119 results on '"Cattini PA"'
Search Results
2. Diastolic Dysfunction with Normal Ejection Fraction and Reduced Heart Rate in Mice Expressing Human Growth Hormone and Displaying Signs of Growth Hormone Insufficiency.
- Author
-
Jin Y, Xiang B, Dolinsky VW, Kardami E, and Cattini PA
- Subjects
- Animals, Mice, Humans, Male, Echocardiography, Diet, High-Fat adverse effects, Growth Hormone metabolism, Mice, Transgenic, Human Growth Hormone metabolism, Stroke Volume, Heart Rate
- Abstract
Growth hormone (GH) signaling is essential for heart development. Both GH deficiency and excess raise cardiovascular risk. Human (h) and mouse (m) GH differ structurally and functionally: hGH binds both the GH receptor (GHR) and prolactin receptor (PRLR), whereas mGH binds only GHR; thus, there is the potential for differential effects. We generated transgenic (hGH-TG) mice that produce pituitary hGH in response to hypothalamic signaling. These mice grow at the same rate as mGH-expressing wild-type (mGH-WT) mice but are smaller and have higher body fat. Echocardiography was used here to compare hGH-TG and mGH-WT mouse hearts. Male hGH-TG mice show a 48% lower left ventricular mass, 36% lower stroke volume, and 48% reduced cardiac output, resembling GH deficiency. Diastolic dysfunction, restrictive ventricular filling, and lower heart rate are suggested in hGH-TG mice. No significant differences in ejection fraction or fractional shortening were observed, even after high-fat diet (HFD) stress. HFD did not affect RNA markers of cardiac damage, although a possible association between B-type natriuretic peptide RNA levels and heart rate was detected. These observations suggest that diastolic dysfunction related to hGH and/or low GH might be offset by a lower heart rate, while structural changes precede functional effects.
- Published
- 2024
- Full Text
- View/download PDF
3. Effect of high fat diet on maternal behavior, brain-derived neurotrophic factor and neural stem cell proliferation in mice expressing human placental lactogen during pregnancy.
- Author
-
Moazzam S, Noorjahan N, Jin Y, Nagy JI, Kardami E, and Cattini PA
- Subjects
- Animals, Female, Pregnancy, Mice, Humans, Neurogenesis physiology, Brain-Derived Neurotrophic Factor metabolism, Brain-Derived Neurotrophic Factor genetics, Diet, High-Fat adverse effects, Maternal Behavior physiology, Placental Lactogen metabolism, Cell Proliferation, Neural Stem Cells metabolism, Mice, Transgenic
- Abstract
Maternal obesity is a serious health concern because it increases risks of neurological disorders, including anxiety and peripartum depression. In mice, a high fat diet (HFD) in pregnancy can negatively affect placental structure and function as well as maternal behavior reflected by impaired nest building and pup-retrieval. In humans, maternal obesity in pregnancy is associated with reduced placental lactogen (PL) gene expression, which has been linked to a higher risk of depression. PL acting predominantly through the prolactin receptor maintains energy homeostasis and is a marker of placenta villous trophoblast differentiation during pregnancy. Impaired neurogenesis and low serum levels of brain-derived neurotrophic factor (BDNF) have also been implicated in depression. Augmented neurogenesis in brain during pregnancy was reported in the subventricular zone (SVZ) of mice at gestation day 7 and linked to increased prolactin receptor signaling. Here, we used transgenic CD-1 mice that express human (h) PL during pregnancy to investigate whether the negative effects of diet on maternal behavior are mitigated in these (CD-1[hGH/PL]) mice. Specifically, we examined the effect of a HFD on nest building prepartum and pup retrieval postpartum, as well as on brain BDNF levels and neurogenesis. In contrast to wild-type CD-1[WT]mice, CD-1[hGH/PL] mice displayed significantly less anxiety-like behavior, and showed no impairment in prepartum nest building or postpartum pup-retrieval when fed a HFD. Furthermore, the HFD decreased prepartum and increased postpartum BDNF levels in CD-1[WT] but not CD-1[hGH/PL] mice. Finally, neurogenesis in the SVZ as well as phosphorylated mitogen-activated protein kinase, indicative of lactogenic signaling, appeared unaffected by pregnancy and diet at gestation day 7 in CD-1[hGH/PL] mice. These observations indicate that CD-1[hGH/PL] mice are resistant to the negative effects of HFD reported for CD-1[WT] mice, including effects on maternal behaviors and BDNF levels, and potentially, neurogenesis. This difference probably reflects a direct or indirect effect of the products of the hGH/PL transgene., (© 2023 The Authors. Journal of Neuroendocrinology published by John Wiley & Sons Ltd on behalf of British Society for Neuroendocrinology.)
- Published
- 2024
- Full Text
- View/download PDF
4. Increased capacity to maintain glucose homeostasis in a transgenic mouse expressing human but not mouse growth hormone with developing high-fat diet-related insulin resistance, hepatic steatosis and adipose dysfunction.
- Author
-
Jin Y, Jarmasz JS, Sultana S, Cordero-Monroy L, Taylor CG, Zahradka P, Kardami E, and Cattini PA
- Subjects
- Animals, Humans, Mice, Male, Human Growth Hormone metabolism, Human Growth Hormone genetics, Growth Hormone metabolism, Growth Hormone genetics, Prolactin metabolism, Leptin metabolism, Adipocytes metabolism, Adipose Tissue, White metabolism, Mice, Transgenic, Diet, High-Fat adverse effects, Insulin Resistance genetics, Fatty Liver metabolism, Fatty Liver etiology, Fatty Liver genetics, Homeostasis, Glucose metabolism, Adipose Tissue metabolism
- Abstract
The objective was to assess the potential differential effects of human versus mouse growth hormone in vivo, given that human unlike mouse growth hormone can bind prolactin as well as the growth hormone receptor. To this end, a transgenic CD-1 mouse expressing human but not mouse growth hormone was generated, and the phenotypes of male mice fed with a regular chow or high-fat diet were assessed. Pancreas and epididymal white adipose tissue gene expression and/or related function were targeted as the pancreas responds to both prolactin and growth hormone receptor signaling, and catabolic effects like lipolytic activity are more directly attributable to growth hormone and growth hormone receptor signaling. The resulting human growth hormone-expressing mice are smaller than wild-type CD-1 mice, despite higher body fat and larger adipocytes, but both mouse types grow at the same rate with similar bone densities. Unlike wild-type mice, there was no significant delay in glucose clearance in human growth hormone-expressing mice when assessed at 8 versus 24 weeks on a high-fat diet. However, both mouse types showed signs of hepatic steatosis that correlated with elevated prolactin but not growth hormone RNA levels. The larger adipocytes in human growth hormone-expressing mice were associated with modified leptin (higher) and adiponectin (lower) RNA levels. Thus, while limited to observations in the male, the human growth hormone-expressing mice exhibit signs of growth hormone insufficiency and adipocyte dysfunction as well as an initial resistance to the negative effects of high-fat diet on glucose clearance.
- Published
- 2024
- Full Text
- View/download PDF
5. Neurogenesis in the Maternal Rodent Brain: Impacts of Gestation-Related Hormonal Regulation, Stress, and Obesity.
- Author
-
Noorjahan N and Cattini PA
- Subjects
- Animals, Brain, Female, Hormones pharmacology, Humans, Mice, Neurogenesis, Obesity, Pregnancy, Rats, Placenta, Rodentia
- Abstract
In order to maintain maternal behavior, it is important that the maternal rodent brain promotes neurogenesis. Maternal neurogenesis is altered by the dynamic shifts in reproductive hormone levels during pregnancy. Thus, lifestyle events such as gestational stress and obesity that can affect hormone production will affect neuroendocrine control of maternal neurogenesis. However, there is a lack of information about the regulation of maternal neurogenesis by placental hormones, which are key components of the reproductive hormonal profile during pregnancy. There is also little known about how maternal neurogenesis can be affected by health concerns such as gestational stress and obesity, and its relationship to peripartum mental health disorders. This review summarizes the changing levels of neurogenesis in mice and rats during gestation and postpartum as well as regulation of neurogenesis by pregnancy-related hormones. The influence of neurogenesis on maternal behavior is also discussed while bringing attention to the effect of health-related concerns during gestation, such as stress and obesity on neuroendocrine control of maternal neurogenesis. In doing so, this review identifies the gaps in the literature and specifically emphasizes the importance of further research on maternal brain physiology to address them., (© 2021 S. Karger AG, Basel.)
- Published
- 2022
- Full Text
- View/download PDF
6. Elimination of endogenous high molecular weight FGF2 prevents pressure-overload-induced systolic dysfunction, linked to increased FGFR1 activity and NR1D1 expression.
- Author
-
Koleini N, Nickel BE, Nagalingam RS, Landry NM, Fandrich RR, Cheung DYC, Dixon IM, Czubryt MP, Jassal DS, Cattini PA, and Kardami E
- Subjects
- Animals, Male, Mice, Mice, Knockout, Rats, Signal Transduction, Blood Pressure genetics, Fibroblast Growth Factor 2 therapeutic use, Nuclear Receptor Subfamily 1, Group D, Member 1 metabolism
- Abstract
Fibroblast growth factor 2 (FGF2), produced as high (Hi-) and low (Lo-) molecular weight isoforms, is implicated in cardiac response to injury. The role of endogenous FGF2 isoforms during chronic stress is not well defined. We investigated the effects of endogenous Hi-FGF2 in a mouse model of simulated pressure-overload stress achieved by transverse aortic constriction (TAC) surgery. Hi-FGF2 knockout mice, expressing only Lo-FGF2, FGF2(Lo), and wild-type mice, FGF2(WT), expressing both Hi-FGF2 and Lo-FGF2, were used. By echocardiography, a decline in systolic function was observed in FGF2(WT) but not FGF2(Lo) mice compared to corresponding sham-operated animals at 4-8 weeks post-TAC surgery. TAC surgery increased markers of myocardial stress/damage including B-type natriuretic peptide (BNP) and the pro-cell death protein BCL2/adenovirus E1B 19 kDa protein-interacting protein-3 (Bnip3) in FGF2(WT) but not FGF2(Lo) mice. In FGF2(Lo) mice, cardiac levels of activated FGF receptor 1 (FGFR1), and downstream signals, including phosphorylated mTOR and p70S6 kinase, were elevated post-TAC. Finally, NR1D1 (nuclear receptor subfamily 1 group D member 1), implicated in cardioprotection from pressure-overload stress, was downregulated or upregulated in the presence or absence, respectively, of Hi-FGF2 expression, post-TAC surgery. In wild-type cardiomyocyte cultures, endothelin-1 (added to simulate pressure-overload signals) caused NR1D1 downregulation and BNP upregulation, similar to the effect of TAC surgery on the FGF2(WT) mice. The NR1D1 agonist SR9009 prevented BNP upregulation, simulating post-TAC findings in FGF2(Lo) mice. We propose that elimination of Hi-FGF2 is cardioprotective during pressure-overload by increasing FGFR1-associated signaling and NR1D1 expression., (© 2021. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.)
- Published
- 2021
- Full Text
- View/download PDF
7. Effects of high fat diet-induced obesity and pregnancy on prepartum and postpartum maternal mouse behavior.
- Author
-
Moazzam S, Jarmasz JS, Jin Y, Siddiqui TJ, and Cattini PA
- Subjects
- Animals, Female, Humans, Lactation, Mice, Pregnancy, Anhedonia, Diet, High-Fat adverse effects, Maternal Behavior, Obesity etiology
- Abstract
Obesity before and during pregnancy negatively affects the mental and physical health of the mother. A diet high in fat also increases the risk for anxiety, depression and cognitive dysfunction. We examined the effects of high fat diet (HFD) -induced obesity and pregnancy on maternal behavior, cognitive function and anxiety- and depression-like behaviors in mice. Four-week-old female CD-1 mice were placed on a HFD or regular chow diet (RCD) for 5 weeks. Mice were maintained on either diet as non-pregnant HFD and RCD groups, or allowed to breed, and then fed these diets throughout gestation, lactation and after weaning, as pregnant HFD and RCD groups. Mice on HFD but not on RCD for 5 weeks pre-pregnancy significantly gained weight and had impaired glucose clearance. Maternal behavior was assessed by nest building prepartum and pup-retrieval postpartum. Anxiety-like behavior was evaluated both prepartum and postpartum by elevated plus maze and cognitive function was assessed by the novel object recognition test postpartum. Anhedonia, a measure of impaired reward function, is an endophenotype of depression and was assessed by sucrose preference test pre- and post-weaning in dams. Mice on HFD in pregnancy exhibited both impaired maternal behavior and cognitive function in the postpartum period. We did not detect measurable differences between the HFD and RCD groups in anxiety-like behavior in the prepartum period. In contrast, HFD was also associated with anhedonia in pregnant mice pre-weaning, and anxiety-like behavior post-weaning. Thus, HFD has a negative effect on maternal behavior in the outbred CD-1 mouse, which provides a model to study associated outcomes and related mechanisms., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
8. A potential role for insulin treatment during pregnancy in reducing postpartum psychological distress in maternal obesity: an administrative population health study.
