scholarly journals Androgen Receptor Localizes to Plasma Membrane by Binding to Caveolin-1 in Mouse Sertoli Cells

2017 ◽  
Vol 2017 ◽  
pp. 1-8 ◽  
Author(s):  
Qiong Deng ◽  
Yong Wu ◽  
Zeng Zhang ◽  
Yue Wang ◽  
Minghua Li ◽  
...  

The nonclassical androgen signaling pathway translates signals into alterations in cellular function within minutes, and this action is proposed to be mediated by an androgen receptor (AR) localized to the plasma membrane. This study was designed to determine the mechanism underlying the membrane association of androgen receptor in TM4 cells, a mouse Sertoli cell line. Western blot analysis indicated testosterone-induced AR translocation to the cell membrane. Data from coimmunoprecipitation indicated that AR is associated with caveolin-1, and testosterone enhanced this association. Knockdown of caveolin-1 by shRNA decreased the amount of AR localized to membrane fraction and prevented AR membrane trafficking after being exposed to testosterone at physiological concentration. The palmitoylation inhibitor 2-bromopalmitate decreased AR membrane localization in basal condition and completely blocked testosterone-induced AR translocation to membrane fraction. These data suggested that AR localized to membrane fraction by binding with caveolin-1 through palmitoylation of the cysteine residue. This study provided a new evidence for AR membrane localization and its application for clarifying the nonclassical signaling pathway of androgens.

2018 ◽  
Vol 47 (6) ◽  
pp. 2360-2368 ◽  
Author(s):  
Qiong Deng ◽  
Chihua He ◽  
Yong Wu ◽  
Jianwen Zhang ◽  
Ying Zhang ◽  
...  

Background/Aims: Androgen and its receptor (AR) play an important role in maintaining spermatogenesis and male fertility. Our previous studies showed that testosterone at a physiological concentration induces cytoplasmic AR translocation to the Sertoli cell plasma membrane of within 5 minutes. Methods: In this study, mass spectrometry (MS) and bioinformatic analyses were applied to identify candidate proteins mediating AR trafficking. The candidate proteins were knocked down by shRNA transfection. Results: Nine candidate proteins were identified by MS. The data was verified by co-immunoprecipitation and Western blot. Of the candidates, CSN6 regulated AR transport through the phosphorylation signaling pathway and Rab34 affected AR trafficking by regulating Ras activity. Conclusions: CSN6 and Rab34 are involved in AR trafficking by regulating the phosphorylation signaling pathway. These findings provide new insights into the testosterone signaling pathway in Sertoli cells that mediates spermatogenesis.


2005 ◽  
Vol 16 (1) ◽  
pp. 231-237 ◽  
Author(s):  
Filippo Acconcia ◽  
Paolo Ascenzi ◽  
Alessio Bocedi ◽  
Enzo Spisni ◽  
Vittorio Tomasi ◽  
...  

A fraction of the nuclear estrogen receptor α (ERα) is localized to the plasma membrane region of 17β-estradiol (E2) target cells. We previously reported that ERα is a palmitoylated protein. To gain insight into the molecular mechanism of ERα residence at the plasma membrane, we tested both the role of palmitoylation and the impact of E2 stimulation on ERα membrane localization. The cancer cell lines expressing transfected or endogenous human ERα (HeLa and HepG2, respectively) or the ERα nonpalmitoylable Cys447Ala mutant transfected in HeLa cells were used as experimental models. We found that palmitoylation of ERα enacts ERα association with the plasma membrane, interaction with the membrane protein caveolin-1, and nongenomic activities, including activation of signaling pathways and cell proliferation (i.e., ERK and AKT activation, cyclin D1 promoter activity, DNA synthesis). Moreover, E2 reduces both ERα palmitoylation and its interaction with caveolin-1, in a time- and dose-dependent manner. These data point to the physiological role of ERα palmitoylation in the receptor localization to the cell membrane and in the regulation of the E2-induced cell proliferation.


