Skip to main content
Top
Published in: Endocrine 3/2012

Open Access 01-12-2012 | Original Article

Cyclic AMP signaling in bone marrow stromal cells has reciprocal effects on the ability of mesenchymal stem cells to differentiate into mature osteoblasts versus mature adipocytes

Authors: Richard Kao, Weidar Lu, Alyssa Louie, Robert Nissenson

Published in: Endocrine | Issue 3/2012

Login to get access

Abstract

Stimulatory G protein-mediated cAMP signaling is intimately involved in skeletal homeostasis. However, limited information is available on the role of the cAMP signaling in regulating the differentiation of mesenchymal stem cells into mature osteoblasts and adipocytes. To investigate this, we treated primary mouse bone marrow stromal cells (BMSCs) with forskolin to stimulate cAMP signaling and determined the effect on osteoblast and adipocyte differentiation. Exposure of differentiating osteoblasts to forskolin markedly inhibited progression to the late stages of osteoblast differentiation, and this effect was replicated by continuous exposure to PTH. Strikingly, forskolin activation of cAMP signaling in BMSCs conditioned mesenchymal stem cells (MSCs) to undergo increased osteogenic differentiation and decreased adipogenic differentiation. PTH treatment of BMSCs also enhanced subsequent osteogenesis, but promoted an increased adipogenesis as well. Thus, activation of cAMP signaling alters the lineage commitment of MSCs, favoring osteogenesis at the expense of adipogenesis.
Literature
1.
go back to reference L.S. Weinstein et al., Activating mutations of the stimulatory G protein in the McCune–Albright syndrome. N. Engl. J. Med. 325(24), 1688–1695 (1991)PubMedCrossRef L.S. Weinstein et al., Activating mutations of the stimulatory G protein in the McCune–Albright syndrome. N. Engl. J. Med. 325(24), 1688–1695 (1991)PubMedCrossRef
2.
go back to reference W.F. Schwindinger, C.A. Francomano, M.A. Levine, Identification of a mutation in the gene encoding the alpha subunit of the stimulatory G protein of adenylyl cyclase in McCune–Albright syndrome. Proc. Natl. Acad. Sci. USA 89(11), 5152–5156 (1992)PubMedCrossRef W.F. Schwindinger, C.A. Francomano, M.A. Levine, Identification of a mutation in the gene encoding the alpha subunit of the stimulatory G protein of adenylyl cyclase in McCune–Albright syndrome. Proc. Natl. Acad. Sci. USA 89(11), 5152–5156 (1992)PubMedCrossRef
3.
go back to reference A. Shenker et al., Severe endocrine and nonendocrine manifestations of the McCune–Albright syndrome associated with activating mutations of stimulatory G protein GS. J. Pediatr. 123(4), 509–518 (1993)PubMedCrossRef A. Shenker et al., Severe endocrine and nonendocrine manifestations of the McCune–Albright syndrome associated with activating mutations of stimulatory G protein GS. J. Pediatr. 123(4), 509–518 (1993)PubMedCrossRef
4.
go back to reference C.D. Malchoff et al., An unusual presentation of McCune–Albright syndrome confirmed by an activating mutation of the Gs alpha-subunit from a bone lesion. J. Clin. Endocrinol. Metab. 78(3), 803–806 (1994)PubMedCrossRef C.D. Malchoff et al., An unusual presentation of McCune–Albright syndrome confirmed by an activating mutation of the Gs alpha-subunit from a bone lesion. J. Clin. Endocrinol. Metab. 78(3), 803–806 (1994)PubMedCrossRef
5.
go back to reference A. Shenker et al., An activating Gs alpha mutation is present in fibrous dysplasia of bone in the McCune–Albright syndrome. J. Clin. Endocrinol. Metab. 79(3), 750–755 (1994)PubMedCrossRef A. Shenker et al., An activating Gs alpha mutation is present in fibrous dysplasia of bone in the McCune–Albright syndrome. J. Clin. Endocrinol. Metab. 79(3), 750–755 (1994)PubMedCrossRef
6.
