Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 11(1): 19773, 2021 Oct 05.
Article in English | MEDLINE | ID: mdl-34611261

ABSTRACT

This work presents the simulated and measured performance of single- and double-layer frequency selective surface filters for operation at sub-THz frequencies (250 GHz center frequency). They were composed of concentric square loops with a split as a unit cell resonator on top of a low dielectric permittivity, low thickness material (RT5880). Both a single layer filter and a cascaded two layer filter with varied distances were investigated. The simulated bandwidth for the cascaded filter was 27 GHz and 16 GHz and 9 GHz bandwidth measured with a THz-TDS and microwave system.

2.
Oncogene ; 36(41): 5681-5694, 2017 10 12.
Article in English | MEDLINE | ID: mdl-28604746

ABSTRACT

In many cancer types, integrin-mediated signaling regulates proliferation, survival and invasion of tumorigenic cells. However, it is still unclear how integrins crosstalk with oncogenes to regulate tumorigenesis and metastasis. Here we show that oncogenic K-RasV12 upregulates α6-integrin expression in Madin-Darby canine kidney (MDCK) cells via activation of the mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK)/Fos-related antigen 1-signaling cascade. Activated α6-integrins promoted metastatic capacity and anoikis resistance, and led to perturbed growth of MDCK cysts. Transcriptomic analysis of K-RasV12-transformed MDCK cells also revealed robust downregulation of αV-class integrins. Re-expression of αV-integrin in K-RasV12-transformed MDCK cells synergistically upregulated the expression of Zinc finger E-box-binding homeobox 1 and Twist-related protein 1 and triggered epithelial-mesenchymal transition leading to induced cell motility and invasion. These results delineate the signaling cascades connecting oncogenic K-RasV12 with α6- and αV-integrin functions to modulate cancer cell survival and tumorigenesis, and reveal new possible strategies to target highly oncogenic K-RasV12 mutants.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Integrin alphaV/genetics , Neoplasms/genetics , Proto-Oncogene Proteins c-fos/genetics , Animals , Anoikis/genetics , Cell Proliferation/genetics , Dogs , Humans , Integrin alpha6/genetics , Madin Darby Canine Kidney Cells , Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics
3.
Int J Androl ; 29(2): 313-22, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16533353

ABSTRACT

Exposure of adult male animals to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) decreases serum androgen concentrations. Reduction in androgen levels after maternal exposure has also been reported, but these results have not been reproduced. We have earlier shown that TCDD stimulates rather than inhibits testosterone synthesis in the prenatal rat testis. The aim of the present study was to elucidate in utero-induced effects of TCDD on testicular steroidogenesis in the 14-day-old infant rats. At that time the foetal Leydig cell population is still the prevailing source of androgens. Pregnant Sprague-Dawley dams were given a single oral dose of TCDD (0, 0.04, 0.2, or 1.0 microg/kg) on day 13 of pregnancy. On postnatal day 14, the body weight of male offspring was reduced after exposure to 1.0 microg/kg TCDD (from 33.9 +/- 1.66 g to 31.6 +/- 2.67 g). Relative testis weight, plasma testosterone, luteinizing hormone and follicle-stimulating hormone levels remained unaltered in all exposure groups. Moreover, in ex vivo incubations, testosterone and cAMP production was not affected. StAR protein level in the freshly isolated testes was increased in the 0.2 microg/kg group, and seminiferous cord diameter in the 0.04 microg/kg group. The present study confirms our earlier findings in in utero TCDD-exposed foetal testis indicating that maternal TCDD exposure does not negatively influence the developmental testosterone production of foetal type Leydig cells in rats.


Subject(s)
Environmental Pollutants/toxicity , Fetus/drug effects , Maternal Exposure , Polychlorinated Dibenzodioxins/toxicity , Testis/drug effects , Testosterone/blood , Animals , Animals, Newborn , Cyclic AMP/biosynthesis , Dose-Response Relationship, Drug , Female , Fetus/metabolism , Follicle Stimulating Hormone/blood , Leydig Cells/drug effects , Leydig Cells/metabolism , Luteinizing Hormone/blood , Male , Organ Size/drug effects , Pregnancy , Rats , Rats, Sprague-Dawley , Testis/anatomy & histology
4.
Toxicol Sci ; 88(2): 534-44, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16141434