2008 ◽  
Vol 182 (6) ◽  
pp. 1153-1169 ◽  
Author(s):  
Ting Cai ◽  
Haojie Wang ◽  
Yiliang Chen ◽  
Lijun Liu ◽  
William T Gunning ◽  
...  

Here, we show that the Na/K-ATPase interacts with caveolin-1 (Cav1) and regulates Cav1 trafficking. Graded knockdown of Na/K-ATPase decreases the plasma membrane pool of Cav1, which results in a significant reduction in the number of caveolae on the cell surface. These effects are independent of the pumping function of Na/K-ATPase, and instead depend on interaction between Na/K-ATPase and Cav1 mediated by an N-terminal caveolin-binding motif within the ATPase α1 subunit. Moreover, knockdown of the Na/K-ATPase increases basal levels of active Src and stimulates endocytosis of Cav1 from the plasma membrane. Microtubule-dependent long-range directional trafficking in Na/K-ATPase–depleted cells results in perinuclear accumulation of Cav1-positive vesicles. Finally, Na/K-ATPase knockdown has no effect on processing or exit of Cav1 from the Golgi. Thus, the Na/K-ATPase regulates Cav1 endocytic trafficking and stabilizes the Cav1 plasma membrane pool.


2003 ◽  
Vol 278 (29) ◽  
pp. 27208-27215 ◽  
Author(s):  
So-ching W. Brazer ◽  
Brij B. Singh ◽  
Xibao Liu ◽  
William Swaim ◽  
Indu S. Ambudkar

2007 ◽  
Vol 330 (2) ◽  
pp. 231-245 ◽  
Author(s):  
Valentina Kubale ◽  
Zrinka Abramović ◽  
Azra Pogačnik ◽  
Anders Heding ◽  
Marjeta Šentjurc ◽  
...  

2018 ◽  
Vol 2018 ◽  
pp. 1-6 ◽  
Author(s):  
Qiong Deng ◽  
Jianwen Zhang ◽  
Zhu Wang ◽  
Shengping Zhang ◽  
Fan Zhi ◽  
...  

Androgen and its receptor (AR) play an important role in maintaining spermatogenesis and male fertility. The nonclassical androgen signaling pathway is proposed to be mediated by an AR in plasma membrane in Sertoli cells. Our previous studies showed that testosterone induces cytoplasmic AR translocation to plasma membrane by binding with caveolin-1. This study was conducted to the underlying molecular mechanism mediating AR trafficking. Data from mass spectrometry using membrane coimmunoprecipitation sample by anti-AR antibody indicated VAPA is a candidate protein. Knockdown of VAPA by shRNA decreased the amount of AR localized to membrane and nuclear fraction and prevented AR trafficking after being exposed to testosterone. Further studies indicated AR trafficking in Sertoli cells might be mediated by VAPA via association with vesicle transport protein OSBP. This study can enrich the mechanism of the androgen actions and will be helpful for further clarifying the nonclassical signaling pathway of androgens in Sertoli cells.


2020 ◽  
Vol 41 (Supplement_2) ◽  
Author(s):  
S Tomita ◽  
N Nakanishi ◽  
T Ogata ◽  
Y Tsuji ◽  
A Sakamoto ◽  
...  

Abstract Background Pulmonary hypertension (PH) is a progressive disease associated with poor outcome. Caveolin-1 (Cav1) is a component of caveolae and classified as a related gene of pulmonary arterial hypertension (PAH). Gene mutations of bone morphogenetic protein type II receptor (BMPRII) is a most common cause of PAH. BMPRII is localized in caveolae and associates with Cav1. However, the role of the Caveolin-Cavin system on the BMP/Smad signaling and the PAH progression has not been well-known. Purpose The aim of our study is to investigate the relationship between Caveolin-Cavin system and BMP/Smad signaling pathway and explore the mechanism of downstream signal transduction of BMP signaling by the interaction between Caveolin and BMPRII. Methods Cav1 knockout mice were used to assess PH and caveolae in pulmonary artery endothelial cells were observed by electron microscope. Cav1 and Cavin-1, which is a component of caveolae and form a complex with Cav1, were knocked-down in human pulmonary artery endothelial cell (hPAEC) using siRNA and phosphorylation of Smad signal was evaluated. Apoptosis of these cells was explored by flow cytometry. We investigated the interaction between Cav1 and BMPRII, and evaluated whether Cavin-1 affects this interaction and signal transduction of BMP signaling. Results As previously described, deletion of Cav1 revealed disappearance of caveolae in pulmonary artery endothelial cells (PAECs), and Cav1 knockout mice exhibited PH with pulmonary vascular remodeling and right ventricular hypertrophy. We then examined roles of Cav1 in human PAECs (hPAECs). Cav1 knockdown in hPAECs reduced phosphorylation of Smad 1/5/9. In addition, Cav1 knockdown significantly increased hypoxia-induced apoptosis in hPAEC. Knockdown of Cavin-1 reversed phosphorylation of Smad 1/5/9 decreased by Cav1 knockdown in BMP9 stimulation. Cavin-1 reversed the expression of BMPRII decreased by overexpression of Cav1. Cav1 was associated with Cavin-1 at the plasma membrane in PAECs. Cav1 also associated with BMPRII at the membrane of hPAECs that was inhibited by Cavin-1, and Cavin-1 reduced the localization of BMPRII to the membrane of hPAECs. These results suggest that BMPRII interacts with Cav1 via Cavin-1-associated localization at the plasma membrane in hPAECs, resulting in regulating BMP/Smad signaling pathway and involving in the development of PAH. Conclusions Cavin-1 affects the interaction of Cav1 with BMPRII at the membrane of PAECs, and regulates BMP/Smad signaling. These results reveal a previously undescribed function of Cavin-Caveolin system in the development of PAH through regulation of BMP/Smad signaling. Funding Acknowledgement Type of funding source: None


2015 ◽  
Vol 89 (16) ◽  
pp. 8162-8181 ◽  
Author(s):  
Audrey Thomas ◽  
Charlotte Mariani-Floderer ◽  
Maria Rosa López-Huertas ◽  
Nathalie Gros ◽  
Elise Hamard-Péron ◽  
...  

ABSTRACTDuring HIV-1 assembly, the Gag viral proteins are targeted and assemble at the inner leaflet of the cell plasma membrane. This process could modulate the cortical actin cytoskeleton, located underneath the plasma membrane, since actin dynamics are able to promote localized membrane reorganization. In addition, activated small Rho GTPases are known for regulating actin dynamics and membrane remodeling. Therefore, the modulation of such Rho GTPase activity and of F-actin by the Gag protein during virus particle formation was considered. Here, we studied the implication of the main Rac1, Cdc42, and RhoA small GTPases, and some of their effectors, in this process. The effect of small interfering RNA (siRNA)-mediated Rho GTPases and silencing of their effectors on Gag localization, Gag membrane attachment, and virus-like particle production was analyzed by immunofluorescence coupled to confocal microscopy, membrane flotation assays, and immunoblot assays, respectively. In parallel, the effect of Gag expression on the Rac1 activation level was monitored by G-LISA, and the intracellular F-actin content in T cells was monitored by flow cytometry and fluorescence microscopy. Our results revealed the involvement of activated Rac1 and of the IRSp53-Wave2-Arp2/3 signaling pathway in HIV-1 Gag membrane localization and particle release in T cells as well as a role for actin branching and polymerization, and this was solely dependent on the Gag viral protein. In conclusion, our results highlight a new role for the Rac1-IRSp53-Wave2-Arp2/3 signaling pathway in the late steps of HIV-1 replication in CD4 T lymphocytes.IMPORTANCEDuring HIV-1 assembly, the Gag proteins are targeted and assembled at the inner leaflet of the host cell plasma membrane. Gag interacts with specific membrane phospholipids that can also modulate the regulation of cortical actin cytoskeleton dynamics. Actin dynamics can promote localized membrane reorganization and thus can be involved in facilitating Gag assembly and particle formation. Activated small Rho GTPases and effectors are regulators of actin dynamics and membrane remodeling. We thus studied the effects of the Rac1, Cdc42, and RhoA GTPases and their specific effectors on HIV-1 Gag membrane localization and viral particle release in T cells. Our results show that activated Rac1 and the IRSp53-Wave2-Arp2/3 signaling pathway are involved in Gag plasma membrane localization and viral particle production. This work uncovers a role for cortical actin through the activation of Rac1 and the IRSp53/Wave2 signaling pathway in HIV-1 particle formation in CD4 T lymphocytes.


2003 ◽  
Vol 23 (5) ◽  
pp. 1633-1646 ◽  
Author(s):  
Mahnaz Razandi ◽  
Gordon Alton ◽  
Ali Pedram ◽  
Sanjiv Ghonshani ◽  
Paul Webb ◽  
...  

ABSTRACT Estrogen receptors (ER) have been localized to the cell plasma membrane (PM), where signal transduction mediates some estradiol (E2) actions. However, the precise structural features of ER that result in membrane localization have not been determined. We obtained a partial tryptic peptide/mass spectrometry analysis of membrane mouse ERα protein. Based on this, we substituted alanine for the determined serine at amino acid 522 within the E domain of wild-type (wt) ERα. Upon transfection in CHO cells, the S522A mutant ERα resulted in a 62% decrease in membrane receptor number and reduced colocalization with caveolin 1 relative to those with expression of wt ERα. E2 was significantly less effective in stimulating multiple rapid signals from the membranes of CHO cells expressing ERα S522A than from those of CHO cells expressing wt ERα. In contrast, nuclear receptor expression and transcriptional function were very similar. The S522A mutant was also 60% less effective than wt ERα in binding caveolin 1, which facilitates ER transport to the PM. All functions of ERα mutants with other S-to-A substitutions were comparable to those of wt ER, and deletion of the A/B or C domain had little consequence for membrane localization or function. Transfection of ERα S522A into breast cancer cells that express native ER downregulated E2 binding at the membrane, signaling to ERK, and G1/S cell cycle events and progression. However, there was no effect on the E2 transactivation of an ERE-luciferase reporter. In summary, serine 522 is necessary for the efficient translocation and function of ERα at the PM. The S522A mutant also serves as a dominant-negative construct, identifying important functions of E2 that originate from activating PM ER.


2002 ◽  
Vol 115 (15) ◽  
pp. 3119-3130 ◽  
Author(s):  
Inmaculada Navarro-Lérida ◽  
Alberto Álvarez-Barrientos ◽  
Francisco Gavilanes ◽  
Ignacio Rodriguez-Crespo

Using recursive PCR, we created an artificial protein sequence that consists of a consensus myristoylation motif (MGCTLS) followed by the triplet AGS repeated nine times and fused to the GFP reporter. This linker-GFP sequence was utilized as a base to produce multiple mutants that were used to transfect COS-7 cells. Constructs where a `palmitoylable' cysteine residue was progressively moved apart from the myristoylation site to positions 3, 9, 15 and 21 of the protein sequence were made, and these mutants were used to investigate the effect of protein myristoylation on subsequent palmitoylation,subcellular localization, membrane association and caveolin-1 colocalization. In all cases, dual acylation of the GFP chimeras correlated with translocation to Triton X-100-insoluble cholesterol/sphingomyelin-enriched subdomains. Whereas a strong Golgi labeling was observed in all the myristoylated chimeras, association with the plasma membrane was only observed in the dually acylated constructs. Taking into account the conflicting data regarding the existence and specificity of cellular palmitoyl-transferases, our results provide evidence that de-novo-designed sequences can be efficiently S-acylated with palmitic acid in vivo, strongly supporting the hypothesis that non-enzymatic protein palmitoylation can occur within mammalian cells. Additionally, this palmitoylation results in the translocation of the recombinant construct to low-fluidity domains in a myristate-palmitate distance-dependent manner.


Sign in / Sign up

Export Citation Format

Share Document