go back to reference P.J. Marie et al., Increased proliferation of osteoblastic cells expressing the activating Gs alpha mutation in monostotic and polyostotic fibrous dysplasia. Am. J. Pathol. 150(3), 1059–1069 (1997)PubMed P.J. Marie et al., Increased proliferation of osteoblastic cells expressing the activating Gs alpha mutation in monostotic and polyostotic fibrous dysplasia. Am. J. Pathol. 150(3), 1059–1069 (1997)PubMed
7.
go back to reference B.A. Alman, D.A. Greel, H.J. Wolfe, Activating mutations of Gs protein in monostotic fibrous lesions of bone. J. Orthop. Res. 14(2), 311–315 (1996)PubMedCrossRef B.A. Alman, D.A. Greel, H.J. Wolfe, Activating mutations of Gs protein in monostotic fibrous lesions of bone. J. Orthop. Res. 14(2), 311–315 (1996)PubMedCrossRef
8.
go back to reference M. Riminucci et al., Fibrous dysplasia of bone in the McCune–Albright syndrome: abnormalities in bone formation. Am. J. Pathol. 151(6), 1587–1600 (1997)PubMed M. Riminucci et al., Fibrous dysplasia of bone in the McCune–Albright syndrome: abnormalities in bone formation. Am. J. Pathol. 151(6), 1587–1600 (1997)PubMed
9.
go back to reference S. Piersanti et al., Transfer, analysis, and reversion of the fibrous dysplasia cellular phenotype in human skeletal progenitors. J. Bone Miner. Res. 25(5), 1103–1116 (2010)PubMed S. Piersanti et al., Transfer, analysis, and reversion of the fibrous dysplasia cellular phenotype in human skeletal progenitors. J. Bone Miner. Res. 25(5), 1103–1116 (2010)PubMed
10.
go back to reference A. Sakamoto et al., Deficiency of the G-protein alpha-subunit G(s)alpha in osteoblasts leads to differential effects on trabecular and cortical bone. J. Biol. Chem. 280(22), 21369–21375 (2005)PubMedCrossRef A. Sakamoto et al., Deficiency of the G-protein alpha-subunit G(s)alpha in osteoblasts leads to differential effects on trabecular and cortical bone. J. Biol. Chem. 280(22), 21369–21375 (2005)PubMedCrossRef
11.
go back to reference E.C. Hsiao et al., Osteoblast expression of an engineered Gs-coupled receptor dramatically increases bone mass. Proc. Natl. Acad. Sci. USA 105(4), 1209–1214 (2008)PubMedCrossRef E.C. Hsiao et al., Osteoblast expression of an engineered Gs-coupled receptor dramatically increases bone mass. Proc. Natl. Acad. Sci. USA 105(4), 1209–1214 (2008)PubMedCrossRef
12.
go back to reference L.M. Calvi et al., Activated parathyroid hormone/parathyroid hormone-related protein receptor in osteoblastic cells differentially affects cortical and trabecular bone. J. Clin. Invest. 107(3), 277–286 (2001)PubMedCrossRef L.M. Calvi et al., Activated parathyroid hormone/parathyroid hormone-related protein receptor in osteoblastic cells differentially affects cortical and trabecular bone. J. Clin. Invest. 107(3), 277–286 (2001)PubMedCrossRef
13.
go back to reference J. Peng et al., Conditional expression of a Gi-coupled receptor in osteoblasts results in trabecular osteopenia. Endocrinology 149(3), 1329–1337 (2007)PubMedCrossRef J. Peng et al., Conditional expression of a Gi-coupled receptor in osteoblasts results in trabecular osteopenia. Endocrinology 149(3), 1329–1337 (2007)PubMedCrossRef
14.
