Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
Article in English | MEDLINE | ID: mdl-35886217

ABSTRACT

BACKGROUND: Mifepristone (RU-486) has been approved for abortion in Taiwan since 2000. Mifepristone was the first non-addictive medicine to be classified as a schedule IV controlled drug. As a case of the "misuse" of "misuse of drugs laws," the policy and consequences of mifepristone-assisted abortion for pregnant women could be compared with those of illicit drug use for drug addicts. METHODS: The rule-making process of mifepristone regulation was analyzed from various aspects of legitimacy, social stigma, women's human rights, and access to health care. RESULTS AND DISCUSSION: The restriction policy on mifepristone regulation in Taiwan has raised concerns over the legitimacy of listing a non-addictive substance as a controlled drug, which may produce stigma and negatively affect women's reproductive and privacy rights. Such a restriction policy and social stigma may lead to the unwillingness of pregnant women to utilize safe abortion services. Under the threat of the COVID-19 pandemic, the US FDA's action on mifepristone prescription and dispensing reminds us it is time to consider a change of policy. CONCLUSIONS: Listing mifepristone as a controlled drug could impede the acceptability and accessibility of safe mifepristone use and violates women's right to health care.


Subject(s)
Mifepristone , Public Policy , Abortion, Induced/methods , COVID-19/epidemiology , Female , Humans , Mifepristone/therapeutic use , Pandemics , Pregnancy , Women's Health , COVID-19 Drug Treatment
3.
Viruses ; 13(5)2021 04 30.
Article in English | MEDLINE | ID: mdl-33946474

ABSTRACT

Human immunodeficiency virus (HIV) is associated with neuroendocrine dysfunction which may contribute to co-morbid stress-sensitive disorders. The hypothalamic-pituitary-adrenal (HPA) or -gonadal (HPG) axes are perturbed in up to 50% of HIV patients. The mechanisms are not known, but we have found the HIV-1 trans-activator of transcription (Tat) protein to recapitulate the clinical phenotype in male mice. We hypothesized that HPA and/or HPG dysregulation contributes to Tat-mediated interactions with oxycodone, an opioid often prescribed to HIV patients, in females. Female mice that conditionally-expressed the Tat1-86 protein [Tat(+) mice] or their counterparts that did not [Tat(-) control mice] were exposed to forced swim stress (or not) and behaviorally-assessed for motor and anxiety-like behavior. Some mice had glucocorticoid receptors (GR) or corticotropin-releasing factor receptors (CRF-R) pharmacologically inhibited. Some mice were ovariectomized (OVX). As seen previously in males, Tat elevated basal corticosterone levels and potentiated oxycodone's psychomotor activity in females. Unlike males, females did not demonstrate adrenal insufficiency and oxycodone potentiation was not regulated by GRs or CRF-Rs. Rather OVX attenuated Tat/oxycodone interactions. Either Tat or oxycodone increased anxiety-like behavior and their combination increased hypothalamic allopregnanolone. OVX increased basal hypothalamic allopregnanolone and obviated Tat or oxycodone-mediated fluctuations. Together, these data provide further evidence for Tat-mediated dysregulation of the HPA axis and reveal the importance of HPG axis regulation in females. HPA/HPG disruption may contribute vulnerability to affective and substance use disorders.


Subject(s)
HIV Infections/complications , HIV Infections/virology , HIV-1/physiology , Neurosecretory Systems/metabolism , Neurosecretory Systems/physiopathology , Oxycodone/pharmacology , Psychomotor Performance/drug effects , tat Gene Products, Human Immunodeficiency Virus/metabolism , Animals , Behavior, Animal , Disease Models, Animal , Estrous Cycle , Female , Humans , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Mice , Motor Activity , Neurosecretory Systems/drug effects , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Pregnanolone/blood , Pregnanolone/metabolism , Steroids/blood
4.
J Steroid Biochem Mol Biol ; 208: 105823, 2021 04.
Article in English | MEDLINE | ID: mdl-33484844

