A novel regulatory function of sweet taste-sensing receptor in adipogenic differentiation of 3T3-L1 cells

Yosuke Masubuchi, Yuko Nakagawa, Jinhui Ma, Tsutomu Sasaki, Tadahiro Kitamura, Yoritsuna Yamamoto, Hitoshi Kurose, Itaru Kojima, Hiroshi Shibata, Yosuke Masubuchi, Yuko Nakagawa, Jinhui Ma, Tsutomu Sasaki, Tadahiro Kitamura, Yoritsuna Yamamoto, Hitoshi Kurose, Itaru Kojima, Hiroshi Shibata

Abstract

Background: Sweet taste receptor is expressed not only in taste buds but also in nongustatory organs such as enteroendocrine cells and pancreatic beta-cells, and may play more extensive physiological roles in energy metabolism. Here we examined the expression and function of the sweet taste receptor in 3T3-L1 cells.

Methodology/principal findings: In undifferentiated preadipocytes, both T1R2 and T1R3 were expressed very weakly, whereas the expression of T1R3 but not T1R2 was markedly up-regulated upon induction of differentiation (by 83.0 and 3.8-fold, respectively at Day 6). The α subunits of Gs (Gαs) and G14 (Gα14) but not gustducin were expressed throughout the differentiation process. The addition of sucralose or saccharin during the first 48 hours of differentiation considerably reduced the expression of peroxisome proliferator activated receptor γ (PPARγ and CCAAT/enhancer-binding protein α (C/EBPα at Day 2, the expression of aP2 at Day 4 and triglyceride accumulation at Day 6. These anti-adipogenic effects were attenuated by short hairpin RNA-mediated gene-silencing of T1R3. In addition, overexpression of the dominant-negative mutant of Gαs but not YM-254890, an inhibitor of Gα14, impeded the effects of sweeteners, suggesting a possible coupling of Gs with the putative sweet taste-sensing receptor. In agreement, sucralose and saccharin increased the cyclic AMP concentration in differentiating 3T3-L1 cells and also in HEK293 cells heterologously expressing T1R3. Furthermore, the anti-adipogenic effects of sweeteners were mimicked by Gs activation with cholera toxin but not by adenylate cyclase activation with forskolin, whereas small interfering RNA-mediated knockdown of Gαs had the opposite effects.