- Author
-
Jarmasz JS, Anderson A, Bock ME, Jin Y, Cattini PA, and Ruth C
- Subjects
- Adolescent, Female, Humans, Placenta, Postpartum Period, Pregnancy, Risk Factors, Depression, Postpartum epidemiology, Insulins, Pregnancy in Obesity, Population Health, Psychological Distress
- Abstract
Background: Studies have found an association between obesity and an increased risk for peripartum depression, which has also been linked to decreased placental lactogen levels. In addition, women with obesity treated for gestational diabetes with insulin were found to have increased levels of placental lactogen. Treatment options exist for perinatal and postpartum depression however they pose a risk to the developing offspring. Thus, prevention as well as markers for early identification of peripartum depression are needed. Therefore, our study objective is to identify the association between insulin treatment in pregnancy and the risk of postpartum psychological distress (abbreviated here as PPD) among cohorts of women with and without obesity., Methods: Administrative health data (2002/03-2018/19) were used to identify a cohort of women (age 15+ years) who gave birth (N = 250,746) and had no pre-existing mood/anxiety disorders or diabetes (N = 222,863 excluded). Women were then divided into two groups: lean (N = 17,975) and with obesity (N = 9908), which was identified by a recorded maternal weight of > 38 to < 65.6 kg and ≥ 85 to < 186 kg (respectively). The risk of PPD within one year after delivery with and without insulin treatment was assessed by Poisson regression analysis. Models were adjusted for maternal age group (at pregnancy start date) and area-level income (at delivery)., Results: The unadjusted risk of PPD was higher in the obesity group (8.56%; 95% CI 8.00-9.15) than in the lean group (6.93%; 95% CI 6.56-7.33). When no insulin treatment was given during pregnancy, mothers with obesity had a significantly higher risk of PPD than the lean group (aRR 1.27; 95% CI 1.17-1.39; p < 0.0001). However, when women with obesity and insulin treatment were compared to the lean group with no insulin treatment, no significant difference in the risk of PPD was observed between the groups (aRR 1.30; 95% CI 0.83-2.02; p = 0.248)., Conclusion: This is the first study to demonstrate a positive association between insulin treatment in pregnancy among women with obesity and reduced PPD rates, suggesting insulin as a possible preventative measure. However, the biological mechanism behind the observed positive effect of insulin on PPD rates remains to be investigated.
- Published
- 2021
- Full Text
- View/download PDF
9. Dexamethasone Rescues an Acute High-Fat Diet-Induced Decrease in Human Growth Hormone Gene Expression in Male Partially Humanized CD-1 Mice.
- Author
-
Jin Y, Jarmasz JS, and Cattini PA
- Subjects
- Animals, Human Growth Hormone genetics, Male, Mice, Mice, Transgenic, Time Factors, Dexamethasone pharmacology, Diet, High-Fat adverse effects, Gene Expression Regulation drug effects, Human Growth Hormone biosynthesis
- Abstract
Obesity in puberty, already a time of insulin resistance, increases the risk of developing type 2 diabetes. Human (h) growth hormone (GH) levels also peak during puberty, where it contributes to growth and energy homeostasis through positive effects on maintaining pancreatic β cell mass. Thus, it is important to understand the effects of overeating and obesity on hGH production in puberty. Three days of overeating in young male adults or high-fat diet (HFD) in pubescent male transgenic ( 171hGH/CS ) CD-1 mice containing the hGH gene ( hGH-N ) results in excess insulin and a decrease in hGH production. This reduction in these mice occurred during the light phase of the daily cycle, and was associated with decreased availability of the clock-related transcription factor Brain and Muscle ARNT-Like 1 (Bmal1). However, the HFD-induced decrease in hGH-N expression was blocked by forced daily swim activity, which is expected to increase glucocorticoid (GC) levels. The aim of the study was to assess whether GCs, specifically daily injections with a pharmacological dose of dexamethasone (DEX) in the light or dark phase of the daily cycle, can limit the negative effect of HFD for 3 days on hGH-N expression in male 171hGH/CS mice. DEX treatment increased or rescued hGH-N RNA levels, and was associated with elevated Bmal1 transcripts when assessed 12 h after final treatment, and at a time when serum corticosterone levels were suppressed >90%. In addition, a diet-dependent effect on hGH-N RNA levels was observed at 36 h after final treatment, but only in the light stage, presumably due to residual effects of DEX treatment and/or recovery of endogenous corticosterone levels. This is the first evidence for a direct effect of GCs on hGH-N expression in vivo and the ability to potentially limit the negative effect of overeating/obesity on hGH production in puberty.
- Published
- 2021
- Full Text
- View/download PDF
10. Sleep deprivation and diet affect human GH gene expression in transgenic mice in vivo.
- Author
-
Jarmasz JS, Jin Y, Vakili H, and Cattini PA
- Abstract
Human (h) growth hormone (GH) production studies are largely limited to effects on secretion. How pituitary hGH gene (hGH-N/GH1) expression is regulated is important in our understanding of the role hGH plays in physiology and disease. Here we assess for the first time the effect of sleep deprivation (SD) and high-fat diet (HFD) on hGH-N expression in vivo using partially humanized 171hGH/CS transgenic (TG) mice, and attempted to elucidate a role for DNA methylation. Activation of hGH-N expression requires interactions between promoter and upstream locus control region (LCR) sequences including pituitary-specific hypersensitive site (HS) I/II. Both SD and diet affect hGH secretion, but the effect of SD on hGH-N expression is unknown. Mice fed a HFD or regular chow diet for 3 days underwent SD (or no SD) for 6 h at Zeitgeber time (ZT) 3. Serum and pituitaries were assessed over 24 h at 6-h intervals beginning at ZT 14. SD and HFD caused significant changes in serum corticosterone and insulin, as well as hGH and circadian clock-related gene RNA levels. No clear association between DNA methylation and the negative effects of SD or diet on hGH RNA levels was observed. However, a correlation with increased methylation at a CpG (cytosine paired with a guanine) in a putative E-box within the hGH LCR HS II was suggested in situ. Methylation at this site also increased BMAL1/CLOCK-related nuclear protein binding in vitro. These observations support an effect of SD on hGH synthesis at the level of gene expression.
- Published
- 2020
- Full Text
- View/download PDF
11. Obesity and regulation of human placental lactogen production in pregnancy.
- Author
-
Cattini PA, Jin Y, Jarmasz JS, Noorjahan N, and Bock ME
- Subjects
- Animals, Female, Gene Expression Regulation, Humans, Obesity genetics, Placenta metabolism, Placental Lactogen genetics, Obesity metabolism, Placental Lactogen biosynthesis, Pregnancy metabolism
- Abstract
The four genes coding for placental members of the human (h) growth hormone (GH) family include two that code independently for placental lactogen (PL), also known as chorionic somatomammotrophin hormone, one that codes for placental growth hormone (PGH) and a pseudogene for which RNA but no protein product is reported. These genes are expressed preferentially in the villus syncytiotrophoblast of the placenta in pregnancy. In higher primates, the placental members, including hPL and PGH, are the result of multiple duplication events of the GH gene. This contrasts with rodents and ruminants, where PLs result from duplication of the prolactin (PRL) gene. Thus, unlike their mouse counterparts, the hPL and PGH hormones bind both lactogenic and somatogenic receptors with varying affinity. Roles influenced by nutrient availability in both metabolic control in pregnancy and maternal behaviour are supported. However, the effect maternal obesity has on the activation of placental members of the hGH gene family, particularly the expression and function of those genes, is poorly understood. Evidence from partially humanised hGH/PL transgenic mice indicates that both the remote upstream hPL locus control region (LCR) and more gene-related regulatory regions are required for placental expression in vivo. Furthermore, a specific pattern of interactions between the LCR and hPL gene promoter regions is detected in term placenta chromatin from women with a normal body mass index (BMI) in the range 18.5-25 kg m
-2 by chromosome conformation capture assay. This pattern is disrupted with maternal obesity (class II BMI > 35 kg m-2 ) and associated with a > 40% decrease in term hPL RNA levels, as well as serum hPL but not PRL levels, during pregnancy. The relative importance of the chromosomal architecture and predicted properties for transcription factor participation in terms of hPL production and response to obesity are considered, based on comparison with components required for efficient human pituitary GH gene expression., (© 2020 British Society for Neuroendocrinology.)- Published
- 2020
- Full Text
- View/download PDF
12. Negative Effects of Cyclic Palmitate Treatment on Glucose Responsiveness and Insulin Production in Mouse Insulinoma Min6 Cells Are Reversible.
- Author
-
Nie Y, Li J, Jin Y, Nyomba BLG, Cattini PA, and Vakili H
- Subjects
- Animals, Cell Death drug effects, Cell Line, Tumor, Dose-Response Relationship, Drug, Mice, Glucose pharmacology, Insulin biosynthesis, Insulinoma pathology, Palmitates pharmacology
- Abstract
Pancreatic β-cell failure is characterized by compromised insulin secretion in response to glucose, which ultimately results in hyperglycemia, the clinical hallmark of type 2 diabetes mellitus (T2DM). Acute exposure to plasma free fatty acids (FFAs) potentiates glucose stimulated insulin secretion (GSIS), while chronic exposure impairs GSIS, and the latter has been associated with the mechanism of β cell failure in obesity linked T2DM. By contrast, growth hormone (GH) signaling has been linked positively to GSIS in β cells. Numerous studies have examined chronic exposure of β cells to elevated FFAs both with in vivo cohorts and in vitro models. Little attention, however, has been given to the fluctuation of plasma FFA levels due to rhythmic effects that are affected by daily diet and fat intake. Mouse insulinoma Min6 cells were exposed to cyclic/daily palmitate treatment over 2 and 3 days to assess effects on GSIS. Cyclic/daily palmitate treatment with a period of recovery negatively affected GSIS in a dose-dependent manner. Removal of palmitate after two cycles/day resulted in reversal of the effect on GSIS, which was also reflected by relative gene expression involved in insulin biosynthesis (Ins1, Ins2, Pdx1, and MafA) and GSIS (glucose 2 transporter and glucokinase). Modest positive effects on GSIS and glucokinase transcript levels were also observed when Min6 cells were cotreated with human GH and palmitate. These observations indicate that like continuous palmitate treatment, cyclic exposure to palmitate can acutely impair GSIS over 48 and 72 h. However, they also suggest that the negative effects of short periods of exposure to FFAs on β cell function remain reversible.
- Published
- 2019
- Full Text
- View/download PDF
13. Elimination or neutralization of endogenous high-molecular-weight FGF2 mitigates doxorubicin-induced cardiotoxicity.
- Author
-
Koleini N, Santiago JJ, Nickel BE, Sequiera GL, Wang J, Fandrich RR, Jassal DS, Dhingra S, Kirshenbaum LA, Cattini PA, and Kardami E
- Subjects
- Animals, Cardiac Output, Cardiotoxicity, Cells, Cultured, Culture Media, Conditioned pharmacology, Female, Fibroblast Growth Factor 2 genetics, Heart physiology, Humans, Male, Mice, Rats, Doxorubicin toxicity, Fibroblast Growth Factor 2 metabolism, Heart drug effects, Myocytes, Cardiac drug effects, Myofibroblasts metabolism
- Abstract
Cardiac fibroblast growth factor 2 (FGF2) exerts multiple paracrine activities related to cardiac response to injury. Endogenous FGF2 is composed of a mixture of 70% high- and 30% low-molecular-weight isoforms (Hi-FGF2 and Lo-FGF2, respectivley); although exogenously added Lo-FGF2 is cardioprotective, the roles of endogenous Hi-FGF2 or Lo-FGF2 have not been well defined. Therefore, we investigated the effect of elimination of Hi-FGF2 expression on susceptibility to acute cardiac damage in vivo caused by an injection of the genotoxic drug doxorubicin (Dox). Mice genetically depleted of endogenous Hi-FGF2 and expressing only Lo-FGF2 [FGF2(Lo) mice] were protected from the Dox-induced decline in ejection fraction displayed by their wild-type FGF2 [FGF2(WT)] mouse counterparts, regardless of sex, as assessed by echocardiography for up to 10 days post-Dox treatment. Because cardiac FGF2 is produced mainly by nonmyocytes, we next addressed potential contribution of fibroblast-produced FGF2 on myocyte vulnerability to Dox. In cocultures of neonatal rat cardiomyocytes (r-cardiomyocytes) with mouse fibroblasts from FGF2(WT) or FGF2(Lo) mice, only the FGF2(Lo)-fibroblast cocultures protected r-cardiomyocytes from Dox-induced mitochondrial and cellular damage. When r-cardiomyocytes were cocultured with or exposed to conditioned medium from human fibroblasts, neutralizing antibodies for human Hi-FGF-2, but not total FGF2, mitigated Dox-induced injury of cardiomyocytes. We conclude that endogenous Hi-FGF2 reduces cardioprotection by endogenous Lo-FGF2. Antibody-based neutralization of endogenous Hi-FGF2 may offer a prophylactic treatment against agents causing acute cardiac damage. NEW & NOTEWORTHY Cardiomyocytes, in vivo and in vitro, were protected from the deleterious effects of the anticancer drug doxorubicin by the genetic elimination or antibody-based neutralization of endogenous paracrine high-molecular-weight fibroblast growth factor 2 isoforms. These findings have a translational potential for mitigating doxorubicin-induced cardiac damage in patients with cancer by an antibody-based treatment.
