Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 43
1.
Biochem Biophys Res Commun ; 525(2): 313-318, 2020 04 30.
Article En | MEDLINE | ID: mdl-32087973

For achieving efficient cancer treatment, it is important to elucidate the mechanism responsible for the accumulation of nanoparticles in tumor tissue. Recent studies suggest that nanoparticles are not delivered merely through gaps between tumor endothelial cells. We previously reported that the maturation of the vascular structure by the vascular endothelial cell growth factor receptor 2 (VEGFR2) using a previously developed siRNA delivery technology (RGD-MEND) significantly enhanced the accumulation of nanoparticles in types of cancers that area vessel-rich (renal cell carcinoma). This result was completely inconsistent with the generally accepted theory of the enhanced permeability and retention (EPR) effect. We hypothesized that a caveolin-1 (Cav1)-mediated transcellular route would be involved with the penetration of nanoparticles into tumor vasculature. To reveal the exact mechanism responsible for this enhancement, we observed the delivery of long-circulating liposomes (LPs) after Cav1 was co-suppressed by RGD-MEND with VEGFR2. The enhanced delivery of LPs by siRNA against VEGFR2 (siVEGFR2) was accompanied by the elevated expression of the Cav1 protein. In addition, Cav1 knockdown by siRNA against Cav1 (siCav1) canceled the enhanced delivery of LPs by siVEGFR2. The injection of siCav1 had no effect on the formation of alpha smooth muscle actin or vascular endothelial cell adhesion molecules. These results suggest that a Cav1-induced transcellular route and not a paracellular route, at least partially, contributes to the accumulation of nanoparticles in tumors.


Caveolin 1/physiology , Liposomes/metabolism , Neoplasms/metabolism , Transcytosis , Animals , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/metabolism , Caveolin 1/drug effects , Humans , Nanoparticles/metabolism , Neoplasms/blood supply , RNA, Small Interfering/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
2.
Mol Cancer Res ; 18(4): 644-656, 2020 04.
Article En | MEDLINE | ID: mdl-31900313

The antibody-drug conjugate trastuzumab-emtansine (T-DM1) offers an additional treatment option for patients with HER2-amplified tumors. However, primary and acquired resistance is a limiting factor in a significant subset of patients. Hypoxia, a hallmark of cancer, regulates the trafficking of several receptor proteins with potential implications for tumor targeting. Here, we have investigated how hypoxic conditions may regulate T-DM1 treatment efficacy in breast cancer. The therapeutic effect of T-DM1 and its metabolites was evaluated in conjunction with biochemical, flow cytometry, and high-resolution imaging studies to elucidate the functional and mechanistic aspects of hypoxic regulation. HER2 and caveolin-1 expression was investigated in a well-annotated breast cancer cohort. We find that hypoxia fosters relative resistance to T-DM1 in HER2+ cells (SKBR3 and BT474). This effect was not a result of deregulated HER2 expression or resistance to emtansine and its metabolites. Instead, we show that hypoxia-induced translocation of caveolin-1 from cytoplasmic vesicles to the plasma membrane contributes to deficient trastuzumab internalization and T-DM1 chemosensitivity. Caveolin-1 depletion mimicked the hypoxic situation, indicating that vesicular caveolin-1 is indispensable for trastuzumab uptake and T-DM1 cytotoxicity. In vitro studies suggested that HER2 and caveolin-1 are not coregulated, which was supported by IHC analysis in patient tumors. We find that phosphorylation-deficient caveolin-1 inhibits trastuzumab internalization and T-DM1 cytotoxicity, suggesting a specific role for caveolin-1 phosphorylation in HER2 trafficking. IMPLICATIONS: Together, our data for the first time identify hypoxic regulation of caveolin-1 as a resistance mechanism to T-DM1 with potential implications for individualized treatment of breast cancer.


Antineoplastic Agents, Immunological/therapeutic use , Breast Neoplasms/drug therapy , Caveolin 1/drug effects , Cell Hypoxia/drug effects , Maytansine/therapeutic use , Trastuzumab/therapeutic use , Antineoplastic Agents, Immunological/pharmacology , Breast Neoplasms/pathology , Female , Humans , Maytansine/pharmacology , Transfection , Trastuzumab/pharmacology
3.
J Neurosurg Anesthesiol ; 32(4): 359-366, 2020 Oct.
Article En | MEDLINE | ID: mdl-31107683

BACKGROUND: Ketamine, a commonly used nonbarbiturate anesthetic drug, possesses antidepressant properties at subanesthetic doses; however, the underlying mechanisms remain unclear. MATERIALS AND METHODS: The analgesic and antidepressant effects of ketamine were explored using a complete Freund adjuvant (CFA)-induced peripheral inflammatory pain model in vivo. Mice were first divided into sham or CFA injection group randomly, and were observed for mechanical hyperalgesia, depression-like behavior, and mRNA expression of caveolin-1. Then ketamine was administered in CFA-treated mice at day 7. RESULTS: The behavioral testing results revealed mechanical hyperalgesia and depression in mice from days 7 to 21 after CFA injection. Ketamine reversed depression-like behaviors induced by CFA injection. It also restored the brain-regional expression levels of caveolin-1 in CFA-treated mice. In addition, caveolin-1 mRNA and protein expression were increased in the prefrontal cortex and nucleus accumbens of CFA-treated mice. However, ketamine reversed the increase in caveolin-1 expression in the ipsilateral and contralateral prefrontal cortex and nucleus accumbens, supporting the distinct roles of specific brain regions in the regulation of pain and depression-like behaviors. CONCLUSIONS: In CFA-treated mice that exhibited pain behavior and depression-like behavior, ketamine reversed depression-like behavior. The prefrontal cortex and nucleus accumbens are the important brain regions in this regulation network. Despite these findings, other molecules and their mechanisms in the signal pathway, as well as other regions of the brain in the pain matrix, require further exploration.


