Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
2.
Br J Cancer ; 116(10): 1294-1301, 2017 May 09.
Article in English | MEDLINE | ID: mdl-28359078

ABSTRACT

BACKGROUND: Investigating tumour evolution and acquired chemotherapy resistance requires analysis of sequential tumour material. We describe the feasibility of obtaining research biopsies in women with relapsed ovarian high-grade serous carcinoma (HGSC). METHODS: Women with relapsed ovarian HGSC underwent either image-guided biopsy or intra-operative biopsy during secondary debulking, and samples were fixed in methanol-based fixative. Tagged-amplicon sequencing was performed on biopsy DNA. RESULTS: We screened 519 patients in order to enrol 220. Two hundred and two patients underwent successful biopsy, 118 of which were image-guided. There were 22 study-related adverse events (AE) in the image-guided biopsies, all grades 1 and 2; pain was the commonest AE. There were pre-specified significant AE in 3/118 biopsies (2.5%). 87% biopsies were fit-for-purpose for genomic analyses. Median DNA yield was 2.87 µg, and was higher in biopsies utilising 14 G or 16 G needles compared to 18 G. TP53 mutations were identified in 94.4% patients. CONCLUSIONS: Obtaining tumour biopsies for research in relapsed HGSC is safe and feasible. Adverse events are rare. The large majority of biopsies yield sufficient DNA for genomic analyses-we recommend use of larger gauge needles and methanol fixation for such biopsies, as DNA yields are higher but with no increase in AEs.


Subject(s)
Carcinoma/genetics , Carcinoma/secondary , DNA, Neoplasm/analysis , Image-Guided Biopsy , Liver Neoplasms/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Class I Phosphatidylinositol 3-Kinases , DNA Mutational Analysis , DNA, Neoplasm/isolation & purification , ErbB Receptors/genetics , Feasibility Studies , Female , Humans , Image-Guided Biopsy/adverse effects , Image-Guided Biopsy/instrumentation , Liver/pathology , Liver Neoplasms/secondary , Lymph Nodes/pathology , Lymphatic Metastasis , Middle Aged , Neoplasm Grading , Omentum/pathology , PTEN Phosphohydrolase/genetics , Pain/etiology , Peritoneal Neoplasms/secondary , Peritoneum/pathology , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Suppressor Protein p53/genetics
3.
Ann Oncol ; 27(3): 532-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26681675

ABSTRACT

BACKGROUND: Next-generation sequencing (NGS) of tumour samples is a critical component of personalised cancer treatment, but it requires high-quality DNA samples. Routine neutral-buffered formalin (NBF) fixation has detrimental effects on nucleic acids, causing low yields, as well as fragmentation and DNA base changes, leading to significant artefacts. PATIENTS AND METHODS: We have carried out a detailed comparison of DNA quality from matched samples isolated from high-grade serous ovarian cancers from 16 patients fixed in methanol and NBF. These experiments use tumour fragments and mock biopsies to simulate routine practice, ensuring that results are applicable to standard clinical biopsies. RESULTS: Using matched snap-frozen tissue as gold standard comparator, we show that methanol-based fixation has significant benefits over NBF, with greater DNA yield, longer fragment size and more accurate copy-number calling using shallow whole-genome sequencing (WGS). These data also provide a new approach to understand and quantify artefactual effects of fixation using non-negative matrix factorisation to analyse mutational spectra from targeted and WGS data. CONCLUSION: We strongly recommend the adoption of methanol fixation for sample collection strategies in new clinical trials. This approach is immediately available, is logistically simple and can offer cheaper and more reliable mutation calling than traditional NBF fixation.