Conclusions: 3T3-L1 cells express a functional sweet taste-sensing receptor presumably as a T1R3 homomer, which mediates the anti-adipogenic signal by a Gs-dependent but cAMP-independent mechanism.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Expression of T1Rs during differentiation…
Figure 1. Expression of T1Rs during differentiation of 3T3-L1 cells.
A. The total RNAs prepared from 3T3-L1 cells at the indicated time points during differentiation were subjected to quantitative RT-PCR using mouse ribosomal protein S18 as an internal control as described in`Materials and Methods'. The mRNA levels of T1R2 and T1R3 are shown as the percentage of that of T1R3 at Day 6. Results are shown as the mean ± SE (n = 3–6). B. Immumoblot data for T1R3 and actin in undifferentiated (Day 0) and differentiated (Day 7) 3T3-L1 cells. C. Immunofluorescence staining images for T1R3 (a and b, red) and GLUT4 (c and d, green) in undifferentiated (Day 0, a and c) and differentiated (Day 7, b and d) 3T3-L1 cells. Nuclei were visualized with DAPI (blue). e, Subcellular distribution of T1R3 (red) in Day 7 cells. Arrowheads indicate peripheral localization of T1R3.
Figure 2. Effects of Sweeteners on Adipogenesis.
Figure 2. Effects of Sweeteners on Adipogenesis.
A. 3T3-L1 cells were differentiated in the presence of the indicated concentrations of sucralose or saccharin, and the expression levels of PPARγ and C/EBPα at Day 2 (48 hours) were examined by immunoblotting. Representative immunoblot data for PPARγ and C/EBPα (upper panel) and the relative amounts of the proteins normalized with β-tubulin (lower panel) are shown. Results are shown as the mean values of two independent experiments. B. 3T3-L1 cells were differentiated without (control) or with D-mannitol (20 mM), sucralose (20 mM), or saccharin (20 mM) during the first 48 hours of differentiation. The expression levels of PPARγ and C/EBPα at Day 2, and aP2 at Day 4 were examined by immunoblotting. Representative immunoblot data (upper panel) and the relative amounts of the proteins normalized with β-tubulin (lower panel) are shown. Results are shown as the mean ± SE (n = 3). ?, Pa), D-mannitol (20 mM) (b), sucralose (20 mM) (c) or saccharin (20 mM) (d) during the first 48 hours are shown (right panel). D. Adipose tissue-derived stem cells were differentiated to adipocytes as described in ‘Materials and Methods’ in the presence of none (control) (a), D-mannitol (20 mM) (b), sucralose (20 mM) (c) or saccharin (20 mM) (d) for 6 days, and were stained with Oil Red-O. The amounts of Oil Red-O (left panel) and microscopic images (right panel) are shown. Results are presented as the mean ± SE (n = 3). ?, P<0.001; ??, P<0.01 (vs. control). E. Undifferentiated 3T3-L1 cells were transfected with the pGIPz expression vectors containing non-silencing or T1R3-targeting shRNA sequences (30 μg each) by electroporation as described in ‘Materials and Methods’. Transfected cells were seeded on a 12-well dish and cultured to confluence before induction of differentiation without (control) or with sucralose (20 mM) or saccharin (20 mM). The expression level of T1R3 was measured by quantitative RT-PCR immediately before induction of differentiation (at Day 0) (left panel). The expression levels of PPARγ and C/EBPα at Day 2 (48 hours) were measured by immunoblotting. Representative immunoblot data and the relative amounts of the proteins normalized with β-tubulin are shown (middle panel). Results are presented as the mean ± SE (n = 3). ?, P<0.05 (vs. shControl). The amounts of Oil Red-O and the microscopic images of stained cells at Day 6 are shown (right panel). Cells transfected with non-silencing (ac) or T1R3-targeting shRNA (d-f) were differentiated in the absence (a and d) or the presence of sucralose (20 mM) (b and e) or saccharin (20 mM) (c and f) during the first 48 hours. Results are presented as the mean ± SE (n = 3). ?, P<0.01; ??, P<0.05 (vs. shControl).
Figure 3. Roles for G proteins in…
Figure 3. Roles for G proteins in Sweeteners Effects on Differentiation of 3T3-L1 cells.
A. Expression profiles of Gαgust, Gα14 and Gαs during differentiation of 3T3-L1 cells. The total RNAs were prepared from 3T3-L1 cells as described in Fig. 1 and the mRNA levels of Gαgust, Gα14 and Gαs were measured by quantitative RT-PCR using mouse ribosomal protein S18 as an internal control. Results are shown as the mean ± SE (n = 3–6). B. 3T3-L1 cells were differentiated without (control) or with sucralose (20 mM), saccharin (20 mM), or endothelin-1 (20 nM) in the absence (0.1% DMSO) or the presence of YM-254890 (10 µM). The expression levels of PPARγ and C/EBPα at Day 2 (48 hours) were measured by immunoblotting. Representative immunoblot data (upper panel) and the relative amounts of the proteins normalized with β-tubulin (lower panel) are shown. Gray and black bars show the control and the plus YM-254890 data, respectively. Results are shown as the mean values from two independent experiments. C. Undifferentiated 3T3-L1 cells were detached and transfected with the expression vectors containing wild-type or G226A mutant Gαs cDNAs (20 μg each) by electroporation as described in ‘Materials and Methods’. Transfected cells were seeded on a 6-well culture dish and cultured to confluence before induction of differentiation without (control) or with sucralose (20 mM) or saccharin (20 mM). The expression levels of PPARγ and C/EBPα were measured by immunoblotting at Day 2 (48 hours). Representative immunoblot data (upper panel) and the relative amounts of the proteins normalized with β-tubulin (lower panel) are shown. Gray and black bars show the control and the Gαs-G226A data, respectively. Results are shown as the mean ± SE (n = 3). P

Figure 4. Effects of Sweeteners on cAMP.