Analgesics/pharmacology , Behavior, Animal/drug effects , Caveolin 1/blood , Depression/prevention & control , Hyperalgesia/prevention & control , Ketamine/pharmacology , Analgesics/blood , Animals , Caveolin 1/drug effects , Depression/blood , Disease Models, Animal , Freund's Adjuvant , Ketamine/blood , Mice
4.
Zhonghua Jie He He Hu Xi Za Zhi ; 42(11): 845-851, 2019 Nov 12.
Article Zh | MEDLINE | ID: mdl-31694095

Objective: To explore the role of S100A8, the receptor for advanced glycation endproducts (RAGE) and Caveolin-1 in neutrophilic asthmatic rats, and to further study the intervention of roxithromycin and the possible mechanisms. Methods: Male Brown Norway rats were randomly assigned to a control group, an asthma group and a Roxithromycin group. The asthmatic rat model was established by intraperitoneal injection of ovalbumin (OVA) and Freund's complete adjuvant (FCA) mixture, and aerosol inhalation of OVA. Rats in the Roxithromycin group were given roxithromycin injection 30 mg/kg 30 minutes before each challenge. Rats in the control and the asthma groups were replaced with equal volumes of saline, respectively. Bronchoalveolar lavage fluid (BALF) neutrophil percentage (Neu%) and pathological changes of pulmonary tissue (hematoxylin-eosin, HE staining) were measured to confirm the establishment of asthmatic models. The concentration of inflammatory cytokines and S100A8 were quantified by enzyme-linked immunosorbent assay (ELISA), and the expression of Caveolin-1 and RAGE at protein levels were detected by immunohistochemistry and Western blot. Results: Neu% in BALF of the asthma group was significantly higher than those of the control group, and Neu% in the Roxithromycin group was lower than the asthma group (all P<0.01). Pulmonary histology revealed that there were a large number of inflammatory cells infiltrated in the bronchial and perivascular, pulmonary interstitial and alveolar spaces, and the bronchial wall and smooth muscles were thickened obviously in the asthma group. Rats in the Roxithromycin group showed milder inflammation and airway remodeling change than the asthma group. There was no obvious pathological damage in the control group. The concentration of IL-6 and IL-17 in BALF and serum of rats in the asthma group were significantly higher than those in the control group (P<0.01), and Roxithromycin inhibited the high expression of these cytokines (P<0.05). The expression of S100A8 and RAGE in the asthma group were significantly higher than those in the control group [(20.6±4.4) vs (7.1±2.0) ng/L; (885±118) vs (462±102) ng/L; (14.2±1.7) vs (7.6±1.8) ng/L; (774±166) vs (406±69) ng/L, all P<0.05], and Roxithromycin inhibited the high expression of these proteins [(14.3±3.7) vs (20.6±4.4) ng/L; (650±53) vs (885±118) ng/L; (10.4±1.2) vs (14.2±1.7) ng/L; (560±64) vs (728±72) ng/L] (all P<0.05). Meanwhile, the expression of Caveolin-1 in the asthma group was significantly lower than that in the control group (P<0.01), and Roxithromycin up-regulated its expression (P<0.01). Correlation analysis showed that there was a significantly positive correlation between the expression of S100A8 and RAGE (r=0.706, P<0.01), while there was a significantly negative correlation between the expression of S100A8 and Caveolin-1 (r=-0.775, P<0.01), and between the expression of Caveolin-1 and RAGE (r=-0.919, P<0.01). Conclusion: S100A8 and Caveolin-1 may play an important role in neutrophilic asthma via RAGE, and Roxithromycin may exerts anti-inflammatory effects and inhibition of airway remodeling partly through this signaling pathway.


Anti-Bacterial Agents/pharmacology , Asthma/drug therapy , Calgranulin A/drug effects , Caveolin 1/drug effects , Roxithromycin/pharmacology , Airway Remodeling , Animals , Anti-Bacterial Agents/administration & dosage , Blotting, Western , Bronchoalveolar Lavage Fluid , Calgranulin A/metabolism , Caveolin 1/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Lung/physiopathology , Male , Neutrophils/drug effects , Neutrophils/metabolism , Ovalbumin , Rats , Receptor for Advanced Glycation End Products , Roxithromycin/administration & dosage
5.
Toxicol Appl Pharmacol ; 355: 43-51, 2018 09 15.
Article En | MEDLINE | ID: mdl-29940203

Toluene diisocyanate (TDI) is a reactive chemical used in manufacturing plastics. TDI exposure adversely affects workers' health, causing occupational asthma, but individuals differ in susceptibility. We recently suggested a role for signalling mediated by the enzyme autotaxin (ATX) and its product, lysophosphatidic acid (LPA), in TDI toxicity. Here we genotyped 118 TDI-exposed workers for six single-nucleotide polymorphisms (SNPs) in genes encoding proteins implicated in ATX-LPA signalling: purinergic receptor P2X7 (P2RX7), CC motif chemokine ligand 2 (CCL2), interleukin 1ß (IL1B), and caveolin 1 (CAV1). Two P2RX7 SNPs (rs208294 and rs2230911) significantly modified the associations between a biomarker of TDI exposure (urinary 2,4-toluene diamine) and plasma LPA; two IL1B SNPs (rs16944 and rs1143634) did not. CAV1 rs3807989 modified the associations, but the effect was not statistically significant (p = 0.05-0.09). In vitro, TDI-exposed bronchial epithelial cells (16HBE14o-) rapidly released ATX and IL-1ß. P2X7 inhibitors attenuated both responses, but confocal microscopy showed non-overlapping localizations of ATX and IL-1ß, and down-regulation of CAV1 inhibited the ATX response but not the IL-1ß response. This study indicates that P2X7 is pivotal for TDI-induced ATX-LPA signalling, which was modified by genetic variation in P2RX7. Furthermore, our data suggest that the TDI-induced ATX and IL-1ß responses occur independently.