Subject(s)
DNA/drug effects , Formaldehyde/chemistry , Methanol/chemistry , Neoplasms/diagnosis , Tissue Fixation/methods , Base Sequence , DNA/analysis , High-Throughput Nucleotide Sequencing , Humans , Middle Aged , Paraffin Embedding , Sequence Analysis, DNA
4.
Br J Cancer ; 111(12): 2297-307, 2014 Dec 09.
Article in English | MEDLINE | ID: mdl-25349970

ABSTRACT

BACKGROUND: Folate receptor 1 (FOLR1) is expressed in the majority of ovarian carcinomas (OvCa), making it an attractive target for therapy. However, clinical trials testing anti-FOLR1 therapies in OvCa show mixed results and require better understanding of the prognostic relevance of FOLR1 expression. We conducted a large study evaluating FOLR1 expression with survival in different histological types of OvCa. METHODS: Tissue microarrays composed of tumour samples from 2801 patients in the Ovarian Tumour Tissue Analysis (OTTA) consortium were assessed for FOLR1 expression by centralised immunohistochemistry. We estimated associations for overall (OS) and progression-free (PFS) survival using adjusted Cox regression models. High-grade serous ovarian carcinomas (HGSC) from The Cancer Genome Atlas (TCGA) were evaluated independently for association between FOLR1 mRNA upregulation and survival. RESULTS: FOLR1 expression ranged from 76% in HGSC to 11% in mucinous carcinomas in OTTA. For HGSC, the association between FOLR1 expression and OS changed significantly during the years following diagnosis in OTTA (Pinteraction=0.01, N=1422) and TCGA (Pinteraction=0.01, N=485). In OTTA, particularly for FIGO stage I/II tumours, patients with FOLR1-positive HGSC showed increased OS during the first 2 years only (hazard ratio=0.44, 95% confidence interval=0.20-0.96) and patients with FOLR1-positive clear cell carcinomas (CCC) showed decreased PFS independent of follow-up time (HR=1.89, 95% CI=1.10-3.25, N=259). In TCGA, FOLR1 mRNA upregulation in HGSC was also associated with increased OS during the first 2 years following diagnosis irrespective of tumour stage (HR: 0.48, 95% CI: 0.25-0.94). CONCLUSIONS: FOLR1-positive HGSC tumours were associated with an increased OS in the first 2 years following diagnosis. Patients with FOLR1-negative, poor prognosis HGSC would be unlikely to benefit from anti-FOLR1 therapies. In contrast, a decreased PFS interval was observed for FOLR1-positive CCC. The clinical efficacy of FOLR1-targeted interventions should therefore be evaluated according to histology, stage and time following diagnosis.


Subject(s)
Biomarkers, Tumor/biosynthesis , Folate Receptor 1/biosynthesis , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/metabolism , Carcinoma, Ovarian Epithelial , Disease-Free Survival , Female , Humans , Immunohistochemistry , Middle Aged , Survival Analysis , Tissue Array Analysis
5.
Br J Cancer ; 104(2): 361-8, 2011 Jan 18.
Article in English | MEDLINE | ID: mdl-21063398

ABSTRACT

BACKGROUND: Intra-tumour genetic heterogeneity has been reported in both leukaemias and solid tumours and is implicated in the development of drug resistance in CML and AML. The role of genetic heterogeneity in drug response in solid tumours is unknown. METHODS: To investigate intra-tumour genetic heterogeneity and chemoradiation response in advanced cervical cancer, we analysed 10 cases treated on the CTCR-CE01 clinical study. Core biopsies for molecular profiling were taken from four quadrants of the cervix pre-treatment, and weeks 2 and 5 of treatment. Biopsies were scored for cellularity and profiled using Agilent 180k human whole genome CGH arrays. We compared genomic profiles from 69 cores from 10 patients to test for genetic heterogeneity and treatment effects at weeks 0, 2 and 5 of treatment. RESULTS: Three patients had two or more distinct genetic subpopulations pre-treatment. Subpopulations within each tumour showed differential responses to chemoradiotherapy. In two cases, there was selection for a single intrinsically resistant subpopulation that persisted at detectable levels after 5 weeks of chemoradiotherapy. Phylogenetic analysis reconstructed the order in which genomic rearrangements occurred in the carcinogenesis of these tumours and confirmed gain of 3q and loss of 11q as early events in cervical cancer progression. CONCLUSION: Selection effects from chemoradiotherapy cause dynamic changes in genetic subpopulations in advanced cervical cancers, which may explain disease persistence and subsequent relapse. Significant genetic heterogeneity in advanced cervical cancers may therefore be predictive of poor outcome.