- Published
- 2019
- Full Text
- View/download PDF
14. Cardiac Fgf-16 Expression Supports Cardiomyocyte Survival and Increases Resistance to Doxorubicin Cytotoxicity.
- Author
-
Wang J, Xiang B, Dolinsky VW, Kardami E, and Cattini PA
- Subjects
- Adenoviridae genetics, Adenoviridae metabolism, Animals, Animals, Newborn, Apoptosis drug effects, Atrial Natriuretic Factor genetics, Atrial Natriuretic Factor metabolism, Biological Transport drug effects, Cardiomyopathies diagnostic imaging, Cardiomyopathies pathology, Cardiomyopathies prevention & control, Cell Survival drug effects, Echocardiography, Fibroblast Growth Factors agonists, Fibroblast Growth Factors antagonists & inhibitors, Fibroblast Growth Factors metabolism, Gene Expression Regulation, Genetic Vectors chemistry, Genetic Vectors metabolism, Heart Function Tests, Injections, Intraperitoneal, Myocardium metabolism, Myocardium pathology, Myocytes, Cardiac drug effects, Myocytes, Cardiac pathology, Primary Cell Culture, Pyrroles pharmacology, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Rats, Rats, Sprague-Dawley, Receptor, Fibroblast Growth Factor, Type 1 antagonists & inhibitors, Receptor, Fibroblast Growth Factor, Type 1 metabolism, Signal Transduction, Cardiomyopathies genetics, Cytotoxins toxicity, Doxorubicin toxicity, Fibroblast Growth Factors genetics, Myocytes, Cardiac metabolism, Receptor, Fibroblast Growth Factor, Type 1 genetics
- Abstract
The fibroblast growth factor (FGF) 16 gene is preferentially expressed by cardiomyocytes after birth with levels increasing into adulthood. Null mice and isolated heart studies suggest a role for FGF-16 in cardiac maintenance and survival, including increased resistance to doxorubicin (DOX)-induced injury. A single treatment with DOX was also shown to rapidly deplete endogenous rat FGF-16 mRNA at 6 h in both adult heart and neonatal cardiomyocytes. However, the effect of DOX on rat cardiac function at the time of decreased FGF-16 gene expression and the effect of FGF-16 availability on cardiomyocyte survival, including in the context of acute DOX cytotoxicity, have not been reported. The objective was to assess the effect of acute (6 and 24 h) DOX treatment on cardiac function and the effects of FGF-16 small interfering RNA "knockdown," as well as adenoviral overexpression, in the context of acute DOX cytotoxicity, including cardiomyocyte survival and DOX efflux transport. A significant decrease in heart systolic function was detected by echocardiography in adult rats treated with 15 mg DOX/kg at 6 h; however, unlike FGF-16, there was no change in atrial natriuretic peptide transcript levels. Both systolic and diastolic dysfunctions were observed at 24 h. In addition, specific FGF-16 "knockdown" in neonatal rat cardiomyocytes results in a significant increase in cell death. Conversely, adenoviral FGF-16 overexpression was associated with a significant decrease in cardiomyocyte injury as a result of 1 μM DOX treatment. A specific increase in efflux transporter gene expression and DOX efflux was also seen, which is consistent with a reduction in DOX cytotoxicity. Finally, the increased efflux and decreased DOX-induced damage with FGF-16 overexpression were blunted by inhibition of FGF receptor signaling. These observations are consistent with FGF-16 serving as an endogenous cardiomyocyte survival factor, which may involve a positive effect on regulating efflux transport to reduce cardiotoxicity.
- Published
- 2018
- Full Text
- View/download PDF
15. Chromosomal architecture and placental expression of the human growth hormone gene family are targeted by pre-pregnancy maternal obesity.
- Author
-
Jin Y, Vakili H, Liu SY, Menticoglou S, Bock ME, and Cattini PA
- Subjects
- Body Mass Index, Chromatin Immunoprecipitation, Chromosomes, Human metabolism, Female, Gene Expression, Growth Hormone metabolism, Human Growth Hormone metabolism, Humans, Immunoblotting, Insulin Resistance, Obesity metabolism, Placental Hormones metabolism, Placental Lactogen metabolism, Pregnancy, Pregnancy Complications metabolism, Promoter Regions, Genetic, Pseudogenes, RNA, Messenger metabolism, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Chromosomes, Human genetics, Growth Hormone genetics, Human Growth Hormone genetics, Obesity genetics, Placenta metabolism, Placental Hormones genetics, Placental Lactogen genetics, Pregnancy Complications genetics
- Abstract
The human (h) placental lactogenic hormone chorionic somatomammotropin (CS) is highly produced during pregnancy and acts as a metabolic adaptor in response to maternal insulin resistance. Maternal obesity can exacerbate this "resistance", and a >75% decrease in CS RNA levels was observed in term placentas from obese vs. lean women. The genes coding for hCS ( hCS-A and hCS-B) and placental growth hormone ( hGH-V) as well as the hCS-L pseudogene and pituitary growth hormone (GH) gene ( hGH-N) are located at a single locus on chromosome 17. Three remote hypersensitive sites (HS III-V) located >28 kb upstream of hGH-N as well as local hCS gene promoter and enhancer regions are implicated in hCS gene expression. A placenta-specific chromosomal architecture, including interaction between HS III-V and hCS but not hGH gene promoters, was detected in placentas from lean women (BMI <25 kg/m
2 ) by using the chromosome conformation capture assay. This architecture was disrupted by pre-pregnancy maternal obesity (BMI >35 kg/m2 ), resulting in a predominant interaction between HS III and the hGH-N promoter, which was also observed in nonplacental tissues. This was accompanied by a decrease in hCS levels, which was consistent with reduced RNA polymerase II and CCAAT/enhancer-binding protein-β association with individual hCS promoter and enhancer sequences, respectively. Thus, pre-pregnancy maternal obesity disrupts the placental hGH/CS gene locus chromosomal architecture. However, based on data from obese women who develop GDM, insulin treatment partially recapitulates the chromosomal architecture seen in lean women and positively affects hCS production, presumably facilitating prolactin receptor-related signaling by hCS.- Published
- 2018
- Full Text
- View/download PDF
16. A useful model to compare human and mouse growth hormone gene chromosomal structure, expression and regulation, and immune tolerance of human growth hormone analogues.
- Author
-
Cattini PA, Bock ME, Jin Y, Zanghi JA, and Vakili H
- Subjects
- Animals, Growth Hormone immunology, Humans, Mice, Chromosomes chemistry, Gene Expression Regulation, Growth Hormone analogs & derivatives, Growth Hormone genetics, Immune Tolerance, Models, Biological
- Abstract
Human (h) pituitary growth hormone (GH) is both physiologically and clinically important. GH reaches its highest circulatory levels in puberty, where it contributes to energy homeostasis and somatogenic growth. GH also helps to maintain tissues and organs and, thus, health and homeostasis. A reduction in the rate of hGH production begins in middle age but if GH insufficiency occurs this may result in tissue degenerative and metabolic diseases. As a consequence, hGH is prescribed under conditions of GH deficiency and, because of its lipolytic activity, stimulation of hGH release has also been used to treat obesity. However, studies of normal GH production and particularly synthesis versus secretion are not feasible in humans as they require sampling normal pituitaries from living subjects. Furthermore, human (or primate) GH structure and, as such, regulation and potential function, is distinct from non-primate rodent GH. As a result, most information about hGH regulation comes from measurements of secreted levels of GH in humans. Thus, partially humanized hGH transgenic mice, generated containing fragments of human chromosome 17 that include the intact hGH gene locus and many thousands of flanking base pairs as well as the endogenous mouse (m) GH gene provide a potentially useful model. Here we review this mouse model in terms of its ability to allow comparison of hGH versus mGH gene expression, and specifically: (i) GH locus structure as well as regulated and rhythmic expression; (ii) their ability to model a clinical assessment of hGH production in response to overeating and hyperinsulinemia as well as a possible effect of exercise, and (iii) their hGH-related immune tolerance and thus potential for testing hGH-related analogue immunogenicity., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
17. Fibroblast growth factor-2-mediated protection of cardiomyocytes from the toxic effects of doxorubicin requires the mTOR/Nrf-2/HO-1 pathway.
- Author
-
Koleini N, Nickel BE, Wang J, Roveimiab Z, Fandrich RR, Kirshenbaum LA, Cattini PA, and Kardami E
- Abstract
Background: Cardiotoxic side effects impose limits to the use of anti-tumour chemotherapeutic drugs such as doxorubicin (Dox). There is a need for cardioprotective strategies to prevent the multiple deleterious effects of Dox. Here, we examined the ability of administered fibroblast growth factor-2 (FGF-2), a cardioprotective protein that is synthesized as high and low molecular weight (Hi-, Lo-FGF-2) isoforms, to prevent Dox-induced: oxidative stress; cell death; lysosome dysregulation; and inactivation of potent endogenous protective pathways, such as the anti-oxidant/detoxification nuclear factor erythroid-2-related factor (Nrf-2), heme oxygenase-1 (HO-1) axis., Methods and Results: Brief pre-incubation of neonatal rat cardiomyocyte cultures with either Hi- or Lo-FGF-2 reduced the Dox-induced: oxidative stress; apoptotic/necrotic cell death; lysosomal dysregulation; decrease in active mammalian target of Rapamycin (mTOR). FGF-2 isoforms prevented the Dox-induced downregulation of Nrf-2, and promoted robust increases in the Nrf-2-downstream targets including the cardioprotective protein HO-1, and p62/SQSTM1, a multifunctional scaffold protein involved in autophagy. Chloroquine, an autophagic flux inhibitor, caused a further increase in p62/SQSTM1, indicating intact autophagic flux in the FGF-2-treated groups. A selective inhibitor for HO-1, Tin-Protoporphyrin, prevented the FGF-2 protection against cell death. The mTOR inhibitor Rapamycin prevented FGF-2 protection, and blocked the FGF-2 effects on Nrf-2, HO-1 and p62/SQSTM1., Conclusions: In an acute setting Hi- or Lo-FGF-2 protect cardiomyocytes against multiple Dox-induced deleterious effects, by a mechanism dependent on preservation of mTOR activity, Nrf-2 levels, and the upregulation of HO-1. Preservation/activation of endogenous anti-oxidant/detoxification defences by FGF-2 is a desirable property in the setting of Dox-cardiotoxicity., Competing Interests: CONFLICTS OF INTEREST None declared.
- Published
- 2017
- Full Text
- View/download PDF
18. Expression of the Cardiac Maintenance and Survival Factor FGF-16 Gene Is Regulated by Csx/Nkx2.5 and Is an Early Target of Doxorubicin Cardiotoxicity.
- Author
-
Wang J, Jin Y, and Cattini PA
- Subjects
- Animals, Cardiotoxicity genetics, Cardiotoxicity metabolism, Gene Knockdown Techniques, Homeobox Protein Nkx-2.5 deficiency, Homeobox Protein Nkx-2.5 genetics, Mice, Promoter Regions, Genetic genetics, Rats, Time Factors, Doxorubicin adverse effects, Fibroblast Growth Factors genetics, Gene Expression Regulation drug effects, Homeobox Protein Nkx-2.5 metabolism, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism
- Abstract
The fibroblast growth factor (FGF) 16 gene (Fgf-16) is preferentially expressed by neonatal cardiomyocytes after birth, with levels increasing into adulthood. Null mice and isolated heart studies suggest a role for FGF-16 in cardiac maintenance and survival, including increased resistance to doxorubicin (DOX)-induced injury. However, the effect of DOX on endogenous FGF-16 synthesis and specifically regulation of cardiac Fgf-16 expression has not been reported. Here we assess the effect of DOX on FGF-16 RNA levels and stability as well as promoter activity and use sequence analysis, knockdown, and overexpression to investigate the role of cardiac transcription factor(s) implicated in the response. Endogenous FGF-16 RNA levels were reduced >70% in 8-week-old rats treated with 15 mg DOX/kg for 6 h. This was modeled in neonatal rat cardiomyocyte cultures, where an equivalent decrease was also seen within 6 h of 1 μM DOX treatment. Six kilobases of mouse Fgf-16 upstream flanking and promoter DNA was also assessed for DOX responsiveness in transfected cardiomyocytes. A decrease in FGF-16 promoter activity was seen with only 747 base pairs containing the Fgf-16 TATA box that includes a putative and highly conserved binding site for the cardiac transcription factor Csx/Nkx2.5. There was also no effect of DOX on FGF-16 RNA stability, consistent with transcriptional control. Levels and binding of Csx/Nkx2.5 to the FGF-16 promoter were reduced with DOX treatment. Knockdown of Csx/Nkx2.5 specifically decreased endogenous FGF-16 RNA and protein levels, whereas Csx/Nkx2.5 overexpression stimulated levels, and increased resistance to the rapid DOX-induced depletion of FGF-16. These observations indicate that Fgf-16 expression is directly regulated by Csx/Nkx2.5 in neonatal cardiomyocytes, and a negative effect of DOX on Csx/Nkx2.5 and, thus, endogenous FGF-16 synthesis may contribute indirectly to its cardiotoxic effects. Targeting FGF-16 levels could, however, offer increased resistance to cardiac injury.
- Published
- 2017
- Full Text
- View/download PDF
19. Evidence for a Circadian Effect on the Reduction of Human Growth Hormone Gene Expression in Response to Excess Caloric Intake.
- Author
-
Vakili H, Jin Y, and Cattini PA
- Subjects
- ARNTL Transcription Factors genetics, ARNTL Transcription Factors metabolism, Animals, CLOCK Proteins genetics, CLOCK Proteins metabolism, HEK293 Cells, Human Growth Hormone genetics, Humans, Mice, Mice, Transgenic, Circadian Rhythm, Energy Intake, Gene Expression Regulation, Human Growth Hormone biosynthesis, Pituitary Gland metabolism, Promoter Regions, Genetic
- Abstract
Rhythmicity of biological functions is fundamental for optimal adaptations to environmental cues. Growth hormone (GH) is a major metabolic homeostatic factor that is secreted with a circadian pattern, but whether it is synthesized rhythmically is unknown. We used transgenic mice containing the human (h) GH gene (hGH1) locus to investigate the rhythmicity of hGH synthesis and secretion and to show that RNA and secreted protein levels oscillate over a 24-h cycle. Analysis of hGH1 promoter sequences revealed an enhancer motif (E-box) element that binds the circadian transcriptional machinery (Bmal1 and Clock). Furthermore, Bmal1/Clock were able to transactivate the hGH1 promoter, and mutation of this E-box element adversely affected basal activity after gene transfer. The ability of Bmal1 to bind the hGH1 promoter region containing the E-box element was confirmed in the hGH1 transgenic mouse pituitary in situ Occupancy was reduced in mice fed a high fat diet during the light (inactive) stage of the daily cycle in mice and corresponded to a decrease in hGH1 RNA levels. The decreases in occupancy and RNA levels were not seen, however, during the dark (active) stage. A chromatin loop required for efficient postnatal hGH1 expression was negatively affected by the high fat diet in the light but not dark stage similar to the pattern observed with Bmal1 association with the promoter region. This is the first evidence that hGH synthesis follows a diurnal rhythm and of dynamic associations of the circadian machinery with a component of a chromosomal structure of the hGH1 locus that is essential for efficient expression., (© 2016 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
20. Expression of Placental Members of the Human Growth Hormone Gene Family Is Increased in Response to Sequential Inhibition of DNA Methylation and Histone Deacetylation.