Lysophospholipids/metabolism , Phosphoric Diester Hydrolases/drug effects , Signal Transduction/drug effects , Toluene 2,4-Diisocyanate/toxicity , Adolescent , Adult , Biomarkers , Caveolin 1/drug effects , Caveolin 1/genetics , Cell Line , Chemical Industry , Female , Genotype , Humans , Male , Middle Aged , Occupational Exposure/adverse effects , Phosphoric Diester Hydrolases/genetics , Polymorphism, Single Nucleotide/genetics , RNA, Small Interfering/pharmacology , Receptors, Purinergic P2X7/drug effects , Receptors, Purinergic P2X7/genetics , Signal Transduction/genetics , Young Adult
6.
Acta Biochim Pol ; 65(1): 111-118, 2018.
Article En | MEDLINE | ID: mdl-29549671

This study was undertaken to verify whether simvastatin modulates Cav-1/eNOS expression, and if this modulation is associated with changes in pro- and anti-inflammatory cytokine and Toll-like receptor 4 (TLR4) level in abdominal aortic aneurysm (AAA). It is a 1:2 case-control study of non-statin (n=12) and simvastatin-treated patients (n=24) who underwent open AAA repair. Simvastatin treatment decreased Cav-1 (p<0.05) and increased eNOS expression (p<0.01) in the AAA wall. These changes might be dose dependent. The changes in Cav-1 and eNOS were associated with a trend towards decreased IL-6 and IL-17 concentration (p>0.05) and increased IL-10 concentration (p=0.055); however, TLR4 expression was unaffected, suggesting that simvastatin influences Cav-1 and eNOS in the AAA wall by other mechanisms. Simvastatin may modulate Cav-1 and eNOS expression in the aneurysmal wall, indicating a potentially beneficial role for statins in AAA patients.


Aortic Aneurysm, Abdominal/metabolism , Caveolin 1/metabolism , Nitric Oxide Synthase Type III/metabolism , Simvastatin/therapeutic use , Aged , Aortic Aneurysm, Abdominal/drug therapy , Aortic Aneurysm, Abdominal/surgery , Case-Control Studies , Caveolin 1/drug effects , Cytokines/drug effects , Female , Humans , Interleukin-17/metabolism , Interleukin-6/metabolism , Male , Middle Aged , Nitric Oxide Synthase Type III/drug effects , Simvastatin/pharmacology , Toll-Like Receptor 4/drug effects
7.
Life Sci ; 192: 270-277, 2018 Jan 01.
Article En | MEDLINE | ID: mdl-29129771

Tumor metastasis leads to a poor prognosis in breast cancer, yet the mechanisms remain unclear. Docosahexaenoic acid (DHA) extracted from Antarctic krill is an optical isomer of common DHA and has a much stronger anti-neoplastic effect. In this work, the migration and invasion abilities of MCF-7 cells treated with low concentrations of Antarctic krill DHA were evaluated. Low concentrations of Antarctic krill DHA significantly reduced the numbers of migrating and invasive MCF-7 cells, whereas the cell numbers decreased slowly in the CD95-silenced MCF-7 cells, which implies that CD95 might be involved in cell migration and invasion. Additionally, co-immunoprecipitation and Western blotting demonstrated that Antarctic krill DHA induced the accumulation of CD95 and caveolin-1 interaction, resulting in the down-regulation of MMP2 expression through the FAK/SRC/PI3K/AKT signaling pathway. In conclusion, Antarctic krill DHA enhanced the interaction between CD95 and caveolin-1, which may led to an inhibitory effect on cell migration and invasion via the FAK/SRC/PI3K/AKT signaling pathway. Our study indicates that Antarctic krill DHA has great potential for tumor therapy and has revealed a new metastatic mechanism mediated by the interaction of CD95 with caveolin-1.


Caveolin 1/drug effects , Cell Movement/drug effects , Docosahexaenoic Acids/pharmacology , Euphausiacea/chemistry , Neoplasm Invasiveness , fas Receptor/drug effects , Animals , Antarctic Regions , Cell Count , Cell Survival , Female , Humans , MCF-7 Cells , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 2/genetics , Neoplasm Metastasis , Signal Transduction/drug effects
8.
J Clin Endocrinol Metab ; 102(6): 2100-2110, 2017 06 01.
Article En | MEDLINE | ID: mdl-28368445