Subject(s)
Antineoplastic Agents/therapeutic use , Genetic Heterogeneity , Uterine Cervical Neoplasms/genetics , Adult , Aged , Combined Modality Therapy , Female , Humans , Middle Aged , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/radiotherapy
6.
Acta Radiol ; 49(7): 848-51, 2008 Sep.
Article in English | MEDLINE | ID: mdl-19143068

ABSTRACT

Müllerian adenosarcoma of the uterus is a rare biphasic tumor, which was first described in 1974. Recent studies have suggested an association with tamoxifen therapy, but there have been few reports with detailed imaging findings. We present a case with magnetic resonance imaging (MRI) findings of this rare tumor in a woman who received long-term tamoxifen therapy for breast cancer. In addition, myometrial invasion was detected more accurately with MRI compared to ultrasound in this one single case.


Subject(s)
Adenosarcoma/chemically induced , Adenosarcoma/diagnosis , Antineoplastic Agents, Hormonal/adverse effects , Magnetic Resonance Imaging/methods , Mullerian Ducts/pathology , Tamoxifen/adverse effects , Uterine Neoplasms/chemically induced , Uterine Neoplasms/diagnosis , Adenosarcoma/pathology , Adenosarcoma/surgery , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Female , Humans , Hysterectomy , Middle Aged , Neoplasm Invasiveness , Tamoxifen/therapeutic use , Uterine Neoplasms/pathology , Uterine Neoplasms/surgery
7.
J Neuroendocrinol ; 19(9): 708-16, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17680886

ABSTRACT

The hypothalamic decapeptide gonadotrophin-releasing hormone (GnRH), modulates gonadotrophin synthesis and secretion and is essential for the preovulatory luteinising hormone (LH) surge. As females age, there is a gradual attenuation and eventual loss of the preovulatory LH surge and oestrous cyclicity. Data from previous studies have demonstrated evidence of compromised GnRH neuronal function at this time. The present study begins to explore the hypothesis that the age-related attenuation of the LH surge and decline in GnRH neuronal function are due, in part, to increased inhibitory influences on GnRH neurones. In situ hybridisation (ISH) was used to assess relative levels of mRNA for one isoform of glutamic acid decarboxylase (GAD), the rate-limiting enzyme for GABA synthesis. Ovariectomised young and middle-aged rats were injected with oestradiol benzoate and progesterone in a regimen for LH surge induction. Animals were killed at time points prior to, during the ascending phase, and during the peak and early descending phase of the LH surge. Dynamic changes in GAD(67) mRNA levels were observed in young but not middle-aged females in two regions known to be important for LH surge induction, the rostral proeptic area in the region of the organum vasculosum of the lamina terminalis (OVLT) and in the ventral periventricular preoptic area. Furthermore, GAD(67) mRNA levels were elevated in middle-aged relative to young females in the region of the OVLT at the time of LH surge induction and in the ventral periventricular preoptic area prior to surge induction. Age-related differences were not observed in other brain regions analysed. These data suggest that GABA synthesis may be elevated in middle-aged relative to young females in specific brain regions at critical times in conjunction with the LH surge, and that the lack of dynamic changes in GABA levels in these regions may contribute to the attenuated LH surge observed in middle-aged females.