ABSTRACT

Polycystic ovarian syndrome (PCOS) is a multi-factorial gynecological endocrine disorder. It affects fertility in women and also predisposes to insulin resistance, type 2 diabetes mellitus, obesity etc. Earlier, significance of autophagy has been explored in PCOS-related metabolic disorders and during normal folliculogenesis. Increasing evidences reveal connection of autophagy with chronic inflammatory behaviour, an associated phenomena in polycystic ovaries. However, understanding of the association of autophagy with PCOS is still obscure. This study reveals that increased autophagy in mifepristone (RU486) treated KK-1 cells and in vivo PCO rat model is characterized by upregulated Androgen Receptor (AR) expression and downregulated PCO biomarker aromatase. The prevalence of autophagy has been observed to be concomitant with increased expression of two autophagic markers Beclin1 and MAP-LC3-II while the autophagy substrate p62/SQSTM1 was downregulated. Immunohistochemical staining revealed increased localization of MAP-LC3 in the compacted granulosa layers of the follicular cysts in the PCO model. The PCO rat models also demonstrated augmented levels of p65, the active subunit of NF-κB, which acts as a transcriptional regulator of several pro-inflammatory factors. NF-κB repressor and anti-inflammatory herbal drug thymoquinone, known to alleviate PCO condition, downregulated autophagy modules substantially. Pre-treatment with thymoquinone upregulated aromatase, reduced AR levels and decreased autophagic markers as well as p65 levels, simulating super-ovulated condition. In conclusion, the anti-inflammatory phytochemical thymoquinone alleviated PCO condition.


Subject(s)
Autophagy/drug effects , Benzoquinones/pharmacology , Mifepristone/pharmacology , Polycystic Ovary Syndrome/drug therapy , Receptors, Androgen/genetics , Androgens/metabolism , Animals , Autophagy/genetics , Beclin-1/genetics , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Granulosa Cells/drug effects , Humans , Insulin Resistance/genetics , Ovary/drug effects , Ovary/growth & development , Ovulation/drug effects , Ovulation/genetics , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/pathology , Rats , eIF-2 Kinase/genetics
5.
Behav Brain Res ; 347: 436-445, 2018 07 16.
Article in English | MEDLINE | ID: mdl-29580890

ABSTRACT

Increased proinflammatory cytokines, such as interleukin (IL)-1ß, may play an important role in the etiology of depression because they cause the hypothalamic-pituitary-adrenal axis to release glucocorticoids (GC) and induce dysfunction of serotonin and norepinephrine neurotransmission. Sustained increase in GC may activate microglia to induce neuroinflammation, and suppress astrocytes to produce neurotrophins, which lead to neuronal apoptosis. Here, we tested the hypothesis that glucocorticoid receptor (GR) antagonist mifepristone (RU486) may attenuate IL-1ß-induced depression-like behavior by regulating the neuroinflammation and neurotrophin functions of microglia and astrocytes. Rats received intracerebroventricular injections of IL-1ß (10 ng) and/or subcutaneous injections of RU486 for 14 days. Then animal depression-like behaviors, serum corticosterone concentration, the levels of pro-inflammatory cytokines (TNF-α, IL-6), mRNA and protein expressions of CD11b, GFAP and neurotrophins (pro-BDNF, BDNF, GDNF and their receptors TrkB, p75, GFRα-1 and GFRα-2) in the amygdala were studied. Compared to controls, significantly decreased rearing score and increased defecation in the open field test, decreases in ratio of open/closed time in the elevated plus maze and in sucrose preference, while increased level of corticosterone in the serum were found in the rats administrated with IL-1ß. IL-1ß administration also reduced the expressions of GFAP, BDNF, GDNF and its receptor GFR-α1, but increased the expressions of CD11b, pro-BDNF, p75 and pro-inflammatory cytokines (TNF-α, IL-6) concentrations. RU486 treatment markedly attenuated these changes induced by IL-1ß, except for the expressions of GFR-α1. In conclusion, RU486 may improve depression-like changes by suppressing microglia and inflammation and promoting astrocytes to restore neurotrophin function.


