Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Gynecol Endocrinol ; 36(3): 226-232, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31389293

ABSTRACT

Polycystic ovary syndrome (PCOS) is associated with increased risk of endometrial cancer. There is growing evidence that prolactin and its receptor (PRLR) are involved in the development of cancer. We assessed endometrial expression of PRLR mRNA, and immunostaining of PRLR and the proliferation marker Ki67 on different cycle days in obese (OB-PCOS) and normal-weight women with PCOS and body mass index-matched controls. The OB-PCOS group underwent a 3 months lifestyle intervention. Prior to intervention, obese women with PCOS and controls had lower endometrial levels of PRLR mRNA in proliferative endometrium than the normal-weight groups (p < .05). After intervention, six OB-PCOS women had confirmed ovulation, while 12 remained anovulatory. Both these subgroups displayed higher immunostaining of PRLR in endometrial stroma, and in the anovulatory subgroup also increased Ki67, on cycle days 21-23 compared with controls (p < .05). In obese controls, the PRLR mRNA expression was decreased in secretory endometrium compared with proliferative endometrium (p = .004). A corresponding change within the cycle was not found in OB-PCOS women. Immunostaining of PRLR in the secretory phase correlated positively with Ki67 (p < .05) in the endometrium. These observations suggest that short-term lifestyle intervention can restore ovulation but not normalize PRLR expression in the endometrium of obese women with PCOS. Trial registration: ISRCTN, ISRCTN18400086, https://doi.org/10.1186/ISRCTN18400086.


Subject(s)
Cell Proliferation/genetics , Endometrium/metabolism , Obesity/genetics , Polycystic Ovary Syndrome/genetics , Receptors, Prolactin/genetics , Adult , Case-Control Studies , Diet, Reducing , Female , Follicular Phase/genetics , Follicular Phase/metabolism , Humans , Ki-67 Antigen/metabolism , Luteal Phase/genetics , Luteal Phase/metabolism , Obesity/metabolism , Obesity/therapy , Ovulation , Polycystic Ovary Syndrome/metabolism , RNA, Messenger/metabolism , Receptors, Prolactin/metabolism , Treatment Outcome , Weight Reduction Programs , Young Adult
2.
Carcinogenesis ; 35(1): 24-33, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24031028

ABSTRACT

UNLABELLED: Anabolic signals such as androgens and the growth hormone/insulin-like growth factor 1 (GH/IGF-1) axis play an essential role in the normal development of the prostate but also in its malignant transformation. In this study, we investigated the role of suppressor of cytokine signaling 2 (SOCS2) as mediator of the cross talk between androgens and GH signals in the prostate and its potential role as tumor suppressor in prostate cancer (PCa). We observed that SOCS2 protein levels assayed by immunohistochemistry are elevated in hormone therapy-naive localized prostatic adenocarcinoma in comparison with benign tissue. In contrast, however, castration-resistant bone metastases exhibit reduced levels of SOCS2 in comparison with localized or hormone naive, untreated metastatic tumors. In PCa cells, SOCS2 expression is induced by androgens through a mechanism that requires signal transducer and activator of transcription 5 protein (STAT5) and androgen receptor-dependent transcription. Consequentially, SOCS2 inhibits GH activation of Janus kinase 2, Src and STAT5 as well as both cell invasion and cell proliferation in vitro. In vivo, SOCS2 limits proliferation and production of IGF-1 in the prostate in response to GH. Our results suggest that the use of GH-signaling inhibitors could be of value as a complementary treatment for castration-resistant PCa. SUMMARY: Androgen induced SOCS2 ubiquitin ligase expression and inhibited GH signaling as well as cell proliferation and invasion in PCa, whereas reduced SOCS2 was present in castration-resistant cases. GH-signaling inhibitors might be a complementary therapeutic option for advanced PCa.


Subject(s)
Androgens/metabolism , Human Growth Hormone/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Suppressor of Cytokine Signaling Proteins/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Aged , Animals , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Human Growth Hormone/pharmacology , Humans , Insulin-Like Growth Factor I/metabolism , Male , Metribolone/pharmacology , Mice, Inbred C57BL , Mice, Mutant Strains , Middle Aged , Predictive Value of Tests , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Suppressor of Cytokine Signaling Proteins/analysis , Suppressor of Cytokine Signaling Proteins/genetics
3.
FASEB J ; 26(8): 3282-91, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22562833