Figure 4. Effects of Sweeteners on cAMP.

A. 3T3-L1 cells at Day 0 (undifferentiated), Day…

Figure 4. Effects of Sweeteners on cAMP.
A. 3T3-L1 cells at Day 0 (undifferentiated), Day 2 and Day 6 of differentiation were stimulated without (control) or with sucralose (20 mM) or saccharin (20 mM) for 30 min at 37°C, and the cellular cAMP contents were measured as described in ‘Materials and Methods’. Results are shown as the mean ± SE (n = 3)., Pc). At the time point indicated with the arrow, cells were stimulated with 20 mM of sucralose. The [cAMP]c was shown as the reciprocal of the emission ratio of EYFP/ECFP (i.e. ECFP/EYFP). C. HEK293 cells were transfected with pcDNA3.1 vector (20 μg) alone (mock) or with the vector containing mouse T1R3 cDNA (20 μg) by electroporation and seeded on a 12-well culture plate. After incubation for 24 hours, cells were stimulated without (control) or with sucralose (20 mM) or saccharin (20 mM) for 30 minutes at 37°C and the cellular contents of cAMP were measured as described in ‘Materials and Methods’. Results are shown as the mean ± SE (n = 4–6)., P<0.05, P<0.01 (vs. control). D. HEK293 cells transfected by electroporation with pcDNA3.1 vector (mock), T1R2 (20 μg), T1R3 (20 μg), or both T1R2 and T1R3 (10 μg, each) were seeded on a 35 mm glass bottom dish, cultured for 24 hours and stimulated with 20 mM of sucralose at the time point indicated with the arrow. The [cAMP]c were monitored as described in B.

Figure 5. Role for Gαs in adipogenesis.

Figure 5. Role for Gαs in adipogenesis.

A. 3T3-L1 cells were differentiated for 48 hours…

Figure 5. Role for Gαs in adipogenesis.
A. 3T3-L1 cells were differentiated for 48 hours in the absence (control) or the presence of cholera toxin (0.1 μg/ml) or forskolin (20 μM). Then the expression levels of PPARγ and C/EBPα were measured by immunoblotting. The representative immunoblot data (left panel) and the relative amounts of the proteins normalized with β-tubulin (right panel) are shown. Data are shown as the mean ± SE (n = 3)., P

Figure 6. A model for signal transduction…

Figure 6. A model for signal transduction mechanism downstream of the sweet taste-sensing receptors in…