Context: Increased microparticle (MP) shedding by placental trophoblasts contributes to maternal vascular inflammatory response and endothelial dysfunction in preeclampsia. Vitamin D has beneficial effects in pregnancy; however, its effect on trophoblast MP release has not been investigated. Objective: To investigate if vitamin D could protect trophoblasts from oxidative stress-induced MP release. Design: Placental trophoblasts were isolated from uncomplicated and preeclamptic placentas. Effects of vitamin D on MP release induced by oxidative stress inducer CoCl2 were studied. Main Outcome Measures: Annexin V+ MPs were assessed by flow cytometry. Expression of caveolin-1, endothelial nitric oxide synthase (eNOS), procaspase-3, cleaved caspase-3, and Rho-associated coiled-coil protein kinase 1 (ROCK1) in trophoblasts and trophoblast-derived MPs were determined by Western blot. Results: Trophoblasts from preeclamptic pregnancies released significantly more MPs than cells from uncomplicated pregnancies (P < 0.01). CoCl2-induced increase in MP release was associated with upregulation of caveolin-1 and downregulation of eNOS expression in trophoblasts (P < 0.05), which could be attenuated by 1,25(OH)2D3. Moreover, 1,25(OH)2D3 could also inhibit CoCl2-induced procaspase-3 cleavage and ROCK1 activation in trophoblasts. Consistently, CoCl2-induced upregulation of procaspase-3, cleaved caspase-3, and ROCK1 expression in trophoblast-derived MPs were also reduced in cells treated with 1,25(OH)2D3. Conclusions: Placental trophoblasts from preeclamptic pregnancies released more MP than cells from uncomplicated pregnancies. Oxidative stress-induced increase in MP shedding is associated with upregulation of caveolin-1 and downregulation of eNOS expression in placental trophoblasts. Inhibition of caspase-3 cleavage and ROCK1 activation, together with upregulation of eNOS expression, could be the potential cellular/molecular mechanism(s) of vitamin D protective effects on placental trophoblasts.


Calcitriol/pharmacology , Caspase 3/drug effects , Oxidative Stress/drug effects , Trophoblasts/drug effects , Vitamins/pharmacology , rho-Associated Kinases/drug effects , Adolescent , Adult , Blotting, Western , Case-Control Studies , Caspase 3/metabolism , Caveolin 1/drug effects , Caveolin 1/metabolism , Cell-Derived Microparticles/drug effects , Cell-Derived Microparticles/metabolism , Cobalt/pharmacology , Female , Humans , Nitric Oxide Synthase Type III/drug effects , Nitric Oxide Synthase Type III/metabolism , Pre-Eclampsia , Pregnancy , Trophoblasts/metabolism , Young Adult , rho-Associated Kinases/metabolism
9.
Psychopharmacology (Berl) ; 234(7): 1069-1077, 2017 Apr.
Article En | MEDLINE | ID: mdl-28233032

OBJECTIVE: Here, we present the data indicating that chronic treatment with fluoxetine regulates Cav-1/PTEN/PI3K/AKT/GSK-3ß signalling pathway and glycogen content in primary cultures of astrocytes with bi-phasic concentration dependence. RESULTS: At lower concentrations, fluoxetine downregulates gene expression of Cav-1, decreases membrane content of PTEN, increases activity of PI3K/AKT, and elevates GSK-3ß phosphorylation thus suppressing its activity. At higher concentrations, fluoxetine acts in an inverse fashion. As expected, fluoxetine at lower concentrations increased while at higher concentrations decreased glycogen content in astrocytes. CONCLUSIONS: Our findings indicate that bi-phasic regulation of glycogen content via Cav-1/PTEN/PI3K/AKT/GSK-3ß pathway by fluoxetine may be responsible for both therapeutic and side effects of the drug.


Astrocytes/drug effects , Astrocytes/metabolism , Caveolin 1/drug effects , Fluoxetine/pharmacology , Glycogen Synthase Kinase 3 beta/drug effects , Glycogen/metabolism , PTEN Phosphohydrolase/drug effects , Phosphatidylinositol 3-Kinases/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Signal Transduction/drug effects , Animals , Class I Phosphatidylinositol 3-Kinases , Gene Knockdown Techniques , Mice , Phosphorylation , Primary Cell Culture , RNA, Small Interfering/pharmacology
10.
Diabetes Obes Metab ; 18(12): 1236-1243, 2016 12.
Article En | MEDLINE | ID: mdl-27460695

AIM: To test the effect of linagliptin in non-obese diabetic (NOD) mice, a murine model of type 1 diabetes, to unveil a possible direct cardiovascular action of dipeptidyl peptidase 4 (DPP-4) inhibitors beyond glycaemia control. METHODS: NOD mice were grouped according to glycosuria levels as NODI: none; NODII: high; NODIII: severe. Linagliptin treatment was initiated once they reached NODII levels. Vascular reactivity was assessed ex vivo on aorta harvested from mice upon reaching NODIII level. In a separate set of experiments, the effect of linagliptin was tested directly in vitro on vessels harvested from untreated NODIII, glucagon-like peptide-1 (GLP-1) receptor knockout and soluble guanylyl cyclase-α1 knockout mice. Molecular and cellular studies were performed on endothelial and endothelial nitric oxide synthase (eNOS)-transfected cells. RESULTS: In this ex vivo vascular study, endothelium-dependent vasorelaxation was ameliorated and eNOS/nitric oxide (NO)/soluble guanylyl cyclase (sGC) signalling was enhanced. In the in vitro vascular study, linagliptin exerted a direct vasodilating activity on vessels harvested from both normo- or hyperglycaemic mice. The effect was independent from GLP-1/GLP-1 receptor (GLP-1R) interaction and required eNOS/NO/sGC pathway activation. Molecular studies performed on endothelial cells show that linagliptin rescues eNOS from caveolin-1 (CAV-1)-binding in a calcium-independent manner. CONCLUSION: Linagliptin, by interfering with the protein-protein interaction CAV-1/eNOS, led to an increased eNOS availability, thus enhancing NO production. This mechanism accounts for the vascular effect of linagliptin that is independent from glucose control and GLP-1/GLP-1R interaction.