ABSTRACT

Hepatic steatosis is a prominent feature in patients with growth hormone (GH) deficiency. The ubiquitin ligase SOCS2 attenuates hepatic GH signaling by inhibiting the Janus kinase 2 (JAK2)-signal transducer and activator of transcription 5b (STAT5b) axis. Here, we investigated the role of SOCS2 in the development of diet-induced hepatic steatosis and insulin resistance. SOCS2-knockout (SOCS2(-/-)) mice and wild-type littermates were fed for 4 mo with control or high-fat diet, followed by assessment of insulin sensitivity, hepatic lipid content, and expression of inflammatory cytokines. SOCS2(-/-) mice exhibited increased hepatic TG secretion by 77.6% (P<0.001) as compared with wild-type control mice and were protected from high-fat-diet (HFD)-induced hepatic steatosis, showing 49.3% (P<0.01) reduction in liver TG levels compared to HFD-fed wild-type littermates. In contrast, we found that HFD-triggered attenuation of systemic insulin sensitivity was more marked in SOCS2(-/-) mice. Livers from the HFD-fed SOCS2(-/-) mice showed increased NF-κB activity as well as elevated expression of genes for the inflammatory cytokines IFN-γ and IL-6. An inhibitory role of SOCS2 on Toll-like receptor 4 signaling was demonstrated in macrophages obtained from the SOCS2(-/-) and wild-type mice. This study identified SOCS2 as an important regulator of hepatic homeostasis under conditions of high-fat dietary stress.


Subject(s)
Diet, High-Fat , Fatty Liver/prevention & control , Insulin Resistance/physiology , Suppressor of Cytokine Signaling Proteins/deficiency , Suppressor of Cytokine Signaling Proteins/physiology , Animals , Interferon-gamma/metabolism , Interleukin-6/metabolism , Lipid Metabolism , Liver/metabolism , Male , Mice , Mice, Knockout , NF-kappa B/metabolism , Toll-Like Receptor 4/antagonists & inhibitors , Triglycerides/metabolism
4.
Clin Endocrinol (Oxf) ; 77(2): 288-95, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22372591

ABSTRACT

OBJECTIVES: Metabolite profiles of body fluids or tissue extracts can be regarded as important indicators of physiological or pathological states. Whether hormone-specific alterations of the serum metabolome can be identified using this technique has not been tested yet. The aim of this study was to investigate metabolic responses during hormone therapy in postmenopausal women by a nontargeted metabolomics approach. METHODS: Sixty naturally postmenopausal women were randomly assigned to treatment with testosterone undecanoate 40 mg every second day; estradiol valerate 2 mg daily; or the combination of both. Serum metabolites were determined by gas chromatography-mass spectrometry (GC-MS) before and after 3 months of treatment. Metabolites affected by the treatment were identified and correlated with changes in insulin sensitivity and lipid profiles. RESULTS: Treatment-dependent and hormone-specific effects on serum metabolites were observed, ranging between 69% reduction and 184% increase, but the metabolites that best explained the differences could not be structurally identified. Effects on annotated metabolites were less associated with clinical parameters as compared to established serum markers for adverse lipid and carbohydrate metabolism, such as cholesterol and triglycerides. However, cystine, lysine and tyrosine were shown to change in correlation with insulin sensitivity and high-density lipoprotein cholesterol levels in response to testosterone, indicating that those responses were somehow related to each other. CONCLUSIONS: Oestrogen- and androgen-specific alterations in the serum metabolome could be identified using GC-MS, reflecting hormone-specific effects on whole body metabolism. New knowledge regarding steroid-mediated metabolic responses within different tissues might be obtained using a similar approach on tissue extracts.


Subject(s)
Estrogens/therapeutic use , Testosterone/therapeutic use , Adult , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Female , Gas Chromatography-Mass Spectrometry , Humans , Lipid Metabolism/drug effects , Lipoprotein(a)/blood , Middle Aged , Postmenopause/blood , Postmenopause/drug effects
5.
Phytother Res ; 26(2): 259-64, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21656602

ABSTRACT

Extracts from Serenoa repens are widely used for the treatment of benign prostatic hyperplasia (BPH) and traditionally for prostatitis. In the present study we evaluated the biological effects of Serenoa repens extract (Prostasan®) on prostate cells beyond its known antiandrogenic actions. Prostasan® inhibited epidermal growth factor (EGF) and lipopolysaccharide (LPS) induced proliferation of the prostatic epithelial, androgen independent cell line PC-3. At effective concentrations of 50 µg/mL, Prostasan® partly displaced EGF from EGF receptor (EGFR) but fully blocked EGF-induced cell proliferation of PC-3 cells. Similarly, Prostasan® inhibited LPS-induced proliferation of PC-3 cells without affecting LPS activation of the NFĸB pathway via toll-like receptor-4 (TLR-4). Additionally, Prostasan® reduced the constitutive secretion of monocyte chemotactic protein-1 (MCP-1), the LPS-induced secretion of IL-12 and inhibited MCP-1 and granulocyte-macrophage colony-stimulating factor (GM-CSF) production in the presence of LPS on PC-3 cells. Taken together, our results suggest that S. repens extracts, in addition to other reported effects on BPH development and prostatitis, inhibits EGF-dependent growth and proinflammatory responses of the prostate epithelial cells.