Figure 6. A model for signal transduction mechanism downstream of the sweet taste-sensing receptors in taste cells and 3T3-L1 cells.
In taste cells (in the left side), T1R2+T1R3 heterodimeric sweet receptor activates PLCβ via gustducin (Ggust) or other G proteins, leading to [Ca2+]c elevation and membrane depolarization. In 3T3-L1 cells (in the right side), T1R3 homomeric receptor may activates Gs, which mediates the anti-adipogenic signal by a cAMP-independent mechanism. PLCβ: phospholipase C-β; DAG: diacylglycerol; IP3: inositol 1,4,5-trisphosphate; Px1: pannexin 1; AC: adenylate cyclase; PDE: cAMP phosphodiesterase.
Similar articles
Cited by
References
    1. Nelson G, Hoon MA, Chandrashekar J, Zhang Y, Ryba NJ, et al. (2001) Mammalian sweet taste receptors. Cell 106: 381–390. - PubMed
    1. Kojima I, Nakagawa Y (2011) The Role of the Sweet Taste Receptor in Enteroendocrine Cells and Pancreatic beta-Cells. Diabetes Metab J 35: 451–457. - PMC - PubMed
    1. Roper SD (2007) Signal transduction and information processing in mammalian taste buds. Pflugers Arch 454: 759–776. - PMC - PubMed
    1. Damak S, Rong M, Yasumatsu K, Kokrashvili Z, Varadarajan V, et al. (2003) Detection of sweet and umami taste in the absence of taste receptor T1r3. Science 301: 850–853. - PubMed
    1. Zhao GQ, Zhang Y, Hoon MA, Chandrashekar J, Erlenbach I, et al. (2003) The receptors for mammalian sweet and umami taste. Cell 115: 255–266. - PubMed
Show all 53 references
Publication types
MeSH terms
Grant support
This work was supported by the Global Centers of Excellence Program “Signal Transduction in the Regulatory System and Its Disorders” and grant-in-aid from the Ministry of Education, Culture, Sports, Science and Technology of Japan. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 4. Effects of Sweeteners on cAMP.
Figure 4. Effects of Sweeteners on cAMP.
A. 3T3-L1 cells at Day 0 (undifferentiated), Day 2 and Day 6 of differentiation were stimulated without (control) or with sucralose (20 mM) or saccharin (20 mM) for 30 min at 37°C, and the cellular cAMP contents were measured as described in ‘Materials and Methods’. Results are shown as the mean ± SE (n = 3)., Pc). At the time point indicated with the arrow, cells were stimulated with 20 mM of sucralose. The [cAMP]c was shown as the reciprocal of the emission ratio of EYFP/ECFP (i.e. ECFP/EYFP). C. HEK293 cells were transfected with pcDNA3.1 vector (20 μg) alone (mock) or with the vector containing mouse T1R3 cDNA (20 μg) by electroporation and seeded on a 12-well culture plate. After incubation for 24 hours, cells were stimulated without (control) or with sucralose (20 mM) or saccharin (20 mM) for 30 minutes at 37°C and the cellular contents of cAMP were measured as described in ‘Materials and Methods’. Results are shown as the mean ± SE (n = 4–6)., P<0.05, P<0.01 (vs. control). D. HEK293 cells transfected by electroporation with pcDNA3.1 vector (mock), T1R2 (20 μg), T1R3 (20 μg), or both T1R2 and T1R3 (10 μg, each) were seeded on a 35 mm glass bottom dish, cultured for 24 hours and stimulated with 20 mM of sucralose at the time point indicated with the arrow. The [cAMP]c were monitored as described in B.
Figure 5. Role for Gαs in adipogenesis.
Figure 5. Role for Gαs in adipogenesis.
A. 3T3-L1 cells were differentiated for 48 hours in the absence (control) or the presence of cholera toxin (0.1 μg/ml) or forskolin (20 μM). Then the expression levels of PPARγ and C/EBPα were measured by immunoblotting. The representative immunoblot data (left panel) and the relative amounts of the proteins normalized with β-tubulin (right panel) are shown. Data are shown as the mean ± SE (n = 3)., P

Figure 6. A model for signal transduction…

Figure 6. A model for signal transduction mechanism downstream of the sweet taste-sensing receptors in…

Figure 6. A model for signal transduction mechanism downstream of the sweet taste-sensing receptors in taste cells and 3T3-L1 cells.
In taste cells (in the left side), T1R2+T1R3 heterodimeric sweet receptor activates PLCβ via gustducin (Ggust) or other G proteins, leading to [Ca2+]c elevation and membrane depolarization. In 3T3-L1 cells (in the right side), T1R3 homomeric receptor may activates Gs, which mediates the anti-adipogenic signal by a cAMP-independent mechanism. PLCβ: phospholipase C-β; DAG: diacylglycerol; IP3: inositol 1,4,5-trisphosphate; Px1: pannexin 1; AC: adenylate cyclase; PDE: cAMP phosphodiesterase.
Figure 6. A model for signal transduction…
Figure 6. A model for signal transduction mechanism downstream of the sweet taste-sensing receptors in taste cells and 3T3-L1 cells.
In taste cells (in the left side), T1R2+T1R3 heterodimeric sweet receptor activates PLCβ via gustducin (Ggust) or other G proteins, leading to [Ca2+]c elevation and membrane depolarization. In 3T3-L1 cells (in the right side), T1R3 homomeric receptor may activates Gs, which mediates the anti-adipogenic signal by a cAMP-independent mechanism. PLCβ: phospholipase C-β; DAG: diacylglycerol; IP3: inositol 1,4,5-trisphosphate; Px1: pannexin 1; AC: adenylate cyclase; PDE: cAMP phosphodiesterase.

References

    1. Nelson G, Hoon MA, Chandrashekar J, Zhang Y, Ryba NJ, et al. (2001) Mammalian sweet taste receptors. Cell 106: 381–390.
    1. Kojima I, Nakagawa Y (2011) The Role of the Sweet Taste Receptor in Enteroendocrine Cells and Pancreatic beta-Cells. Diabetes Metab J 35: 451–457.
    1. Roper SD (2007) Signal transduction and information processing in mammalian taste buds. Pflugers Arch 454: 759–776.
    1. Damak S, Rong M, Yasumatsu K, Kokrashvili Z, Varadarajan V, et al. (2003) Detection of sweet and umami taste in the absence of taste receptor T1r3. Science 301: 850–853.
    1. Zhao GQ, Zhang Y, Hoon MA, Chandrashekar J, Erlenbach I, et al. (2003) The receptors for mammalian sweet and umami taste. Cell 115: 255–266.
    1. Delay ER, Hernandez NP, Bromley K, Margolskee RF (2006) Sucrose and monosodium glutamate taste thresholds and discrimination ability of T1R3 knockout mice. Chem Senses 31: 351–357.
    1. Wong GT, Gannon KS, Margolskee RF (1996) Transduction of bitter and sweet taste by gustducin. Nature 381: 796–800.
    1. Danilova V, Damak S, Margolskee RF, Hellekant G (2006) Taste responses to sweet stimuli in alpha-gustducin knockout and wild-type mice. Chem Senses 31: 573–580.
    1. Damak S, Rong M, Yasumatsu K, Kokrashvili Z, Perez CA, et al. (2006) Trpm5 null mice respond to bitter, sweet, and umami compounds. Chem Senses 31: 253–264.
    1. Margolskee RF, Dyer J, Kokrashvili Z, Salmon KS, Ilegems E, et al. (2007) T1R3 and gustducin in gut sense sugars to regulate expression of Na+-glucose cotransporter 1. Proc Natl Acad Sci U S A 104: 15075–15080.
    1. Nakagawa Y, Nagasawa M, Yamada S, Hara A, Mogami H, et al. (2009) Sweet taste receptor expressed in pancreatic beta-cells activates the calcium and cyclic AMP signaling systems and stimulates insulin secretion. PLoS One 4: e5106.
    1. Jang HJ, Kokrashvili Z, Theodorakis MJ, Carlson OD, Kim BJ, et al. (2007) Gut-expressed gustducin and taste receptors regulate secretion of glucagon-like peptide-1. Proc Natl Acad Sci U S A 104: 15069–15074.
    1. Kokrashvili Z, Mosinger B, Margolskee RF (2009) Taste signaling elements expressed in gut enteroendocrine cells regulate nutrient-responsive secretion of gut hormones. Am J Clin Nutr 90: 822S–825S.
    1. Shibata H, Suzuki Y, Omata W, Tanaka S, Kojima I (1995) Dissection of GLUT4 recycling pathway into exocytosis and endocytosis in rat adipocytes. Evidence that GTP-binding proteins are involved in both processes. J Biol Chem 270: 11489–11495.
    1. Maruyama Y, Yasuda R, Kuroda M, Eto Y (2012) Kokumi substances, enhancers of basic tastes, induce responses in calcium-sensing receptor expressing taste cells. PLoS One 7: e34489.
    1. Djian P, Phillips M, Green H (1985) The activation of specific gene transcription in the adipose conversion of 3T3 cells. J Cell Physiol 124: 554–556.
    1. Liu LB, Omata W, Kojima I, Shibata H (2007) The SUMO conjugating enzyme Ubc9 is a regulator of GLUT4 turnover and targeting to the insulin-responsive storage compartment in 3T3-L1 adipocytes. Diabetes 56: 1977–1985.
    1. Rodbell M (1964) Metabolism of Isolated Fat Cells. I. Effects of Hormones on Glucose Metabolism and Lipolysis. J Biol Chem 239: 375–380.
    1. Hattori H, Ishihara M, Fukuda T, Suda T, Katagiri T (2006) Establishment of a novel method for enriching osteoblast progenitors from adipose tissues using a difference in cell adhesive properties. Biochem Biophys Res Commun 343: 1118–1123.
    1. Jones DT, Reed RR (1987) Molecular cloning of five GTP-binding protein cDNA species from rat olfactory neuroepithelium. J Biol Chem 262: 14241–14249.
    1. Miller RT, Masters SB, Sullivan KA, Beiderman B, Bourne HR (1988) A mutation that prevents GTP-dependent activation of the alpha chain of Gs. Nature 334: 712–715.
    1. Lee E, Taussig R, Gilman AG (1992) The G226A mutant of Gs alpha highlights the requirement for dissociation of G protein subunits. J Biol Chem 267: 1212–1218.
    1. Nikolaev VO, Bunemann M, Hein L, Hannawacker A, Lohse MJ (2004) Novel single chain cAMP sensors for receptor-induced signal propagation. J Biol Chem 279: 37215–37218.
    1. Bystrova MF, Romanov RA, Rogachevskaja OA, Churbanov GD, Kolesnikov SS (2010) Functional expression of the extracellular-Ca2+-sensing receptor in mouse taste cells. J Cell Sci 123: 972–982.
    1. Ohsu T, Amino Y, Nagasaki H, Yamanaka T, Takeshita S, et al. (2010) Involvement of the calcium-sensing receptor in human taste perception. J Biol Chem 285: 1016–1022.
    1. San Gabriel A, Uneyama H, Maekawa T, Torii K (2009) The calcium-sensing receptor in taste tissue. Biochem Biophys Res Commun 378: 414–418.
    1. Shindo Y, Miura H, Carninci P, Kawai J, Hayashizaki Y, et al. (2008) G alpha14 is a candidate mediator of sweet/umami signal transduction in the posterior region of the mouse tongue. Biochem Biophys Res Commun 376: 504–508.
    1. Tizzano M, Dvoryanchikov G, Barrows JK, Kim S, Chaudhari N, et al. (2008) Expression of Galpha14 in sweet-transducing taste cells of the posterior tongue. BMC Neurosci 9: 110.
    1. Kusakabe Y, Yasuoka A, Asano-Miyoshi M, Iwabuchi K, Matsumoto I, et al. (2000) Comprehensive study on G protein alpha-subunits in taste bud cells, with special reference to the occurrence of Galphai2 as a major Galpha species. Chem Senses 25: 525–531.
    1. Trubey KR, Culpepper S, Maruyama Y, Kinnamon SC, Chaudhari N (2006) Tastants evoke cAMP signal in taste buds that is independent of calcium signaling. Am J Physiol Cell Physiol 291: C237–244.
    1. Striem BJ, Pace U, Zehavi U, Naim M, Lancet D (1989) Sweet tastants stimulate adenylate cyclase coupled to GTP-binding protein in rat tongue membranes. Biochem J 260: 121–126.
    1. Naim M, Ronen T, Striem BJ, Levinson M, Zehavi U (1991) Adenylate cyclase responses to sucrose stimulation in membranes of pig circumvallate taste papillae. Comp Biochem Physiol B 100: 455–458.
    1. Nakashima K, Ninomiya Y (1999) Transduction for sweet taste of saccharin may involve both inositol 1,4,5-trisphosphate and cAMP pathways in the fungiform taste buds in C57BL mice. Cell Physiol Biochem 9: 90–98.
    1. Nishimura A, Kitano K, Takasaki J, Taniguchi M, Mizuno N, et al. (2010) Structural basis for the specific inhibition of heterotrimeric Gq protein by a small molecule. Proc Natl Acad Sci U S A 107: 13666–13671.
    1. Takasaki J, Saito T, Taniguchi M, Kawasaki T, Moritani Y, et al. (2004) A novel Galphaq/11-selective inhibitor. J Biol Chem 279: 47438–47445.
    1. Tanahashi T, Yamaguchi K, Ishikawa S, Kusuhara M, Adachi I, et al. (1991) Endothelin-1 inhibits adipogenic differentiation of 3T3-L1 preadipocytes. Biochem Biophys Res Commun 177: 854–860.
    1. Bhattacharya I, Ullrich A (2006) Endothelin-1 inhibits adipogenesis: role of phosphorylation of Akt and ERK1/2. FEBS Lett 580: 5765–5771.
    1. Imamura T, Ishibashi K, Dalle S, Ugi S, Olefsky JM (1999) Endothelin-1-induced GLUT4 translocation is mediated via Galpha(q/11) protein and phosphatidylinositol 3-kinase in 3T3-L1 adipocytes. J Biol Chem 274: 33691–33695.
    1. Bose A, Cherniack AD, Langille SE, Nicoloro SM, Buxton JM, et al. (2001) G(alpha)11 signaling through ARF6 regulates F-actin mobilization and GLUT4 glucose transporter translocation to the plasma membrane. Mol Cell Biol 21: 5262–5275.
    1. Lawrence JT, Birnbaum MJ (2001) ADP-ribosylation factor 6 delineates separate pathways used by endothelin 1 and insulin for stimulating glucose uptake in 3T3-L1 adipocytes. Mol Cell Biol 21: 5276–5285.
    1. Gether U (2000) Uncovering molecular mechanisms involved in activation of G protein-coupled receptors. Endocr Rev 21: 90–113.
    1. Olansky L, Myers GA, Pohl SL, Hewlett EL (1983) Promotion of lipolysis in rat adipocytes by pertussis toxin: reversal of endogenous inhibition. Proc Natl Acad Sci U S A 80: 6547–6551.
    1. Reusch JE, Colton LA, Klemm DJ (2000) CREB activation induces adipogenesis in 3T3-L1 cells. Mol Cell Biol 20: 1008–1020.
    1. Petersen RK, Madsen L, Pedersen LM, Hallenborg P, Hagland H, et al. (2008) Cyclic AMP (cAMP)-mediated stimulation of adipocyte differentiation requires the synergistic action of Epac- and cAMP-dependent protein kinase-dependent processes. Mol Cell Biol 28: 3804–3816.
    1. Wang HY, Watkins DC, Malbon CC (1992) Antisense oligodeoxynucleotides to GS protein alpha-subunit sequence accelerate differentiation of fibroblasts to adipocytes. Nature 358: 334–337.
    1. Liu X, Malbon CC, Wang HY (1998) Identification of amino acid residues of Gsalpha critical to repression of adipogenesis. J Biol Chem 273: 11685–11694.
    1. Wang H, Johnson GL, Liu X, Malbon CC (1996) Repression of adipogenesis by adenylyl cyclase stimulatory G-protein alpha subunit is expressed within region 146-220. J Biol Chem 271: 22022–22029.
    1. Zhang L, Paddon C, Lewis MD, Grennan-Jones F, Ludgate M (2009) Gsalpha signalling suppresses PPARgamma2 generation and inhibits 3T3L1 adipogenesis. J Endocrinol 202: 207–215.
    1. Hewavitharana T, Wedegaertner PB (2012) Non-canonical signaling and localizations of heterotrimeric G proteins. Cell Signal 24: 25–34.
    1. Dave RH, Saengsawang W, Yu JZ, Donati R, Rasenick MM (2009) Heterotrimeric G-proteins interact directly with cytoskeletal components to modify microtubule-dependent cellular processes. Neurosignals 17: 100–108.
    1. Zheng B, Lavoie C, Tang TD, Ma P, Meerloo T, et al. (2004) Regulation of epidermal growth factor receptor degradation by heterotrimeric Galphas protein. Mol Biol Cell 15: 5538–5550.
    1. Krendel M, Zenke FT, Bokoch GM (2002) Nucleotide exchange factor GEF-H1 mediates cross-talk between microtubules and the actin cytoskeleton. Nat Cell Biol 4: 294–301.
    1. Sordella R, Jiang W, Chen GC, Curto M, Settleman J (2003) Modulation of Rho GTPase signaling regulates a switch between adipogenesis and myogenesis. Cell 113: 147–158.

Source: PubMed

3
Iratkozz fel