Subject(s)
Antidepressive Agents/pharmacology , Depressive Disorder/drug therapy , Depressive Disorder/immunology , Interleukin-1beta/metabolism , Mifepristone/pharmacology , Amygdala/drug effects , Amygdala/metabolism , Anhedonia/drug effects , Anhedonia/physiology , Animals , Corticosterone/blood , Disease Models, Animal , Hormone Antagonists/pharmacology , Interleukin-1beta/administration & dosage , Male , Motor Activity/drug effects , Motor Activity/physiology , Nerve Growth Factors/metabolism , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/metabolism , Recombinant Proteins/administration & dosage
6.
Oncotarget ; 8(35): 59123-59135, 2017 Aug 29.
Article in English | MEDLINE | ID: mdl-28938623

ABSTRACT

SDF-1/CXCR4 signaling axis determines the proliferative potential and site-specific cancer metastasis. Recent studies suggest involvement of the axis and steroidal hormone in ovarian cancer metastasis. Here we hypothesize that mifepristone (RU486), a well-known progesterone-based abortifacient, might interfere this axis and inhibit ovarian cancer metastasis. Mifepristone at concentrations < IC50 inhibited expression of CXCR4 on cell surface of ovarian cancer SKOV-3 and IGROV-1, and reduced expression of the intracellular CXCR4 protein and its related mRNA activated by SDF-1. SDF-1 significantly stimulated proliferation of SKOV-3 and IGROV-1 cells with concomitant increases in intracellular phosphorylation of Akt and ERK. SDF-1 activated cell chemotatic migration and actin polymerization, and up-regulated expression of MMP-2, MMP-9, COX-2, VEGF without influencing the adhesion molecules ICAM-1 and integrins ß1, α1, α3, α5, and α6. The above-mentioned effects of SDF-1 could be antagonized by mifepristone concentration-dependently, and CXCR4 antagonist AMD3100. Mifepristone suppressed the SDF-1-induced migration, invasion and adhesion of the cancer cells to extracellular matrixes. Three-day pretreatment of nude mice with mifepristone (5 and 20 mg/kg/day) followed by a single intraperitoneal IGROV-1 inoculation, along with repeated SDF-1 and mifepristone administrations in turn every other day for 36 days significantly reduced ascitic fluid, metastatic foci, tumor weight and immunoreactivity of CXCR4 in comparison with the SDF-1-treated control. Our results suggest that mifepristone inhibit SDF-1/CXCR4 signaling axis, may have preventive and therapeutic effects on ovarian cancer metastasis.

7.
Biomed Pharmacother ; 90: 339-349, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28376402

ABSTRACT

Uncontrolled cell proliferation and metastasis are the two well-known manifestations of melanoma. We hypothesized that metapristone, a potential cancer metastatic chemopreventive agent derived from mifepristone (RU486), had a dual function to fight cancer. In the present study, our findings clearly demonstrated that metapristone had modest cytostatic effect in melanoma cells. Metapristone inhibited cell viability and induced both early and late apoptosis in B16F10 and A375 cells in a time- and concentrate-dependent manner. Metapristone-treatment caused the cell arrest at the G0/G1 stage, and the inhibition of colony formation in B16F10 cells. Western blot analysis further revealed that metapristone treatment elicited a decline of Akt and ERK phosphorylation and Bcl-2, and facilitated expression of total P53 and Bax in A375 cells. In addition, cell migration and invasion were significantly suppressed by metapristone through down-regulating the expression of MMP-2, MMP-9, N-cadherin and vimentin, whereas up-regulating E-cadherin expression. Notably, metapristone exhibited anti-metastatic activity in melanoma B16F10 cells in vivo. Our results reveal metapristone, having the dual function of anti-proliferation and anti-migration for melanoma cell lines, may be a useful chemopreventive agent to reduce the risk of melanoma cancer metastasis.