Subject(s)
Cell Proliferation/drug effects , Epithelial Cells/drug effects , Inflammation/pathology , Plant Extracts/pharmacology , Prostate/cytology , Serenoa/chemistry , Cell Line/drug effects , Cytokines/metabolism , Epidermal Growth Factor/antagonists & inhibitors , ErbB Receptors/metabolism , Humans , Inflammation/drug therapy , Lipopolysaccharides , Male
6.
PLoS One ; 16(8): e0255701, 2021.
Article in English | MEDLINE | ID: mdl-34358244

ABSTRACT

Ovarian cancer (OC) is characterized by a high morbidity and mortality, highlighting a great need for a better understanding of biological mechanisms that affect OC progression and improving its early detection methods. This study investigates effects of prolactin (PRL) on ovarian cancer cells, analyzes PRL receptors (PRLR) in tissue micro arrays and relates PRLR expression to survival of ovarian cancer. A database, composed of transcript profiles from OC, was searched for PRLR expression and results were put in relation to survival. Expression of PRLR in OC tissue sections and OC cell lines SKOV3, OV2008 and OVSAHO was assessed using immunohistochemistry, western blots and quantitative real-time PCR. The biological function of PRLR was evaluated by proliferation, colony formation and wound healing assays. Levels of PRLR mRNA are related to survival; in epithelial OC a high PRLR mRNA expression is related to a shorter survival. Analysis of a tissue micro array consisting of 84 OC showed that 72% were positive for PRLR immuno-staining. PRLR staining tended to be higher in OC of high grade tumors compared to lower grades. PRLR mRNA and protein can further be detected in OC cell lines. Moreover, in vitro treatment with PRL significantly activated the JAK/STAT pathway. PRLR expression is associated with OC survivals. PRL and its receptor may play an onco-modulatory role and promote tumor aggressiveness in OC. Alternatively, increased PRLR levels may form a base for the development of PRLR antagonist or PRLR antagonist-drug conjugate to increase selective uptake of anti-cancer drugs.


Subject(s)
Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/mortality , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/mortality , Prolactin/pharmacology , Receptors, Prolactin/metabolism , Signal Transduction/drug effects , Carcinoma, Ovarian Epithelial/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , MCF-7 Cells , Ovarian Neoplasms/pathology , Phosphorylation/drug effects , Progression-Free Survival , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Prolactin/genetics , Recombinant Proteins/pharmacology , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Survival Rate
7.
BMC Biochem ; 11: 38, 2010 Sep 23.
Article in English | MEDLINE | ID: mdl-20863371

ABSTRACT

BACKGROUND: Genes involved in hepatic metabolism have a sex-different expression in rodents. To test whether male and female rat livers differ regarding lipid and carbohydrate metabolism, whole-genome transcript profiles were generated and these were complemented by measurements of hepatic lipid and glycogen content, fatty acid (FA) oxidation rates and hepatic glucose output (HGO). The latter was determined in perfusates from in situ perfusion of male and female rat livers. These perfusates were also analysed using nuclear magnetic resonance (NMR) spectroscopy to identify putative sex-differences in other liver-derived metabolites. Effects of insulin were monitored by analysis of Akt-phosphorylation, gene expression and HGO after s.c. insulin injections. RESULTS: Out of approximately 3 500 gene products being detected in liver, 11% were significantly higher in females, and 11% were higher in males. Many transcripts for the production of triglycerides (TG), cholesterol and VLDL particles were female-predominant, whereas genes for FA oxidation, gluconeogenesis and glycogen synthesis were male-predominant. Sex-differences in mRNA levels related to metabolism were more pronounced during mild starvation (12 h fasting), as compared to the postabsorptive state (4 h fasting). No sex-differences were observed regarding hepatic TG content, FA oxidation rates or blood levels of ketone bodies or glucose. However, males had higher hepatic glycogen content and higher HGO, as well as higher ratios of insulin to glucagon levels. Based on NMR spectroscopy, liver-derived lactate was also higher in males. HGO was inhibited by insulin in parallel with increased phosphorylation of Akt, without any sex-differences in insulin sensitivity. However, the degree of Thr172-phosphorylated AMP kinase (AMPK) was higher in females, indicating a higher degree of AMPK-dependent actions. CONCLUSIONS: Taken together, males had higher ratios of insulin to glucagon levels, higher levels of glycogen, lower degree of AMPK phosphorylation, higher expression of gluconeogenic genes and higher hepatic glucose output. Possibly these sex-differences reflect a higher ability for the healthy male rat liver to respond to increased energy demands.