- Author
-
Ganguly E, Bock ME, and Cattini PA
- Abstract
The genes coding for human (h) chorionic somatomammotropin (CS), hCS-A and hCS-B, and placental growth hormone (GH-V), hGH-V, are located at a single locus on chromosome 17. Efficient expression of these placental genes has been linked to local regulatory (5' P and 3' enhancer) sequences and a remote locus control region (LCR), in part, through gene transfer in placental and nonplacental tumor cells. However, low levels of endogenous hCS/GH-V transcripts are reported in the same cells compared with term placenta, suggesting that chromatin structure, or regulatory region accessibility, versus transcription factor availability contributes to the relatively low levels. To assess individual hCS-A, CS-B, and GH-V gene expression in placental and nonplacental tumor cells and the effect of increasing chromatin accessibility by inhibiting DNA methylation and histone deacetylation using 5-aza-2'-deoxycytidine (azadC) and trichostatin A (TSA). Low levels of hCS-A, CS-B, and GH-V were detected in placental and nonplacental tumor cells compared with term placenta. A significant >5-fold increase in activity was seen in placental, but not nonplacental, cells transfected with hybrid hCS promoter luciferase genes containing 3' enhancer sequences. Pretreatment of placental JEG-3 cells with azadC resulted in a >10-fold increase in hCS-A, CS-B, and GH-V RNA levels with TSA treatment compared with TSA treatment alone. This effect was specific as reversing the treatment regimen did not have the same effect. An assessment of hyperacetylated H3/H4 in JEG-3 cells treated with azadC and TSA versus TSA alone revealed significant increases consistent with a more open chromatin structure, including the hCS 3' enhancer sequences and LCR. These observations suggest that accessibility of remote and local regulatory regions required for efficient placental hGH/CS expression can be restricted by DNA methylation and histone acetylation status. This includes restricting access of the hCS 3' enhancer sequences to available placental enhancer transcription factors.
- Published
- 2015
- Full Text
- View/download PDF
21. Heart-specific expression of FGF-16 and a potential role in postnatal cardioprotection.
- Author
-
Wang J, Sontag D, and Cattini PA
- Subjects
- Animals, Cardiovascular Diseases genetics, Cardiovascular Diseases therapy, Codon, Nonsense, Embryonic Development genetics, Female, Fibroblast Growth Factors metabolism, Fibroblast Growth Factors pharmacology, Heart embryology, Humans, Male, Mice, Mice, Knockout, Pregnancy, Promoter Regions, Genetic, Rats, Risk Factors, TATA Box, Cardiotonic Agents pharmacology, Fibroblast Growth Factors genetics, Gene Expression Regulation, Heart growth & development, Myocardium metabolism
- Abstract
Fibroblast growth factor 16 (FGF-16) was originally cloned from rat heart. Subsequent investigation of mouse FGF-16, including generation of null mice, revealed a specific pattern of expression in the endocardium and epicardium, and role for FGF-16 during embryonic heart development. FGF-16 is expressed mainly in brown adipose tissue during rat embryonic development, but is expressed mainly in the murine heart after birth. There is also an apparent switch from limited endocardial and epicardial expression in the embryo to the myocardium in the perinatal period. The FGF-16 gene and its location on the X chromosome are conserved between human and murine species, and no other member of the FGF family shows this pattern of spatial and temporal expression. The human and murine FGF-16 gene promoter regions also share an equivalent location for TATA sequences, as well as adjacent putative binding sites for transcription factors linked to cardiac expression and response to stress. Recent evidence has implicated nonsense mutation of FGF-16 with increased cardiovascular risk, and FGF-16 supplementation with cardioprotection. Here we review the important role of FGF-16 in embryonic heart development, its gene regulation, and evidence for FGF-16 as an endogenous and exogenous cardiac-specific and protective factor in the postnatal heart. Moreover, given the conservation of the FGF-16 gene and its chromosomal location between species, the question of support for a cardiac role in the human population is also considered., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
22. Energy homeostasis targets chromosomal reconfiguration of the human GH1 locus.
- Author
-
Vakili H, Jin Y, and Cattini PA
- Subjects
- Acetylation, Animals, Diet, High-Fat, Energy Intake, Gene Silencing, Growth Hormone-Releasing Hormone genetics, Growth Hormone-Releasing Hormone metabolism, Histones metabolism, Human Growth Hormone biosynthesis, Human Growth Hormone metabolism, Humans, Mice, Transgenic, Motor Activity, Nuclear Receptor Co-Repressor 1 metabolism, Pituitary Gland metabolism, Promoter Regions, Genetic, Protein Processing, Post-Translational, RNA Polymerase II metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Energy Metabolism, Homeostasis, Human Growth Hormone genetics
- Abstract
Levels of pituitary growth hormone (GH), a metabolic homeostatic factor with strong lipolytic activity, are decreased in obese individuals. GH declines prior to the onset of weight gain in response to excess caloric intake and hyperinsulinemia; however, the mechanism by which GH is reduced is not clear. We used transgenic mice expressing the human GH (hGH) gene, GH1, to assess the effect of high caloric intake on expression as well as the local chromosome structure of the intact GH1 locus. Animals exposed to 3 days of high caloric intake exhibited hyperinsulinemia without hyperglycemia and a decrease in both hGH synthesis and secretion, but no difference in endogenous production of murine GH. Efficient GH1 expression requires a long-range intrachromosomal interaction between remote enhancer sequences and the proximal promoter region through "looping" of intervening chromatin. High caloric intake disrupted this interaction and decreased both histone H3/H4 hyperacetylation and RNA polymerase II occupancy at the GH1 promoter. Incorporation of physical activity muted the effects of excess caloric intake on insulin levels, GH1 promoter hyperacetylation, chromosomal architecture, and expression. These results indicate that energy homeostasis alters postnatal hGH synthesis through dynamic changes in the 3-dimensional chromatin structure of the GH1 locus, including structures required for cell type specificity during development.
- Published
- 2014
- Full Text
- View/download PDF
23. Identification of three novel FGF16 mutations in X-linked recessive fusion of the fourth and fifth metacarpals and possible correlation with heart disease.
- Author
-
Laurell T, Nilsson D, Hofmeister W, Lindstrand A, Ahituv N, Vandermeer J, Amilon A, Annerén G, Arner M, Pettersson M, Jäntti N, Rosberg HE, Cattini PA, Nordenskjöld A, Mäkitie O, Grigelioniene G, and Nordgren A
- Abstract
Nonsense mutations in FGF16 have recently been linked to X-linked recessive hand malformations with fusion between the fourth and the fifth metacarpals and hypoplasia of the fifth digit (MF4; MIM#309630). The purpose of this study was to perform careful clinical phenotyping and to define molecular mechanisms behind X-linked recessive MF4 in three unrelated families. We performed whole-exome sequencing, and identified three novel mutations in FGF16. The functional impact of FGF16 loss was further studied using morpholino-based suppression of fgf16 in zebrafish. In addition, clinical investigations revealed reduced penetrance and variable expressivity of the MF4 phenotype. Cardiac disorders, including myocardial infarction and atrial fibrillation followed the X-linked FGF16 mutated trait in one large family. Our findings establish that a mutation in exon 1, 2 or 3 of FGF16 results in X-linked recessive MF4 and expand the phenotypic spectrum of FGF16 mutations to include a possible correlation with heart disease.
- Published
- 2014
- Full Text
- View/download PDF
24. FGF-2 and FGF-16 protect isolated perfused mouse hearts from acute doxorubicin-induced contractile dysfunction.
- Author
-
Sontag DP, Wang J, Kardami E, and Cattini PA
- Subjects
- Animals, Anti-Arrhythmia Agents, Blood Pressure drug effects, Coronary Circulation drug effects, Enzyme Activation drug effects, Fibroblast Growth Factor 2 biosynthesis, Fibroblast Growth Factor 2 genetics, Fibroblast Growth Factors biosynthesis, Fibroblast Growth Factors genetics, In Vitro Techniques, L-Lactate Dehydrogenase metabolism, Mice, Myocardial Reperfusion Injury physiopathology, Protein Kinase C metabolism, Recombinant Proteins pharmacology, Reperfusion Injury prevention & control, Ventricular Function, Left drug effects, Antibiotics, Antineoplastic antagonists & inhibitors, Antibiotics, Antineoplastic toxicity, Cardiotonic Agents, Doxorubicin antagonists & inhibitors, Doxorubicin toxicity, Fibroblast Growth Factor 2 pharmacology, Fibroblast Growth Factors pharmacology, Heart Diseases chemically induced, Heart Diseases prevention & control, Myocardial Contraction drug effects
- Abstract
The anti-cancer drug doxorubicin is associated with an increased risk of cardiac damage and dysfunction, which can be acute as well as chronic. Fibroblast growth factor 2 (FGF-2) provides cardioprotection from ischemia-reperfusion injury but its effects on doxorubicin-induced damage are not known. We investigated the acute effects of doxorubicin administered in the absence and presence of FGF-2 pre-treatment, on isolated mouse perfused heart function over a period of 120 min. Doxorubicin elicited a significant decrease in left ventricular developed pressure (DP) at 30 min that persisted throughout the study. No effect on lactate dehydrogenase levels was detected in the perfusate, suggesting a lack of significant plasma membrane damage. FGF-2 pre-treatment lessened the deleterious effect of doxorubicin on DP significantly, and this beneficial effect of FGF-2 was blunted by protein kinase C inhibition with chelerythrine. Pre-treatment with a non-mitogenic FGF-2 mutant or FGF-16 also protected against a doxorubicin-induced decrease in DP. FGF-16 as well as FGF-2 pre-treatment elicited a small and transient negative inotropic effect. In conclusion, FGF-2 and FGF-16 increase resistance to acute doxorubicin-induced cardiac dysfunction, and protein kinase C activation is implicated in this response.
- Published
- 2013
- Full Text
- View/download PDF
25. CCAAT-enhancer-binding protein β (C/EBPβ) and downstream human placental growth hormone genes are targets for dysregulation in pregnancies complicated by maternal obesity.
- Author
-
Vakili H, Jin Y, Menticoglou S, and Cattini PA
- Subjects
- Adult, Animals, Base Sequence, DNA Primers, Female, Humans, Mice, Molecular Sequence Data, Obesity complications, Placenta Growth Factor, Polymerase Chain Reaction, Pregnancy, Sequence Homology, Nucleic Acid, Young Adult, CCAAT-Enhancer-Binding Protein-beta genetics, Obesity genetics, Pregnancy Complications genetics, Pregnancy Proteins genetics
- Abstract
Human chorionic somatomammotropin (CS) and placental growth hormone variant (GH-V) act as metabolic adaptors in response to maternal insulin resistance, which occurs in "normal" pregnancy. Maternal obesity can exacerbate this "resistance," suggesting that CS, GH-V, or transcription factors that regulate their production might be targets. The human CS genes, hCS-A and hCS-B, flank the GH-V gene. A significant decrease in pre-term placental CS/GH-V RNA levels was observed in transgenic mice containing the CS/GH-V genes in a model of high fat diet (HFD)-induced maternal obesity. Similarly, a decrease in CS/GH-V RNA levels was detected in term placentas from obese (body mass index (BMI) ≥ 35 kg/m(2)) versus lean (BMI 20-25 kg/m(2)) women. A specific decrease in transcription factor CCAAT-enhancer-binding protein β (C/EBPβ) RNA levels was also seen with obesity; C/EBPβ is required for mouse placenta development and is expressed, like CS and GH-V, in syncytiotrophoblasts. Binding of C/EBPβ to the CS gene downstream enhancer regions, which by virtue of their position distally flank the GH-V gene, was reduced in placenta chromatin from mice on a HFD and in obese women; a corresponding decrease in RNA polymerase II associated with CS/GH-V promoters was also observed. Detection of decreased endogenous CS/GH-V RNA levels in human placental tumor cells treated with C/EBPβ siRNA is consistent with a direct effect. These data provide evidence for CS/GH-V dysregulation in acute HFD-induced obesity in mouse pregnancy and chronic obesity in human pregnancy and implicate C/EBPβ, a factor associated with CS regulation and placental development.
- Published
- 2013
- Full Text
- View/download PDF
26. FGF-2 protects cardiomyocytes from doxorubicin damage via protein kinase C-dependent effects on efflux transporters.
- Author
-
Wang J, Nachtigal MW, Kardami E, and Cattini PA
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 1 genetics, ATP Binding Cassette Transporter, Subfamily B, Member 1 physiology, ATP-Binding Cassette Transporters genetics, ATP-Binding Cassette Transporters physiology, Animals, Cyclosporine pharmacology, L-Lactate Dehydrogenase metabolism, Multidrug Resistance-Associated Proteins genetics, Multidrug Resistance-Associated Proteins physiology, Protein Kinase C antagonists & inhibitors, Rats, Rats, Sprague-Dawley, Antibiotics, Antineoplastic toxicity, Doxorubicin toxicity, Fibroblast Growth Factor 2 pharmacology, Membrane Transport Proteins physiology, Myocytes, Cardiac drug effects, Protein Kinase C physiology
- Abstract
Aims: The anti-cancer anthracycline doxorubicin (DOX) increases the risk of cardiac damage, indicating a need to protect the heart and still allow the benefits of drug treatment. Fibroblast growth factor-2 (FGF-2) is cardioprotective against ischaemia-reperfusion injury. Our aim is to investigate: (i) the ability of FGF-2 to protect against DOX-induced cardiomyocyte damage and (ii) the contribution of efflux drug transport to any increase in injury-resistance., Methods and Results: Neonatal rat cardiomyocyte damage was assessed by measuring cell death markers and lactate dehydrogenase (LDH) activity in the culture medium. LDH activity was increased significantly after incubation with 0.5 μM DOX for 24 h in the absence but not presence of 10 nM FGF-2; this beneficial effect of FGF-2 was blocked by tyrosine kinase (FGF) receptor inhibition. An increase in efflux drug transporter RNA levels was also detected after FGF-2 treatment in the presence of DOX. The beneficial effect of FGF-2 against cell damage and increased transporter RNA levels were blunted with protein kinase C (PKC) inhibition. Finally, FGF-2 stimulated efflux transport of calcein and DOX, and treatment with efflux transporter inhibitors significantly attenuated the protective effect of FGF-2 from DOX-induced injury., Conclusion: Administered FGF-2 increases resistance to DOX-induced cardiomyocyte damage, by a mechanism dependent on PKC as well as regulation of efflux transporter production and/or function.