go back to reference R.C. Gensure, T.J. Gardella, H. Juppner, Parathyroid hormone and parathyroid hormone-related peptide, and their receptors. Biochem. Biophys. Res. Commun. 328(3), 666–678 (2005)PubMedCrossRef R.C. Gensure, T.J. Gardella, H. Juppner, Parathyroid hormone and parathyroid hormone-related peptide, and their receptors. Biochem. Biophys. Res. Commun. 328(3), 666–678 (2005)PubMedCrossRef
15.
go back to reference H. Juppner et al., A G protein-linked receptor for parathyroid hormone and parathyroid hormone-related peptide. Science 254(5034), 1024–1026 (1991)PubMedCrossRef H. Juppner et al., A G protein-linked receptor for parathyroid hormone and parathyroid hormone-related peptide. Science 254(5034), 1024–1026 (1991)PubMedCrossRef
16.
go back to reference W.F. Schwindinger et al., Coupling of the PTH/PTHrP receptor to multiple G-proteins. Direct demonstration of receptor activation of Gs, Gq/11, and Gi(1) by [alpha-32P]GTP-gamma-azidoanilide photoaffinity labeling. Endocrine 8(2), 201–209 (1998)PubMedCrossRef W.F. Schwindinger et al., Coupling of the PTH/PTHrP receptor to multiple G-proteins. Direct demonstration of receptor activation of Gs, Gq/11, and Gi(1) by [alpha-32P]GTP-gamma-azidoanilide photoaffinity labeling. Endocrine 8(2), 201–209 (1998)PubMedCrossRef
17.
go back to reference C. Ge et al., Critical role of the extracellular signal-regulated kinase-MAPK pathway in osteoblast differentiation and skeletal development. J. Cell Biol. 176(5), 709–718 (2007)PubMedCrossRef C. Ge et al., Critical role of the extracellular signal-regulated kinase-MAPK pathway in osteoblast differentiation and skeletal development. J. Cell Biol. 176(5), 709–718 (2007)PubMedCrossRef
18.
go back to reference A. Rey et al., Proline-rich motifs in the parathyroid hormone (PTH)/PTH-related protein receptor C terminus mediate scaffolding of c-Src with beta-arrestin2 for ERK1/2 activation. J. Biol. Chem. 281(50), 38181–38188 (2006)PubMedCrossRef A. Rey et al., Proline-rich motifs in the parathyroid hormone (PTH)/PTH-related protein receptor C terminus mediate scaffolding of c-Src with beta-arrestin2 for ERK1/2 activation. J. Biol. Chem. 281(50), 38181–38188 (2006)PubMedCrossRef
19.
go back to reference R. Armamento-Villareal et al., An intact N terminus is required for the anabolic action of parathyroid hormone on adult female rats. J. Bone Miner. Res. 12(3), 384–392 (1997)PubMedCrossRef R. Armamento-Villareal et al., An intact N terminus is required for the anabolic action of parathyroid hormone on adult female rats. J. Bone Miner. Res. 12(3), 384–392 (1997)PubMedCrossRef
20.
go back to reference S. Hilliker et al., Truncation of the amino terminus of PTH alters its anabolic activity on bone in vivo. Bone 19(5), 469–477 (1996)PubMedCrossRef S. Hilliker et al., Truncation of the amino terminus of PTH alters its anabolic activity on bone in vivo. Bone 19(5), 469–477 (1996)PubMedCrossRef
21.
go back to reference R.H. Rixon et al., Parathyroid hormone fragments may stimulate bone growth in ovariectomized rats by activating adenylyl cyclase. J. Bone Miner. Res. 9(8), 1179–1189 (1994)PubMedCrossRef R.H. Rixon et al., Parathyroid hormone fragments may stimulate bone growth in ovariectomized rats by activating adenylyl cyclase. J. Bone Miner. Res. 9(8), 1179–1189 (1994)PubMedCrossRef
22.
