not in their adipose tissues. Finally, we demonstrate that male EST knockout mice developed increased epididymal fat accumulation with enlarged adipocyte size. We conclude that EST is expressed in adipose tissues in a sexually dimorphic manner, is regulated by testosterone, and plays a physiological role in regulating adipose tissue accumulation in male mice.


Subject(s)
Adipose Tissue/metabolism , Gene Expression Regulation, Enzymologic , Sex Characteristics , Sulfotransferases/genetics , Adipose Tissue/drug effects , Adipose Tissue/enzymology , Adiposity/genetics , Animals , Female , Gene Expression Regulation, Enzymologic/drug effects , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity/drug effects , Organ Specificity/genetics , Sulfotransferases/metabolism , Testosterone/pharmacology
13.
Mol Biochem Parasitol ; 140(1): 107-17, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15694492

ABSTRACT

We have previously reported the presence of a DNA gyrase-like topoisomerase activity associated with the 35kb apicoplast DNA in the malarial parasite Plasmodium falciparum [Weissig V, Vetro-Widenhouse TS, Rowe TC. Topoisomerase II inhibitors induce cleavage of nuclear and 35kb plastid DNAs in the malarial parasite Plasmodium falciparum. DNA Cell Biol 1997;16:1483]. Sequences encoding polypeptides homologous to both the A and B subunits of bacterial DNA gyrase have been identified in the genome sequence of P. falciparum among data produced by the Malaria Genome Consortium and the University of Florida Malaria Gene Sequence Tag Project. Based on these findings, we have cloned and expressed a region of the Plasmodium vivax GyrB gene encoding a 43kDa polypeptide homologous to the ATP-binding domain of Escherichia coli DNA gyrase. The 43kDa PvGyrB polypeptide was found to have intrinsic ATPase activity with a K(m) of 0.27mM and a k(cat) of 0.051s(-1). The PvGyrB ATPase was also sensitive to the bacterial DNA gyrase inhibitor coumermycin. The implications of these findings are discussed.


Subject(s)
Adenosine Triphosphate/metabolism , DNA Gyrase/genetics , Plasmodium vivax/genetics , Aminocoumarins , Animals , Cloning, Molecular , Coumarins/pharmacology , DNA Gyrase/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression , Plasmodium vivax/enzymology , Protein Subunits/metabolism , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...