- Published
- 2013
- Full Text
- View/download PDF
27. The hidden but positive role for glucocorticoids in the regulation of growth hormone-producing cells.
- Author
-
Vakili H and Cattini PA
- Subjects
- Animals, Cell Differentiation, Glucocorticoids pharmacology, Human Development, Human Growth Hormone genetics, Humans, Hypothalamo-Hypophyseal System cytology, Hypothalamo-Hypophyseal System metabolism, Pituitary Gland cytology, Pituitary Gland metabolism, Pituitary-Adrenal System cytology, Pituitary-Adrenal System metabolism, Somatotrophs physiology, Glucocorticoids physiology, Human Growth Hormone metabolism, Somatotrophs metabolism
- Abstract
Growth hormone (GH) is a prominent metabolic factor that is targeted by glucocorticoids; however, their role in GH production remains controversial. This is explained in part by discrepancies between in vitro and in vivo, short-term versus long-term exposure and even species-specific effects. The prevailing view, however, is that glucocorticoids are negative modulators of growth and GH production. An examination of recent findings from elegant avian and gene ablation in mice studies as well as clinical case reports, suggests this is not the case. The evidence suggests that the effect of glucocorticoids on growth and GH production can be uncoupled, and reveals they play a crucial and positive role in maturation of functional somatotrophs, the GH-producing cells of the anterior pituitary. Here, we provide an overview and insights into the possible roles of glucocorticoids in the development of somatotrophs before birth as well as regulation of GH production in infancy (neonatal) and adulthood (postnatal). A fully functional glucocorticoid-signaling pathway appears to be required for establishment of somatotrophs before birth, and glucocorticoids continue to be required for maintenance of GH production in the newborn. There is evidence to suggest progenitor somatotrophs may persist after birth, and perhaps account for the ability of glucocorticoid therapy to correct some cases of GH deficiency as a result of compromised glucocorticoid signaling. Finally, there is support for positive regulation of avian, murine and human GH gene activation and/or expression by glucocorticoids, however, there appears to be no common mechanism and the contribution of direct versus indirect effects remains unclear. Thus, our observations reveal a largely hidden face of glucocorticoids, specifically, a positive role in somatotroph development and GH gene activation/expression, which may enable us to better understand the differential effect of glucocorticoids on growth and GH production in human studies., (Copyright © 2012 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
28. Negative regulation of human growth hormone gene expression by insulin is dependent on hypoxia-inducible factor binding in primary non-tumor pituitary cells.
- Author
-
Vakili H, Jin Y, and Cattini PA
- Subjects
- Animals, Base Sequence, Chromatin metabolism, Female, Gene Expression Regulation, Humans, Hyperinsulinism metabolism, Hypoxia-Inducible Factor 1 metabolism, Male, Mice, Mice, Transgenic, Models, Biological, Molecular Sequence Data, Protein Binding, RNA metabolism, Sequence Homology, Nucleic Acid, Human Growth Hormone biosynthesis, Hypoxia metabolism, Insulin metabolism, Pituitary Gland metabolism
- Abstract
Insulin controls growth hormone (GH) production at multiple levels, including via a direct effect on pituitary somatotrophs. There are no data, however, on the regulation of the intact human (h) GH gene (hGH1) by insulin in non-tumor pituitary cells, but the proximal promoter region (nucleotides -496/+1) responds negatively to insulin in transfected pituitary tumor cells. A DNA-protein interaction was also induced by insulin at nucleotides -308/-235. Here, we confirmed the presence of a hypoxia-inducible factor 1 (HIF-1) binding site within these sequences (-264/-259) and investigated whether HIF-1 is associated with insulin regulation of "endogenous" hGH1. In the absence of primary human pituitary cells, transgenic mice expressing the intact hGH locus in a somatotroph-specific manner were generated. A significant and dose-dependent decrease in hGH and mouse GH RNA levels was detected in primary pituitary cell cultures from these mice with insulin treatment. Increasing HIF-1α availability with a hypoxia mimetic significantly decreased hGH RNA levels and was accompanied by recruitment of HIF-1α to the hGH1 promoter in situ as seen with insulin. Both inhibition of HIF-1 DNA binding by echinomycin and RNA interference of HIF-1α synthesis blunted the negative effect of insulin on hGH1 but not mGH. The insulin response is also sensitive to histone deacetylase inhibition/trichostatin A and associated with a decrease in H3/H4 hyperacetylation in the proximal hGH1 promoter region. These data are consistent with HIF-1-dependent down-regulation of hGH1 by insulin via chromatin remodeling specifically in the proximal promoter region.
- Published
- 2012
- Full Text
- View/download PDF
29. Enhancer-blocking activity is associated with hypersensitive site V sequences in the human growth hormone locus control region.
- Author
-
Jin Y, Oomah K, and Cattini PA
- Subjects
- Base Sequence, CCCTC-Binding Factor, Chromatin Immunoprecipitation, CpG Islands, DNA Methylation genetics, DNA, Recombinant genetics, Female, Gene Expression Regulation, HEK293 Cells, HeLa Cells, Humans, Insulator Elements, Molecular Sequence Data, Pituitary Gland metabolism, Placenta metabolism, Pregnancy, Protein Binding, Protein Interaction Mapping, RNA Interference, RNA, Small Interfering pharmacology, Repressor Proteins antagonists & inhibitors, Repressor Proteins genetics, Repressor Proteins metabolism, Transfection, YY1 Transcription Factor antagonists & inhibitors, YY1 Transcription Factor genetics, YY1 Transcription Factor metabolism, Enhancer Elements, Genetic genetics, Human Growth Hormone genetics, Locus Control Region genetics
- Abstract
Activation of the human growth hormone gene (hGH-N) is linked to a locus control region (LCR) containing four (I-III, V) hypersensitive sites (HS). Pit-1 binding to HS I/II is required for efficient pituitary expression. However, inclusion of HS III and V, located about 28 and 32 kb upstream of the hGH-N gene, respectively, is also required for consistent hGH-N expression levels in vivo. HS V is referred to as a boundary for the hGH LCR, but no specific enhancer blocking or barrier function is reported. We examined a 547 bp fragment containing HS V sequences (nucleotides -32,718/-32,172 relative to hGH-N) for enhancer-blocking activity using a well-established transient gene transfer system and assessed these sequences for CCCTC binding factor (CTCF), which is linked to enhancer-blocking activity. The 547 bp HS V fragment decreased enhancer activity with a reverse-orientation preference when inserted between HS III enhancer sequences and a minimal thymidine kinase promoter (TKp). These sequences are associated with CTCF in human pituitary and nonpituitary chromatin. Enhancer-blocking activity with an orientation preference was further localized to a 45 bp sub-fragment, with evidence of CTCF and upstream binding factor 1 (USF1) binding; USF1 is linked more closely with barrier function. The presence of yin and yang 1 (Yy1) that cooperates with CTCF in the regulation of X-chromosome inactivation was also seen. A decrease in CTCF and Yy1 RNA levels was associated with a significant reduction in enhancer-blocking activity. Assessment of CpG-dinucleotides in the TKp indicates that the presence of HS V sequences are associated with an increased incidence of CpG-dinucleotide methylation of the GC box region. These data support association of CTCF and enhancer-blocking activity with HS V that is consistent with a role as a (LCR) boundary element and also implicates Yy1 in this process.
- Published
- 2011
- Full Text
- View/download PDF
30. Protection by endogenous FGF-2 against isoproterenol-induced cardiac dysfunction is attenuated by cyclosporine A.
- Author
-
Jimenez SK, Jassal DS, Kardami E, and Cattini PA
- Subjects
- Adrenergic beta-Agonists pharmacology, Animals, Fibroblast Growth Factor 2 genetics, Isoproterenol pharmacology, Mice, Mice, Transgenic, Ultrasonography, Doppler, Ventricular Dysfunction diagnostic imaging, Cyclosporine pharmacology, Fibroblast Growth Factor 2 metabolism, Ventricular Dysfunction chemically induced, Ventricular Dysfunction metabolism
- Abstract
Fibroblast growth factor-2 (FGF-2) is implicated in cardioprotection. However, previously we found that chronic elevation in cardiac FGF-2 levels in transgenic mice was associated with exaggerated, cyclosporine A-preventable, cellular infiltration after isoproterenol-induced injury, suggestive of an adverse outcome, although this was not examined with functional studies. We have now used highly sensitive tissue Doppler imaging (TDI) to evaluate cardiac functional parameters after isoproterenol administration in transgenic mice overexpressing the 18 kDa FGF-2 in the heart in vivo. Cardiac function was assessed in conscious FGF-2 transgenic and non-transgenic mice at 24 h as well as 2 and 4 weeks after isoproterenol administration, and in the absence or presence of either cyclosporine A or anti-CD3ε treatments. Isoproterenol decreased left ventricular endocardial velocity and strain rate by 47-51% at 24 h in non-transgenic mice, but to a significantly lesser extent (by 24%) in transgenic mice. While additional decreases were seen in non-transgenic mice at 2 weeks, there was no further reduction in ventricular endocardial velocity or strain rate up to 4 weeks post-treatment in FGF-2 transgenic mice. Functional improvement at 2 and 4 weeks post-isoproterenol was reduced significantly by treatment with cyclosporine A but not anti-CD3ε; the latter targets T lymphocyte activation more specifically. TDI values in the presence of chronic FGF-2 overexpression are prognostic of an improved cardiac outcome and protection from isoproterenol induced cardiac dysfunction in vivo. Our data also suggest that cyclosporine A-sensitive infiltrating cell population(s) may contribute to the sustained beneficial effect of FGF-2 in vivo.
- Published
- 2011
- Full Text
- View/download PDF
31. Transgenic mice expressing the human growth hormone gene provide a model system to study human growth hormone synthesis and secretion in non-tumor-derived pituitary cells: differential effects of dexamethasone and thyroid hormone.
- Author
-
Vakili H, Jin Y, Nagy JI, and Cattini PA
- Subjects
- Animals, Cells, Cultured, Female, Ghrelin pharmacology, Growth Hormone-Releasing Hormone pharmacology, Humans, Immunohistochemistry, Male, Mice, Mice, Transgenic, Pituitary Gland drug effects, RNA, Messenger genetics, RNA, Messenger metabolism, Receptors, Ghrelin genetics, Receptors, Ghrelin metabolism, Receptors, Neuropeptide genetics, Receptors, Neuropeptide metabolism, Receptors, Pituitary Hormone-Regulating Hormone genetics, Receptors, Pituitary Hormone-Regulating Hormone metabolism, Triiodothyronine pharmacology, Dexamethasone pharmacology, Human Growth Hormone genetics, Human Growth Hormone metabolism, Models, Biological, Pituitary Gland cytology, Pituitary Gland metabolism, Thyroid Hormones pharmacology
- Abstract
Growth hormone (GH) is regulated by pituitary and hypothalamic factors as well as peripheral endocrine factors including glucocorticoids and thyroid hormone. Studies on human GH are limited largely to the assessment of plasma levels in endocrine disorders. Thus, insight into the regulation of synthesis versus secretion has come mainly from studies done on non-human GH and/or pituitary tumor cells. However, primate and non-primate GH gene loci have differences in their structure and, by extension, regulation. We generated transgenic (171hGH/CS-TG) mice containing the intact hGH1 gene and locus control region, including sequences required for integration-independent and preferential pituitary expression. Here, we show hGH co-localizes with mouse (m) GH in somatotrophs in situ and in primary pituitary cells. Dexamethasone treatment increased hGH and mGH, as well as GH releasing hormone (GHRH) receptor RNA levels, and hGH release was stimulated by GHRH treatment. By contrast, triiodothyronine decreased or had no effect on hGH and mGH production, respectively, and the negative effect on hGH was also seen in the presence of dexamethasone. Thus, 171hGH/CS-TG mouse pituitary cultures represent a model system to investigate hormonal control of hGH synthesis and secretion., (Copyright © 2011 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
32. Identification of functional CCAAT/enhancer-binding protein and Ets protein binding sites in the human chorionic somatomammotropin enhancer sequences.
- Author
-
Lytras A, Detillieux K, and Cattini PA
- Subjects
- Binding Sites, Cell Line, Tumor, Chromatin Immunoprecipitation, Electrophoretic Mobility Shift Assay, Humans, Promoter Regions, Genetic genetics, Protein Binding genetics, CCAAT-Enhancer-Binding Proteins metabolism, Enhancer Elements, Genetic genetics, Placental Lactogen genetics, Proto-Oncogene Proteins c-ets metabolism
- Abstract
The human chorionic somatomammotropin (CS) A and B genes (listed as CSH1 and CSH2 in the HUGO database) are highly expressed in placenta. A 241 bp potent enhancer, nucleotides (nts) 1-241, located at the 3' end of the CS-B gene (CS-Benh) stimulates promoter activity specifically in placental trophoblast cells in vitro. Strong activity is exerted by a 23 bp element within the CS-Benh (nts 117-139), shown to interact with transcription enhancer factor (TEF) members of the transcription enhancer activator (TEA) DNA-binding domain-containing family. An identical TEF element is present in the homologous (97.5%) CS-Aenh; however, a few nucleotide differences suppress its activity. Previously, we identified regulatory sequences distinct from the TEF element within an 80 bp modulatory domain (nts 1-80) in the CS-Benh. Using structural and functional assays we now show that CCAAT/enhancer-binding protein (C/EBP) binding sites exist in the 80 bp modulatory domains of both enhancers, and an Elk-1 binding site exists in the modulatory domain of the CS-Aenh. C/EBPα or C/EBPβ strongly repressed CSp.CAT activity but stimulated CSp.CAT.CS-Benh activity. In contrast, the equivalent CS-A enhancer sequences were unable to relieve promoter repression. Elk-1 overexpression also resulted in differential effects on the CS-Aenh versus CS-Benh. Finally, we provide evidence for the association of C/EBPβ with the CS-A and CS-B genes in human placental chromatin, including differential involvement of C/EBPβ with the CS-Aenh versus the CS-Benh, and therefore consistent with the notion that these are regions of regulatory significance in vivo. We conclude that members of the C/EBP and Ets families can differentially modulate CS-Benh and CS-Aenh activity.
- Published
- 2011
- Full Text
- View/download PDF
33. A single bout of exercise promotes sustained left ventricular function improvement after isoproterenol-induced injury in mice.
- Author
-
Jimenez SK, Jassal DS, Kardami E, and Cattini PA
- Subjects
- Animals, Echocardiography, Doppler, Female, Heart drug effects, Heart physiopathology, Heart Injuries pathology, Heart Injuries physiopathology, Male, Mice, Mice, Inbred Strains, Myocardium pathology, Swimming physiology, Ventricular Dysfunction, Left physiopathology, Heart Injuries chemically induced, Heart Injuries prevention & control, Isoproterenol pharmacology, Physical Conditioning, Animal physiology, Ventricular Dysfunction, Left chemically induced, Ventricular Dysfunction, Left prevention & control
- Abstract
We have investigated whether acute (swimming) exercise is sufficient to have sustained beneficial effects against cardiac functional decline observed after high-dose isoproterenol administration. Mice were subjected to one bout of swimming for 30 min ("swim" group). Twenty-four hours later, they were given isoproterenol (160 mg/kg) to cause injury. Two control groups were included, a shallow "water" group, for which no swimming took place, and a "cage" group; they were both given isoproterenol as in the "swim" group. Cardiac function was assessed by tissue Doppler imaging (TDI) 24 h, 2 weeks, and 4 weeks post-isoproterenol. Left ventricular (LV) systolic function including endocardial velocity and radial strain rate declined significantly in all groups at all time points after isoproterenol, compared with their pre-isoproterenol treatment values. The "swim" group, however, had significantly higher LV systolic function compared with either of the control groups at 24 h, and this improvement persisted 2 and 4 weeks post-treatment. There were no significant differences between the control groups at any time point. In conclusion, a single bout of swimming has sustained beneficial effects against injury, as measured by TDI, after administration of isoproterenol.
- Published
- 2011
- Full Text
- View/download PDF
34. Embryonic survival and severity of cardiac and craniofacial defects are affected by genetic background in fibroblast growth factor-16 null mice.
- Author
-
Lu SY, Jin Y, Li X, Sheppard P, Bock ME, Sheikh F, Duckworth ML, and Cattini PA
- Subjects
- Animals, Craniofacial Abnormalities genetics, DNA Helicases genetics, DNA Helicases metabolism, Embryo, Mammalian metabolism, Female, Fibroblast Growth Factors deficiency, Heart Diseases genetics, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Nuclear Proteins genetics, Nuclear Proteins metabolism, Phenotype, X Chromosome, X-linked Nuclear Protein, Craniofacial Abnormalities embryology, Craniofacial Abnormalities metabolism, Fibroblast Growth Factors metabolism, Heart Diseases embryology, Heart Diseases metabolism
- Abstract
Disruption of the X-chromosome fibroblast growth factor 16 (Fgf-16) gene, a member of the FGF-9 subfamily with FGF-20, was linked with an effect on cardiac development in two independent studies. However, poor trabeculation with lethality by embryonic day (E) 11.5 was associated with only one, involving maintenance in Black Swiss (Bsw) versus C57BL/6 mice. The aim of this study was to examine the potential influence of genetic background through breeding the null mutation onto an alternate (C57BL/6) background. After three generations, 25% of Fgf-16(-/Y) mice survived to adulthood, which could be reversed by reducing the contribution of the C57BL/6 genetic background by back crossing to another strain. There was no significant difference between FGF-9 and FGF-20 RNA levels in Fgf-16 null versus wild-type mice regardless of strain. However, FGF-8 RNA levels were reduced significantly in Bsw but not C57BL/6 mice. FGF-8 is linked to anterior heart development and like the FGF-9 subfamily is reportedly expressed at E10.5. Like FGF-16, neuregulin as well as signaling via ErbB2 and ErbB4 receptors have been linked to trabeculae formation and cardiac development around E10.5. Basal neuregulin, ErbB2, and ErbB4 as well as FGF-8, FGF-9, and FGF-16 RNA levels varied in Bsw versus C57BL/6 mice. These data are consistent with the ability of genetic background to modify the phenotype and affect embryonic survival in Fgf-16 null mice.
- Published
- 2010
- Full Text
- View/download PDF
35. Appearance of the pituitary factor Pit-1 increases chromatin remodeling at hypersensitive site III in the human GH locus.
- Author
-
Yang X, Jin Y, and Cattini PA
- Subjects
- Animals, Base Sequence, Cell Line, Gene Expression Regulation, Human Growth Hormone metabolism, Humans, Molecular Sequence Data, Pituitary Gland metabolism, RNA genetics, RNA metabolism, Transcription Factor Pit-1 genetics, ets-Domain Protein Elk-1 genetics, ets-Domain Protein Elk-1 metabolism, Chromatin Assembly and Disassembly, Human Growth Hormone genetics, Locus Control Region, Transcription Factor Pit-1 metabolism
- Abstract
Expression of pituitary and placental members of the human GH and chorionic somatomammotropin (CS) gene family is directed by an upstream remote locus control region (LCR). Pituitary-specific expression of GH requires direct binding of Pit-1 (listed as POU1F1 in the HUGO database) to sequences marked by a hypersensitive site (HS) region (HS I/II) 14.6 kb upstream of the GH-N gene (listed as GH1 in the HUGO database). We used human embryonic kidney 293 (HEK293) cells overexpressing wild-type and mutant Pit-1 proteins as a model system to gain insight into the mechanism by which Pit-1 gains access to the GH LCR. Addition of Pit-1 to these cells increased DNA accessibility at HS III, located 28 kb upstream of the human GH-N gene, in a POU homeodomain-dependent manner, as reflected by effects on histone hyperacetylation and RNA polymerase II activity. Direct binding of Pit-1 to HS III sequences is not supported. However, the potential for binding of ETS family members to this region has been demonstrated, and Pit-1 association with this ETS element in HS III sequences requires the POU homeodomain. Also, both ETS1 and ELK1 co-precipitate from human pituitary extracts using two independent sources of Pit-1 antibodies. Finally, overexpression of ELK1 or Pit-1 expression in HEK293 cells increased GH-N RNA levels. However, while ELK1 overexpression also stimulated placental CS RNA levels, the effect of Pit-1 appeared to correlate with ETS factor levels and target GH-N preferentially. These data are consistent with recruitment and an early role for Pit-1 in remodeling of the GH LCR at the constitutively open HS III through protein-protein interaction., Competing Interests: Declaration of interest The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.
- Published
- 2010
- Full Text
- View/download PDF
36. FGF-16 is a target for adrenergic stimulation through NF-kappaB activation in postnatal cardiac cells and adult mouse heart.
- Author
-
Sofronescu AG, Detillieux KA, and Cattini PA
- Subjects
- Adult, Aged, Animals, Animals, Newborn, Base Sequence, Binding Sites, Cells, Cultured, Conserved Sequence, Female, Fibroblast Growth Factors genetics, Gene Expression Regulation drug effects, Humans, Male, Mice, Molecular Sequence Data, Myocytes, Cardiac metabolism, Promoter Regions, Genetic drug effects, RNA, Messenger metabolism, Rats, Time Factors, Transfection, Young Adult, Adrenergic beta-Agonists pharmacology, Fibroblast Growth Factors metabolism, Isoproterenol pharmacology, Myocytes, Cardiac drug effects, NF-kappa B metabolism
- Abstract
Aims: The fibroblast growth factor (FGF) family plays an important role in cardiac growth and development. However, only FGF-16 RNA levels are reported to increase during the perinatal period and to be expressed preferentially in the myocardium, suggesting control at the transcriptional level and a role for FGF-16 in the postnatal heart. Beyond the identification of two TATA-like elements (TATA1 and TATA2) in the mouse FGF-16 promoter region and the preferential cardiac activity of TATA2, there is no report of Fgf-16 gene regulation. Assessment of promoter sequences, however, reveals putative nuclear factor-kappaB (NF-kappaB) elements, suggesting that Fgf-16 is regulated via NF-kappaB activation and thereby implicated in a number of cardiac events. Thus, the Fgf-16 gene was investigated as a target for NF-kappaB activation in cardiac cells., Methods and Results: Assessments of Fgf-16 promoter activity were made using truncated and transfected hybrid genes with NF-kappaB inhibitors and/or beta-adrenergic stimulation via isoproterenol (IsP) treatment (a known NF-kappaB activator) in culture, and on endogenous mouse and human Fgf-16 genes in situ. The mouse Fgf-16 promoter region was stimulated in response to IsP treatment, but this response was lost with NF-kappaB inhibitor pretreatment. Deletion analysis revealed IsP responsiveness linked to sequences between TATA2 and TATA1 and, more specifically, a NF-kappaB element upstream and adjacent to TATA1 that associates with NF-kappaB p50/p65 subunits in chromatin. Finally, TATA1 and the proximal NF-kappaB element are conserved in the human genome and responsive to IsP., Conclusion: The mouse and human Fgf-16 gene is a target for NF-kappaB activation in the postnatal heart., Competing Interests: Conflicts of Interest: None declared.
- Published
- 2010
- Full Text
- View/download PDF
37. Mutation of a gene essential for ribosome biogenesis, EMG1, causes Bowen-Conradi syndrome.
- Author
-
Armistead J, Khatkar S, Meyer B, Mark BL, Patel N, Coghlan G, Lamont RE, Liu S, Wiechert J, Cattini PA, Koetter P, Wrogemann K, Greenberg CR, Entian KD, Zelinski T, and Triggs-Raine B
- Subjects
- Amino Acid Sequence, Animals, Case-Control Studies, Cell Line, Cricetinae, DNA-Directed RNA Polymerases metabolism, Female, Fetal Growth Retardation metabolism, Fetal Growth Retardation pathology, Fibroblasts cytology, Fibroblasts metabolism, Genes, Recessive, Humans, Immunoblotting, Male, Models, Molecular, Molecular Sequence Data, Organelle Biogenesis, Pedigree, Protein Conformation, Psychomotor Disorders metabolism, Psychomotor Disorders pathology, RNA, Messenger genetics, RNA, Messenger metabolism, Sequence Homology, Amino Acid, Syndrome, Two-Hybrid System Techniques, Chromosomes, Human, Pair 12 genetics, DNA-Directed RNA Polymerases genetics, Fetal Growth Retardation genetics, Mutation genetics, Psychomotor Disorders genetics, Ribosomes genetics
- Abstract
Bowen-Conradi syndrome (BCS) is an autosomal-recessive disorder characterized by severely impaired prenatal and postnatal growth, profound psychomotor retardation, and death in early childhood. Nearly all reported BCS cases have been among Hutterites, with an estimated birth prevalence of 1/355. We previously localized the BCS gene to a 1.9 Mbp interval on human chromosome 12p13.3. The 59 genes in this interval were ranked as candidates for BCS, and 35 of these, including all of the best candidates, were sequenced. We identified variant NM_006331.6:c.400A-->G, p.D86G in the 18S ribosome assembly protein EMG1 as the probable cause of BCS. This mutation segregated with disease, was not found in 414 non-Hutterite alleles, and altered a highly conserved aspartic acid (D) residue. A structural model of human EMG1 suggested that the D86 residue formed a salt bridge with arginine 84 that would be disrupted by the glycine (G) substitution. EMG1 mRNA was detected in all human adult and fetal tissues tested. In BCS patient fibroblasts, EMG1 mRNA levels did not differ from those of normal cells, but EMG1 protein was dramatically reduced in comparison to that of normal controls. In mammalian cells, overexpression of EMG1 harboring the D86G mutation decreased the level of soluble EMG1 protein, and in yeast two-hybrid analysis, the D86G substitution increased interaction between EMG1 subunits. These findings suggested that the D-to-G mutation caused aggregation of EMG1, thereby reducing the level of the protein and causing BCS.
- Published
- 2009
- Full Text
- View/download PDF
38. Differential placental hormone gene expression during pregnancy in a transgenic mouse containing the human growth hormone/chorionic somatomammotropin locus.
- Author
-
Jin Y, Lu SY, Fresnoza A, Detillieux KA, Duckworth ML, and Cattini PA
- Subjects
- Animals, CD79 Antigens genetics, Female, Gene Expression Regulation, Developmental, Human Growth Hormone genetics, Humans, Mice, Mice, Transgenic, NAV1.4 Voltage-Gated Sodium Channel, Pituitary Gland metabolism, Placental Lactogen genetics, Pregnancy, Sodium Channels genetics, Transcription Factor AP-2 metabolism, Transgenes, Human Growth Hormone metabolism, Locus Control Region, Placenta metabolism, Placental Lactogen metabolism, Pregnancy, Animal metabolism
- Abstract
The human (h) growth hormone/chorionic somatomammotropin (GH/CS) gene locus presents a unique model to gain insight into the molecular mechanisms that have allowed a closely related family of genes to be expressed in two distinct cell lineages/tissues: pituitary somatotrophs and placental syncytiotrophoblasts. However, studies of external factors that regulate gene expression have been somewhat limited by (i) a lack of human cell lines expressing endogenous GH or CS appropriately; and (ii) the fact that the GH/CS locus is unique to primates and thus does not exist in rodents. In the current study, a transgenic (171 h GH/CS-TG) mouse was generated containing the intact hGH/CS gene cluster and hGH locus control region (LCR) in a 171-kilobase DNA fragment. Pituitary and placental-specific expression of hGH/CS RNA was detected at embryonic day (E) 18.5. Immunostaining of hGH was seen in somatotrophs of the anterior pituitary beginning in late gestation. The presence of hCS protein was detected in the placental labyrinth in trophoblasts functionally analogous to the syncytiotrophoblast of the chorionic villi. This pattern of gene expression is consistent with the presence of essential components of the hGH/CS LCR. Transcript levels for hCS-A, hCS-B and placental hGH-variant increased in 171 hGH/CS-TG placenta during gestation (E11.5-E18.5), as previously observed in human placental development. Throughout gestation, hCS-A RNA levels were proportionately higher, accounting for 91% of total CS RNA by E18.5, comparable to term human placenta. Finally, the previous correlation between the transcription factor AP-2alpha and hCS RNA expression observed in developing primary human cytotrophoblast cultures, was extended to pregnancy in the 171 hGH/CS-TG mouse. The 171 hGH/CS-TG mouse thus provides a model to investigate hGH/CS gene expression, including in pregnancy.