Subject(s)
Aging/physiology , Estrous Cycle/physiology , Glutamate Decarboxylase/genetics , Isoenzymes/genetics , Luteinizing Hormone/blood , RNA, Messenger/metabolism , Animals , Female , Hypothalamus/cytology , Hypothalamus/metabolism , In Situ Hybridization , Rats , Rats, Sprague-Dawley , gamma-Aminobutyric Acid/metabolism
8.
Breast Cancer Res Treat ; 73(1): 23-9, 2002 May.
Article in English | MEDLINE | ID: mdl-12083628

ABSTRACT

A single intragastric administration of 7,12-dimethylbenz(a)anthracene (DMBA) has been shown to induce mammary tumors in young cycling female Sprague-Dawley rats. The appearance of these tumors is preceded by a series of neuroendocrine disturbances, including attenuation of the preovulatory luteinizing hormone (LH) surge and amplification of the preovulatory 17beta-estradiol surge, and gonadotropin-releasing hormone (GnRH) released in vitro. In this study, we examined the hypothesis that DMBA administration decreases levels of GnRH mRNA in the preoptic area-anterior hypothalamus (POA-AH) and GnRH receptor (GnRH Rc) mRNA and protein in the anterior pituitary gland. Sprague-Dawley rats, 55-60 days of age with regular estrous cycles, received a single dose of 15 mg DMBA in 1 ml sesame oil delivered by intragastric intubation. A first series of experiments was performed for the measurement of hypothalamic GnRH mRNA and pituitary GnRH Rc mRNA levels. A second series of experiments was performed for the measurement of pituitary GnRH receptor. In both experiments, animals were sacrificed by decapitation at 11.00, 16.00, 18.00 and 20.00 h on each day of the 7th or 8th estrous cycle (28-32 days) after treatment. GnRH and GnRH receptor mRNAs were quantified using solution hybridization-RNase protection assay. The GnRH Rc was quantified using the 125I-D-Ala6-N-Met-Leu6-des-Gly10-ethylamide GnRH. DMBA-treatment produced no significant effect on the overall mean values of GnRH mRNA. GnRH mRNA levels in control rats rose significantly between 16.00 and 20.00 h on proestrus and between 18.00 and 20.00 h on diestrus I. DMBA-treated rats had a surge in GnRH mRNA levels at 18.00 h on proestrus, and showed additional surges at 18.00h on diestrus II and estrus. GnRH receptor mRNA content in the anterior pituitary gland surged at 16.00h on certain days of the cycle in both groups of rats. In control rats, only the surge on diestrus II proved significant, whereas DMBA-treated rats exhibited significant surges on diestrus I, diestrus II and proestrus. GnRH receptor mRNA values were significantly lower on both days of diestrus in DMBA-treated rats compared with controls. GnRH Rc peptide content, like GnRH receptor in RNA surged at 16.00h in both groups with the exception of a marked fall on proestrus day for DMBA treated rats. A reduction in the amplitude of the surge was also seen on the day of estrous and to a lesser extend on the day of diestrus DII in DMBA treated animal. Overall, there was a disruption of the GnRH Rc pattern which culminate on the day of proestrus in DMBA-treated animals. Interestingly, the daily rise between 11.00 and 16.00h which is the more pronounced on the day of proestrus in control animals, was completely blunted in DMBA-treated rats. Overall, the results are consistent with the hypothesis that the carcinogen attenuates, directly or indirectly, preovulatory biosynthesis of the GnRH receptor and LH release. Obviously, the changes in GnRH might occur simultaneously, independently from mammary tumorigenesis, but may play a role, in association with others DMBA-induced neuroendocrine disorders, in the promotion stage of mammary tumors in the Sprague-Dawley female rat.