go back to reference J.F. Whitfield et al., Stimulation of the growth of femoral trabecular bone in ovariectomized rats by the novel parathyroid hormone fragment, hPTH-(1-31)NH2 (Ostabolin). Calcif. Tissue Int. 58(2), 81–87 (1996)PubMedCrossRef J.F. Whitfield et al., Stimulation of the growth of femoral trabecular bone in ovariectomized rats by the novel parathyroid hormone fragment, hPTH-(1-31)NH2 (Ostabolin). Calcif. Tissue Int. 58(2), 81–87 (1996)PubMedCrossRef
23.
go back to reference A. Hollnagel, M. Ahrens, G. Gross, Parathyroid hormone enhances early and suppresses late stages of osteogenic and chondrogenic development in a BMP-dependent mesenchymal differentiation system (C3H10T1/2). J. Bone Miner. Res. 12(12), 1993–2004 (1997)PubMedCrossRef A. Hollnagel, M. Ahrens, G. Gross, Parathyroid hormone enhances early and suppresses late stages of osteogenic and chondrogenic development in a BMP-dependent mesenchymal differentiation system (C3H10T1/2). J. Bone Miner. Res. 12(12), 1993–2004 (1997)PubMedCrossRef
24.
go back to reference T. Akune et al., PPARgamma insufficiency enhances osteogenesis through osteoblast formation from bone marrow progenitors. J. Clin. Invest. 113(6), 846–855 (2004)PubMed T. Akune et al., PPARgamma insufficiency enhances osteogenesis through osteoblast formation from bone marrow progenitors. J. Clin. Invest. 113(6), 846–855 (2004)PubMed
25.
go back to reference R.F. Klein et al., Regulation of bone mass in mice by the lipoxygenase gene Alox15. Science 303(5655), 229–232 (2004)PubMedCrossRef R.F. Klein et al., Regulation of bone mass in mice by the lipoxygenase gene Alox15. Science 303(5655), 229–232 (2004)PubMedCrossRef
26.
go back to reference D.J. Rickard et al., Intermittent treatment with parathyroid hormone (PTH) as well as a non-peptide small molecule agonist of the PTH1 receptor inhibits adipocyte differentiation in human bone marrow stromal cells. Bone 39(6), 1361–1372 (2006)PubMedCrossRef D.J. Rickard et al., Intermittent treatment with parathyroid hormone (PTH) as well as a non-peptide small molecule agonist of the PTH1 receptor inhibits adipocyte differentiation in human bone marrow stromal cells. Bone 39(6), 1361–1372 (2006)PubMedCrossRef
27.
go back to reference Y. Tintut et al., cAMP stimulates osteoblast-like differentiation of calcifying vascular cells. Potential signaling pathway for vascular calcification. J. Biol. Chem. 273(13), 7547–7553 (1998)PubMedCrossRef Y. Tintut et al., cAMP stimulates osteoblast-like differentiation of calcifying vascular cells. Potential signaling pathway for vascular calcification. J. Biol. Chem. 273(13), 7547–7553 (1998)PubMedCrossRef
28.
go back to reference R. Siddappa et al., cAMP/PKA pathway activation in human mesenchymal stem cells in vitro results in robust bone formation in vivo. Proc. Natl. Acad. Sci. USA 105(20), 7281–7286 (2008)PubMedCrossRef R. Siddappa et al., cAMP/PKA pathway activation in human mesenchymal stem cells in vitro results in robust bone formation in vivo. Proc. Natl. Acad. Sci. USA 105(20), 7281–7286 (2008)PubMedCrossRef
29.
go back to reference J. Doorn et al., Forskolin enhances in vivo bone formation by human mesenchymal stromal cells. Tissue Eng. Part A 18(5–6), 558–567 (2012)PubMedCrossRef J. Doorn et al., Forskolin enhances in vivo bone formation by human mesenchymal stromal cells. Tissue Eng. Part A 18(5–6), 558–567 (2012)PubMedCrossRef
30.
