24 research outputs found
Evaluation of chidamide and PFI-1 as a combination therapy for triple-negative breast cancer
Purpose: To evaluate the in vitro and in vivo effects of the combination therapy of histone deacetylases (HDAsC) inhibitor, chidamide, and bromodomain-containing proteins (BETs) inhibitor, PFI-1, on triplenegative breast cancer (TNBC).
Methods: Four distinct breast cancer cell lines and one TNBC mouse model were treated with vehicle, chidamide, PFI-1 alone, or chidamide and PFI-1. The inhibitory effect of chidamide or PFI-1 on HDACs and BETs was assessed by HDAC enzyme inhibition and AlphaScreen assays. Cell viability was determined by MTT assay while protein expression of p-STAT3 was evaluated by western blotting and immunohistochemistry (IHC) staining assay.
Results: Chidamide exerted inhibitory effect on HDACs while PFI-1 inhibited BET proteins. The threedimensional model demonstrated the interactions between chidamide and HDAC2, and between PFI-1 and BRD4. Chidamide or PFI-1 exerted inhibitory effects on breast cancer cell proliferation in vitro. However, the combination of PFI-1 and chidamide significantly inhibit MDA-MB-231 cell viability, and decrease the expression of p-STAT3, when compared to that treated with chidamide or PFI-1 alone. Moreover, the combined inhibitory effect of PFI-1 and chidamide on tumor growth was also found in the in vivo mice experiments.
Conclusion: The combination of chidamide and PFI-1 is a potential is a potential therapeutic strategy for the management of TNBC.
Keywords: Triple-negative breast cancer, Histone deacetylases, Bromodomai
Two novel TSC2 mutations in renal epithelioid angiomyolipoma sensitive to everolimus.
People who suffers renal angiomyolipoma (AML) has a low quality of life. It is widely known that genetic factors including TSC2 mutation contribute to certain populations of renal AML-bearing patients. In this study, we are the first to identify novel TSC2 mutations in one Chinese renal epithelioid AML patient: c.2652C>A; c.2688G>A based on sequencing result from biopsy tissue. These two somatic mutations cause a translational stop of TSC2, which leads to mTORC1 activation. Given the fact that activation of mTORC1 ensures cell growth and survival, we applied its inhibitor, FDA-approved everolimus, to this woman. After months of treatment with everolimus, Computer-Tomography (CT) scan results showed that everolimus successfully reduced tumor growth and distal metastasis and achieved partial response (PR) to everolimu according to Response Evaluation Criteria in Solid Tumors (RECIST version 1.1). Further Blood Routine Examination results showed the concentration of red cell mass, hemoglobin, white blood cell (WBC), platelets and hematocrit (HCT) significantly returned to normal levels indicating patients with these two TSC2 mutations could be effectively treated by everolimus
Estrogen Receptor-Alpha 36 Mediates Mitogenic Antiestrogen Signaling in ER-Negative Breast Cancer Cells
It is prevailingly thought that the antiestrogens tamoxifen and ICI 182, 780 are competitive antagonists of the estrogen-binding site of the estrogen receptor-alpha (ER-α). However, a plethora of evidence demonstrated both antiestrogens exhibit agonist activities in different systems such as activation of the membrane-initiated signaling pathways. The mechanisms by which antiestrogens mediate estrogen-like activities have not been fully established. Previously, a variant of ER-α, EP–α36, has been cloned and showed to mediate membrane-initiated estrogen and antiestrogen signaling in cells only expressing ER-α36. Here, we investigated the molecular mechanisms underlying the antiestrogen signaling in ER-negative breast cancer MDA-MB-231 and MDA-MB-436 cells that express high levels of endogenous ER-α36. We found that the effects of both 4-hydoxytamoxifen (4-OHT) and ICI 182, 780 (ICI) exhibited a non-monotonic, or biphasic dose response curve; antiestrogens at low concentrations, elicited a mitogenic signaling pathway to stimulate cell proliferation while at high concentrations, antiestrogens inhibited cell growth. Antiestrogens at l nM induced the phosphorylation of the Src-Y416 residue, an event to activate Src, while at 5 µM induced Src-Y527 phosphorylation that inactivates Src. Antiestrogens at 1 nM also induced phosphorylation of the MAPK/ERK and activated the Cyclin D1 promoter activity through the Src/EGFR/STAT5 pathways but not at 5 µM. Knock-down of ER-α36 abrogated the biphasic antiestrogen signaling in these cells. Our results thus indicated that ER-α36 mediates biphasic antiestrogen signaling in the ER-negative breast cancer cells and Src functions as a switch of antiestrogen signaling dependent on concentrations of antiestrogens through the EGFR/STAT5 pathway