Subject(s)
9,10-Dimethyl-1,2-benzanthracene/toxicity , Carcinogens/toxicity , Gonadotropin-Releasing Hormone/genetics , Hypothalamus, Anterior/metabolism , Mammary Neoplasms, Experimental/metabolism , Pituitary Gland, Anterior/metabolism , Receptors, LHRH/genetics , Animals , Female , Gonadotropin-Releasing Hormone/metabolism , In Vitro Techniques , Mammary Neoplasms, Experimental/pathology , Proestrus/physiology , RNA Probes , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, LHRH/metabolism , Ribonuclease, Pancreatic/metabolism , Time Factors
9.
Brain Res ; 922(1): 71-9, 2001 Dec 13.
Article in English | MEDLINE | ID: mdl-11730703

ABSTRACT

Luteinizing hormone releasing hormone (LHRH) gene transcription was examined in ovariectomized female rats on the day of a steroid-induced LH surge using the RNase protection assay. LHRH mRNA levels were measured in cytosolic extracts, and LHRH primary transcript levels were measured in nuclear extracts prepared from tissue fragments that contained the organum vasculosum of the lamina terminalis (OVLT) and the preoptic area (POA). Measurements of both mature and primary transcript levels demonstrated modest but significant changes over time. Alterations in LHRH primary transcript levels preceded changes in levels of mature mRNA suggesting a delay in the detectable response of the cytoplasmic pool of LHRH mRNA to changes in gene transcription at this time. When viewed in relation to circulating LH titers, LHRH primary transcript levels were high prior to the start of the LH surge and after peak levels of LH were attained, and they declined during the ascending phase of the LH surge. These findings suggest a potential role for increased LHRH gene transcription in the accumulation of LHRH prior to the start of the LH surge and in the replenishment of LHRH stores depleted during the surge. Moreover, the decrease in LHRH gene transcription during the ascending phase of the LH surge may be important for limiting surge duration. The data presented are consistent with a role for dynamic changes in LHRH transcriptional activity in modulating parameters of the steroid-induced LH surge and in replenishing the releasable pool of this essential decapeptide.


Subject(s)
Gonadotropin-Releasing Hormone/biosynthesis , Steroids/pharmacology , Transcription, Genetic/drug effects , Animals , Autoradiography , Circadian Rhythm/drug effects , Circadian Rhythm/physiology , Cytoplasm/metabolism , Female , Gonadotropin-Releasing Hormone/genetics , In Situ Hybridization , Luteinizing Hormone/blood , Nuclease Protection Assays , Ovariectomy , RNA Probes , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley
10.
Endocrinology ; 138(10): 4123-30, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9322920

ABSTRACT

We sequenced the complementary DNA (cDNA) encoding guinea pig LHRH from an expression library derived from the preoptic area-anterior hypothalamus. Data from in situ hybridization and RNase protection assays verified that the cloned cDNA was complementary to guinea pig LHRH messenger RNA. The architecture of the deduced precursor resembles that of LHRH precursors identified in other species. In contrast, the predicted sequence of the decapeptide differs from mammalian LHRH by two amino acid substitutions in positions 2 and 7. This is a novel finding, because the amino acid sequence that comprises LHRH decapeptide is identical in all mammals studied to date. Moreover, the predicted substitution in amino acid position 2 is unique among vertebrates. A second observation of potential significance is the existence of two subspecies of LHRH messenger RNA differing only in the length of their 3' untranslated regions. These two transcripts were verified by sequence analysis of positive clones from the cDNA library and by RNase protection analysis of preoptic area-anterior hypothalamus extracts, and their presence is consistent with the two polyadenylation signals identified in the untranslated regions of the LHRH gene. Future studies will examine LHRH gene expression in guinea pigs, which like primates but unlike rats, have a true luteal phase as a component of their reproductive cycle.