go back to reference A. Scutt, N. Beier, C. Fittschen, EMD273316 & EMD95833, type 4 phosphodiesterase inhibitors, stimulate fibroblastic-colony formation by bone marrow cells via direct inhibition of PDE4 and the induction of endogenous prostaglandin synthesis. BMC Pharmacol. 4, 10 (2004)PubMedCrossRef A. Scutt, N. Beier, C. Fittschen, EMD273316 & EMD95833, type 4 phosphodiesterase inhibitors, stimulate fibroblastic-colony formation by bone marrow cells via direct inhibition of PDE4 and the induction of endogenous prostaglandin synthesis. BMC Pharmacol. 4, 10 (2004)PubMedCrossRef
31.
go back to reference Y. Zhao, S. Ding, A high-throughput siRNA library screen identifies osteogenic suppressors in human mesenchymal stem cells. Proc. Natl. Acad. Sci. USA 104(23), 9673–9678 (2007)PubMedCrossRef Y. Zhao, S. Ding, A high-throughput siRNA library screen identifies osteogenic suppressors in human mesenchymal stem cells. Proc. Natl. Acad. Sci. USA 104(23), 9673–9678 (2007)PubMedCrossRef
32.
go back to reference D.C. Yang et al., cAMP/PKA regulates osteogenesis, adipogenesis and ratio of RANKL/OPG mRNA expression in mesenchymal stem cells by suppressing leptin. PLoS One 3(2), e1540 (2008)PubMedCrossRef D.C. Yang et al., cAMP/PKA regulates osteogenesis, adipogenesis and ratio of RANKL/OPG mRNA expression in mesenchymal stem cells by suppressing leptin. PLoS One 3(2), e1540 (2008)PubMedCrossRef
33.
go back to reference R. Siddappa et al., cAMP/PKA signaling inhibits osteogenic differentiation and bone formation in rodent models. Tissue Eng. Part A 15(8), 2135–2143 (2009)PubMedCrossRef R. Siddappa et al., cAMP/PKA signaling inhibits osteogenic differentiation and bone formation in rodent models. Tissue Eng. Part A 15(8), 2135–2143 (2009)PubMedCrossRef
34.
go back to reference Y. Gong et al., LDL receptor-related protein 5 (LRP5) affects bone accrual and eye development. Cell 107(4), 513–523 (2001)PubMedCrossRef Y. Gong et al., LDL receptor-related protein 5 (LRP5) affects bone accrual and eye development. Cell 107(4), 513–523 (2001)PubMedCrossRef
35.
go back to reference L.M. Boyden et al., High bone density due to a mutation in LDL-receptor-related protein 5. N. Engl. J. Med. 346(20), 1513–1521 (2002)PubMedCrossRef L.M. Boyden et al., High bone density due to a mutation in LDL-receptor-related protein 5. N. Engl. J. Med. 346(20), 1513–1521 (2002)PubMedCrossRef
36.
go back to reference R.D. Little et al., A mutation in the LDL receptor-related protein 5 gene results in the autosomal dominant high-bone-mass trait. Am. J. Hum. Genet. 70(1), 11–19 (2002)PubMedCrossRef R.D. Little et al., A mutation in the LDL receptor-related protein 5 gene results in the autosomal dominant high-bone-mass trait. Am. J. Hum. Genet. 70(1), 11–19 (2002)PubMedCrossRef
37.
go back to reference T.F. Day et al., Wnt/beta-catenin signaling in mesenchymal progenitors controls osteoblast and chondrocyte differentiation during vertebrate skeletogenesis. Dev. Cell 8(5), 739–750 (2005)PubMedCrossRef T.F. Day et al., Wnt/beta-catenin signaling in mesenchymal progenitors controls osteoblast and chondrocyte differentiation during vertebrate skeletogenesis. Dev. Cell 8(5), 739–750 (2005)PubMedCrossRef
38.
go back to reference T. Gaur et al., Canonical WNT signaling promotes osteogenesis by directly stimulating Runx2 gene expression. J. Biol. Chem. 280(39), 33132–33140 (2005)PubMedCrossRef T. Gaur et al., Canonical WNT signaling promotes osteogenesis by directly stimulating Runx2 gene expression. J. Biol. Chem. 280(39), 33132–33140 (2005)PubMedCrossRef
39.