Subject(s)
Blood Glucose/analysis , Glycogen/analysis , Liver/metabolism , AMP-Activated Protein Kinase Kinases , Animals , Blood Glucose/genetics , Fasting , Fatty Acids/chemistry , Fatty Acids/metabolism , Female , Gene Expression Regulation , Glycogen/blood , Glycogen/genetics , Insulin/metabolism , Male , Protein Kinases/metabolism , Rats , Rats, Sprague-Dawley , Sex Factors , Triglycerides/analysis , Triglycerides/blood
8.
Nucleic Acids Res ; 36(9): e53, 2008 May.
Article in English | MEDLINE | ID: mdl-18420656

ABSTRACT

Although RNA interference as a tool for gene knockdown is a great promise for future applications, the specificity of small interfering RNA (siRNA)-mediated gene silencing needs to be thoroughly investigated. Most research regarding siRNA specificity has involved analysis of affected off-target genes instead of exploring the specificity of the siRNA itself. In this study we have developed an efficient method for generating a siRNA target library by combining a siRNA target validation vector with a nucleotide oligomix. We have used this library to perform an analysis of the silencing effects of a functional siRNA towards its target site with double-nucleotide mismatches. The results indicated that not only the positions of the mismatched base pair have an impact on silencing efficiency but also the identity of the mismatched nucleotide. Our data strengthen earlier observations of widespread siRNA off-target effects and shows that approximately 35% of the double-mutated target sites still causes knockdown efficiency of >50%. We also provide evidence that there may be substantial differences in knockdown efficiency depending on whether the mutations are positioned within the siRNA itself or in the corresponding target site.


Subject(s)
Base Pair Mismatch , RNA Interference , RNA, Small Interfering/chemistry , Cell Line , Gene Library , Humans , Membrane Cofactor Protein/genetics , Mutation , Nucleotides/chemistry
9.
Biology (Basel) ; 9(3)2020 Feb 27.
Article in English | MEDLINE | ID: mdl-32121009

ABSTRACT

One of the potential biomarkers for ovarian cancer patients is high serum level of prolactin (PRL), which is a growth factor that may promote tumor cell growth. The prolactin receptor (PRLR) and human cytomegalovirus (HCMV) proteins are frequently detected in ovarian tumor tissue specimens, but the potential impact of HCMV infection on the PRL system have so far not been investigated. In this study, HCMV's effects on PRL and PRLR expression were assessed in infected ovarian cancer cells (SKOV3) by PCR and Western blot techniques. The levels of both PRL and PRLR transcripts as well as the corresponding proteins were highly increased in HCMV-infected SKOV3 cells. Tissue specimens obtained from 10 patients with ovarian cancer demonstrated high expression of PRLR, HCMV-IE, and pp65 proteins. Extensive expression of PRLR was detected in all examined ovarian tumor tissue specimens except for one from a patient who had focal expression of PRLR and this patient was HCMV-negative in her tumor. In conclusion, PRL and PRLR were induced to high levels in HCMV-infected ovarian cancer cells and PRLR expression was extensively detected in HCMV-infected ovarian tissue specimens. Highly induced PRL and PRLR by HCMV infection may be of relevance for the oncomodulatory role of this virus in ovarian cancer.

10.
Heliyon ; 6(4): e03797, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32322744

ABSTRACT

The physiological role of prolactin (PRL) in the heart, and in particular the diabetic heart, are largely unknown. The effects of PRL on ventricular myocyte shortening and Ca2+ transport in the streptozotocin (STZ) - induced diabetic and in age-matched control rats were investigated. PRL receptor protein, myocyte shortening, intracellular [Ca2+], L-type Ca2+ current were measured by Western blot, cell imaging, fluorescence photometry and whole-cell patch-clamp techniques, respectively. Compared to normal Tyrode solution (NT), PRL (50 ng/ml) significantly (p < 0.05) increased the amplitude of shortening in myocytes from control (7.43 ± 0.38 vs. 9.68 ± 0.46 %) and diabetic (6.57 ± 0.24 vs. 8.91 ± 0.44 %) heart (n = 44-49 cells). Compared to NT, PRL (50 ng/ml) significantly increased the amplitude of Ca2+ transients in myocytes from control (0.084 ± 0.004 vs. 0.115 ± 0.007 Fura-2 ratio units) and diabetic (0.087 ± 0.007 vs. 0.112 ± 0.006 Fura-2 ratio units) heart (n = 36-50 cells). PRL did not significantly alter the amplitude of caffeine-evoked Ca2+ transients however, PRL significantly increased the fractional release of Ca2+ in myocytes from control (21 %) and diabetic (14 %) and heart. The rate of Ca2+ transient recovery following PRL treatment was significantly increased in myocytes from diabetic and control heart. Amplitude of L-type Ca2+ current was not significantly altered by diabetes or by PRL. PRL increased the amplitude of shortening and Ca2+ transients in myocytes from control and diabetic heart. Increased fractional release of sarcoplasmic reticulum Ca2+ may partly underlie the positive inotropic effects of PRL in ventricular myocytes from control and STZ-induced diabetic rat.

11.
BMC Mol Biol ; 10: 13, 2009 Feb 23.
Article in English | MEDLINE | ID: mdl-19236699

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) are short non-coding RNAs playing an important role in post-transcriptional regulation of gene expression. We have previously shown that hepatic transcript profiles are different between males and females; that some of these differences are under the regulation of growth hormone (GH); and that mild starvation diminishes some of the differences. In this study, we tested if hepatic miRNAs are regulated in a similar manner. RESULTS: Using microarrays, miRNA screening was performed to identify sex-dependent miRNAs in rat liver. Out of 324 unique probes on the array, 254 were expressed in the liver and eight (3% of 254) of those were found to be different between the sexes. Among the eight putative sex-different miRNAs, only one female-predominant miRNA (miR-29b) was confirmed using quantitative real-time PCR. Furthermore, 1 week of continuous GH-treatment in male rats reduced the levels of miR-451 and miR-29b, whereas mild starvation (12 hours) raised the levels of miR-451, miR-122a and miR-29b in both sexes. The biggest effects were obtained on miR-29b with GH-treatment. CONCLUSION: We conclude that hepatic miRNA levels depend on the hormonal and nutritional status of the animal and show that miR-29b is a female-predominant and GH-regulated miRNA in rat liver.


Subject(s)
Gene Expression Regulation , Growth Hormone/physiology , Liver/drug effects , Liver/metabolism , MicroRNAs/metabolism , Animal Nutritional Physiological Phenomena , Animals , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Male , Rats , Sex Factors , Starvation/physiopathology
12.
PLoS One ; 14(5): e0215831, 2019.
Article in English | MEDLINE | ID: mdl-31063493

ABSTRACT

Increasing evidence suggests that signaling through the prolactin/prolactin receptor axis is important for stimulation the growth of many cancers including glioblastoma multiforme, breast and ovarian carcinoma. Efficient inhibitors of signaling have previously been developed but their applicability as cancer drugs is limited by the short in vivo half-life. In this study, we show that a fusion protein, consisting of the prolactin receptor antagonist PrlRA and an albumin binding domain for half-life extension can be expressed as inclusion bodies in Escherichia coli and efficiently refolded and purified to homogeneity. The fusion protein was found to have strong affinity for the two intended targets: the prolactin receptor (KD = 2.3±0.2 nM) and mouse serum albumin (KD = 0.38±0.01 nM). Further investigation showed that it could efficiently prevent prolactin mediated phosphorylation of STAT5 at 100 nM concentration and above, similar to the PrlRA itself, suggesting a potential as drug for cancer therapy in the future. Complexion with HSA weakened the affinity for the receptor to 21±3 nM, however the ability to prevent phosphorylation of STAT5 was still prominent. Injection into rats showed a 100-fold higher concentration in blood after 24 h compared to PrlRA itself.


Subject(s)
Prolactin/pharmacology , Receptors, Prolactin/antagonists & inhibitors , STAT5 Transcription Factor/metabolism , Animals , Cell Line, Tumor , Half-Life , Humans , Male , Phosphorylation/drug effects , Prolactin/pharmacokinetics , Rats , Rats, Wistar , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/pharmacology , Tissue Distribution
13.
J Clin Invest ; 115(2): 397-406, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15690087

ABSTRACT

Mice deficient in SOCS2 display an excessive growth phenotype characterized by a 30-50% increase in mature body size. Here we show that the SOCS2-/- phenotype is dependent upon the presence of endogenous growth hormone (GH) and that treatment with exogenous GH induced excessive growth in mice lacking both endogenous GH and SOCS2. This was reflected in terms of overall body weight, body and bone lengths, and the weight of internal organs and tissues. A heightened response to GH was also measured by examining GH-responsive genes expressed in the liver after exogenous GH administration. To further understand the link between SOCS2 and the GH-signaling cascade, we investigated the nature of these interactions using structure/function and biochemical interaction studies. Analysis of the 3 structural motifs of the SOCS2 molecule revealed that each plays a crucial role in SOCS2 function, with the conserved SOCS-box motif being essential for all inhibitory function. SOCS2 was found to bind 2 phosphorylated tyrosines on the GH receptor, and mutational analysis of these amino acids showed that both were essential for SOCS2 function. Together, the data provide clear evidence that SOCS2 is a negative regulator of GH signaling.