Subject(s)
DNA/analysis , Gonadotropin-Releasing Hormone/chemistry , Gonadotropin-Releasing Hormone/genetics , Peptides/analysis , Preoptic Area/chemistry , Transcription, Genetic , Amino Acid Sequence , Animals , Base Sequence , Blotting, Southern , DNA/chemistry , DNA/genetics , Female , Gonadotropin-Releasing Hormone/analysis , Guinea Pigs , Hypothalamus/chemistry , Hypothalamus/cytology , In Situ Hybridization , Molecular Sequence Data , Peptides/chemistry , Polymerase Chain Reaction , Preoptic Area/cytology , RNA, Messenger/analysis , RNA, Messenger/chemistry , RNA, Messenger/genetics
11.
J Clin Invest ; 99(10): 2416-22, 1997 May 15.
Article in English | MEDLINE | ID: mdl-9153284

ABSTRACT

The peroxisome proliferator activated receptor (PPAR gamma) plays a key role in adipogenesis and adipocyte gene expression and is the receptor for the thiazolidinedione class of insulin-sensitizing drugs. The tissue expression and potential for regulation of human PPAR gamma gene expression in vivo are unknown. We have cloned a partial human PPAR gamma cDNA, and established an RNase protection assay that permits simultaneous measurements of both PPAR gamma1 and PPAR gamma2 splice variants. Both gamma1 and gamma2 mRNAs were abundantly expressed in adipose tissue. PPAR gamma1 was detected at lower levels in liver and heart, whereas both gamma1 and gamma2 mRNAs were expressed at low levels in skeletal muscle. To examine the hypothesis that obesity is associated with abnormal adipose tissue expression of PPAR gamma, we quantitated PPARgamma mRNA splice variants in subcutaneous adipose tissue of 14 lean and 24 obese subjects. Adipose expression of PPARgamma 2 mRNA was increased in human obesity (14.25 attomol PPAR gamma2/18S in obese females vs 9.9 in lean, P = 0.003). This increase was observed in both male and females. In contrast, no differences were observed in PPAR gamma1/18S mRNA expression. There was a strong positive correlation (r = 0.70, P < 0.001) between the ratio of PPAR gamma2/gamma1 and the body mass index of these patients. We also observed sexually dimorphic expression with increased expression of both PPAR gamma1 and PPAR gamma2 mRNAs in the subcutaneous adipose tissue of women compared with men. To determine the effect of weight loss on PPAR gamma mRNA expression, seven additional obese subjects were fed a low calorie diet (800 Kcal) until 10% weight loss was achieved. Mean expression of adipose PPAR gamma2 mRNA fell 25% (P = 0.0250 after a 10% reduction in body weight), but then increased to pretreatment levels after 4 wk of weight maintenance. Nutritional regulation of PPAR gamma1 was not seen. In vitro experiments revealed a synergistic effect of insulin and corticosteroids to induce PPAR gamma expression in isolated human adipocytes in culture. We conclude that: (a) human PPAR gamma mRNA expression is most abundant in adipose tissue, but lower level expression of both splice variants is seen in skeletal muscle; to an extent that is unlikely to be due to adipose contamination. (b) RNA derived from adipose tissue of obese humans has increased expression of PPAR gamma 2 mRNA, as well as an increased ratio of PPAR gamma2/gamma1 splice variants that is proportional to the BMI; (c) a low calorie diet specifically down-regulates the expression of PPAR gamma2 mRNA in adipose tissue of obese humans; (d) insulin and corticosteroids synergistically induce PPAR gamma mRNA after in vitro exposure to isolated human adipocytes; and (e) the in vivo modulation of PPAR gamma2 mRNA levels is an additional level of regulation for the control of adipocyte development and function, and could provide a molecular mechanism for alterations in adipocyte number and function in obesity.