- Published
- 2009
- Full Text
- View/download PDF
39. FGF-16 is required for embryonic heart development.
- Author
-
Lu SY, Sheikh F, Sheppard PC, Fresnoza A, Duckworth ML, Detillieux KA, and Cattini PA
- Subjects
- Animals, Craniofacial Abnormalities genetics, Embryo Loss genetics, Female, Gene Targeting, Heart Defects, Congenital pathology, Mice, Mice, Knockout, Pregnancy, Pregnancy Trimester, Second, X Chromosome genetics, Fibroblast Growth Factors genetics, Heart embryology, Heart Defects, Congenital genetics
- Abstract
Fibroblast growth factor 16 (FGF-16) expression has previously been detected in mouse heart at mid-gestation in the endocardium and epicardium, suggesting a role in embryonic heart development. More specifically, exogenously applied FGF-16 has been shown to stimulate growth of embryonic myocardial cells in tissue explants. We have generated mice lacking FGF-16 by targeting the Fgf16 locus on the X chromosome. Elimination of Fgf16 expression resulted in embryonic death as early as day 11.5 (E11.5). External abnormalities, including hemorrhage in the heart and ventral body region as well as facial defects, began to appear in null embryos from E11.5. Morphological analysis of FGF-16 null hearts revealed cardiac defects including chamber dilation, thinning of the atrial and ventricular walls, and poor trabeculation, which were visible at E10.5 and more pronounced at E11.5. These findings indicate FGF-16 is required for embryonic heart development in mid-gestation through its positive effect on myocardial growth.
- Published
- 2008
- Full Text
- View/download PDF
40. FGF-16 is released from neonatal cardiac myocytes and alters growth-related signaling: a possible role in postnatal development.
- Author
-
Lu SY, Sontag DP, Detillieux KA, and Cattini PA
- Subjects
- Aging physiology, Animals, Animals, Newborn, Cell Culture Techniques, Cell Cycle physiology, Cell Division, Enzyme Activation, Fibroblast Growth Factors genetics, Glyceraldehyde-3-Phosphate Dehydrogenases genetics, Kinetics, Protein Kinase C metabolism, Rats, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction, Fibroblast Growth Factors metabolism, Myocytes, Cardiac cytology, Myocytes, Cardiac physiology
- Abstract
FGF-16 has been reported to be preferentially expressed in the adult rat heart. We have investigated the expression of FGF-16 in the perinatal and postnatal heart and its functional significance in neonatal rat cardiac myocytes. FGF-16 mRNA accumulation was observed by quantitative RT-PCR between neonatal days 1 and 7, with this increased expression persisting into adulthood. FGF-2 has been shown to increase neonatal rat cardiac myocyte proliferative potential via PKC activation. Gene array analysis revealed that FGF-16 inhibited the upregulation by FGF-2 of cell cycle promoting genes including cyclin F and Ki67. Furthermore, the CDK4/6 inhibitor gene Arf/INK4A was upregulated with the combination of FGF-16 and FGF-2 but not with either factor on its own. The effect on Ki67 was validated by protein immunodetection, which also showed that FGF-16 significantly decreased FGF-2-induced Ki67 labeling of cardiac myocytes, although it alone had no effect on Ki67 labeling. Inhibition of p38 MAPK potentiated cardiac myocyte proliferation induced by FGF-2 but did not alter the inhibitory action of FGF-16. Receptor binding assay showed that FGF-16 can compete with FGF-2 for binding sites including FGF receptor 1. FGF-16 had no effect on activated p38, ERK1/2, or JNK/SAPK after FGF-2 treatment. However, FGF-16 inhibited PKC-alpha and PKC-epsilon activation induced by FGF-2 and, importantly, IGF-1. Collectively, these data suggest that expression and release of FGF-16 in the neonatal myocardium interfere with cardiac myocyte proliferative potential by altering the local signaling environment via modulation of PKC activation and cell cycle-related gene expression.
- Published
- 2008
- Full Text
- View/download PDF
41. A myocyte enhancer factor 2 (MEF2) site located in a hypersensitive region of the FGF16 gene locus is required for preferential promoter activity in neonatal cardiac myocytes.
- Author
-
Sofronescu AG, Jin Y, and Cattini PA
- Subjects
- Animals, Animals, Newborn, Cells, Cultured, Chromatin genetics, Deoxyribonuclease I metabolism, Kidney cytology, MEF2 Transcription Factors, Mice, Mice, Transgenic, Mutation, Myocardium cytology, Rats, Recombinant Proteins genetics, Recombinant Proteins metabolism, Transfection, Fibroblast Growth Factors genetics, Gene Expression Regulation, Developmental, Myocytes, Cardiac metabolism, Myogenic Regulatory Factors metabolism, Promoter Regions, Genetic
- Abstract
Fibroblast growth factor 16 (FGF16) is preferentially expressed in the heart after birth, suggesting its regulation is associated with tissue-specific chromatin remodeling and DNA-protein interactions. Here we have mapped the transcription initiation site of murine FGF16 to approximately 1.1 kilobases (kb) upstream of the translation start codon (ATG). Hybrid reporter genes directed by about 4.7 kb of upstream FGF16 DNA were expressed specifically in transfected neonatal rat cardiac myocytes, as well as in the heart of transgenic mice. A DNaseI hypersensitive site was mapped to a region about 1.2 kb upstream of the transcription initiation site in heart but not kidney tissue, and a nuclease protection assay gave evidence of a cardiac-specific protein-DNA interaction in this region. Deletion analysis indicated that a hybrid gene with 1205 bp but not 1054 bp of upstream DNA directed FGF16 promoter activity in transfected neonatal rat cardiac myocytes. We identified a putative myocyte enhancer factor 2 (MEF2)-binding site at nucleotides -1159/-1148, confirmed by electrophoretic mobility shift assay and MEF2 antibody binding. Mutation of the MEF2 site resulted in a blunting of FGF16 promoter activity in transfected neonatal rat cardiac myocytes. These data suggest that chromatin remodeling and MEF2 binding in the FGF16 promoter contribute to expression in the postnatal heart.
- Published
- 2008
- Full Text
- View/download PDF
42. Chromatin compaction and cell death by high molecular weight FGF-2 depend on its nuclear localization, intracrine ERK activation, and engagement of mitochondria.
- Author
-
Ma X, Dang X, Claus P, Hirst C, Fandrich RR, Jin Y, Grothe C, Kirshenbaum LA, Cattini PA, and Kardami E
- Subjects
- Adenoviridae genetics, Antibodies, Monoclonal metabolism, Cell Death drug effects, Cell Line, Cell Nucleus enzymology, Chromatin drug effects, Cytochromes c metabolism, Cytoplasm metabolism, Enzyme Activation drug effects, Extracellular Signal-Regulated MAP Kinases antagonists & inhibitors, Fibroblast Growth Factor 2 chemistry, Fibroblast Growth Factor 2 genetics, Flavonoids pharmacology, Fluorescein, Fluorescent Dyes, Humans, In Situ Nick-End Labeling, Kidney cytology, Molecular Weight, Plasmids, Protein Isoforms chemistry, Protein Isoforms genetics, Cell Nucleus metabolism, Chromatin physiology, Extracellular Signal-Regulated MAP Kinases metabolism, Fibroblast Growth Factor 2 pharmacology, Mitochondria metabolism
- Abstract
Fibroblast growth factor 2 (FGF-2) is produced as CUG-initiated, 22-34 kDa or AUG-initiated 18 kDa isoforms (hi- and lo-FGF-2, respectively), with potentially distinct functions. We report that expression of hi-FGF-2 in HEK293 cells elicited chromatin compaction preceding cell death with apoptotic features. Nuclear localization of the intact protein was required as expression of a non-nuclear hi-FGF-2 mutant failed to elicit chromatin compaction. Equally ineffective, despite nuclear localization, was the over-expression of the 18 kDa core sequence (lo-FGF-2). Chromatin compaction by hi-FGF-2 was accompanied by increased cytosolic cytochrome C, and was attenuated either by over-expression of Bcl-2 or by a peptide inhibitor of the pro-apoptotic protein Bax. In addition hi-FGF-2 elicited sustained activation of total and nuclear extracellular signal regulated kinase (ERK1/2) by an intracrine route, as it was not prevented by neutralizing anti-FGF-2 antibodies. Inhibition of the ERK1/2 activating pathway by dominant negative upstream activating kinase, or by PD 98059, prevented chromatin compaction by hi-FGF-2. ERK1/2 activation was not affected by the Bax-inhibiting peptide suggesting that it occurred upstream of mitochondrial involvement. We conclude that the hi-FGF-2-induced chromatin compaction and cell death requires its nuclear localization, intracrine ERK1/2 activation and mitochondrial engagement., (2007 Wiley-Liss, Inc.)
- Published
- 2007
- Full Text
- View/download PDF
43. Fibroblast growth factor-2 and cardioprotection.
- Author
-
Kardami E, Detillieux K, Ma X, Jiang Z, Santiago JJ, Jimenez SK, and Cattini PA
- Subjects
- Humans, Myocytes, Cardiac, Signal Transduction, Fibroblast Growth Factor 2 physiology, Intercellular Signaling Peptides and Proteins, Ischemic Preconditioning, Myocardial, Myocardial Ischemia prevention & control, Myocardium, Receptor, Fibroblast Growth Factor, Type 2 physiology
- Abstract
Boosting myocardial resistance to acute as well as chronic ischemic damage would ameliorate the detrimental effects of numerous cardiac pathologies and reduce the probability of transition to heart failure. Experimental cardiology has pointed to ischemic and pharmacological pre- as well as post-conditioning as potent acute cardioprotective manipulations. Additional exciting experimental strategies include the induction of true regenerative and/or angiogenic responses to the damaged heart, resulting in sustained structural and functional beneficial effects. Fibroblast growth factor-2 (FGF-2), an endogenous multifunctional protein with strong affinity for the extracellular matrix and basal lamina and well-documented paracrine, autocrine and intracellular modes of action, has been shown over the years to exert acute and direct pro-survival effects, irrespectively of whether it is administered before, during or after an ischemic insult to the heart. FGF-2 is also a potent angiogenic protein and a crucial agent for the proliferation, expansion, and survival of several cell types including those with stem cell properties. Human clinical trials have pointed to a good safety record for this protein. In this review, we will present a case for the low molecular weight isoform of fibroblast growth factor-2 (lo-FGF-2) as a very promising therapeutic agent to achieve powerful acute as well as sustained benefits for the heart, due to its cytoprotective and regenerative properties.
- Published
- 2007
- Full Text
- View/download PDF
44. Transforming growth factor-beta as a differentiating factor for cultured smooth muscle cells.
- Author
-
Gawaziuk JP, X, Sheikh F, Cheng ZQ, Cattini PA, and Stephens NL
- Subjects
- Animals, Cell Differentiation, Cell Proliferation, Cells, Cultured, Colorimetry methods, Culture Media, Conditioned pharmacology, Decorin, Dogs, Extracellular Matrix Proteins biosynthesis, Microscopy, Confocal, Models, Biological, Myocytes, Smooth Muscle metabolism, Myosin Heavy Chains metabolism, Phenotype, Phosphorylation, Proteoglycans biosynthesis, Receptor, Transforming Growth Factor-beta Type II, Transforming Growth Factor beta metabolism, Gene Expression Regulation, Myocytes, Smooth Muscle cytology, Protein Serine-Threonine Kinases metabolism, Receptors, Transforming Growth Factor beta metabolism, Smad2 Protein physiology, Transforming Growth Factor beta physiology
- Abstract
The aim of the present study was to determine whether the development of supercontractile smooth muscle cells, contributing to the nonspecific hyperreactivity of airways in asthmatic patients, is due to transforming growth factor (TGF)-beta. In cultured smooth muscle cells starved by removal of 10% foetal bovine serum for 7 days, growth arrest was seen; 30% became elongated and demonstrated super contractility. Study of conditioned medium suggested that the differentiating factor was TGF-beta. Sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE) was carried out on conditioned medium from the arrested cells. Two protein bands were identified as matrix metalloproteinase (MMP)-2 and TGF-beta1. To determine second messenger signalling by SMAD2, Western blotting and confocal microscopy were employed. Conditioned medium from arrested cultures showed the presence of MMP-2 and TGF-beta1, as revealed by SDS-PAGE; 68- and 25-kDa bands were seen. Differentiation was confirmed by upregulation of marker proteins, smooth muscle type myosin heavy chain and myosin light chain kinase. Confirmation was obtained by downregulating these proteins with decorin treatment, which reduces the levels of active TGF-beta and an adenoviral dominant-negative vector coding for a mutated type II TGF-beta-receptor. Activation of second messenger signalling was demonstrated immunocytochemically by the presence of phosphorylated SMAD2 and SMAD4. Transforming growth factor-beta is likely to be the differentiating factor responsible for the development of these supercontractile smooth muscle cells. The development of such cells in vivo after cessation of an asthmatic attack could contribute to the nonspecific hyperreactivity of airways seen in patients.