Subject(s)
DNA-Binding Proteins/metabolism , Growth Hormone/physiology , Receptors, Somatotropin/metabolism , Repressor Proteins/metabolism , Signal Transduction/physiology , Trans-Activators/metabolism , Amino Acid Motifs/genetics , Animals , Body Weight/drug effects , Body Weight/genetics , Body Weight/physiology , DNA-Binding Proteins/genetics , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Growth Hormone/administration & dosage , Growth Hormone/genetics , Insulin-Like Growth Factor I/physiology , Liver/metabolism , Liver/pathology , Mice , Mice, Knockout , Phosphorylation , Protein Binding/genetics , Protein Binding/physiology , Receptors, Somatotropin/genetics , Repressor Proteins/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Suppressor of Cytokine Signaling Proteins , Trans-Activators/genetics , Tyrosine/metabolism
14.
Mol Endocrinol ; 21(1): 293-311, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17008382

ABSTRACT

The GH-activated signal transducer and activator of transcription 5b (STAT5b) is an essential regulator of somatic growth. The transcriptional response to STAT5b in liver is poorly understood. We have combined microarray-based expression profiling and phylogenetic analysis of gene regulatory regions to study the interplay between STAT5b and GH in the regulation of hepatic gene expression. The acute transcriptional response to GH in vivo after a single pulse of GH was studied in the liver of hypophysectomized rats in the presence of either constitutively active or a dominant-negative STAT5b delivered by adenoviral gene transfer. Genes showing differential expression in these two situations were analyzed for the presence of STAT5b binding sites in promoter and intronic regions that are phylogenetically conserved between rats and humans. Using this approach, we showed that most rapid transcriptional effects of GH in the liver are not results of direct actions of STAT5b. In addition, we identified novel STAT5b cis regulatory elements in genes such as Frizzled-4, epithelial membrane protein-1, and the suppressor of cytokine signaling 2 (SOCS2). Detailed analysis of SOCS2 promoter demonstrated its direct transcriptional regulation by STAT5b upon GH stimulation. A novel response element was identified within the first intron of the human SOCS2 gene composed of an E-box followed by tandem STAT5b binding sites, both of which are required for full GH responsiveness. In summary, we demonstrate the power of combining transcript profiling with phylogenetic sequence analysis to define novel regulatory paradigms.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation , Liver/metabolism , STAT5 Transcription Factor/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Base Sequence , Growth Hormone/metabolism , Humans , Male , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Phylogeny , Rats , Rats, Sprague-Dawley , Signal Transduction
15.
Pediatrics ; 142(3)2018 09.
Article in English | MEDLINE | ID: mdl-30093539

ABSTRACT

BACKGROUND: Prenatal exposure to metabolic disturbances is associated with increased risk of offspring neurodevelopmental impairment and autism spectrum disorder, while little is known about the joint effect of maternal obesity and diabetes. With this study, we aim to assess the joint effect of maternal obesity and diabetes on the risk for offspring psychiatric and mild neurodevelopmental disorders. METHODS: Nationwide registries were used to link data of all live births in Finland between 2004 and 2014 (n = 649 043). Cox proportional hazards modeling adjusting for potential confounders was applied to estimate the effect of maternal obesity, pregestational diabetes mellitus (PGDM), and gestational diabetes mellitus, as well as their joint effects, on the outcomes of offspring psychiatric and mild neurodevelopmental diagnoses and offspring prescription of psychotropic drugs. RESULTS: Among mothers without diabetes, severely obese mothers had 67% to 88% increased risk of having a child with mild neurodevelopmental disorders (hazard risk ratio [HR] = 1.69; 95% confidence interval [CI] = 1.54-1.86), attention-deficit/hyperactivity disorder or conduct disorder (HR = 1.88; 95% CI = 1.58-2.23), and psychotic, mood, and stress-related disorders (HR = 1.67; 95% CI = 1.31-2.13) compared with mothers with a normal BMI. PGDM implied a further risk increase for all groups of psychiatric diagnoses with onset in childhood or adolescence in mothers with severe obesity. Marked effects were found particularly for autism spectrum disorder (HR = 6.49; 95% CI = 3.08-13.69), attention-deficit/hyperactivity disorder and conduct disorder (HR = 6.03; 95% CI = 3.23-11.24), and mixed disorders of conduct and emotions (HR = 4.29; 95% CI = 2.14-8.60). Gestational diabetes mellitus did not increase the risk highly for these offspring disorders. CONCLUSIONS: Maternal PGDM combined with severe maternal obesity markedly increases the risk of several children's psychiatric and mild neurodevelopmental disorders.


Subject(s)
Diabetes, Gestational/physiopathology , Neurodevelopmental Disorders/etiology , Obesity/complications , Prenatal Exposure Delayed Effects/etiology , Adult , Child , Child, Preschool , Cohort Studies , Female , Finland/epidemiology , Humans , Infant , Infant, Newborn , Male , Neurodevelopmental Disorders/epidemiology , Pregnancy , Prenatal Exposure Delayed Effects/epidemiology , Prospective Studies , Registries , Risk Factors
16.
BMC Mol Biol ; 8: 60, 2007 Jul 17.
Article in English | MEDLINE | ID: mdl-17640331