Aorta/drug effects , Caveolin 1/drug effects , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Linagliptin/pharmacology , Nitric Oxide Synthase Type III/drug effects , Vasodilation/drug effects , Animals , Blood Glucose/metabolism , Blood Vessels/drug effects , Caveolin 1/metabolism , Glucagon-Like Peptide-1 Receptor/genetics , Mice , Mice, Inbred NOD , Mice, Knockout , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Signal Transduction , Soluble Guanylyl Cyclase/genetics
11.
Future Microbiol ; 11: 1227-1248, 2016 10.
Article En | MEDLINE | ID: mdl-26986451

AIM: To investigate the detailed mechanism of Japanese encephalitis virus (JEV) cell entry. MATERIALS & METHODS: Utilize a siRNA library targeting cellular membrane trafficking genes to identify key molecules that mediate JEV entry into human neuronal cells. RESULTS: JEV enters human neuronal cells by caveolin-1-mediated endocytosis, which depends on a two-step regulation of actin cytoskeleton remodeling triggered by RhoA and Rac1: RhoA activation promoted the phosphorylation of caveolin-1, and then Rac1 activation facilitated caveolin-associated viral internalization. Specifically, virus attachment activates the EGFR-PI3K signaling pathway, thereby leading to RhoA activation. CONCLUSION: This work provides a detailed picture of the entry route and intricate cellular events following the entry of JEV into human neuronal cells, and promotes a better understanding of JEV entry.


Actin Cytoskeleton/metabolism , Actin Cytoskeleton/virology , Caveolin 1/metabolism , Encephalitis Virus, Japanese/metabolism , Encephalitis Virus, Japanese/physiology , Virus Internalization/drug effects , Actin Cytoskeleton/drug effects , Actin Depolymerizing Factors/metabolism , Actin Depolymerizing Factors/pharmacology , Animals , Caveolin 1/drug effects , Caveolin 1/genetics , Cell Line , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Membrane/virology , Cholesterol/metabolism , Cricetinae , Dynamin II/genetics , Dynamin II/metabolism , Encephalitis Virus, Japanese/genetics , Encephalitis Virus, Japanese/pathogenicity , Encephalitis, Japanese/virology , Endocytosis/physiology , ErbB Receptors/metabolism , HEK293 Cells , Host-Pathogen Interactions/physiology , Humans , Life Cycle Stages/physiology , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Neurons/virology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/pharmacology , RNA, Small Interfering/genetics , Signal Transduction , Transfection , Virus Attachment , rac1 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/pharmacology , rhoA GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/pharmacology
12.
Am J Physiol Endocrinol Metab ; 310(3): E225-37, 2016 Feb 01.
Article En | MEDLINE | ID: mdl-26646098

Salidroside (SAL) is a phenylethanoid glycoside isolated from the medicinal plant Rhodiola rosea. R. rosea has been reported to have beneficial effects on diabetic nephropathy (DN) and high-glucose (HG)-induced mesangial cell proliferation. Given the importance of caveolin-1 (Cav-1) in transcytosis of albumin across the endothelial barrier, the present study was designed to elucidate whether SAL could inhibit Cav-1 phosphorylation and reduce the albumin transcytosis across glomerular endothelial cells (GECs) to alleviate diabetic albuminuria as well as to explore its upstream signaling pathway. To assess the therapeutic potential of SAL and the mechanisms involved in DN albuminuria, we orally administered SAL to db/db mice, and the effect of SAL on the albuminuria was measured. The albumin transcytosis across GECs was explored in a newly established in vitro cellular model. The ratio of albumin to creatinine was significantly reduced upon SAL treatment in db/db mice. SAL decreased the albumin transcytosis across GECs in both normoglycemic and hyperglycemic conditions. SAL reversed the HG-induced downregulation of AMP-activated protein kinase and upregulation of Src kinase and blocked the upregulation Cav-1 phosphorylation. Meanwhile, SAL decreased mitochondrial superoxide anion production and moderately depolarized mitochondrial membrane potential. We conclude that SAL exerts its proteinuria-alleviating effects by downregulation of Cav-1 phosphorylation and inhibition of albumin transcytosis across GECs. These studies provide the first evidence of interference with albumin transcytosis across GECs as a novel approach to the treatment of diabetic albuminuria.


Albumins/drug effects , Albuminuria/metabolism , Caveolin 1/drug effects , Diabetic Nephropathies/metabolism , Endothelial Cells/drug effects , Glucosides/pharmacology , Kidney Glomerulus/drug effects , Phenols/pharmacology , Transcytosis/drug effects , AMP-Activated Protein Kinases/drug effects , AMP-Activated Protein Kinases/metabolism , Albumins/metabolism , Animals , Caveolin 1/metabolism , Creatinine/metabolism , Endothelial Cells/metabolism , Glucose/metabolism , In Vitro Techniques , Kidney Glomerulus/metabolism , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Mice , Phosphorylation/drug effects , src-Family Kinases/drug effects , src-Family Kinases/metabolism
14.
Am J Respir Crit Care Med ; 191(11): 1273-86, 2015 Jun 01.
Article En | MEDLINE | ID: mdl-25853696