TiO2 nanoparticles and BPA are combined to impair the development of offspring zebrafish after parental coexposure
Titanium dioxide (TiO2) nanoparticles and bisphenol A (BPA) in aquatic environments interact reciprocally to enhance the maternal transfer of pollutants to offspring, thus varying the innate toxicities during early embryonic development. However, it remains unexplored regarding the molecular mechanisms of developmental toxicity in offspring after parental coexposure. In the present study, adult zebrafish were exposed to TiO2 nanoparticles (100 mu g/L), BPA (20 mu g/L) or their binary mixture for four months. Then, eggs of F1 generation were collected and reared in clean water until 5 days post-fertilization. In characteristic of larval survival and growth, parental coexposure to TiO2 particles and BPA caused a severer inhibition of F1 offspring larvae compared with single exposure. Mechanistic investigation by shotgun proteomics found that development of larval offspring from coexposed parents was impaired through a distinct mode of toxicity, that is, specifically altering the activity of phagosome and lysosome. Single exposure of adult zebrafish to TiO2 mainly affected insulin-responsive compartment; and BPA parental exposure mainly affected carbohydrate metabolism and calcium signaling of larval offspring. Furthermore, considering the tight regulation of sex hormones in the expression of vitellogenin (VTG), addition of nanoparticles during parental exposure led to inconsistencies between VTG induction and endogenous levels of sex hormones (estradiol and testosterone) in F1 offspring fish. This implied that transfer of nanoparticles to offspring larvae may change the availability of hormonal molecules and BPA at target tissues. Overall, current results provided mechanistic clues into the multigenerational developmental toxicity by parental coexposure to TiO2 particles and BPA. (C) 2018 Elsevier Ltd. All rights reserved
Optical toxicity of triphenyl phosphate in zebrafish larvae
Triphenyl phosphate (TPhP) has been shown to cause developmental neurotoxicty. Considering the visual system is a sensitive target, in the present study, we investigated the potential toxicity of TPhP on the visual development and function in zebrafish larvae. Embryos were exposed to 0, 0.1, 1, 10, and 30 mu g/L TPhP from 2 to 144 h post-fertilization (hpf). The transcription of photoreceptor opsin genes, and histopathological changes in the retina and visual behavior (optokinetic and phototactic responses) were evaluated. TPhP significantly downregulated the transcription of opsin genes (zfrho, opn1sw1, opn1sw2, opn1mw1, opn1mw2, opn1mw3, opn1mw4, opn1lw1 and opn1lw2) in all exposure groups. Histopathological analysis revealed that the areas of the outer nuclear layer (ONL), inner nuclear layer (INL), and inner plexiform layer (IPL) of the retina were significantly reduced in the 10 and 30 mu g/L TPhP groups. The number of ganglion cells was reduced significantly in the 30 mu g/L group. The optokinetic response (OKR) and phototactic response showed dose-dependent decreases caused by impaired visual function, which was confirmed by unchanged locomotor activity. The results indicated that exposure to environmentally relevant concentrations of TPhP could inhibit the transcription of genes related to visual function and impair retinal development, thus leading to visual impairment in zebrafish larvae
CRITICAL APPRAISAL OF EFFECTIVENESS OF ORAL FINGOLIMOD ON RELAPSING MULTIPLE SCLEROSIS
Oral fingolimod has been recently accepted as a treatment for relapsing Multiple Sclerosis (MS) by the Indonesian Food and Drug Administration. The number of MS patients is increasing in Indonesia. There is a critical need of systematically reviewing the new medication for both efficacy and safety. This review aimed to appraise the clinical evidences of oral fingolimod for the effective treatment of relapsing MS. We searched in Pubmed database. We limited our search to only articles that were in full text, published within the last 10 years, and in English. We used the Jadad scale to measure the quality of the evidences. We only found 3 trials, all conducted with randomized and