- Published
- 2007
- Full Text
- View/download PDF
45. High- but not low-molecular weight FGF-2 causes cardiac hypertrophy in vivo; possible involvement of cardiotrophin-1.
- Author
-
Jiang ZS, Jeyaraman M, Wen GB, Fandrich RR, Dixon IM, Cattini PA, and Kardami E
- Subjects
- Animals, Cardiomegaly pathology, Cardiomegaly physiopathology, Cardiotonic Agents chemistry, Cardiotonic Agents pharmacology, Cells, Cultured, Fibroblast Growth Factor 2 chemistry, Male, Molecular Weight, Myocardial Infarction drug therapy, Myocardial Infarction metabolism, Myocardial Infarction pathology, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Rats, Rats, Sprague-Dawley, Recombinant Proteins chemistry, Recombinant Proteins pharmacology, Cardiomegaly etiology, Cardiomegaly metabolism, Cytokines metabolism, Fibroblast Growth Factor 2 pharmacology
- Abstract
The heart expresses high and low molecular weight (hmw, lmw) fibroblast growth factor 2 (FGF-2) isoforms. While the injury-repair-related activities of lmw-FGF-2 have been studied extensively, those of hmw-FGF-2 have not. Thus, we investigated the effects of hmw-FGF-2 on acute as well as chronic responses to myocardial infarction (MI) induced by irreversible coronary occlusion in the rat. Hmw- or lmw-FGF-2 was injected into the ischemic zone during acute evolving MI. Both isoforms were equally effective in reducing infarct size (at 24 h post-MI) and improving heart function up to 6 weeks post-MI, compared to a vehicle-treated infarcted group. Lmw-FGF-2 alone upregulated vascularization in the infarct. Hmw-FGF-2 elicited significant hypertrophy, compared to the vehicle-treated group, at 4-8 weeks post-MI, assessed by ultrasound, heart morphometry and cardiomyocyte cross-sectional area. In addition, hmw- (but not lmw-) FGF-2-treated hearts displayed increased accumulation of the cytokine cardiotrophin-1 and its signal transducer gp130. In culture, hmw- (but not lmw-) FGF-2 increased cardiomyocyte protein synthesis and cell size as well as upregulated cardiotrophin-1 released by cardiac fibroblasts, pointing to similar activities in vivo. Thus, hmw- and lmw-FGF-2 exert isoform-specific effects in the heart and only hmw-FGF-2 triggers cardiomyocyte hypertrophic growth. Direct effects of hmw-FGF-2 on cardiomyocytes, becoming reinforced and sustained by upregulation of cardiotrophin-1 and acting in concert with other factors, are likely to contribute to post-MI hypertrophy.
- Published
- 2007
- Full Text
- View/download PDF
46. Hepatocyte nuclear factor-3alpha binding at P sequences of the human growth hormone locus is associated with pituitary repressor function.
- Author
-
Norquay LD, Yang X, Jin Y, Detillieux KA, and Cattini PA
- Subjects
- Animals, Base Sequence, Binding Sites, CCAAT-Enhancer-Binding Proteins genetics, CCAAT-Enhancer-Binding Proteins metabolism, Cells, Cultured, Chromatin genetics, Chromatin metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Hepatocyte Nuclear Factor 3-alpha genetics, Human Growth Hormone genetics, Humans, Molecular Sequence Data, Multiprotein Complexes, NFI Transcription Factors genetics, NFI Transcription Factors metabolism, Pituitary Gland cytology, Placental Lactogen genetics, Placental Lactogen metabolism, Promoter Regions, Genetic, Rats, Regulatory Factor X Transcription Factors, Regulatory Sequences, Nucleic Acid, Transcription Factor Pit-1 genetics, Transcription Factor Pit-1 metabolism, Transcription Factors genetics, Transcription Factors metabolism, Hepatocyte Nuclear Factor 3-alpha metabolism, Human Growth Hormone metabolism, Pituitary Gland metabolism
- Abstract
The human GH family consists of five genes, including the placental chorionic somatomammotropins (CS), within a single locus on chromosome 17. Based on nuclease sensitivity, the entire GH/CS locus is accessible in pituitary chromatin, yet only GH-N is expressed. Previously, we reported a P sequence element (263P) capable of repressing placental CS-A promoter activity in transfected pituitary (GC) cells, and our data indicated a possible role for nuclear factor-1 (NF-1) and regulatory factor X1 in this repression. In this study we show the formation of two independent pituitary complexes in vitro: a repressor complex containing NF-1 and a nonfunctional complex containing regulatory factor X1. In vitro repressor function is stabilized by the presence of P sequence element C (PSE-C), downstream of the previously characterized PSE-A and PSE-B. Repressor function is also dependent on an intact Pit-1 binding site in the CS-A promoter. EMSAs with PSE-C reveal binding of the hepatocyte nuclear factor-3/forkhead (HNF-3/fkh) family of transcription factors in rat pituitary GC cells. This observation is extended to human pituitary tissue, where HNF-3alpha's association with P sequences is confirmed by chromatin immunoprecipitation. Furthermore, protein-protein interactions between HNF-3alpha and NF-1 family members are demonstrated. These results identify HNF-3alpha as an additional member of the pituitary P sequence regulatory complex, implicating it in tissue-specific expression of the human GH/CS family.
- Published
- 2006
- Full Text
- View/download PDF
47. Regulation of the human growth hormone gene family: possible role for Pit-1 in early stages of pituitary-specific expression and repression.
- Author
-
Cattini PA, Yang X, Jin Y, and Detillieux KA
- Subjects
- Chromatin Assembly and Disassembly genetics, Chromosomes, Human, Pair 17 genetics, Chromosomes, Human, Pair 17 metabolism, Human Growth Hormone genetics, Humans, Organ Specificity genetics, Pituitary Gland cytology, Chromatin Assembly and Disassembly physiology, Gene Expression Regulation physiology, Human Growth Hormone metabolism, Pituitary Gland metabolism, Transcription Factor Pit-1 metabolism
- Abstract
The somatic cells of a multicellular organism contain an identical complement of genes that need to be expressed specifically and appropriately to allow the normal development and functions associated with an organism. In the eukaryotic cell nucleus, genes are packaged with nucleoprotein histones into chromatin. The human growth hormone (GH)/chorionic somatomammotropin (CS) gene family offers an excellent model to study the relationship between chromatin structure and transcription factor binding in terms of tissue-specific gene expression. The GH/CS gene family consists of five genes (GH-N, GH-V, CS-A, CS-B and CS-L), contained in a single locus on chromosome 17. Although they share approximately 94% sequence similarity, GH-N expression is restricted to pituitary somatotropes while the four placental GH/CS genes are expressed in the villus syncytiotrophoblast. Appropriate expression in vivo is dependent on remote sequences found 14-32 kb upstream of GH-N in the loci of adjacent genes, and these sequences are characterized by five (I-V) nuclease-hypersensitive sites (HS). Pituitary-specific factor Pit-1 binds at HS I/II and plays an essential role in chromatin remodeling and GH-N expression; however, the processes that lead to HS I/II accessibility are unknown. We discuss the possibility that Pit-1-driven remodeling at HS III may precede that at HS I/II in the pituitary. Also, in pituitary chromatin, all five GH/CS genes share similar nuclease sensitivity, suggesting that the conformation of the placental genes is not inhibitory to transcription. Given that the promoters of both GH-N and the placental GH/CS genes contain Pit-1-binding sites, possible mechanisms to restrict placenta GH/CS promoter activity in the pituitary are discussed, including active repression via P sequences located upstream of each of the placental GH/CS genes. Positively or negatively influencing those components known to be important for pituitary transcription may link epigenetic events to key transcription factors in the overall picture of tissue-specific control of gene expression.
- Published
- 2006
- Full Text
- View/download PDF
48. Inhibition of TGFbeta signaling potentiates the FGF-2-induced stimulation of cardiomyocyte DNA synthesis.
- Author
-
Sheikh F, Hirst CJ, Jin Y, Bock ME, Fandrich RR, Nickel BE, Doble BW, Kardami E, and Cattini PA
- Subjects
- Animals, Cells, Cultured, Gene Expression, Protein Serine-Threonine Kinases, Rats, Rats, Sprague-Dawley, Receptor, Transforming Growth Factor-beta Type II, Receptors, Transforming Growth Factor beta genetics, Transfection methods, DNA biosynthesis, Fibroblast Growth Factor 2 metabolism, Myocytes, Cardiac metabolism, Signal Transduction, Transforming Growth Factor beta metabolism
- Abstract
Objective: Added transforming growth factor beta (TGFbeta) inhibits the proliferation of immature cardiomyocytes. We have now examined the hypothesis that suppression of endogenous TGFbeta signaling will boost the proliferative response (DNA synthesis) of cardiac myocytes to serum and/or to the mitogenic factor fibroblast growth factor-2 (FGF-2)., Methods and Results: Overexpression of a kinase-deficient TGFbeta type II receptor (TGFbetaRIIDeltaKD) resulted in a 2.8-fold increase in cardiomyocyte DNA synthesis in serum-rich cultures, an effect requiring active FGFR-1 since it was not observed in the presence of excess kinase-deficient FGFR-1. This finding suggested that endogenous TGFbeta-TGFbetaRII suppressed endogenous FGFR-1-mediated signals that stimulate or are permissive for DNA synthesis. TGFbeta had no effect, however, on the FGF-2-induced acute stimulation of extracellular signal regulated kinase1/2. FGF-2, added in the absence or presence of TGFbeta inhibition, elicited a 3- or a 13-fold stimulation of DNA synthesis, respectively, pointing to a synergistic effect., Conclusion: Inhibition of TGFbetaRII-transduced signaling upregulates the proliferative response of cardiomyocytes to serum, and greatly potentiates the stimulatory effect of FGF-2. A combinatorial strategy including activation of FGF-2 and inhibition of TGFbeta-triggered signal transduction may be required for maximal stimulation of immature cardiomyocyte DNA synthesis.
- Published
- 2004
- Full Text
- View/download PDF
49. Beyond angiogenesis: the cardioprotective potential of fibroblast growth factor-2.
- Author
-
Detillieux KA, Cattini PA, and Kardami E
- Subjects
- Animals, Humans, Ischemic Preconditioning, Myocardial, Fibroblast Growth Factor 2 therapeutic use, Heart Diseases prevention & control, Neovascularization, Pathologic prevention & control
- Abstract
In the field of cardiovascular research, a number of independent approaches have been explored to protect the heart from acute and chronic ischemic damage. Fibroblast growth factor-2 (FGF-2) recently has received considerable attention with respect to its angiogenic potential. While therapeutic angiogenesis may serve to salvage chronically ischemic myocardium, more acute treatments are in demand to increase cardiac resistance to injury (preconditioning) and to guard against secondary injury after an acute ischemic insult. Here, we look beyond the angiogenic potential of FGF-2 and examine its acute cardioprotective activity as demonstrated under experimental conditions, both as an agent of a preconditioning-like response and for secondary injury prevention at the time of reperfusion. Factors to consider in moving to the clinical setting will be discussed, including issues of dosage, treatment duration, and routes of administration. Finally, issues of safety and clinical trial design will be considered. The prospect of such a multipotent growth factor having clinical usefulness opens the door to effective treatment of both acute and chronic ischemic heart disease, something well worth the attention of the cardiovascular community.
- Published
- 2004
- Full Text
- View/download PDF
50. Cell transplantation for treatment of acute myocardial infarction: unique capacity for repair by skeletal muscle satellite cells.
- Author
-
Horackova M, Arora R, Chen R, Armour JA, Cattini PA, Livingston R, and Byczko Z
- Subjects
- Age Factors, Animals, Cell Differentiation, Electrocardiography, Fibroblasts physiology, Fibroblasts transplantation, Guinea Pigs, Male, Myoblasts, Cardiac physiology, Myoblasts, Skeletal cytology, Myoblasts, Skeletal physiology, Satellite Cells, Skeletal Muscle cytology, Transfection, beta-Galactosidase genetics, Myoblasts, Cardiac transplantation, Myoblasts, Skeletal transplantation, Myocardial Infarction therapy, Satellite Cells, Skeletal Muscle physiology
- Abstract
An adult heart injured by an ischemic episode has a limited capacity to regenerate. We administered three types of adult guinea pig cells [cardiomyocytes (CMs), cardiac fibroblasts (CFs), and skeletal myoblasts (Mbs)] to compare their suitability for repair of acute myocardial infarction. We used confocal fluorescent microscopy and a variety of specific immunomarkers and echocardiography to provide anatomic evidence for the viability of such cells and their possible functional beneficial effects. All cells were transfected with adenovirus-containing beta-galactosidase gene so that migration from the injection sites could be traced. Both freshly isolated CMs as well as CFs were found concentrated in the infarcted zone; these cells survived for at least 2 wk posttransplantation. Transplanted CMs were regularly striated and grew long projections that could form gap junctions with native CMs, which was evidenced by connexin43 labeling. In addition, CM transplantation resulted in increased angiogenesis in the infarcted areas. In contrast, transplanted CFs did not appear to make any gap junctional contacts with native CMs nor did they enhance local angiogenesis. Mbs cultured for 7 days and transfected Mbs were identified 7 days posttransplantation in the infarcted area. During that time and thereafter, Mbs proliferated and differentiated into myotubes that formed new, regularly striated myofibers that occupied most (50-70%) of the infarcted area by 2-3 wk. These newly formed myofibers maintained their Mb skeletal muscle origin as evidenced by their capacity to express myogenin and fast skeletal myosin. This skeletal phenotype appeared to downregulate with time, and Mbs partially transdifferentiated into a cardiac phenotype as indicated by labeling for cardiac-specific troponin T and cardiac myosin heavy chain. By the third week posttransplantation, new myofibers formed apparent contacts with the native CMs via putative gap junctions that expressed connexin43. Myocardial performance of animals that were successfully transplanted with Mbs was improved.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.