go back to reference R.A. Terkeltaub, Inorganic pyrophosphate generation and disposition in pathophysiology. Am. J. Physiol. Cell Physiol. 281(1), C1–C11 (2001)PubMed R.A. Terkeltaub, Inorganic pyrophosphate generation and disposition in pathophysiology. Am. J. Physiol. Cell Physiol. 281(1), C1–C11 (2001)PubMed
40.
go back to reference A. Baba et al., PKA-dependent regulation of the histone lysine demethylase complex PHF2-ARID5B. Nat. Cell Biol. 13(6), 668–675 (2011)PubMedCrossRef A. Baba et al., PKA-dependent regulation of the histone lysine demethylase complex PHF2-ARID5B. Nat. Cell Biol. 13(6), 668–675 (2011)PubMedCrossRef
41.
go back to reference J. Guo et al., Suppression of Wnt signaling by Dkk1 attenuates PTH-mediated stromal cell response and new bone formation. Cell. Metab. 11(2), 161–171 (2010)PubMedCrossRef J. Guo et al., Suppression of Wnt signaling by Dkk1 attenuates PTH-mediated stromal cell response and new bone formation. Cell. Metab. 11(2), 161–171 (2010)PubMedCrossRef
42.
go back to reference C. O’Brien et al., Activation of PTH receptor 1 specifically in osteocytes suppresses Sost expression and increases bone mass in transgenic mice. J. Bone Miner. Res. 20, S4 (2006) C. O’Brien et al., Activation of PTH receptor 1 specifically in osteocytes suppresses Sost expression and increases bone mass in transgenic mice. J. Bone Miner. Res. 20, S4 (2006)
43.
go back to reference N.H. Kulkarni et al., Effects of parathyroid hormone on Wnt signaling pathway in bone. J. Cell. Biochem. 95(6), 1178–1190 (2005)PubMedCrossRef N.H. Kulkarni et al., Effects of parathyroid hormone on Wnt signaling pathway in bone. J. Cell. Biochem. 95(6), 1178–1190 (2005)PubMedCrossRef
44.
go back to reference G. Liu et al., Canonical Wnts function as potent regulators of osteogenesis by human mesenchymal stem cells. J. Cell Biol. 185(1), 67–75 (2009)PubMedCrossRef G. Liu et al., Canonical Wnts function as potent regulators of osteogenesis by human mesenchymal stem cells. J. Cell Biol. 185(1), 67–75 (2009)PubMedCrossRef
45.
go back to reference N. Quarto, B. Behr, M.T. Longaker, Opposite spectrum of activity of canonical Wnt signaling in the osteogenic context of undifferentiated and differentiated mesenchymal cells: implications for tissue engineering. Tissue Eng. Part A 16(10), 3185–3197 (2010)PubMedCrossRef N. Quarto, B. Behr, M.T. Longaker, Opposite spectrum of activity of canonical Wnt signaling in the osteogenic context of undifferentiated and differentiated mesenchymal cells: implications for tissue engineering. Tissue Eng. Part A 16(10), 3185–3197 (2010)PubMedCrossRef
46.
go back to reference G.M. Boland et al., Wnt 3a promotes proliferation and suppresses osteogenic differentiation of adult human mesenchymal stem cells. J. Cell. Biochem. 93(6), 1210–1230 (2004)PubMedCrossRef G.M. Boland et al., Wnt 3a promotes proliferation and suppresses osteogenic differentiation of adult human mesenchymal stem cells. J. Cell. Biochem. 93(6), 1210–1230 (2004)PubMedCrossRef
47.
go back to reference H. Zhou et al., Osteoblasts directly control lineage commitment of mesenchymal progenitor cells through Wnt signaling. J. Biol. Chem. 283(4), 1936–1945 (2008)PubMedCrossRef H. Zhou et al., Osteoblasts directly control lineage commitment of mesenchymal progenitor cells through Wnt signaling. J. Biol. Chem. 283(4), 1936–1945 (2008)PubMedCrossRef
48.
go back to reference M. Terauchi et al., T lymphocytes amplify the anabolic activity of parathyroid hormone through Wnt10b signaling. Cell Metab. 10(3), 229–240 (2009)PubMedCrossRef M. Terauchi et al., T lymphocytes amplify the anabolic activity of parathyroid hormone through Wnt10b signaling. Cell Metab. 10(3), 229–240 (2009)PubMedCrossRef
49.
go back to reference P.H. Luiz de Freitas et al., Intermittent PTH administration stimulates pre-osteoblastic proliferation without leading to enhanced bone formation in osteoclast-less c-fos(−/−) mice. J. Bone Miner. Res. 24(9), 1586–1597 (2009)PubMedCrossRef P.H. Luiz de Freitas et al., Intermittent PTH administration stimulates pre-osteoblastic proliferation without leading to enhanced bone formation in osteoclast-less c-fos(−/−) mice. J. Bone Miner. Res. 24(9), 1586–1597 (2009)PubMedCrossRef
50.
go back to reference S.A. Kuznetsov et al., The interplay of osteogenesis and hematopoiesis: expression of a constitutively active PTH/PTHrP receptor in osteogenic cells perturbs the establishment of hematopoiesis in bone and of skeletal stem cells in the bone marrow. J. Cell Biol. 167(6), 1113–1122 (2004)PubMedCrossRef S.A. Kuznetsov et al., The interplay of osteogenesis and hematopoiesis: expression of a constitutively active PTH/PTHrP receptor in osteogenic cells perturbs the establishment of hematopoiesis in bone and of skeletal stem cells in the bone marrow. J. Cell Biol. 167(6), 1113–1122 (2004)PubMedCrossRef
51.
go back to reference M. Katebi, M. Soleimani, B.N. Cronstein, Adenosine A2A receptors play an active role in mouse bone marrow-derived mesenchymal stem cell development. J. Leukoc. Biol. 85(3), 438–444 (2009)PubMedCrossRef M. Katebi, M. Soleimani, B.N. Cronstein, Adenosine A2A receptors play an active role in mouse bone marrow-derived mesenchymal stem cell development. J. Leukoc. Biol. 85(3), 438–444 (2009)PubMedCrossRef
52.
go back to reference S.H. Carroll et al., The A2B adenosine receptor promotes mesenchymal stem cell differentiation to osteoblasts and bone formation in vivo. J. Biol. Chem. 287(19), 15718–15727 (2012)PubMedCrossRef S.H. Carroll et al., The A2B adenosine receptor promotes mesenchymal stem cell differentiation to osteoblasts and bone formation in vivo. J. Biol. Chem. 287(19), 15718–15727 (2012)PubMedCrossRef
53.
go back to reference B. Gharibi et al., Contrasting effects of A1 and A2b adenosine receptors on adipogenesis. Int. J. Obes. (Lond.) 36(3), 397–406 (2012)CrossRef B. Gharibi et al., Contrasting effects of A1 and A2b adenosine receptors on adipogenesis. Int. J. Obes. (Lond.) 36(3), 397–406 (2012)CrossRef
Metadata
Title
Cyclic AMP signaling in bone marrow stromal cells has reciprocal effects on the ability of mesenchymal stem cells to differentiate into mature osteoblasts versus mature adipocytes
Authors
Richard Kao
Weidar Lu
Alyssa Louie
Robert Nissenson
Publication date
01-12-2012
Publisher
Springer US
Published in
Endocrine / Issue 3/2012
Print ISSN: 1355-008X
Electronic ISSN: 1559-0100
DOI
https://doi.org/10.1007/s12020-012-9717-9

Other articles of this Issue 3/2012

Endocrine 3/2012 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.