RATIONALE: Pulmonary arterial hypertension is characterized by endothelial dysfunction, impaired bone morphogenetic protein receptor 2 (BMPR2) signaling, and increased elastase activity. Synthetic elastase inhibitors reverse experimental pulmonary hypertension but cause hepatotoxicity in clinical studies. The endogenous elastase inhibitor elafin attenuates hypoxic pulmonary hypertension in mice, but its potential to improve endothelial function and BMPR2 signaling, and to reverse severe experimental pulmonary hypertension or vascular pathology in the human disease was unknown. OBJECTIVES: To assess elafin-mediated regression of pulmonary vascular pathology in rats and in lung explants from patients with pulmonary hypertension. To determine if elafin amplifies BMPR2 signaling in pulmonary artery endothelial cells and to elucidate the underlying mechanism. METHODS: Rats with pulmonary hypertension induced by vascular endothelial growth factor receptor blockade and hypoxia (Sugen/hypoxia) as well as lung organ cultures from patients with pulmonary hypertension were used to assess elafin-mediated reversibility of pulmonary vascular disease. Pulmonary arterial endothelial cells from patients and control subjects were used to determine the efficacy and mechanism of elafin-mediated BMPR2 signaling. MEASUREMENTS AND MAIN RESULTS: In Sugen/hypoxia rats, elafin reduced elastase activity and reversed pulmonary hypertension, judged by regression of right ventricular systolic pressure and hypertrophy and pulmonary artery occlusive changes. Elafin improved endothelial function by increasing apelin, a BMPR2 target. Elafin induced apoptosis in human pulmonary arterial smooth muscle cells and decreased neointimal lesions in lung organ culture. In normal and patient pulmonary artery endothelial cells, elafin promoted angiogenesis by increasing pSMAD-dependent and -independent BMPR2 signaling. This was linked mechanistically to augmented interaction of BMPR2 with caveolin-1 via elafin-mediated stabilization of endothelial surface caveolin-1. CONCLUSIONS: Elafin reverses obliterative changes in pulmonary arteries via elastase inhibition and caveolin-1-dependent amplification of BMPR2 signaling.


Bone Morphogenetic Protein Receptors, Type II/drug effects , Caveolin 1/drug effects , Elafin/pharmacology , Hypertension, Pulmonary/drug therapy , Protease Inhibitors/pharmacology , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Humans , Myocytes, Smooth Muscle/drug effects , Pancreatic Elastase/drug effects , Rats
15.
Acta Neurochir (Wien) ; 156(11): 2103-9, 2014 Nov.
Article En | MEDLINE | ID: mdl-25139403

BACKGROUND: Recent evidence has demonstrated that rosiglitazone can attenuate cerebral vasospasm following subarachnoid hemorrhage (SAH). Some studies have shown that rosiglitazone can suppress inflammation and immune responses after SAH. However, the precise molecular mechanisms by which cerebral vasospasm is attenuated is not clear. METHODS: In this study, SAH was created using a "double hemorrhage" injection rat model. Rats were randomly divided into three groups and treated with saline (control group), untreated (SAH group), or treated with rosiglitazone. Using immunocytochemistry, hematoxylin and eosin (HE) staining, and measurement of the basilar artery, we investigated the formation of pathologic changes in the basilar artery, measured the expression of caveolin-1 and proliferating cell nuclear antigen (PCNA), and investigated the role of rosiglitazone in vascular smooth muscle cell (VSMC) proliferation in the basilar artery after SAH. RESULTS: In this study, we observed significant pathologic changes in the basilar artery after experimental SAH. The level of vasospasm gradually increased with time during the 1st week, peaked on day 7, and almost recovered on day 14. After rosiglitazone treatment, the level of vasospasm was significantly attenuated in comparison with the SAH group. Immunocytochemistry staining showed that caveolin-1 expression was significantly increased in the rosiglitazone group, compared with the SAH group. Inversely, the expression of PCNA showed a notable decrease after rosiglitazone treatment. CONCLUSIONS: The results indicate that rosiglitazone can attenuate cerebral vasospasm following SAH. Up-regulation of caveolin-1 by rosiglitazone may be a new molecular mechanism for this response, which is to inhibit proliferation of VSMCs after SAH, and this study may provide a novel insight to prevent delayed cerebral vasospasm (DCVS).


Basilar Artery/drug effects , Cell Proliferation/drug effects , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Subarachnoid Hemorrhage/complications , Thiazolidinediones/pharmacology , Vasoconstriction/drug effects , Vasodilator Agents/pharmacology , Vasospasm, Intracranial/etiology , Animals , Basilar Artery/pathology , Caveolin 1/drug effects , Caveolin 1/metabolism , Disease Models, Animal , Immunohistochemistry , Male , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Proliferating Cell Nuclear Antigen/drug effects , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Sprague-Dawley , Rosiglitazone , Subarachnoid Hemorrhage/pathology , Subarachnoid Hemorrhage/physiopathology , Up-Regulation , Vasospasm, Intracranial/physiopathology , Vasospasm, Intracranial/prevention & control
16.
Am J Obstet Gynecol ; 211(5): 496.e1-496.e13, 2014 Nov.
Article En | MEDLINE | ID: mdl-24909339

OBJECTIVE: Uterine leiomyomas, the most common neoplasms of the female genital tract, are benign tumors of the uterus arising from the smooth muscle cells (SMCs) of the myometrium with an involvement of estrogen. Caveolin-1 (Cav-1), a major protein component in caveolae membrane lipid rafts, is down-regulated in several estrogen-related cancer cells, and overexpression of Cav-1 inhibits proliferation of cancer cells and vascular SMCs as well. Therefore, we hypothesize that Cav-1 is down-regulated in human uterine leiomyoma. RESULTS: Western blot using tissues from clinical patients showed that Cav-1 expression was significantly lower or undetectable in uterine leiomyoma compared with their matched myometrium (P < .001). This finding was confirmed by immunohistochemistry and confocal microscopy. The cav-1 mRNA level in uterine leiomyomas was also significantly lower as detected by reverse transcription-quantitative polymerase chain reaction analysis (P = .001). To further study the underlying mechanism, we performed primary cell culture, and found that the expression of Cav-1 remained low in cultured leiomyoma SMCs (P = .009). Serum withdrawal did not change Cav-1 expression in leiomyoma SMCs, but increased expression in myometrial SMCs (P = .006). 17-ß estradiol inhibited the expression of Cav-1 protein (P = .047) and mRNA (P = .007) in leiomyoma SMCs, whereas it stimulated expression in myometrial SMCs (P = .043). 17-ß estradiol, although activating the mitogen-activated protein kinase pathway in both SMCs, did not stimulate their proliferation. CONCLUSION: We conclude that human uterine leiomyomas in vitro express low levels of Cav-1, which may result from estrogen inhibition. This effect of estrogen may contribute to the pathogenesis of uterine leiomyoma. Further studies in vivo are needed to verify these results.