ABSTRACT

TCDD (2,3,7,8-tetrachlorodibenzo-p-dioxin) has a potency to induce decreased fertility and structural reproductive anomalies in male and female mammals. While the activity profile of sex steroid hormone production distinctly differs in developing males and females, we wanted to analyze sex-specific effects of TCDD introduced in utero and via lactation on gonadal steroidogenesis and gonadotropin levels in male and female rat infant pups. One oral dose of TCDD (0, 0.04, 0.2, or 1.0 microg/kg) was given to dams on gestational day (GD) 13. Plasma testosterone, estradiol, progesterone, follicle stimulating hormone (FSH), luteinizing hormone (LH), and gonadal mRNA levels for steroid acute regulatory protein (StAR), cytochrome P-450 cholesterol side-chain cleavage (P450scc), 3beta-hydroxy-steroid-dehydrogenase/Delta(5)-Delta(4) isomerase type I (3beta-HSD1), P-450 17alpha-hydroxylase/17,20-lyase (P450-17alpha), and cytochrome P-450 aromatase (P450arom) were determined on postnatal days (PND) 10-16. TCDD 1.0 mug/kg reduced body weights but did not affect relative testis weight or alter testicular and ovarian histology. Plasma estradiol levels in dams and female pups were reduced on PND 14 and 16. Progesterone levels remained unaltered, and FSH levels were increased in female pups. In males, testosterone levels were elevated on PND 10. Gonadal mRNA levels for StAR and steroidogenic enzymes increased during the postnatal growth. TCDD caused no changes in relatively low testicular mRNA levels. However, significant reductions in StAR and P450arom mRNA levels were seen in PND 14 ovaries, and P450arom activity was decreased in isolated ovarian follicles. We conclude that developing testis and male gonadotropin secretion are resistant to TCDD-induced toxicity. In female pups, reduced estradiol, ovarian P450arom expression and enzyme activity levels, and elevated FSH levels may have a role in the development of ovarian dysfunction reported in TCDD-exposed females.


Subject(s)
Abnormalities, Drug-Induced , Lactation/drug effects , Maternal Exposure , Ovary/drug effects , Polychlorinated Dibenzodioxins/toxicity , Teratogens/toxicity , Testis/drug effects , Animals , Animals, Newborn , Dose-Response Relationship, Drug , Enzymes/genetics , Enzymes/metabolism , Female , Gene Expression Regulation, Developmental/drug effects , Hormones/blood , Male , Ovary/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Pregnancy/blood , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Testis/metabolism
5.
Toxicol Appl Pharmacol ; 207(1): 59-68, 2005 Aug 22.
Article in English | MEDLINE | ID: mdl-16054900

ABSTRACT

Phenolic compounds, such as 4-tert-octylphenol (OP), have been shown to interfere with rat ovarian steroidogenesis. However, little is known about steroidogenic effects of infantile OP exposure on immature ovary. The aim of the present study was to investigate the effects of infantile OP exposure on plasma FSH, LH, estradiol, and progesterone levels in 14-day-old female rats. The effect on ovarian steroidogenic acute regulatory protein (StAR) and FSH receptor (FSHr) expression was analyzed by Western blotting. Ex vivo analysis was carried out for follicular estradiol, progesterone, testosterone, and cAMP production. Sprague-Dawley rats were given OP (0, 10, 50, or 100 mg/kg) subcutaneously on postnatal days 6, 8, 10, and 12. On postnatal day 14, plasma FSH was decreased and progesterone increased significantly at a dose of 100 mg OP/kg. In addition, the highest OP dose advanced the time of vaginal opening in puberty. OP had no effect on infantile LH and estradiol levels or ovarian FSHr content. Ovarian StAR protein content and ex vivo hormone and cAMP production were decreased at all OP doses compared to controls. However, hormone levels recovered independent on FSH and even increased above the control level during a prolonged culture. On postnatal day 35, no statistically significant differences were seen between control and OP-exposed animals in plasma FSH, LH, estradiol, and progesterone levels, or in ovarian StAR protein content. The results indicate that the effect of OP on the infantile ovary is reversible, while more permanent effects in the hypothalamus and pituitary, as described earlier, are involved in the reduction of circulating FSH levels and premature vaginal opening.


Subject(s)
Ovary/drug effects , Phenols/toxicity , Phosphoproteins/analysis , Steroids/biosynthesis , Animals , Body Weight/drug effects , Cyclic AMP/biosynthesis , Female , Follicle Stimulating Hormone/blood , Gonadal Steroid Hormones/blood , Luteinizing Hormone/blood , Ovary/metabolism , Rats , Rats, Sprague-Dawley , Receptors, FSH/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...