double blind design. The three trials were: the FREEDOMS I, FREEDOMS II, and TRANSFORMS studies. The FREEDOMS studies compared with placebo, and the TRANSFORMS study compared with injectable interferon. All of the studies have good quality in methodology (Jadad scale >3). The results of the three studies showed the benefit of oral fingolimod in reducing MS relapse compared with placebo with relative risk reduction range from 48% to 54%, and also in reducing new lesions in T2 brain MRI with relative risk reduction range from 35% to 74%. Our critical appraisal found that oral fingolimod improved clinical outcomes. The availability of oral fingolimod in Indonesia makes it one of the options in treatment of MS relapse in Indonesia and has been demonstrated to be effective and safe with relatively small risk
Reduction of n-3 PUFAs, specifically DHA and EPA, and enhancement of peroxisomal beta-oxidation in type 2 diabetic rat heart
<p>Abstract</p> <p>Background</p> <p>There is overwhelming evidence that dietary supplementation with n-3 polyunsaturated fatty acids (PUFAs), mainly EPA (C20:5n-3) and DHA (C22:6n-3), has cardiovascular protective effects on patients with type 2 diabetes mellitus (T2DM) but not on healthy people. Because the T2DM heart increases fatty acid oxidation (FAO) to compensate for the diminished utilization of glucose, we hypothesize that T2DM hearts consume more n-3 PUFAs and, therefore, need more n-3 PUFAs. In the present study, we investigated the changes in cardiac n-3 PUFAs and peroxisomal beta-oxidation, which are responsible for the degradation of PUFAs in a high-fat diet (HFD) and low-dose streptozotocin- (STZ) induced type 2 diabetic rat model.</p> <p>Methods and results</p> <p>The capillary gas chromatography results showed that all the n-3 (or omega-3) PUFAs, especially DHA (~50%) and EPA (~100%), were significantly decreased, and the n-6/n-3 ratio (~115%) was significantly increased in the hearts of diabetic rats. The activity of peroxisomal beta-oxidation, which is crucial to very-long-chain and unsaturated FA metabolism (including DHA), was significantly elevated in DM hearts. Additionally, the real-time PCR results showed that the mRNA expression of most peroxisomal beta-oxidation key enzymes were up-regulated in T2DM rat hearts, which might contribute to the reduction of n-3 (or omega-3) PUFAs.</p> <p>Conclusion</p> <p>In conclusion, our results indicate that T2DM hearts consume more n-3 PUFAs, especially DHA and EPA, due to exaggerated peroxisomal beta-oxidation.</p
Accumulation of perfluorobutane sulfonate (PFBS) and impairment of visual function in the eyes of marine medaka after a life-cycle exposure
As an alternative to perfluorooctane sulfonate (PFOS), increasing usage of perfluorobutane sulfonate (PFBS) has led to ubiquitous presence in the environment. PFBS is also shown to potently disrupt the thyroid endocrine system. Considering the regulation of thyroid hormones in visual development, PFBS is likely to adversely affect the development and function of visual systems, which is a sensitive target of environmental pollutants. Therefore, the present study exposed marine medaka embryos to environmentally realistic concentrations of PFBS (0, 1.0, 2.9 and 9.5 mu g/L) for an entire life-cycle. After exposure until sexual maturity, eyes of adult medaka were dissected to directly investigate the ocular accumulation and toxicity of PFBS. For the first time, substantial accumulation of an environmental pollutant (i.e., PFBS) was observed in the eye tissue. PFBS exposure was also found to impair the visual development and function in a sex-dependent manner. In female medaka, weight of eyes was significantly decreased, while content of water was increased, probably resulting in higher intraocular pressure. Multiple neural signaling processes were also disturbed by PFBS life-cycle exposure, including cholinergic, glutamatergic, GABAergic and monoaminergic systems. Increased levels of norepinephrine and epinephrine neurotransmitters may adaptively decrease the intraocular hypertension in female eyes. In addition, proteomic profiling identified the visual proteins of differential expressions (e.g., beta and gamma crystallins, arrestin and lumican), which were significantly associated with visual perception and motor activity of eyes. Overall, this study found that PFBS was able to accumulate in the eyes and induce ocular toxicities. The susceptibility and sex-specific responses of visual systems to environmental pollutants warrants more works for a comprehensive risk assessment.</p
Profile of cardiac lipid metabolism in STZ-induced diabetic mice
Abstract Background Lipotoxicity contributes to diabetic myocardial disease. In this study, we investigated the lipid species contributing to lipotoxicity and the relationship with peroxisomal β-oxidation in the heart of diabetic mice. Methods Male C57BL/6 mice were randomly divided into a Diabetic group (intraperitoneal injection of STZ) and a Control group (saline). Cardiac function indexes [ejection fraction (EF%) and fractional shortening (FS%)] were evaluated by echocardiography. Morphological changes in the myocardial tissues and mitochondria were assessed by electron microscopy following hematoxylin and eosin staining. Blood myocardial injury indexes and lipids were measured using an automatic biochemical analyzer. Cardiac ATP levels were analyzed using a commercially available kit. mRNA levels of glucose transporter 4 (GLUT4), fatty acid binding protein 3 (FABP3), palmitoyl transferase 1α (CPT-1α), acyl-CoA oxidase 1 (AOX1), D-bifunctional protein (DBP), 3-ketoacyl-CoA thiolase A (THLA), uncoupling protein (UCP) 2 and UCP3 were investigated by quantitative reverse-transcription polymerase chain reaction. FABP3 protein expression was analyzed by Western blotting. Non-targeted metabolomics by LC-MS/MS was applied to evaluate profile of lipid metabolism in heart. Results Compared with controls, EF% and FS% were significantly reduced in diabetic mice. Furthermore, blood myocardial injury indexes and lipids, as well as myocardial mitochondrial cristae fusion were significantly increased. In the diabetic heart, GLUT4 expression was decreased, while expression of FABP3, CPT-1α, AOX1, DBP, THLA, UCP2 and UCP3 was increased, and ATP levels were reduced. In total, 113 lipids exhibited significant differential expression (FC > 2, P < 0.05) between the two groups, with sphingolipid metabolism identified as the top-ranking affected canonical pathway. In the diabetic heart, long-chain hydroxyl-acylcarnitines (8/8) and acylcarnitines (6/11), triglycerides (2/5), and diacyglycerol (3/7) were upregulated, while very long-chain polyunsaturated fatty acids (PUFAs) (5/6) including eicosapentaenoate, docosahexaenoate, phosphocholine (11/19), lysophosphocholine (5/9), phosphoethanolamine (7/11), lysophosphoethanolamine (7/10), phosphatidylglycerol (6/8), phosphoserine (6/8), phosphatidylinositol (2/2), phosphatidic acid (1/1), lysophosphatidic acid (1/1) and sphingomyelin (6/6) were downregulated. Conclusions Our data suggest that the increase in toxic lipid species and decreased in PUFAs undergoing peroxisomal β-oxidation, combined with the reduction in phospholipids cause mitochondrial injury and subsequent uncoupling of phosphorylation and ATP deficiency; thereby leading to diabetic heart dysfunction
Multigenerational Disruption of the Thyroid Endocrine System in Marine Medaka after a Life-Cycle Exposure to Perfluorobutanesulfonate
Accumulation of perfluorobutanesulfonate (PFBS) is frequently detected in biota, raising concerns about its ecological safety. However, hazardous effects of PFBS remain largely unexplored, especially for endocrine disrupting potency. In the present study, the multigenerational endocrine disrupting potential of PFBS was investigated by exposing F0 marine medaka eggs to PFBS at different concentrations (0, 1.0, 2.9, and 9.5 mu g/L) until sexual maturity. The F1 and F2 generations were reared without continued exposure. Thyroidal disturbances were examined in all three generations. PFBS exposure decreased the levels of 3,5,3'-triiodothyronine (T3) in F0 female blood; however, it increased T3 or thyroxine (T4) levels in F0 brains, in which hyperthyroidism suppressed the local transcription of 5'-deiodinase 2 (Dio2). Obviously decreased T3 was transferred to F1 eggs, although the parental influences were reversed in F1 larvae. Delayed hatching was coupled with elevated T3 levels in F1 larvae. F1 adults showed comparable symptoms of thyroidal disruption with F0 adults. A slight recovery was noted in the F2 generation, although F2 larvae still exhibited thyroid disruption and synthesized excessive T4. Our results suggested that the offspring suffered more severe dysfunction of the thyroidal axis albeit without direct exposure. This study provided the first molecular insight about PFBS toxicology on the thyroid, beneficial to both human and environmental risk assessment