ABSTRACT

BACKGROUND: CD36 is a multiligand receptor involved in various metabolic pathways, including cellular uptake of long-chain fatty acids. Defect function or expression of CD36 can result in dyslipidemia or insulin resistance. We have previously shown that CD36 expression is female-predominant in rat liver. In the present study, hormonal and nutritional regulation of hepatic CD36 expression was examined in male and female rats. Since alternative transcription start sites have been described in murine and human Cd36, we investigated whether alternative CD36 transcripts are differentially regulated in rat liver during these conditions. RESULTS: Sequence information of the rat Cd36 5'-UTR was extended, showing that the gene structure of Cd36 in rat is similar to that previously described in mouse with at least two alternative first exons. The rat Cd36 exon 1a promoter was sequenced and found to be highly similar to murine and human Cd36. We show that alternative first exon usage is involved in the female-predominant expression of CD36 in rat liver and during certain hormonal states that induce CD36 mRNA abundance. Estrogen treatment or continuous infusion of growth hormone (GH) in male rats induced CD36 expression preferentially through the exon 1a promoter. Old age was associated with increased CD36 expression in male rats, albeit without any preferential first exon usage. Intermittent GH treatment in old male rats reversed this effect. Mild starvation (12 hours without food) reduced CD36 expression in female liver, whereas its expression was increased in skeletal muscle. CONCLUSION: The results obtained in this study confirm and extend our previous observation that GH is an important regulator of hepatic CD36, and depending on the mode of treatment (continuous or intermittent) the gene might be either induced or repressed. We suggest that the effects of continuous GH secretion in females (which is stimulatory) and intermittent GH secretion in males (which is inhibitory) explains the sex-different expression of this gene. Furthermore, a female-specific repression of hepatic CD36 in response to food deprivation was found, which was in contrast to a stimulatory effect in skeletal muscle. This demonstrates a tissue-specific regulation of Cd36.


Subject(s)
CD36 Antigens/genetics , Exons/genetics , Growth Hormone/pharmacology , Liver/drug effects , Alternative Splicing , Animals , Base Sequence , Estrogens/pharmacology , Female , Gene Expression Regulation/drug effects , Humans , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Protein Binding , Rats , Rats, Sprague-Dawley , Sequence Analysis, DNA , Sequence Homology, Nucleic Acid , Sex Factors , Starvation/physiopathology , Transcription Factors/metabolism , Transcription, Genetic/drug effects
17.
Mol Endocrinol ; 20(2): 241-53, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16037128

ABSTRACT

GH has been of significant scientific interest for decades because of its capacity to dramatically change physiological growth parameters. Furthermore, GH interacts with a range of other hormonal pathways and is an established pharmacological agent for which novel therapeutical applications can be foreseen. It is easy to see the requirement for a number of postreceptor mechanisms to regulate and control target tissue sensitivity to this versatile hormone. In recent years, some of the components that take part in the down-regulatory mechanism targeting the activated GH receptor (GHR) have been defined, and the physiological significance of some of these key components has begun to be characterized. Down-regulation of the GHR is achieved through a complex mechanism that involves rapid ubiquitin-dependent endocytosis of the receptor, the action of tyrosine phosphatases, and the degradation by the proteasome. The suppressors of cytokine signaling (SOCS) protein family, particularly SOCS2, plays an important role in regulating GH actions. The aim of this review is to summarize collected knowledge, including very recent findings, regarding the intracellular mechanisms responsible for the GHR signaling down-regulation. Insights into these mechanisms can be of relevance to several aspects of GH research. It can help to understand growth-related disease conditions, to explain GH resistance, and may be used to develop pharmaceuticals that enhance some the beneficial actions of endogenously secreted GH in a tissue-specific manner.


Subject(s)
Down-Regulation , Receptors, Somatotropin/metabolism , Signal Transduction , Animals , Antigens, Differentiation/metabolism , Female , Humans , Male , Mice , Protein Tyrosine Phosphatases/metabolism , Rats , Receptors, Immunologic/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism
18.
Sci Rep ; 7: 42800, 2017 02 20.
Article in English | MEDLINE | ID: mdl-28216640

ABSTRACT

SOCS2 is a pleiotropic E3 ligase. Its deficiency is associated with gigantism and organismal lethality upon inflammatory challenge. However, mechanistic understanding of SOCS2 function is dismal due to our unawareness of its protein substrates. We performed a mass spectrometry based proteomic profiling upon SOCS2 depletion and yield quantitative data for ~4200 proteins. Through this screen we identify a novel target of SOCS2, the serine-threonine kinase NDR1. Over-expression of SOCS2 accelerates turnover, while its knockdown stabilizes, endogenous NDR1 protein. SOCS2 interacts with NDR1 and promotes its degradation through K48-linked ubiquitination. Functionally, over-expression of SOCS2 antagonizes NDR1-induced TNFα-stimulated NF-κB activity. Conversely, depletion of NDR1 rescues the effect of SOCS2-deficiency on TNFα-induced NF-κB transactivation. Using a SOCS2-/- mice model of colitis we show that SOCS2-deficiency is pro-inflammatory and negatively correlates with NDR1 and nuclear p65 levels. Lastly, we provide evidence to suggest that NDR1 acts as an oncogene in prostate cancer. To the best of our knowledge, this is the first report of an identified E3 ligase for NDR1. These results might explain how SOCS2-deficiency leads to hyper-activation of NF-κB and downstream pathological implications and posits that SOCS2 induced degradation of NDR1 may act as a switch in restricting TNFα-NF-κB pathway.


Subject(s)
Colitis/metabolism , NF-kappa B/metabolism , Prostatic Neoplasms/metabolism , Protein Serine-Threonine Kinases/chemistry , Proteomics/methods , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Cell Line, Tumor , Colitis/genetics , Disease Models, Animal , Enzyme Stability , Gene Expression Regulation , HEK293 Cells , Humans , Male , Mass Spectrometry , Mice , Protein Serine-Threonine Kinases/metabolism , Suppressor of Cytokine Signaling Proteins/deficiency , Transcriptional Activation , Tumor Necrosis Factor-alpha/metabolism , Ubiquitination
19.
Mol Endocrinol ; 19(3): 781-93, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15563548

ABSTRACT

Suppressor of cytokine signaling-2 (SOCS2)-deficient (SOCS2-/-) mice grow significantly larger than their littermates, suggesting that SOCS2 is important in the negative regulation of the actions of GH and/or IGF-I. The aim of this study was to identify genes and metabolic parameters that might contribute to the SOCS2-/- phenotype. We demonstrate that although SOCS2 deficiency induces significant changes in hepatic gene expression, only a fraction of these overlap with known GH-induced effects in the liver, suggesting that SOCS2 might be an important regulator of other growth factors and cytokines acting on the liver. However, an important role of GH and IGF-I in the phenotype of these animals was demonstrated by an overexpression of IGF-binding protein-3 mRNA in the liver and increased levels of circulating IGF-binding protein-3. Other GH-like effects included diminished serum triglycerides and down-regulation of lipoprotein lipase in adipose tissue. Interestingly, SOCS2-/- mice did not differ from their wild-type littermates in glucose or insulin tolerance tests, which is in contrast with the known diabetogenic effects of GH. Furthermore, there was no evidence of impaired insulin signaling in primary hepatocytes isolated from SOCS2-/- mice. Moreover, increased expression of peroxisome proliferator-activated receptor-gamma coactivator-1alpha mRNA was detected in skeletal muscle, which might contribute to normal glycemic control despite the apparent overactivity of the GH/IGF-I axis. Our data indicate that SOCS2 deficiency partially mimics a state of increased GH activity, but also results in changes that cannot be related to known GH effects.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Repressor Proteins/genetics , Repressor Proteins/physiology , Trans-Activators/genetics , Trans-Activators/physiology , Adipose Tissue/enzymology , Animals , Cluster Analysis , DNA, Complementary/metabolism , Down-Regulation , Glucose/metabolism , Glucose Tolerance Test , Growth Hormone/metabolism , Hepatocytes/metabolism , Insulin/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor I/metabolism , Lipid Metabolism , Lipoprotein Lipase/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Skeletal/metabolism , Oligonucleotide Array Sequence Analysis , Phenotype , Phosphorylation , Phylogeny , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Suppressor of Cytokine Signaling Proteins , Time Factors , Tissue Distribution
20.
PLoS One ; 11(1): e0146653, 2016.
Article in English | MEDLINE | ID: mdl-26765535

ABSTRACT

Pulmonary lymphangioleiomyomatosis (LAM) is a rare lung disease caused by mutations in the tumor suppressor genes encoding Tuberous Sclerosis Complex (TSC) 1 and TSC2. The protein product of the TSC2 gene is a well-known suppressor of the mTOR pathway. Emerging evidence suggests that the pituitary hormone prolactin (Prl) has both endocrine and paracrine modes of action. Here, we have investigated components of the Prl system in models for LAM. In a TSC2 (+/-) mouse sarcoma cell line, down-regulation of TSC2 using siRNA resulted in increased levels of the Prl receptor. In human LAM cells, the Prl receptor is detectable by immunohistochemistry, and the expression of Prl in these cells stimulates STAT3 and Erk phosphorylation, as well as proliferation. A high affinity Prl receptor antagonist consisting of Prl with four amino acid substitutions reduced phosphorylation of STAT3 and Erk. Antagonist treatment further reduced the proliferative and invasive properties of LAM cells. In histological sections from LAM patients, Prl receptor immuno reactivity was observed. We conclude that the Prl receptor is expressed in LAM, and that loss of TSC2 increases Prl receptor levels. It is proposed that Prl exerts growth-stimulatory effects on LAM cells, and that antagonizing the Prl receptor can block such effects.


Subject(s)
Lymphangioleiomyomatosis/metabolism , Receptors, Prolactin/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Lymphangioleiomyomatosis/genetics , Mice , Receptors, Prolactin/genetics , STAT3 Transcription Factor/metabolism , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL