Increased proliferation of human bladder smooth muscle cells is mediated by physiological cyclic stretch via the PI3K‑SGK1‑Kv1.3 pathway

  • Authors:
    • Ye Tian
    • Xuan Yue
    • Deyi Luo
    • Romel Wazir
    • Jianzhong Wang
    • Tao Wu
    • Lin Chen
    • Banghua Liao
    • Kunjie Wang
  • View Affiliations

  • Published online on: May 13, 2013     https://doi.org/10.3892/mmr.2013.1473
  • Pages: 294-298
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

It is well known that specific mechanical stimuli induce positive changes in the physiological function and status of a number of cell types. However, an in‑depth understanding of the application of mechanical forces has yet to be developed. The aim of the present study was to explore the optimal elongation and frequency of stretch‑induced proliferation of human bladder smooth muscle cells (HBSMCs) and to investigate the mechanism involved in this process. HBSMCs were seeded in a silicone membrane and subjected to cyclic stretch of 2.5, 5, 10 and 15% equibiaxial elongation at frequencies of 0.05, 0.1, 0.2, 0.5 and 1 Hz, respectively. Bromodeoxyuridine (BrdU) assays were used to detect the proliferative activity of each group. To further determine the mechanism of the cell proliferation process triggered by physiological cyclic stretch, the expression of PI3K/SGK1/Akt/Kv1.3 was investigated at the transcriptional and translational levels by RT‑PCR and western blot analysis, respectively. Optimal physiological stretch was established as 5% elongation at a frequency of 0.1 Hz, whereby HBSMCs revealed a marked increase in proliferative activity compared with the other groups, including the non‑stretched group, which served as the control (P<0.05). The expression of PI3K/SGK1/Kv1.3; however, not Akt, were upregulated by cyclic stretch as compared with the control group. When separately treated with inhibitors of SGK1 and Kv1.3, increased stretch‑induced proliferation was largely eliminated. These results markedly indicate that cyclic stretch induces the proliferation of HBSMCs and the PI3K‑SGK1‑Kv1.3 pathway is involved in this process, either fully or at least partially, rather than its related pathway, PI3K‑Akt.

Introduction

Worldwide, patients with end-stage bladder disease are treated with cystoplasty using their own gastrointestinal segments; however, serious complications are attributable to the absorptive, mucus-secreting epithelial lining in the urinary tracts. Practical and functional tissue engineering of the bladder represents an ideal substitute and a number of studies have focused on this process in previous years (1,2). However, simple accumulation of the cells and matrix does not generate satisfactory results (3) and organic combinations of these cells and the scaffolds [extracellular matrix (ECM)] remain to be identified.

It is well documented that appropriate mechanical stimulation is critical for gene expression and cell proliferation, differentiation, migration, function optimization and production of structurally suitable ECM components (49). Human bladder smooth muscle cells (HBSMCs) are constantly subjected to mechanical stimuli, including hydrodynamic pressure and stretch, during filling and voiding cycles. Our previous studies indicated that HBSMC proliferation is stimulated by hydrodynamic pressure (10,11). However, to the best of our knowledge, stretch, which is considerably more important than hydrostatic pressure in physiological conditions, is far from well explored and the mechanisms by which HBSMCs perceive exterior mechanical stimulation remain poorly defined. It is well known that all cellular life recognizes and responds to stimuli from the extracellular environment. Environmental sensing at the cellular level relies on signal transduction involving the binding of extracellular signaling molecules and ligands to cell surface receptors that trigger events inside the cell (12). Elucidation of the interactions between physiological cyclic stretch and these signal transduction pathways may be beneficial or even fundamental to functional tissue engineering of the urinary bladder, and have important implications for the development of interventions for cell remodeling diseases, including incontinence, overactive bladder and bladder outlet obstruction (BOO) (1315). The PI3K pathway is one of the most common physiological and pathological pathways, and is involved in a number of processes, including cell proliferation, metabolism, survival and tumorigenesis. AKT and SGK1 are related downstream effectors of the PI3K cascade, sharing similar downstream targets and 45–55% homology in their catalytic domains (1618). For a specific type of cell, which of the ‘two sisters’ is responsible for the proliferation remains controversial (1924). Cell proliferation requires an increase in the expression and function of potassium (K+) channels. Blockade of K+ channels inhibits the proliferation of a number of cell types (23,25,26). The Kv1.3 channel represents a novel target for vascular diseases due to its important relationship with the proliferation of vascular smooth muscle cells (27). However, the regulation of HBSMC proliferation by cyclic stretch and the mechanism of this process remains undefined.

Based on these previous studies (10), the aim of the present study was to explore the correlation between appropriate cyclic stretch and HBSMC proliferation, and furthermore, to identify changes in PI3K, SGK1, AKT and Kv1.3 expression and activity during regulated proliferation induced by cyclic stretch. The results indicate that HBSMC proliferative activity is upregulated by physiological cyclic stretch and when the stretch was applied by 5% elongation and 0.1 Hz, HBSMCs were demonstrated to exhibit maximum proliferative activity. Expression of PI3K, SGK1 and Kv1.3 was observed to be significantly increased. By contrast, AKT expression was unchanged. Increased proliferative activity was eliminated following blockade of SGK1 and Kv1.3. In addition, increased Kv1.3 expression was downregulated following blockade of SGK1, indicating that the PI3K-SGK1-Kv1.3 pathway is involved, at least in part, in HBSMC proliferation induced by cyclic stretch.

Materials and methods

Cell culture and identification

HBSMCs (ScienCell, Carlsbad, CA, USA; cat. no. 4310) were grown and maintained at 37°C, in a 5% CO2/95% air gas mixture and humidified atmosphere in a cell incubator. Dulbecco’s modified Eagle’s medium (DMEM; low glucose) was supplemented with 10% fetal bovine serum (both Hyclone Laboratories, Inc., Logan, UT, USA), penicillin (100 U/ml) and streptomycin (100 μg/ml). All experiments were performed on cells between passages 3 and 7 with normal morphology and good activity, which were confirmed by immunocytochemistry (data not shown).

Application of cyclic stretch

HBSMCs were seeded onto a silicone membrane for 24 h and then subjected to cyclic stretch. Stretch was applied based on the physiological bladder cycles using the Bose BioDynamic (Bose Corporation, Eden Prairie, MN, USA). In the first 3 h, elongation of stretch rose gradually from 0 to 2.5% and in the next 1 h, stretch rose from 2.5 to 5, 10 or 15%, or was maintained at 2.5%. Following a rapid decrease, this 4-h stretch cycle was repeated 4 times. Next, the silicone membrane was maintained in a relaxed state for 8 h (simulation of bladder cycles during night time). After applied elongations were divided into groups, each group was subjected to 0.05, 0.1, 0.2, 0.5 and 1 Hz with a sine wave stretch pattern by a 1:1 stretch/relaxation ratio (see Table I).

Table I

Absorbance values of BrdU incorporation.

Table I

Absorbance values of BrdU incorporation.

Frequency, Hz0% (control)2.5%5%10%15%
0.050.826±0.019a0.965±0.0351.264±0.0291.281±0.0531.235±0.026
0.101.161±0.0331.460±0.015b1.330±0.0281.226±0.045
0.201.112±0.0231.213±0.0201.204±0.0171.178±0.036
0.501.095±0.0191.240±0.0521.018±0.0251.014±0.041
1.001.048±0.0311.074±0.0740.974±0.0280.981±0.016

a P<0.05, vs. stretch;

b P<0.05, vs. other groups.

{ label (or @symbol) needed for fn[@id='tfn3-mmr-08-01-0294'] } Optimal stretch model for HBSMC proliferation was established. Results of four independent BrdU incorporation assays in each group with various elongations and frequencies 30 min following the addition of anti-BrdU antibody. HBSMC, human bladder smooth muscle cell; BrdU, bromodeoxyuridine.

Proliferation studies

Bromodeoxyuridine (BrdU) incorporation (Roche Diagnostics GmbH, Mannheim, Germany) was employed as a direct parameter of DNA synthesis to quantify cell proliferation according to the manufacturer’s instructions. Briefly, HBSMCs from each group were harvested and suspended at a concentration of 4×105 cells/ml in DMEM. The cell suspension was transferred into a 96-well plate, with 200 μl in each well. BrdU labeling reagent (final concentration, 10 μM) was added and the cells were reincubated for 3 h. Following centrifugation, culture medium was removed and cells were fixed by FixDenat. Next, anti-BrdU antibody (1:100) was added to bind the BrdU incorporated in newly synthesized cellular DNA. Proliferation was quantified by measuring the absorbance value at a wavelength of 450 nm using an uQuant ELISA microplate reader (BioTek Instruments, Inc., Winooski, VT, USA).

RNA expression profile

Total RNA was extracted using TRIzol and cDNA was synthesized with SuperScript II (both Takara Bio, Inc., Shiga, Japan) according to the manufacturer’s instructions, at 37°C for 15 min and 85°C for 5 sec. PI3K, SGK1, AKT and Kv1.3 mRNA were quantified by real-time PCR using the GAPDH housekeeping gene as an internal control. Real-time PCR was performed using the SYBR Premix Ex Taq reagent (Takara Bio, Inc.) and the Bio-Rad iQ5 machine (Hercules, CA, USA). The PCR conditions were programmed as 94°C for 3 min and 40 cycles of 94°C for 5 sec, 54°C for 30 sec and 72°C for 20 sec. PCR product quality was monitored using post-PCR melt curve analysis. The following primer sequences were used: GAPDH forward, 5′-GCTTCGCTCTCTGCTCCT-3′ and reverse, 5′-CGCCCAATACGACCAAAT-3′; PI3K forward, 5′-TGGCCTTAGCTCTTAGCCAAACAC-3′ and reverse, 5′-ATTGGAACACGGCCTTTGACA-3′; SGK1 forward, 5′-CTATGCTGCTGAAATAGC-3′ and reverse, 5′-GTCCGAAGTCAGTAAGG-3′; AKT forward, 5′-TCGGCAAGGTGATCCTGGTGAA-3′ and reverse, 5′-AGGCGGTCGTGGGTCTGGAAAG-3′; Kv1.3 forward, 5′-AGTATATGGTGATCGAAGAGG-3′ and reverse, 5′-AGTGAATATCTTCTTGATGTT-3′.

Western blot analysis

Expression of AKT/p-AKT, SGK1/p-SGK1 and Kv1.3 in HBSMCs was analyzed by western blot analysis, using GAPDH as an internal control. Briefly, total cells on the silicone membrane (with or without stretch) were harvested and then stored at −70°C. Protein extracts were obtained from HBSMC samples treated with cell lysis buffer [50 mM Tris-HCl (pH 8.0), 150 mM NaCl, 100 μg/ml PMSF and 1% Triton X-100] for 30 min on ice. Following removal of cell debris by centrifugation (4°C, 12,000 × g, 5 min), the lysate sample was boiled for 5 min in sample buffer, separated by 10% SDS-PAGE and transferred onto a nitrocellulose membrane. The membrane was incubated with specific primary antibodies at 4°C overnight, followed by secondary anti-rabbit IgG (Jackson Immunoresearch Inc., West Grove, PA, USA) for 1 h. Reactive protein was detected by exposure on BioMax MR-1 film (Kodak, Rochester, NY, USA).

Statistical analysis

BrdU and RT-PCR assays were performed at least in triplicate, yielding similar results. Data are presented as the mean ± SD. Statistical significance was analyzed by one-way ANOVA test. P<0.05 was considered to indicate a statistically significant difference.

Results

Cyclic stretch increases proliferation

To investigate the proliferative activity of HBSMCs with or without stretch, the BrdU assay was performed. The correlation between absorbance values and proliferative activities was investigated in each group with a peak at 30 min following the addition of the anti-BrdU antibody. As demonstrated in Table I, proliferative activity was enhanced in each group compared with the control (elongation = 0). At a specific frequency (e.g., 0.1 Hz), proliferative activity was increased from 2.5 to 5% elongation; however, gradually decreased following 5%, indicating 5% elongation is the optimal magnitude of stretch for HBSMC proliferation (Fig. 1A). Similarly, when cyclic stretch was performed at a specific elongation (e.g., 5%), maximum proliferative activity was identified at 0.1 Hz, indicating that 0.1 Hz is an ideal parameter of cyclic stretch (Fig. 1B). Following the examination of proliferative activity induced by cyclic stretch, the simulated optimal physiological stretch (5% elongation, 0.1 Hz) was established. All subsequent stretches were performed based on these results.

Activation of the PI3K-SGK1-Kv1.3 pathway by cyclic stretch

To determine the possible mechanism of proliferative activity in response to the physiological stretch applied, the expression levels of proteins involved in the PI3K pathway, including AKT and SGK1 (the two main related downstream targets which regulate cell survival and proliferation) were assessed. Significant upregulation of PI3K (3.75±0.56-fold, P<0.05) and SGK1 (11.47±1.09-fold, P<0.05); however, not AKT (1.17±0.14-fold, P>0.05), was observed compared with the non-stretch group. In addition, mRNA expression of Kv1.3, which is responsible for proliferation, was increased by 3.05±0.30-fold (P<0.05; Fig. 2A). mRNA expression levels were verified with western blot analysis. In response to cyclic stretch, SGK1, p-SGK1 and Kv1.3 protein levels were significantly increased; however, AKT gene expression and activation did not respond to stretch stimulation (Fig. 2B). These results indicate that the PI3K-SGK1-Kv1.3 pathway, but not the PI3K-AKT pathway, is involved in stretch-induced proliferation of HBSMCs.

Inhibitors of SGK-1 and Kv1.3 eliminate the increase in proliferative activity

The role of the PI3K-SGK1-Kv1.3 pathway in stretch-induced HBSMC proliferation was further confirmed using SGK-1 and Kv1.3 inhibitors. Cyclic stretch was applied following exposure of the cells to the SKG1 inhibitor, CKI-7 dihydrochloride (Sigma-Aldrich, St. Louis, MO, USA), at a final concentration of 100 μM. Proliferative activity was significantly suppressed compared with the stretch group without SGK1 inhibitor (0.986±0.042 vs. 1.460±0.015, P<0.05; Fig. 3A). In addition, Kv1.3 protein expression levels were analyzed by western blot analysis and no differences between the stretch and non-stretch group was identified (Fig. 3B). Similarly, when the high-affinity blocker of Kv1.3, margatoxin (Sigma-Aldrich), was used at a final concentration of 10 nM, the increased proliferative activity was largely eradicated compared with the same stretch group; however, without the Kv1.3 inhibitor (0.902±0.030 vs. 1.460±0.015, P<0.05; Fig. 3A). These results indicate that Kv1.3 is a downstream target of SGK1 which, in turn, is responsible for HBSMC proliferation induced by physiological cyclic stretch.

Discussion

Due to the unique properties and functions of the human bladder compared with the gastrointestinal segments, the generation of functional bladders using tissue engineering is important for organ regeneration and replacement. However, there are a number of technical limitations associated with bladder tissue engineering (28). HBSMCs are fundamental for bladder tissue engineering, but HBSMCs are an inadequate source and show dysfunctional contractility in the tissue engineered urinary bladder without appropriate external stimuli. Mechanical stimuli, including stretch and hydrostatic and hydrodynamic pressure, are crucial for the maturity of these mechanically sensitive cells. Stretch is considerably more important than hydrostatic pressure and other mechanical stimuli for bladder tissue engineering (9).

Although previous studies have attempted to apply stretch to SMCs, the majority of these stretches were not designed based on physiological conditions (29). The proliferative effects varied with different strains in magnitude and frequency, thus making normative application of stretch imperative. With the two key parameters (elongation and frequency) defined, in the present study, optimal simulated physiological cyclic stretch for HBSMC proliferation was established. When cyclic stretch was applied at 5% elongation and 0.1 Hz, a significantly augmented proliferative activity was detected (1.460±0.015, P<0.05) compared with the other groups. To the best of our knowledge, this study is the first to report the optimal stretch parameters for HBSMC proliferation. Identification of the optimal proliferation stretch model is beneficial to the solution of cell source inadequacy and is also important for the elucidation of the pathophysiological mechanisms involved in diseases associated with HBSMC overproliferation.

A vast number of kinases in the cell constitute a complicated but specific network in which kinases activate their own pathways and interact with other pathways. Located downstream from PI3K, SGK1 and AKT are considered to be extremely similar kinase proteins (19). As proteins implicated as mechanotransduction mediators of HBSMC proliferation, the expression of PI3K, AKT, SGK1 and Kv1.3 was investigated at the transcriptional and translational levels. Results of RT-PCR revealed increased mRNA expression of PI3K, SGK1 and Kv1.3, but not AKT, consistent with the results of western blot analysis. Initially, we hypothesized that PI3K-SGK1-Kv1.3 was responsible for stretch-induced proliferation of HBSMCs. Inhibitors of SGK1 and Kv1.3 largely eliminated the proliferative activity and confirmed the role of this pathway. These results indicated that SGK1, but not AKT, is a mediator of mechanical signaling events in HBSMC proliferation. These observations are consistent with the results of a previous study in which SGK1 predisposed vasculature smooth muscle to an increased proliferative response to mechanical stimuli and this proliferative response was markedly suppressed by SGK1 knockout (24). In addition, in the present study, Kv1.3 was identified to be involved in the stretch-induced proliferation process downstream of SGK1. As a dominant intracellular secondary messenger, increased concentrations of cytoplasmic Ca2+ are involved in the regulation of cell proliferation (30). K+ channels have been hypothesized to be important for the maintenance of cell membrane potential, which, in turn, is required for correct function of the Ca2+ release-activated Ca2+ channel (31). The latter channel, which is highly sensitive to membrane potential, mediates Ca2+ entry upon stimulation of cells, a prerequisite for triggering cell proliferation (32,33).

PI3K-AKT is a critical signaling pathway in the survival and proliferation of a number of cells (21,34) and is relatively well understood. However, SGK1, a ‘sister’ of AKT, has always been neglected. Functional analysis of gene-targeted mice lacking SGK1 provided insight into the functional significance of SGK1-dependent regulation of physiological functions. Knockout of SGK1 led to no severe phenotypes, indicating that SGK1 is not required for survival (35). Results indicated that SGK1 is a stimuli-responsive kinase that may mediate mechanical stretch-induced proliferation of HBSMCs, leading to bladder formation. Therefore, a hypothetical theory is raised: of the two ‘sisters’, AKT is implicated with basal proliferative activity and by contrast, SGK1 is responsible for the stimuli-induced activity. However, further confirmation and efforts must be performed to clearly illustrate this assumption.

In contrast to previous studies, several innovations were employed in this study: i) Cells were obtained from humans instead of animals; ii) physiological cyclic stretch was applied to stimulate the real bladder environment; iii) different elongations and frequencies were well studied, improved representative results were obtained and an optimized stretch model was established. Based on these results, we are likely to be able to provide qualified seed cells to bladder engineering techniques. In addition, observations indicate that the PI3K-SGK1-Kv1.3 pathway, but not PI3K-AKT, is the signal transduction pathway involved in stretch-induced proliferation and represents a promising pathway for novel targeted therapies for specific urinary bladder diseases caused by excessive mechanical forces, including BOO. The use of drugs or inhibitors to inhibit SGK1 or Kv1.3 may provide promising methods to interrupt this pathological process. In particular, in the case of SGK1, which is not required for individual survival (35), ‘block’ treatment is likely be extremely effective, without severe complications.

Acknowledgements

This study was supported by grants from the National Natural Science Foundation of China (no. 30872593 and 31170907), the Technology Support Program of Science and Technology Department of Sichuan Province (no. 2010SZ0163) and the Ph.D. Programs Foundation of Ministry of Education of China (no. 20110181110028).

References

1 

Fraser M, Thomas DF, Pitt E, Harnden P, Trejdosiewicz LK and Southgate J: A surgical model of composite cystoplasty with cultured urothelial cells: a controlled study of gross outcome and urothelial phenotype. BJU Int. 93:609–616. 2004. View Article : Google Scholar : PubMed/NCBI

2 

Southgate J, Gross W, Eardley I, Thomas DF and Trejdosiewicz LK: Bladder reconstruction - from cells to materials. Proc Inst Mech Eng H. 217:311–316. 2003. View Article : Google Scholar : PubMed/NCBI

3 

Zhang Y, Frimberger D, Cheng EY, Lin HK and Kropp BP: Challenges in a larger bladder replacement with cell-seeded and unseeded small intestinal submucosa grafts in a subtotal cystectomy model. BJU Int. 98:1100–1105. 2006. View Article : Google Scholar

4 

Lee DY, Yeh CR, Chang SF, Lee PL, Chien S, Cheng CK and Chiu JJ: Integrin-mediated expression of bone formation-related genes in osteoblast-like cells in response to fluid shear stress: roles of extracellular matrix, Shc and mitogen-activated protein kinase. J Bone Miner Res. 23:1140–1149. 2008. View Article : Google Scholar

5 

Liu D, Genetos DC, Shao Y, Geist DJ, Li J, Ke HZ, Turner CH and Duncan RL: Activation of extracellular-signal regulated kinase (ERK1/2) by fluid shear is Ca(2+)- and ATP-dependent in MC3T3-E1 osteoblasts. Bone. 42:644–652. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Lee AA, Graham DA, Dela CS, Ratcliffe A and Karlon WJ: Fluid shear stress-induced alignment of cultured vascular smooth muscle cells. J Biomech Eng. 124:37–43. 2002. View Article : Google Scholar : PubMed/NCBI

7 

Wei X, Li DB, Xu F, Wang Y, Zhu YC, Li H and Wang KJ: A novel bioreactor to simulate urinary bladder mechanical properties and compliance for bladder functional tissue engineering. Chin Med J (Engl). 124:568–573. 2011.PubMed/NCBI

8 

Ramachandran A, Gong EM, Pelton K, Ranpura SA, Mulone M, Seth A, Gomez P 3rd and Adam RM: FosB regulates stretch-induced expression of extracellular matrix proteins in smooth muscle. Am J Pathol. 179:2977–2989. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Farhat WA and Yeger H: Does mechanical stimulation have any role in urinary bladder tissue engineering? World J Urol. 26:301–305. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Chen L, Wei TQ, Wang Y, Zhang J, Li H and Wang KJ: Simulated bladder pressure stimulates human bladder smooth muscle cell proliferation via the PI3K/SGK1 signaling pathway. J Urol. 188:661–667. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Wu T, Chen L, Wei TQ, Wang Y, Xu F and Wang K: Effect of cyclic hydrodynamic pressure-induced proliferation of human bladder smooth muscle through Ras-related C3 botulinum toxin substrate 1, mitogen-activated protein kinase kinase 1/2 and extracellular regulated protein kinases 1/2. Int J Urol. 19:867–874. 2012. View Article : Google Scholar

12 

Brivanlou AH and Darnell JJ: Signal transduction and the control of gene expression. Science. 295:813–818. 2002. View Article : Google Scholar : PubMed/NCBI

13 

Elbadawi A: Pathology and pathophysiology of detrusor in incontinence. Urol Clin North Am. 22:499–512. 1995.PubMed/NCBI

14 

el-Feky H, Mangoud AM, Aly MA, Eissa MH, Abdel-Wahab RM, Kamhawy M, Ghobish A, Sabry AH, el Zayyat EA and Morsy TA: Detrusor morphology and pathology in relation to bladder outflow obstruction in bilharzial patients. J Egypt Soc Parasitol. 21:699–706. 1991.PubMed/NCBI

15 

Cerruto MA, Asimakopoulos AD, Artibani W, Del Popolo G, La Martina M, Carone R and Finazzi-Agrò E: Insight into new potential targets for the treatment of overactive bladder and detrusor overactivity. Urol Int. 89:1–8. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Webster MK, Goya L, Ge Y, Maiyar AC and Firestone GL: Characterization of sgk, a novel member of the serine/threonine protein kinase gene family which is transcriptionally induced by glucocorticoids and serum. Mol Cell Biol. 13:2031–2040. 1993.PubMed/NCBI

17 

Park J, Leong ML, Buse P, Maiyar AC, Firestone GL and Hemmings BA: Serum and glucocorticoid-inducible kinase (SGK) is a target of the PI 3-kinase-stimulated signaling pathway. EMBO J. 18:3024–3033. 1999. View Article : Google Scholar : PubMed/NCBI

18 

Kobayashi T and Cohen P: Activation of serum- and glucocorticoid-regulated protein kinase by agonists that activate phosphatidylinositide 3-kinase is mediated by 3-phosphoinositide-dependent protein kinase-1 (PDK1) and PDK2. Biochem J. 339:319–328. 1999. View Article : Google Scholar

19 

Lang F, Böhmer C, Palmada M, Seebohm G, Strutz-Seebohm N and Vallon V: (Patho)physiological significance of the serum- and glucocorticoid-inducible kinase isoforms. Physiol Rev. 86:1151–1178. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Liu G, Hitomi H, Hosomi N, Lei B, Nakano D, Deguchi K, Mori H, Masaki T, Ma H, Griendling KK and Nishiyama A: Mechanical stretch augments insulin-induced vascular smooth muscle cell proliferation by insulin-like growth factor-1 receptor. Exp Cell Res. 317:2420–2428. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Stover J and Nagatomi J: Cyclic pressure stimulates DNA synthesis through the PI3K/AKT signaling pathway in rat bladder smooth muscle cells. Ann Biomed Eng. 35:1585–1594. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Sakoda H, Gotoh Y, Katagiri H, Kurokawa M, Ono H, Onishi Y, Anai M, Ogihara T, Fujishiro M, Fukushima Y, Abe M, Shojima N, Kikuchi M, Oka Y, Hirai H and Asano T: Differing roles of AKT and serum- and glucocorticoid-regulated kinase in glucose metabolism, DNA synthesis and oncogenic activity. J Biol Chem. 278:25802–25807. 2003. View Article : Google Scholar : PubMed/NCBI

23 

Gamper N, Fillon S, Huber SM, Feng Y, Kobayashi T, Cohen P and Lang F: IGF-1 up-regulates K+ channels via PI3-kinase, PDK1 and SGK1. Pflugers Arch. 443:625–634. 2002.PubMed/NCBI

24 

Cheng J, Wang Y, Ma Y, Chan BT, Yang M, Liang A, Zhang L, Li H and Du J: The mechanical stress-activated serum-, glucocorticoid-regulated kinase 1 contributes to neointima formation in vein grafts. Circ Res. 107:1265–1274. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Wonderlin WF and Strobl JS: Potassium channels, proliferation and G1 progression. J Membr Biol. 154:91–107. 1996. View Article : Google Scholar : PubMed/NCBI

26 

Pardo LA: Voltage-gated potassium channels in cell proliferation. Physiology (Bethesda). 19:285–292. 2004. View Article : Google Scholar : PubMed/NCBI

27 

Jackson WF: KV1.3: a new therapeutic target to control vascular smooth muscle cell proliferation. Arterioscler Thromb Vasc Biol. 30:1073–1074. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Atala A: Tissue engineering of human bladder. Br Med Bull. 97:81–104. 2011. View Article : Google Scholar

29 

Lang F, Bohmer C, Palmada M, Seebohm G, Strutz-Seebohm N and Vallon V: (Patho)physiological significance of the serum- and glucocorticoid-inducible kinase isoforms. Physiol Rev. 86:1151–1178. 2006. View Article : Google Scholar : PubMed/NCBI

30 

Kahl CR and Means AR: Regulation of cell cycle progression by calcium/calmodulin-dependent pathways. Endocr Rev. 24:719–736. 2003. View Article : Google Scholar : PubMed/NCBI

31 

Parekh AB and Penner R: Store depletion and calcium influx. Physiol Rev. 77:901–930. 1997.PubMed/NCBI

32 

Berridge MJ, Lipp P and Bootman MD: The versatility and universality of calcium signalling. Nat Rev Mol Cell Biol. 1:11–21. 2000. View Article : Google Scholar : PubMed/NCBI

33 

Means AR: Calcium, calmodulin and cell cycle regulation. FEBS Lett. 347:1–4. 1994. View Article : Google Scholar : PubMed/NCBI

34 

Allard D, Figg N, Bennett MR and Littlewood TD: AKT regulates the survival of vascular smooth muscle cells via inhibition of FoxO3a and GSK3. J Biol Chem. 283:19739–19747. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Wu W, Chaudhuri S, Brickley DR, Pang D, Karrison T and Conzen SD: Microarray analysis reveals glucocorticoid-regulated survival genes that are associated with inhibition of apoptosis in breast epithelial cells. Cancer Res. 64:1757–1764. 2004. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July 2013
Volume 8 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tian Y, Yue X, Luo D, Wazir R, Wang J, Wu T, Chen L, Liao B and Wang K: Increased proliferation of human bladder smooth muscle cells is mediated by physiological cyclic stretch via the PI3K‑SGK1‑Kv1.3 pathway. Mol Med Rep 8: 294-298, 2013
APA
Tian, Y., Yue, X., Luo, D., Wazir, R., Wang, J., Wu, T. ... Wang, K. (2013). Increased proliferation of human bladder smooth muscle cells is mediated by physiological cyclic stretch via the PI3K‑SGK1‑Kv1.3 pathway. Molecular Medicine Reports, 8, 294-298. https://doi.org/10.3892/mmr.2013.1473
MLA
Tian, Y., Yue, X., Luo, D., Wazir, R., Wang, J., Wu, T., Chen, L., Liao, B., Wang, K."Increased proliferation of human bladder smooth muscle cells is mediated by physiological cyclic stretch via the PI3K‑SGK1‑Kv1.3 pathway". Molecular Medicine Reports 8.1 (2013): 294-298.
Chicago
Tian, Y., Yue, X., Luo, D., Wazir, R., Wang, J., Wu, T., Chen, L., Liao, B., Wang, K."Increased proliferation of human bladder smooth muscle cells is mediated by physiological cyclic stretch via the PI3K‑SGK1‑Kv1.3 pathway". Molecular Medicine Reports 8, no. 1 (2013): 294-298. https://doi.org/10.3892/mmr.2013.1473