Subject(s)
Adipose Tissue/metabolism , Dexamethasone/pharmacology , Gene Expression , Insulin/pharmacology , Muscle, Skeletal/metabolism , Obesity, Morbid/physiopathology , Receptors, Cytoplasmic and Nuclear/biosynthesis , Transcription Factors/biosynthesis , Transcription, Genetic , Weight Loss/physiology , Adipose Tissue/drug effects , Adult , Cells, Cultured , Cloning, Molecular , DNA Primers , Diet, Reducing , Energy Intake , Female , Gene Expression/drug effects , Humans , Liver/metabolism , Male , Muscle, Skeletal/drug effects , Myocardium/metabolism , Nuclear Proteins/biosynthesis , Obesity, Morbid/diet therapy , Obesity, Morbid/metabolism , Polymerase Chain Reaction , Sex Characteristics , Thinness , Transcription, Genetic/drug effects
12.
J Clin Invest ; 97(11): 2553-61, 1996 Jun 01.
Article in English | MEDLINE | ID: mdl-8647948

ABSTRACT

The orphan nuclear receptor, peroxisome proliferator-activated receptor (PPAR) gamma, is implicated in mediating expression of fat-specific genes and in activating the program of adipocyte differentiation. The potential for regulation of PPAR gamma gene expression in vivo is unknown. We cloned a partial mouse PPAR gamma cDNA and developed an RNase protection assay that permits simultaneous quantitation of mRNAs for both gamma l and gamma 2 isoforms encoded by the PPAR gamma gene. Probes for detection of adipocyte P2, the obese gene product, leptin, and 18S mRNAs were also employed. Both gamma l and gamma 2 mRNAs were abundantly expressed in adipose tissue. PPAR gamma 1 expression was also detected at lower levels in liver, spleen, and heart; whereas, gamma l and gamma 2 mRNA were expressed at low levels in skeletal muscle. Adipose tissue levels of gamma l and gamma 2 were not altered in two murine models of obesity (gold thioglucose and ob/ob), but were modestly increased in mice with toxigene-induced brown fat ablation uncoupling protein diphtheria toxin A mice. Fasting (12-48 h) was associated with an 80% fall in PPAR gamma 2 and a 50% fall in PPAR gamma mRNA levels in adipose tissue. Western blot analysis demonstrated a marked effect of fasting to reduce PPAR gamma protein levels in adipose tissue. Similar effects of fasting on PPAR gamma mRNAs were noted in all three models of obesity. Insulin-deficient (streptozotocin) diabetes suppressed adipose tissue gamma l and gamma 2 expression by 75% in normal mice with partial restoration during insulin treatment. Levels of adipose tissue PPAR gamma 2 mRNA were increased by 50% in normal mice exposed to a high fat diet. In obese uncoupling protein diphtheria toxin A mice, high fat feeding resulted in de novo induction of PPAR gamma 2 expression in liver. We conclude (a) PPAR gamma 2 mRNA expression is most abundant in adipocytes in normal mice, but lower level expression is seen in skeletal muscle; (b) expression of adipose tissue gamma1 or gamma2 mRNAs is increased in only one of the three models of obesity; (c) PPAR gamma 1 and gamma 2 expression is downregulated by fasting and insulin-deficient diabetes; and (d) exposure of mice to a high fat diet increases adipose tissue expression of PPAR gamma (in normal mice) and induces PPAR gamma 2 mRNA expression in liver (in obese mice). These findings demonstrate in vivo modulation of PPAR gamma mRNA levels over a fourfold range and provide an additional level of regulation for the control of adipocyte development and function.


Subject(s)
Dietary Fats , Gene Expression Regulation , Obesity/metabolism , Receptors, Cytoplasmic and Nuclear/biosynthesis , Transcription Factors/biosynthesis , Transcription, Genetic , Adipocytes/metabolism , Adipose Tissue/metabolism , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/physiology , Animals , Aurothioglucose/pharmacology , Base Sequence , DNA Primers , Diabetes Mellitus, Experimental/metabolism , Diphtheria Toxin/toxicity , Female , Gene Expression Regulation/drug effects , Leptin , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Molecular Sequence Data , Muscle, Skeletal/metabolism , Myocardium/metabolism , Obesity/genetics , Organ Specificity , Polymerase Chain Reaction , Protein Biosynthesis , Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Ribosomal, 16S/biosynthesis , Reference Values , Spleen/metabolism , Transcription, Genetic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...