Caveolin 1/genetics , Leiomyoma/genetics , Myocytes, Smooth Muscle/metabolism , Myometrium/metabolism , RNA, Messenger/genetics , Uterine Neoplasms/genetics , Adult , Blotting, Western , Caveolin 1/drug effects , Caveolin 1/metabolism , Cells, Cultured , Estradiol/pharmacology , Estrogens/pharmacology , Female , Gene Expression Profiling , Humans , Leiomyoma/metabolism , Middle Aged , Myocytes, Smooth Muscle/drug effects , Myometrium/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Uterine Neoplasms/metabolism
17.
Cardiovasc J Afr ; 25(5): 212-6, 2014.
Article En | MEDLINE | ID: mdl-25629537

AIM: Endothelial dysfunction, oxidative stress and inflammation are among the most important mechanisms of ischaemia-reperfusion (I/R) injury. Besides their cholesterol-lowering effects, statins are known to provide protection against myocardial dysfunction and vascular endothelial injury via nitric oxide-dependent mechanisms. The aim of this study was to investigate the effects of rosuvastatin on certain intermediates involved in the generation of nitric oxide (asymmetrical dimethyl arginin, ADMA, caveolin-1 and hsp 90), oxidative stress (rhokinase, NADPH oxidase) and inflammation (NFkB), using an in vivo model of myocardial infarction in the rat. METHODS: Adult male Sprague Dawley rats were divided into three groups (control, I/R and I/R after 15 days of rosuvastatin administration). Reperfusion was applied for 120 min following left anterior descending coronary artery ischaemia for 30 min. Caveolin-1, hsp 90 and NFkB levels were evaluated with the quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) and ADMA, rhokinase and NADPH oxidase levels were evaluated with ELISA. RESULTS: While NFkB and hsp 90 levels were higher in the I/R group, their levels were significantly lower in the rosuvastatin group. While ADMA and NADPH oxidase levels significantly increased with I/R, they were lower in the rosuvastatin-treated group, but not statistically significant. Rhokinase levels were significantly lower in the rosuvastatin group. Caveolin-1 levels were not different between the groups. CONCLUSION: Our results suggest that ADMA, rhokinase, NADPH oxidase, hsp 90 and NFkB could facilitate I/R injury, and rosuvastatin significantly reduced levels of these parameters. These results indicate that rosuvastatin may have a protective role in I/R injury via mechanisms targeting inflammation, endothelial dysfunction and oxidative stress.


Fluorobenzenes/pharmacology , Heart/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Oxidative Stress/drug effects , Pyrimidines/pharmacology , RNA, Messenger/drug effects , Sulfonamides/pharmacology , Animals , Arginine/analogs & derivatives , Arginine/drug effects , Arginine/genetics , Arginine/metabolism , Caveolin 1/drug effects , Caveolin 1/genetics , Caveolin 1/metabolism , Disease Models, Animal , HSP90 Heat-Shock Proteins/drug effects , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Inflammation/genetics , Inflammation/metabolism , Myocardial Reperfusion Injury/genetics , NADPH Oxidases/drug effects , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , NF-kappa B/drug effects , NF-kappa B/genetics , NF-kappa B/metabolism , Nitric Oxide/metabolism , Oxidative Stress/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Rosuvastatin Calcium , rho-Associated Kinases/drug effects , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
18.
Asian Pac J Cancer Prev ; 14(9): 4983-8, 2013.
Article En | MEDLINE | ID: mdl-24175763

OBJECTIVES: To establish a taxol-resistant cell line of human ovarian carcinoma (A2780/Taxol) and investigate its biological features. METHODS: The drug-resistant cell line (A2780/Taxol) was established by continuous stepwise selection with increasing concentrations of Taxol. Cell morphology was assessed by microscopy and growth curves were generated with in vitro and in vivo tumor xenograft models. With rhodamine123 (Rh123) assays, cell cycle distribution and the apoptotic rate were analyzed by flow cytometry (FCM). Drug resistance-related and signal associated proteins, including P-gp, MRPs, caveolin-1, PKC-α, Akt, ERK1/2, were detected by Western blotting. RESULTS: A2780/Taxol cells were established with stable resistance to taxol. The drug resistance index (RI) was 430.7. Cross-resistance to other drugs was also shown, but there was no significant change to radioresistance. Compared with parental cells, A2780/Taxol cells were significantly heteromorphous, with a significant delay in population doubling time and reduced uptake of Rh123 (p < 0.01). In vivo, tumor take by A2780 cells was 80%, and tumor volume increased gradually. In contrast, with A2780/Taxol cells in xenograft models there was no tumor development. FCM analysis revealed that A2780/Taxol cells had a higher percentage of G0/G1 and lower S phase, but no changes of G2 phase and the apoptosis rate. Expression of P-gp, MRP1, MRP2, BCRP, LRP, caveolin-1, PKC-α, Phospho-ERK1/2 and Phospho-JNK protein was significantly up-regulated, while Akt and p38 MARK protein expression was not changed in A2780/Taxol cells. CONCLUSION: The A2780/Taxol cell line is an ideal model to investigate the mechanism of muti-drug resistance related to overexpression of drug-resistance associated proteins and activation of the PKC-α/ERK (JNK) signaling pathway.


Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma/genetics , Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Ovarian Neoplasms/genetics , Paclitaxel/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/drug effects , ATP-Binding Cassette Transporters/metabolism , Animals , Apoptosis/drug effects , Carcinoma/metabolism , Caveolin 1/drug effects , Caveolin 1/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Drug Resistance, Multiple/physiology , Drug Resistance, Neoplasm/physiology , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/drug effects , Multidrug Resistance-Associated Proteins/metabolism , Neoplasm Proteins/drug effects , Neoplasm Proteins/metabolism , Ovarian Neoplasms/metabolism , Protein Kinase C-alpha/drug effects , Protein Kinase C-alpha/metabolism , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Xenograft Model Antitumor Assays
19.
Am J Respir Crit Care Med ; 188(7): 831-41, 2013 Oct 01.
Article En | MEDLINE | ID: mdl-23924348

RATIONALE: Alveolar transforming growth factor (TGF)-ß1 signaling and expression of TGF-ß1 target genes are increased in patients with idiopathic pulmonary fibrosis (IPF) and in animal models of pulmonary fibrosis. Internalization and degradation of TGF-ß receptor TßRI inhibits TGF-ß signaling and could attenuate development of experimental lung fibrosis. OBJECTIVES: To demonstrate that after experimental lung injury, human syndecan-2 confers antifibrotic effects by inhibiting TGF-ß1 signaling in alveolar epithelial cells. METHODS: Microarray assays were performed to identify genes differentially expressed in alveolar macrophages of patients with IPF versus control subjects. Transgenic mice that constitutively overexpress human syndecan-2 in macrophages were developed to test the antifibrotic properties of syndecan-2. In vitro assays were performed to determine syndecan-2-dependent changes in epithelial cell TGF-ß1 signaling, TGF-ß1, and TßRI internalization and apoptosis. Wild-type mice were treated with recombinant human syndecan-2 during the fibrotic phase of bleomycin-induced lung injury. MEASUREMENTS AND MAIN RESULTS: We observed significant increases in alveolar macrophage syndecan-2 levels in patients with IPF. Macrophage-specific overexpression of human syndecan-2 in transgenic mice conferred antifibrotic effects after lung injury by inhibiting TGF-ß1 signaling and downstream expression of TGF-ß1 target genes, reducing extracellular matrix production and alveolar epithelial cell apoptosis. In vitro, syndecan-2 promoted caveolin-1-dependent internalization of TGF-ß1 and TßRI in alveolar epithelial cells, which inhibited TGF-ß1 signaling and epithelial cell apoptosis. Therapeutic administration of human syndecan-2 abrogated lung fibrosis in mice. CONCLUSIONS: Alveolar macrophage syndecan-2 exerts antifibrotic effects by promoting caveolin-1-dependent TGF-ß1 and TßRI internalization and inhibiting TGF-ß1 signaling in alveolar epithelial cells. Hence, molecules that facilitate TßRI degradation via endocytosis represent potential therapies for pulmonary fibrosis.


Idiopathic Pulmonary Fibrosis/chemically induced , Macrophages, Alveolar/drug effects , Syndecan-2/therapeutic use , Transforming Growth Factor beta1/drug effects , Animals , Apoptosis , Bleomycin/administration & dosage , Bronchoalveolar Lavage , Caveolin 1/drug effects , Disease Models, Animal , Gene Expression Profiling , Genetic Markers , Humans , Hydroxyproline/analysis , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/genetics , In Vitro Techniques , Mice , Mice, Transgenic , Signal Transduction , Syndecan-2/physiology , Tissue Array Analysis , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/physiology , Up-Regulation/drug effects , Up-Regulation/physiology
20.
J Biomed Biotechnol ; 2011: 625380, 2011.
Article En | MEDLINE | ID: mdl-22162638

We evaluated the in vitro effects of estradiol, progesterone, and testosterone on the molting process, which is the initial and crucial step in the development of the muscular larvae (ML or L1) to adult worm. Testosterone had no significative effect on the molting rate of the parasite, however, progesterone decreased the molting rate about a 50% in a concentration- and time-independent pattern, while estradiol had a slight effect (10%). The gene expression of caveolin-1, a specific gene used as a marker of parasite development, showed that progesterone and estradiol downregulated its expression, while protein expression was unaffected. By using flow citometry, a possible protein that is recognized by a commercial antiprogesterone receptor antibody was detected. These findings may have strong implications in the host-parasite coevolution, in the sex-associated susceptibility to this infection and could point out to possibilities to use antihormones to inhibit parasite development.


Gene Expression/physiology , Gonadal Steroid Hormones/metabolism , Helminthiasis/parasitology , Host-Parasite Interactions/physiology , Molting/physiology , Receptors, Progesterone/metabolism , Trichinella spiralis/growth & development , Animals , Caveolin 1/drug effects , Caveolin 1/metabolism , Down-Regulation/drug effects , Estradiol/metabolism , Estradiol/pharmacology , Female , Gene Expression/drug effects , Gonadal Steroid Hormones/pharmacology , Helminthiasis/drug therapy , Humans , Larva/drug effects , Larva/growth & development , Molting/drug effects , Progesterone/metabolism , Progesterone/pharmacology , Receptors, Progesterone/drug effects , Testosterone/metabolism , Testosterone/pharmacology , Trichinella spiralis/drug effects
...