Subject(s)
Cell Movement/drug effects , Cell Proliferation/drug effects , Melanoma/drug therapy , Mifepristone/analogs & derivatives , Mifepristone/pharmacology , Neoplasm Metastasis/drug therapy , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Down-Regulation/drug effects , Female , G1 Phase/drug effects , Melanoma/metabolism , Mice , Mice, Inbred C57BL , Resting Phase, Cell Cycle/drug effects , Signal Transduction/drug effects , Up-Regulation/drug effects
8.
Mol Carcinog ; 56(8): 1896-1908, 2017 08.
Article in English | MEDLINE | ID: mdl-28277622

ABSTRACT

Recent global epidemiological studies revealed the lower ovarian cancer death from long-term use of oral contraceptives. However, the underlying mechanism of action is not clear. Here, we use the abortifacient metapristone (RU486 derivative) to test the hypothesis that the contraceptives might interrupt CXCL12/CXCR4 chemokine axis to inhibit ovarian cancer metastasis. Metapristone at concentrations (

Subject(s)
Antineoplastic Agents/therapeutic use , Chemokine CXCL12/metabolism , Mifepristone/analogs & derivatives , Neoplasm Invasiveness/prevention & control , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/prevention & control , Peritoneal Neoplasms/secondary , Receptors, CXCR4/metabolism , Abortifacient Agents/chemistry , Abortifacient Agents/pharmacology , Abortifacient Agents/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Carcinoma, Ovarian Epithelial , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Chemoprevention , Female , Humans , Mice, Inbred BALB C , Mice, Nude , Mifepristone/chemistry , Mifepristone/pharmacology , Mifepristone/therapeutic use , Neoplasm Invasiveness/pathology , Neoplasms, Glandular and Epithelial/drug therapy , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovary/drug effects , Ovary/metabolism , Ovary/pathology , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/pathology , Peritoneum/drug effects , Peritoneum/metabolism , Peritoneum/pathology , Signal Transduction/drug effects
9.
Reprod Sci ; 24(7): 976-988, 2017 07.
Article in English | MEDLINE | ID: mdl-27678094

ABSTRACT

Mifepristone (RU 486) is a compound that is structurally related to steroid hormones, which is derived from the estrane progestins. This compound strongly binds the progesterone and glucocorticoid receptor and, to a lesser extent, the androgen receptor. This compound has its effects through different signaling pathways, related to genomic and nongenomic effects. The genomic effect involves the activation or blockage of nuclear or intracellular receptor, that in this case the progesterone, glucocorticoid, and androgen receptors. On the contrary, the nongenomic effect of mifepristone is independent of the activation of these receptors. Regarding the nongenomic, several authors observed that mifepristone induces higher uterine artery blood flow probably due to the decrease in serum nitric oxide level. Moreover, recently it has been demonstrated that mifepristone induces relaxation, and this effect is independent of the endothelium and due to the activation of the calcium channels. The main side effects associated with this pathway are hemorrhage and inhibition of platelet aggregation that can lead to hypovolemia or to hypotension. Concerning the genomic effect, this drug blocks progesterone, androgens, and glucocorticoids receptors and also activates the progesterone receptor and their respective effects. The most frequently reported adverse effects of mifepristone are nausea, vomiting, hypovolemia, hypotension, amenorrhea, and infertility. The main purpose of this review is to describe the genomic and nongenomic effects of mifepristone at vascular level and describe some pathologies in which mifepristone is used as a treatment.


Subject(s)
Endothelium, Vascular/drug effects , Epigenesis, Genetic/drug effects , Hormone Antagonists/pharmacology , Mifepristone/pharmacology , Androgen Receptor Antagonists/pharmacology , Animals , Calcium Channels/metabolism , Endothelium, Vascular/metabolism , Nitric Oxide/metabolism , Receptors, Androgen/metabolism , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/metabolism , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/metabolism , Signal Transduction/drug effects , Uterine Artery/drug effects , Uterine Artery/metabolism , Vasodilation/drug effects , Vasodilation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL