Skip to main content
Erschienen in: Endocrine 1/2016

01.01.2016 | Review

Role of the steroidogenic acute regulatory protein in health and disease

verfasst von: Pulak R. Manna, Cloyce L. Stetson, Andrzej T. Slominski, Kevin Pruitt

Erschienen in: Endocrine | Ausgabe 1/2016

Einloggen, um Zugang zu erhalten

Abstract

Steroid hormones are an important class of regulatory molecules that are synthesized in steroidogenic cells of the adrenal, ovary, testis, placenta, brain, and skin, and influence a spectrum of developmental and physiological processes. The steroidogenic acute regulatory protein (STAR) predominantly mediates the rate-limiting step in steroid biosynthesis, i.e., the transport of the substrate of all steroid hormones, cholesterol, from the outer to the inner mitochondrial membrane. At the inner membrane, cytochrome P450 cholesterol side chain cleavage enzyme cleaves the cholesterol side chain to form the first steroid, pregnenolone, which is converted by a series of enzymes to various steroid hormones in specific tissues. Both basic and clinical evidence have demonstrated the crucial involvement of the STAR protein in the regulation of steroid biosynthesis. Multiple levels of regulation impinge on STAR action. Recent findings demonstrate that hormone-sensitive lipase, through its action on the hydrolysis of cholesteryl esters, plays an important role in regulating STAR expression and steroidogenesis which involve the liver X receptor pathway. Activation of the latter influences macrophage cholesterol efflux that is a key process in the prevention of atherosclerotic cardiovascular disease. Appropriate regulation of steroid hormones is vital for proper functioning of many important biological activities, which are also paramount for geriatric populations to live longer and healthier. This review summarizes the current level of understanding on tissue-specific and hormone-induced regulation of STAR expression and steroidogenesis, and provides insights into a number of cholesterol and/or steroid coupled physiological and pathophysiological consequences.
Literatur
1.
Zurück zum Zitat S. Azhar, E. Reaven, Scavenger receptor class BI and selective cholesteryl ester uptake: partners in the regulation of steroidogenesis. Mol. Cell. Endocrinol. 195, 1–26 (2002)PubMedCrossRef S. Azhar, E. Reaven, Scavenger receptor class BI and selective cholesteryl ester uptake: partners in the regulation of steroidogenesis. Mol. Cell. Endocrinol. 195, 1–26 (2002)PubMedCrossRef
2.
Zurück zum Zitat F.B. Kraemer, W.J. Shen, K. Harada, S. Patel, J. Osuga, S. Ishibashi, S. Azhar, Hormone-sensitive lipase is required for high-density lipoprotein cholesteryl ester-supported adrenal steroidogenesis. Mol. Endocrinol. 18, 549–557 (2004)PubMedCrossRef F.B. Kraemer, W.J. Shen, K. Harada, S. Patel, J. Osuga, S. Ishibashi, S. Azhar, Hormone-sensitive lipase is required for high-density lipoprotein cholesteryl ester-supported adrenal steroidogenesis. Mol. Endocrinol. 18, 549–557 (2004)PubMedCrossRef
3.
Zurück zum Zitat P.R. Manna, M.T. Dyson, D.M. Stocco, Regulation of the steroidogenic acute regulatory protein gene expression: present and future perspectives. Mol. Hum. Reprod. 15, 321–333 (2009)PubMedPubMedCentralCrossRef P.R. Manna, M.T. Dyson, D.M. Stocco, Regulation of the steroidogenic acute regulatory protein gene expression: present and future perspectives. Mol. Hum. Reprod. 15, 321–333 (2009)PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat P.R. Manna, D.M. Stocco, Regulation of the steroidogenic acute regulatory protein expression: functional and physiological consequences. Curr. Drug Targets Immune Endocr. Metabol. Disord. 5, 93–108 (2005)PubMedCrossRef P.R. Manna, D.M. Stocco, Regulation of the steroidogenic acute regulatory protein expression: functional and physiological consequences. Curr. Drug Targets Immune Endocr. Metabol. Disord. 5, 93–108 (2005)PubMedCrossRef
5.
Zurück zum Zitat D.M. Stocco, X. Wang, Y. Jo, P.R. Manna, Multiple signaling pathways regulating steroidogenesis and steroidogenic acute regulatory protein expression: more complicated than we thought. Mol. Endocrinol. 19, 2647–2659 (2005)PubMedCrossRef D.M. Stocco, X. Wang, Y. Jo, P.R. Manna, Multiple signaling pathways regulating steroidogenesis and steroidogenic acute regulatory protein expression: more complicated than we thought. Mol. Endocrinol. 19, 2647–2659 (2005)PubMedCrossRef
7.
Zurück zum Zitat A.F. Castillo, U. Orlando, K.E. Helfenberger, C. Poderoso, E.J. Podesta, The role of mitochondrial fusion and StAR phosphorylation in the regulation of StAR activity and steroidogenesis. Mol. Cell. Endocrinol. 408, 73–79 (2015)PubMedCrossRef A.F. Castillo, U. Orlando, K.E. Helfenberger, C. Poderoso, E.J. Podesta, The role of mitochondrial fusion and StAR phosphorylation in the regulation of StAR activity and steroidogenesis. Mol. Cell. Endocrinol. 408, 73–79 (2015)PubMedCrossRef
8.
Zurück zum Zitat D. Stocco, Star protein and the regulation of steroid hormone biosynthesis. Annu. Rev. Physiol. 63, 193–213 (2001)PubMedCrossRef D. Stocco, Star protein and the regulation of steroid hormone biosynthesis. Annu. Rev. Physiol. 63, 193–213 (2001)PubMedCrossRef
9.
Zurück zum Zitat M. Ascoli, F. Fanelli, D.L. Segaloff, The lutropin/choriogonadotropin receptor, a 2002 perspective. Endocr. Rev. 23, 141–174 (2002)PubMedCrossRef M. Ascoli, F. Fanelli, D.L. Segaloff, The lutropin/choriogonadotropin receptor, a 2002 perspective. Endocr. Rev. 23, 141–174 (2002)PubMedCrossRef
10.
Zurück zum Zitat W.L. Miller, StAR search–what we know about how the steroidogenic acute regulatory protein mediates mitochondrial cholesterol import. Mol. Endocrinol. 21, 589–601 (2007)PubMedCrossRef W.L. Miller, StAR search–what we know about how the steroidogenic acute regulatory protein mediates mitochondrial cholesterol import. Mol. Endocrinol. 21, 589–601 (2007)PubMedCrossRef
11.
Zurück zum Zitat W.L. Miller, R.J. Auchus, The molecular biology, biochemistry, and physiology of human steroidogenesis and its disorders. Endocr. Rev. 32, 81–151 (2011)PubMedPubMedCentralCrossRef W.L. Miller, R.J. Auchus, The molecular biology, biochemistry, and physiology of human steroidogenesis and its disorders. Endocr. Rev. 32, 81–151 (2011)PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat P.R. Manna, J. Cohen-Tannoudji, R. Counis, C.W. Garner, I. Huhtaniemi, F.B. Kraemer, D.M. Stocco, Mechanisms of action of hormone-sensitive lipase in mouse Leydig cells: its role in the regulation of the steroidogenic acute regulatory protein. J. Biol. Chem. 288, 8505–8518 (2013)PubMedPubMedCentralCrossRef P.R. Manna, J. Cohen-Tannoudji, R. Counis, C.W. Garner, I. Huhtaniemi, F.B. Kraemer, D.M. Stocco, Mechanisms of action of hormone-sensitive lipase in mouse Leydig cells: its role in the regulation of the steroidogenic acute regulatory protein. J. Biol. Chem. 288, 8505–8518 (2013)PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat F. Arakane, C.B. Kallen, H. Watari, J.A. Foster, N.B. Sepuri, D. Pain, S.E. Stayrook, M. Lewis, G.L. Gerton, J.F. Strauss 3rd, The mechanism of action of steroidogenic acute regulatory protein (StAR). StAR acts on the outside of mitochondria to stimulate steroidogenesis. J. Biol. Chem. 273, 16339–16345 (1998)PubMedCrossRef F. Arakane, C.B. Kallen, H. Watari, J.A. Foster, N.B. Sepuri, D. Pain, S.E. Stayrook, M. Lewis, G.L. Gerton, J.F. Strauss 3rd, The mechanism of action of steroidogenic acute regulatory protein (StAR). StAR acts on the outside of mitochondria to stimulate steroidogenesis. J. Biol. Chem. 273, 16339–16345 (1998)PubMedCrossRef
14.
Zurück zum Zitat I.P. Artemenko, D. Zhao, D.B. Hales, K.H. Hales, C.R. Jefcoate, Mitochondrial processing of newly synthesized steroidogenic acute regulatory protein (StAR), but not total StAR, mediates cholesterol transfer to cytochrome P450 side chain cleavage enzyme in adrenal cells. J. Biol. Chem. 276, 46583–46596 (2001)PubMedCrossRef I.P. Artemenko, D. Zhao, D.B. Hales, K.H. Hales, C.R. Jefcoate, Mitochondrial processing of newly synthesized steroidogenic acute regulatory protein (StAR), but not total StAR, mediates cholesterol transfer to cytochrome P450 side chain cleavage enzyme in adrenal cells. J. Biol. Chem. 276, 46583–46596 (2001)PubMedCrossRef
15.
Zurück zum Zitat J. Liu, M.B. Rone, V. Papadopoulos, Protein–protein interactions mediate mitochondrial cholesterol transport and steroid biosynthesis. J. Biol. Chem. 281, 38879–38893 (2006)PubMedCrossRef J. Liu, M.B. Rone, V. Papadopoulos, Protein–protein interactions mediate mitochondrial cholesterol transport and steroid biosynthesis. J. Biol. Chem. 281, 38879–38893 (2006)PubMedCrossRef
16.
Zurück zum Zitat J. Fan, J. Liu, M. Culty, V. Papadopoulos, Acyl-coenzyme A binding domain containing 3 (ACBD3; PAP7; GCP60): an emerging signaling molecule. Prog. Lipid Res. 49, 218–234 (2010)PubMedPubMedCentralCrossRef J. Fan, J. Liu, M. Culty, V. Papadopoulos, Acyl-coenzyme A binding domain containing 3 (ACBD3; PAP7; GCP60): an emerging signaling molecule. Prog. Lipid Res. 49, 218–234 (2010)PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat M.B. Rone, A.S. Midzak, L. Issop, G. Rammouz, S. Jagannathan, J. Fan, X. Ye, J. Blonder, T. Veenstra, V. Papadopoulos, Identification of a dynamic mitochondrial protein complex driving cholesterol import, trafficking, and metabolism to steroid hormones. Mol. Endocrinol. 26, 1868–1882 (2012)PubMedCrossRef M.B. Rone, A.S. Midzak, L. Issop, G. Rammouz, S. Jagannathan, J. Fan, X. Ye, J. Blonder, T. Veenstra, V. Papadopoulos, Identification of a dynamic mitochondrial protein complex driving cholesterol import, trafficking, and metabolism to steroid hormones. Mol. Endocrinol. 26, 1868–1882 (2012)PubMedCrossRef
18.
Zurück zum Zitat C. Poderoso, A. Duarte, M. Cooke, U. Orlando, V. Gottifredi, A.R. Solano, J.R. Lemos, E.J. Podesta, The spatial and temporal regulation of the hormonal signal. Role of mitochondria in the formation of a protein complex required for the activation of cholesterol transport and steroids synthesis. Mol. Cell. Endocrinol. 371, 26–33 (2013)PubMedCrossRef C. Poderoso, A. Duarte, M. Cooke, U. Orlando, V. Gottifredi, A.R. Solano, J.R. Lemos, E.J. Podesta, The spatial and temporal regulation of the hormonal signal. Role of mitochondria in the formation of a protein complex required for the activation of cholesterol transport and steroids synthesis. Mol. Cell. Endocrinol. 371, 26–33 (2013)PubMedCrossRef
19.
Zurück zum Zitat U. Orlando, M. Cooke, F. Cornejo Maciel, V. Papadopoulos, P. Maloberti, E.J. Podesta, Characterization of the mouse promoter region of the acyl-CoA synthetase 4 gene: role of Sp1 and CREB. Mol. Cell. Endocrinol. 369, 15–26 (2013)PubMedCrossRef U. Orlando, M. Cooke, F. Cornejo Maciel, V. Papadopoulos, P. Maloberti, E.J. Podesta, Characterization of the mouse promoter region of the acyl-CoA synthetase 4 gene: role of Sp1 and CREB. Mol. Cell. Endocrinol. 369, 15–26 (2013)PubMedCrossRef
20.
Zurück zum Zitat J. Fan, E. Campioli, A. Midzak, M. Culty, V. Papadopoulos, Conditional steroidogenic cell-targeted deletion of TSPO unveils a crucial role in viability and hormone-dependent steroid formation. Proc. Natl. Acad. Sci. 112, 7261–7266 (2015)PubMedPubMedCentralCrossRef J. Fan, E. Campioli, A. Midzak, M. Culty, V. Papadopoulos, Conditional steroidogenic cell-targeted deletion of TSPO unveils a crucial role in viability and hormone-dependent steroid formation. Proc. Natl. Acad. Sci. 112, 7261–7266 (2015)PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat B.J. Clark, J. Wells, S.R. King, D.M. Stocco, The purification, cloning, and expression of a novel luteinizing hormone-induced mitochondrial protein in MA-10 mouse Leydig tumor cells. Characterization of the steroidogenic acute regulatory protein (StAR). J. Biol. Chem. 269, 28314–28322 (1994)PubMed B.J. Clark, J. Wells, S.R. King, D.M. Stocco, The purification, cloning, and expression of a novel luteinizing hormone-induced mitochondrial protein in MA-10 mouse Leydig tumor cells. Characterization of the steroidogenic acute regulatory protein (StAR). J. Biol. Chem. 269, 28314–28322 (1994)PubMed
22.
Zurück zum Zitat D.M. Stocco, B.J. Clark, Regulation of the acute production of steroids in steroidogenic cells. Endocr. Rev. 17, 221–244 (1996)PubMed D.M. Stocco, B.J. Clark, Regulation of the acute production of steroids in steroidogenic cells. Endocr. Rev. 17, 221–244 (1996)PubMed
23.
Zurück zum Zitat D. Lin, T. Sugawara, J.F. Strauss III, B.J. Clark, D.M. Stocco, P. Saenger, A. Rogol, W.L. Miller, Role of steroidogenic acute regulatory protein in adrenal and gonadal steroidogenesis. Science 267, 1828–1831 (1995)PubMedCrossRef D. Lin, T. Sugawara, J.F. Strauss III, B.J. Clark, D.M. Stocco, P. Saenger, A. Rogol, W.L. Miller, Role of steroidogenic acute regulatory protein in adrenal and gonadal steroidogenesis. Science 267, 1828–1831 (1995)PubMedCrossRef
24.
Zurück zum Zitat W.L. Miller, Steroidogenic acute regulatory protein (StAR), a novel mitochondrial cholesterol transporter. Biochim. Biophys. Acta 1771, 663–676 (2007)PubMedCrossRef W.L. Miller, Steroidogenic acute regulatory protein (StAR), a novel mitochondrial cholesterol transporter. Biochim. Biophys. Acta 1771, 663–676 (2007)PubMedCrossRef
25.
Zurück zum Zitat H. Bose, V.R. Lingappa, W.L. Miller, Rapid regulation of steroidogenesis by mitochondrial protein import. Nature 417, 87–91 (2002)PubMedCrossRef H. Bose, V.R. Lingappa, W.L. Miller, Rapid regulation of steroidogenesis by mitochondrial protein import. Nature 417, 87–91 (2002)PubMedCrossRef
26.
Zurück zum Zitat H.S. Bose, T. Sugawara, J.F. Strauss 3rd, W.L. Miller, The pathophysiology and genetics of congenital lipoid adrenal hyperplasia. International Congenital Lipoid Adrenal Hyperplasia Consortium. N. Engl. J. Med. 335, 1870–1878 (1996)PubMedCrossRef H.S. Bose, T. Sugawara, J.F. Strauss 3rd, W.L. Miller, The pathophysiology and genetics of congenital lipoid adrenal hyperplasia. International Congenital Lipoid Adrenal Hyperplasia Consortium. N. Engl. J. Med. 335, 1870–1878 (1996)PubMedCrossRef
27.
Zurück zum Zitat D.M. Stocco, Clinical disorders associated with abnormal cholesterol transport: mutations in the steroidogenic acute regulatory protein. Mol. Cell. Endocrinol. 191, 19–25 (2002)PubMedCrossRef D.M. Stocco, Clinical disorders associated with abnormal cholesterol transport: mutations in the steroidogenic acute regulatory protein. Mol. Cell. Endocrinol. 191, 19–25 (2002)PubMedCrossRef
28.
Zurück zum Zitat S.R. King, A. Bhangoo, D.M. Stocco, Functional and physiological consequences of StAR deficiency: role in lipoid congenital adrenal hyperplasia. Endocr. Dev. 20, 47–53 (2011)PubMed S.R. King, A. Bhangoo, D.M. Stocco, Functional and physiological consequences of StAR deficiency: role in lipoid congenital adrenal hyperplasia. Endocr. Dev. 20, 47–53 (2011)PubMed
29.
Zurück zum Zitat R.J. Auchus, W.L. Miller, Congenital adrenal hyperplasia–more dogma bites the dust. J. Clin. Endocrinol. Metab. 97, 772–775 (2012)PubMedCrossRef R.J. Auchus, W.L. Miller, Congenital adrenal hyperplasia–more dogma bites the dust. J. Clin. Endocrinol. Metab. 97, 772–775 (2012)PubMedCrossRef
30.
Zurück zum Zitat K.M. Caron, S.C. Soo, W.C. Wetsel, D.M. Stocco, B.J. Clark, K.L. Parker, Targeted disruption of the mouse gene encoding steroidogenic acute regulatory protein provides insights into congenital lipoid adrenal hyperplasia. Proc. Natl. Acad. Sci. 94, 11540–11545 (1997)PubMedPubMedCentralCrossRef K.M. Caron, S.C. Soo, W.C. Wetsel, D.M. Stocco, B.J. Clark, K.L. Parker, Targeted disruption of the mouse gene encoding steroidogenic acute regulatory protein provides insights into congenital lipoid adrenal hyperplasia. Proc. Natl. Acad. Sci. 94, 11540–11545 (1997)PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat T. Hasegawa, L. Zhao, K.M. Caron, G. Majdic, T. Suzuki, S. Shizawa, H. Sasano, K.L. Parker, Developmental roles of the steroidogenic acute regulatory protein (StAR) as revealed by StAR knockout mice. Mol. Endocrinol. 14, 1462–1471 (2000)PubMedCrossRef T. Hasegawa, L. Zhao, K.M. Caron, G. Majdic, T. Suzuki, S. Shizawa, H. Sasano, K.L. Parker, Developmental roles of the steroidogenic acute regulatory protein (StAR) as revealed by StAR knockout mice. Mol. Endocrinol. 14, 1462–1471 (2000)PubMedCrossRef
32.
Zurück zum Zitat H.S. Bose, S. Sato, J. Aisenberg, S.A. Shalev, N. Matsuo, W.L. Miller, Mutations in the steroidogenic acute regulatory protein (StAR) in six patients with congenital lipoid adrenal hyperplasia. J. Clin. Endocrinol. Metab. 85, 3636–3639 (2000)PubMed H.S. Bose, S. Sato, J. Aisenberg, S.A. Shalev, N. Matsuo, W.L. Miller, Mutations in the steroidogenic acute regulatory protein (StAR) in six patients with congenital lipoid adrenal hyperplasia. J. Clin. Endocrinol. Metab. 85, 3636–3639 (2000)PubMed
33.
Zurück zum Zitat W.L. Miller, Steroid hormone synthesis in mitochondria. Mol. Cell. Endocrinol. 379, 62–73 (2013)PubMedCrossRef W.L. Miller, Steroid hormone synthesis in mitochondria. Mol. Cell. Endocrinol. 379, 62–73 (2013)PubMedCrossRef
35.
Zurück zum Zitat P.R. Manna, A.T. Slominski, S.R. King, C.L. Stetson, D.M. Stocco, Synergistic activation of steroidogenic acute regulatory protein expression and steroid biosynthesis by retinoids: involvement of cAMP/PKA signaling. Endocrinology 155, 576–591 (2014)PubMedPubMedCentralCrossRef P.R. Manna, A.T. Slominski, S.R. King, C.L. Stetson, D.M. Stocco, Synergistic activation of steroidogenic acute regulatory protein expression and steroid biosynthesis by retinoids: involvement of cAMP/PKA signaling. Endocrinology 155, 576–591 (2014)PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat J. Liu, H. Li, V. Papadopoulos, PAP7, a PBR/PKA-RIalpha-associated protein: a new element in the relay of the hormonal induction of steroidogenesis. J. Steroid Biochem. Mol. Biol. 85, 275–283 (2003)PubMedCrossRef J. Liu, H. Li, V. Papadopoulos, PAP7, a PBR/PKA-RIalpha-associated protein: a new element in the relay of the hormonal induction of steroidogenesis. J. Steroid Biochem. Mol. Biol. 85, 275–283 (2003)PubMedCrossRef
37.
Zurück zum Zitat S. Ghosh, B. Zhao, J. Bie, J. Song, Macrophage cholesteryl ester mobilization and atherosclerosis. Vasc. Pharmacol. 52, 1–10 (2010)CrossRef S. Ghosh, B. Zhao, J. Bie, J. Song, Macrophage cholesteryl ester mobilization and atherosclerosis. Vasc. Pharmacol. 52, 1–10 (2010)CrossRef
38.
Zurück zum Zitat J. Osuga, S. Ishibashi, T. Oka, H. Yagyu, R. Tozawa, A. Fujimoto, F. Shionoiri, N. Yahagi, F.B. Kraemer, O. Tsutsumi, N. Yamada, Targeted disruption of hormone-sensitive lipase results in male sterility and adipocyte hypertrophy, but not in obesity. Proc. Natl. Acad. Sci. 97, 787–792 (2000)PubMedPubMedCentralCrossRef J. Osuga, S. Ishibashi, T. Oka, H. Yagyu, R. Tozawa, A. Fujimoto, F. Shionoiri, N. Yahagi, F.B. Kraemer, O. Tsutsumi, N. Yamada, Targeted disruption of hormone-sensitive lipase results in male sterility and adipocyte hypertrophy, but not in obesity. Proc. Natl. Acad. Sci. 97, 787–792 (2000)PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat F.B. Kraemer, W.J. Shen, Hormone-sensitive lipase: control of intracellular tri-(di-)acylglycerol and cholesteryl ester hydrolysis. J. Lipid Res. 43, 1585–1594 (2002)PubMedCrossRef F.B. Kraemer, W.J. Shen, Hormone-sensitive lipase: control of intracellular tri-(di-)acylglycerol and cholesteryl ester hydrolysis. J. Lipid Res. 43, 1585–1594 (2002)PubMedCrossRef
40.
Zurück zum Zitat B. Zhao, J. Bie, J. Wang, S.A. Marqueen, S. Ghosh, Identification of a novel intracellular cholesteryl ester hydrolase (carboxylesterase 3) in human macrophages: compensatory increase in its expression after carboxylesterase 1 silencing. Am. J. Physiol. Cell Physiol. 303, C427–C435 (2012)PubMedPubMedCentralCrossRef B. Zhao, J. Bie, J. Wang, S.A. Marqueen, S. Ghosh, Identification of a novel intracellular cholesteryl ester hydrolase (carboxylesterase 3) in human macrophages: compensatory increase in its expression after carboxylesterase 1 silencing. Am. J. Physiol. Cell Physiol. 303, C427–C435 (2012)PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat K. Sakai, M. Igarashi, D. Yamamuro, T. Ohshiro, S. Nagashima, M. Takahashi, B. Enkhtuvshin, M. Sekiya, H. Okazaki, J. Osuga, S. Ishibashi, Critical role of neutral cholesteryl ester hydrolase 1 in cholesteryl ester hydrolysis in murine macrophages. J. Lipid Res. 55, 2033–2040 (2014)PubMedPubMedCentralCrossRef K. Sakai, M. Igarashi, D. Yamamuro, T. Ohshiro, S. Nagashima, M. Takahashi, B. Enkhtuvshin, M. Sekiya, H. Okazaki, J. Osuga, S. Ishibashi, Critical role of neutral cholesteryl ester hydrolase 1 in cholesteryl ester hydrolysis in murine macrophages. J. Lipid Res. 55, 2033–2040 (2014)PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat J.J. Repa, K.E. Berge, C. Pomajzl, J.A. Richardson, H. Hobbs, D.J. Mangelsdorf, Regulation of ATP-binding cassette sterol transporters ABCG5 and ABCG8 by the liver X receptors alpha and beta. J. Biol. Chem. 277, 18793–18800 (2002)PubMedCrossRef J.J. Repa, K.E. Berge, C. Pomajzl, J.A. Richardson, H. Hobbs, D.J. Mangelsdorf, Regulation of ATP-binding cassette sterol transporters ABCG5 and ABCG8 by the liver X receptors alpha and beta. J. Biol. Chem. 277, 18793–18800 (2002)PubMedCrossRef
43.
Zurück zum Zitat C.L. Cummins, D.H. Volle, Y. Zhang, J.G. McDonald, B. Sion, A.M. Lefrancois-Martinez, F. Caira, G. Veyssiere, D.J. Mangelsdorf, J.M. Lobaccaro, Liver X receptors regulate adrenal cholesterol balance. J. Clin. Investig. 116, 1902–1912 (2006)PubMedPubMedCentralCrossRef C.L. Cummins, D.H. Volle, Y. Zhang, J.G. McDonald, B. Sion, A.M. Lefrancois-Martinez, F. Caira, G. Veyssiere, D.J. Mangelsdorf, J.M. Lobaccaro, Liver X receptors regulate adrenal cholesterol balance. J. Clin. Investig. 116, 1902–1912 (2006)PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat D.H. Volle, J.M. Lobaccaro, Role of the nuclear receptors for oxysterols LXRs in steroidogenic tissues: beyond the “foie gras”, the steroids and sex? Mol. Cell. Endocrinol. 265–266, 183–189 (2007)PubMedCrossRef D.H. Volle, J.M. Lobaccaro, Role of the nuclear receptors for oxysterols LXRs in steroidogenic tissues: beyond the “foie gras”, the steroids and sex? Mol. Cell. Endocrinol. 265–266, 183–189 (2007)PubMedCrossRef
45.
Zurück zum Zitat P.R. Manna, D.W. Eubank, E. Lalli, P. Sassone-Corsi, D.M. Stocco, Transcriptional regulation of the mouse steroidogenic acute regulatory protein gene by the cAMP response-element binding protein and steroidogenic factor 1. J. Mol. Endocrinol. 30, 381–397 (2003)PubMedCrossRef P.R. Manna, D.W. Eubank, E. Lalli, P. Sassone-Corsi, D.M. Stocco, Transcriptional regulation of the mouse steroidogenic acute regulatory protein gene by the cAMP response-element binding protein and steroidogenic factor 1. J. Mol. Endocrinol. 30, 381–397 (2003)PubMedCrossRef
46.
Zurück zum Zitat P.R. Manna, D.M. Stocco, Crosstalk of CREB and Fos/Jun on a single cis-element: transcriptional repression of the steroidogenic acute regulatory protein gene. J. Mol. Endocrinol. 39, 261–277 (2007)PubMedCrossRef P.R. Manna, D.M. Stocco, Crosstalk of CREB and Fos/Jun on a single cis-element: transcriptional repression of the steroidogenic acute regulatory protein gene. J. Mol. Endocrinol. 39, 261–277 (2007)PubMedCrossRef
47.
Zurück zum Zitat P.R. Manna, M.T. Dyson, D.M. Stocco, Role of basic leucine zipper proteins in transcriptional regulation of the steroidogenic acute regulatory protein gene. Mol. Cell. Endocrinol. 302, 1–11 (2009)PubMedCrossRef P.R. Manna, M.T. Dyson, D.M. Stocco, Role of basic leucine zipper proteins in transcriptional regulation of the steroidogenic acute regulatory protein gene. Mol. Cell. Endocrinol. 302, 1–11 (2009)PubMedCrossRef
48.
Zurück zum Zitat H.A. Lavoie, S.R. King, Transcriptional regulation of steroidogenic genes: sTARD1, CYP11A1 and HSD3B. Exp. Biol. Med. (Maywood) 234, 880–907 (2009)CrossRef H.A. Lavoie, S.R. King, Transcriptional regulation of steroidogenic genes: sTARD1, CYP11A1 and HSD3B. Exp. Biol. Med. (Maywood) 234, 880–907 (2009)CrossRef
49.
Zurück zum Zitat B.J. Clark, S.C. Soo, K.M. Caron, Y. Ikeda, K.L. Parker, D.M. Stocco, Hormonal and developmental regulation of the steroidogenic acute regulatory protein. Mol. Endocrinol. 9, 1346–1355 (1995)PubMed B.J. Clark, S.C. Soo, K.M. Caron, Y. Ikeda, K.L. Parker, D.M. Stocco, Hormonal and developmental regulation of the steroidogenic acute regulatory protein. Mol. Endocrinol. 9, 1346–1355 (1995)PubMed
50.
Zurück zum Zitat S.R. King, T. Ronen-Fuhrmann, R. Timberg, B.J. Clark, J. Orly, D.M. Stocco, Steroid production after in vitro transcription, translation, and mitochondrial processing of protein products of complementary deoxyribonucleic acid for steroidogenic acute regulatory protein. Endocrinology 136, 5165–5176 (1995)PubMed S.R. King, T. Ronen-Fuhrmann, R. Timberg, B.J. Clark, J. Orly, D.M. Stocco, Steroid production after in vitro transcription, translation, and mitochondrial processing of protein products of complementary deoxyribonucleic acid for steroidogenic acute regulatory protein. Endocrinology 136, 5165–5176 (1995)PubMed
51.
Zurück zum Zitat P.R. Manna, S.P. Chandrala, S.R. King, Y. Jo, R. Counis, I.T. Huhtaniemi, D.M. Stocco, Molecular mechanisms of insulin-like growth factor-I mediated regulation of the steroidogenic acute regulatory protein in mouse leydig cells. Mol. Endocrinol. 20, 362–378 (2006)PubMedCrossRef P.R. Manna, S.P. Chandrala, S.R. King, Y. Jo, R. Counis, I.T. Huhtaniemi, D.M. Stocco, Molecular mechanisms of insulin-like growth factor-I mediated regulation of the steroidogenic acute regulatory protein in mouse leydig cells. Mol. Endocrinol. 20, 362–378 (2006)PubMedCrossRef
52.
Zurück zum Zitat B.J. Clark, R. Combs, K.H. Hales, D.B. Hales, D.M. Stocco, Inhibition of transcription affects synthesis of steroidogenic acute regulatory protein and steroidogenesis in MA-10 mouse Leydig tumor cells. Endocrinology 138, 4893–4901 (1997)PubMed B.J. Clark, R. Combs, K.H. Hales, D.B. Hales, D.M. Stocco, Inhibition of transcription affects synthesis of steroidogenic acute regulatory protein and steroidogenesis in MA-10 mouse Leydig tumor cells. Endocrinology 138, 4893–4901 (1997)PubMed
53.
Zurück zum Zitat A.P. Mathieu, A. Fleury, L. Ducharme, P. Lavigne, J.G. LeHoux, Insights into steroidogenic acute regulatory protein (StAR)-dependent cholesterol transfer in mitochondria: evidence from molecular modeling and structure-based thermodynamics supporting the existence of partially unfolded states of StAR. J. Mol. Endocrinol. 29, 327–345 (2002)PubMedCrossRef A.P. Mathieu, A. Fleury, L. Ducharme, P. Lavigne, J.G. LeHoux, Insights into steroidogenic acute regulatory protein (StAR)-dependent cholesterol transfer in mitochondria: evidence from molecular modeling and structure-based thermodynamics supporting the existence of partially unfolded states of StAR. J. Mol. Endocrinol. 29, 327–345 (2002)PubMedCrossRef
54.
Zurück zum Zitat A. Roostaee, E. Barbar, J.G. Lehoux, P. Lavigne, Cholesterol binding is a prerequisite for the activity of the steroidogenic acute regulatory protein (StAR). Biochem. J. 412, 553–562 (2008)PubMedCrossRef A. Roostaee, E. Barbar, J.G. Lehoux, P. Lavigne, Cholesterol binding is a prerequisite for the activity of the steroidogenic acute regulatory protein (StAR). Biochem. J. 412, 553–562 (2008)PubMedCrossRef
55.
Zurück zum Zitat E. Barbar, J.G. Lehoux, P. Lavigne, Toward the NMR structure of StAR. Mol. Cell. Endocrinol. 300, 89–93 (2009)PubMedCrossRef E. Barbar, J.G. Lehoux, P. Lavigne, Toward the NMR structure of StAR. Mol. Cell. Endocrinol. 300, 89–93 (2009)PubMedCrossRef
56.
Zurück zum Zitat B.J. Clark, V. Ranganathan, R. Combs, Steroidogenic acute regulatory protein expression is dependent upon post-translational effects of cAMP-dependent protein kinase A. Mol. Cell. Endocrinol. 173, 183–192 (2001)PubMedCrossRef B.J. Clark, V. Ranganathan, R. Combs, Steroidogenic acute regulatory protein expression is dependent upon post-translational effects of cAMP-dependent protein kinase A. Mol. Cell. Endocrinol. 173, 183–192 (2001)PubMedCrossRef
57.
Zurück zum Zitat L.K. Christenson, J.F. Strauss 3rd, Steroidogenic acute regulatory protein: an update on its regulation and mechanism of action. Arch. Med. Res. 32, 576–586 (2001)PubMedCrossRef L.K. Christenson, J.F. Strauss 3rd, Steroidogenic acute regulatory protein: an update on its regulation and mechanism of action. Arch. Med. Res. 32, 576–586 (2001)PubMedCrossRef
58.
Zurück zum Zitat N. Yivgi-Ohana, N. Sher, N. Melamed-Book, S. Eimerl, M. Koler, P.R. Manna, D.M. Stocco, J. Orly, Transcription of steroidogenic acute regulatory protein in the rodent ovary and placenta: alternative modes of cyclic adenosine 3′, 5′-monophosphate dependent and independent regulation. Endocrinology 150, 977–989 (2009)PubMedPubMedCentralCrossRef N. Yivgi-Ohana, N. Sher, N. Melamed-Book, S. Eimerl, M. Koler, P.R. Manna, D.M. Stocco, J. Orly, Transcription of steroidogenic acute regulatory protein in the rodent ovary and placenta: alternative modes of cyclic adenosine 3′, 5′-monophosphate dependent and independent regulation. Endocrinology 150, 977–989 (2009)PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat F. Arakane, S.R. King, Y. Du, C.B. Kallen, L.P. Walsh, H. Watari, D.M. Stocco, J.F. Strauss 3rd, Phosphorylation of steroidogenic acute regulatory protein (StAR) modulates its steroidogenic activity. J. Biol. Chem. 272, 32656–32662 (1997)PubMedCrossRef F. Arakane, S.R. King, Y. Du, C.B. Kallen, L.P. Walsh, H. Watari, D.M. Stocco, J.F. Strauss 3rd, Phosphorylation of steroidogenic acute regulatory protein (StAR) modulates its steroidogenic activity. J. Biol. Chem. 272, 32656–32662 (1997)PubMedCrossRef
60.
Zurück zum Zitat A. Fleury, A.P. Mathieu, L. Ducharme, D.B. Hales, J.G. LeHoux, Phosphorylation and function of the hamster adrenal steroidogenic acute regulatory protein (StAR). J. Steroid Biochem. Mol. Biol. 91, 259–271 (2004)PubMedCrossRef A. Fleury, A.P. Mathieu, L. Ducharme, D.B. Hales, J.G. LeHoux, Phosphorylation and function of the hamster adrenal steroidogenic acute regulatory protein (StAR). J. Steroid Biochem. Mol. Biol. 91, 259–271 (2004)PubMedCrossRef
61.
Zurück zum Zitat P.R. Manna, S.P. Chandrala, Y. Jo, D.M. Stocco, cAMP-independent signaling regulates steroidogenesis in mouse Leydig cells in the absence of StAR phosphorylation. J. Mol. Endocrinol. 37, 81–95 (2006)PubMedCrossRef P.R. Manna, S.P. Chandrala, Y. Jo, D.M. Stocco, cAMP-independent signaling regulates steroidogenesis in mouse Leydig cells in the absence of StAR phosphorylation. J. Mol. Endocrinol. 37, 81–95 (2006)PubMedCrossRef
62.
Zurück zum Zitat P.R. Manna, M.T. Dyson, Y. Jo, D.M. Stocco, Role of dosage-sensitive sex reversal, adrenal hypoplasia congenita, critical region on the X chromosome, gene 1 in protein kinase A- and protein kinase C-mediated regulation of the steroidogenic acute regulatory protein expression in mouse Leydig tumor cells: mechanism of action. Endocrinology 150, 187–199 (2009)PubMedPubMedCentralCrossRef P.R. Manna, M.T. Dyson, Y. Jo, D.M. Stocco, Role of dosage-sensitive sex reversal, adrenal hypoplasia congenita, critical region on the X chromosome, gene 1 in protein kinase A- and protein kinase C-mediated regulation of the steroidogenic acute regulatory protein expression in mouse Leydig tumor cells: mechanism of action. Endocrinology 150, 187–199 (2009)PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat B.A. Cooke, Signal transduction involving cyclic AMP-dependent and cyclic AMP- independent mechanisms in the control of steroidogenesis. Mol. Cell. Endocrinol. 151, 25–35 (1999)PubMedCrossRef B.A. Cooke, Signal transduction involving cyclic AMP-dependent and cyclic AMP- independent mechanisms in the control of steroidogenesis. Mol. Cell. Endocrinol. 151, 25–35 (1999)PubMedCrossRef
64.
Zurück zum Zitat J.S. Richards, New signaling pathways for hormones and cyclic adenosine 3′,5′-monophosphate action in endocrine cells. Mol. Endocrinol. 15, 209–218 (2001)PubMed J.S. Richards, New signaling pathways for hormones and cyclic adenosine 3′,5′-monophosphate action in endocrine cells. Mol. Endocrinol. 15, 209–218 (2001)PubMed
65.
Zurück zum Zitat P.R. Manna, L. Joshi, V.N. Reinhold, M.L. Aubert, N. Suganuma, K. Pettersson, I.T. Huhtaniemi, Synthesis, purification and structural and functional characterization of recombinant form of a common genetic variant of human luteinizing hormone. Hum. Mol. Genet. 11, 301–315 (2002)PubMedCrossRef P.R. Manna, L. Joshi, V.N. Reinhold, M.L. Aubert, N. Suganuma, K. Pettersson, I.T. Huhtaniemi, Synthesis, purification and structural and functional characterization of recombinant form of a common genetic variant of human luteinizing hormone. Hum. Mol. Genet. 11, 301–315 (2002)PubMedCrossRef
66.
Zurück zum Zitat D.B. Hales, Testicular macrophage modulation of Leydig cell steroidogenesis. J. Reprod. Immunol. 57, 3–18 (2002)PubMedCrossRef D.B. Hales, Testicular macrophage modulation of Leydig cell steroidogenesis. J. Reprod. Immunol. 57, 3–18 (2002)PubMedCrossRef
67.
Zurück zum Zitat P.R. Manna, P. Pakarinen, T. El-Hefnawy, I.T. Huhtaniemi, Functional assessment of the calcium messenger system in cultured mouse Leydig tumor cells: regulation of human chorionic gonadotropin-induced expression of the steroidogenic acute regulatory protein. Endocrinology 140, 1739–1751 (1999)PubMedCrossRef P.R. Manna, P. Pakarinen, T. El-Hefnawy, I.T. Huhtaniemi, Functional assessment of the calcium messenger system in cultured mouse Leydig tumor cells: regulation of human chorionic gonadotropin-induced expression of the steroidogenic acute regulatory protein. Endocrinology 140, 1739–1751 (1999)PubMedCrossRef
68.
Zurück zum Zitat X. Wang, L.P. Walsh, A.J. Reinhart, D.M. Stocco, The role of arachidonic acid in steroidogenesis and steroidogenic acute regulatory (StAR) gene and protein expression. J. Biol. Chem. 275, 20204–20209 (2000)PubMedCrossRef X. Wang, L.P. Walsh, A.J. Reinhart, D.M. Stocco, The role of arachidonic acid in steroidogenesis and steroidogenic acute regulatory (StAR) gene and protein expression. J. Biol. Chem. 275, 20204–20209 (2000)PubMedCrossRef
69.
Zurück zum Zitat P.R. Manna, I.T. Huhtaniemi, X.J. Wang, D.W. Eubank, D.M. Stocco, Mechanisms of epidermal growth factor signaling: regulation of steroid biosynthesis and the steroidogenic acute regulatory protein in mouse leydig tumor cells. Biol. Reprod. 67, 1393–1404 (2002)PubMedCrossRef P.R. Manna, I.T. Huhtaniemi, X.J. Wang, D.W. Eubank, D.M. Stocco, Mechanisms of epidermal growth factor signaling: regulation of steroid biosynthesis and the steroidogenic acute regulatory protein in mouse leydig tumor cells. Biol. Reprod. 67, 1393–1404 (2002)PubMedCrossRef
70.
Zurück zum Zitat F. Cornejo Maciel, P. Maloberti, I. Neuman, F. Cano, R. Castilla, F. Castillo, C. Paz, E.J. Podesta, An arachidonic acid-preferring acyl-CoA synthetase is a hormone-dependent and obligatory protein in the signal transduction pathway of steroidogenic hormones. J. Mol. Endocrinol. 34, 655–666 (2005)PubMedCrossRef F. Cornejo Maciel, P. Maloberti, I. Neuman, F. Cano, R. Castilla, F. Castillo, C. Paz, E.J. Podesta, An arachidonic acid-preferring acyl-CoA synthetase is a hormone-dependent and obligatory protein in the signal transduction pathway of steroidogenic hormones. J. Mol. Endocrinol. 34, 655–666 (2005)PubMedCrossRef
71.
Zurück zum Zitat J.Y. Park, Y.Q. Su, M. Ariga, E. Law, S.L. Jin, M. Conti, EGF-like growth factors as mediators of LH action in the ovulatory follicle. Science 303, 682–684 (2004)PubMedCrossRef J.Y. Park, Y.Q. Su, M. Ariga, E. Law, S.L. Jin, M. Conti, EGF-like growth factors as mediators of LH action in the ovulatory follicle. Science 303, 682–684 (2004)PubMedCrossRef
72.
Zurück zum Zitat M. Jamnongjit, A. Gill, S.R. Hammes, Epidermal growth factor receptor signaling is required for normal ovarian steroidogenesis and oocyte maturation. Proc. Natl. Acad. Sci. 102, 16257–16262 (2005)PubMedPubMedCentralCrossRef M. Jamnongjit, A. Gill, S.R. Hammes, Epidermal growth factor receptor signaling is required for normal ovarian steroidogenesis and oocyte maturation. Proc. Natl. Acad. Sci. 102, 16257–16262 (2005)PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat J. Nakae, T. Tajima, T. Sugawara, F. Arakane, K. Hanaki, T. Hotsubo, N. Igarashi, Y. Igarashi, T. Ishii, N. Koda, T. Kondo, H. Kohno, Y. Nakagawa, K. Tachibana, Y. Takeshima, K. Tsubouchi, J.F. Strauss 3rd, K. Fujieda, Analysis of the steroidogenic acute regulatory protein (StAR) gene in Japanese patients with congenital lipoid adrenal hyperplasia. Hum. Mol. Genet. 6, 571–576 (1997)PubMedCrossRef J. Nakae, T. Tajima, T. Sugawara, F. Arakane, K. Hanaki, T. Hotsubo, N. Igarashi, Y. Igarashi, T. Ishii, N. Koda, T. Kondo, H. Kohno, Y. Nakagawa, K. Tachibana, Y. Takeshima, K. Tsubouchi, J.F. Strauss 3rd, K. Fujieda, Analysis of the steroidogenic acute regulatory protein (StAR) gene in Japanese patients with congenital lipoid adrenal hyperplasia. Hum. Mol. Genet. 6, 571–576 (1997)PubMedCrossRef
74.
Zurück zum Zitat E. Okuyama, N. Nishi, S. Onishi, S. Itoh, Y. Ishii, H. Miyanaka, K. Fujita, Y. Ichikawa, A novel splicing junction mutation in the gene for the steroidogenic acute regulatory protein causes congenital lipoid adrenal hyperplasia. J. Clin. Endocrinol. Metab. 82, 2337–2342 (1997)PubMed E. Okuyama, N. Nishi, S. Onishi, S. Itoh, Y. Ishii, H. Miyanaka, K. Fujita, Y. Ichikawa, A novel splicing junction mutation in the gene for the steroidogenic acute regulatory protein causes congenital lipoid adrenal hyperplasia. J. Clin. Endocrinol. Metab. 82, 2337–2342 (1997)PubMed
75.
Zurück zum Zitat W.L. Miller, Congenital lipoid adrenal hyperplasia: the human gene knockout for the steroidogenic acute regulatory protein. J. Mol. Endocrinol. 19, 227–240 (1997)PubMedCrossRef W.L. Miller, Congenital lipoid adrenal hyperplasia: the human gene knockout for the steroidogenic acute regulatory protein. J. Mol. Endocrinol. 19, 227–240 (1997)PubMedCrossRef
76.
Zurück zum Zitat W.L. Miller, J.F. Strauss 3rd, Molecular pathology and mechanism of action of the steroidogenic acute regulatory protein, StAR. J. Steroid. Biochem. Mol. Biol. 69, 131–141 (1999)PubMedCrossRef W.L. Miller, J.F. Strauss 3rd, Molecular pathology and mechanism of action of the steroidogenic acute regulatory protein, StAR. J. Steroid. Biochem. Mol. Biol. 69, 131–141 (1999)PubMedCrossRef
77.
Zurück zum Zitat K. Fujieda, K. Okuhara, S. Abe, T. Tajima, T. Mukai, J. Nakae, Molecular pathogenesis of lipoid adrenal hyperplasia and adrenal hypoplasia congenita. J. Steroid Biochem. Mol. Biol. 85, 483–489 (2003)PubMedCrossRef K. Fujieda, K. Okuhara, S. Abe, T. Tajima, T. Mukai, J. Nakae, Molecular pathogenesis of lipoid adrenal hyperplasia and adrenal hypoplasia congenita. J. Steroid Biochem. Mol. Biol. 85, 483–489 (2003)PubMedCrossRef
78.
Zurück zum Zitat M.K. Tee, D. Lin, T. Sugawara, J.A. Holt, Y. Guiguen, B. Buckingham, J.F. Strauss 3rd, W.L. Miller, T->A transversion 11 bp from a splice acceptor site in the human gene for steroidogenic acute regulatory protein causes congenial lipoid adrenal hyperplasia. Hum. Mol. Genet. 4, 2299–2305 (1995)PubMedCrossRef M.K. Tee, D. Lin, T. Sugawara, J.A. Holt, Y. Guiguen, B. Buckingham, J.F. Strauss 3rd, W.L. Miller, T->A transversion 11 bp from a splice acceptor site in the human gene for steroidogenic acute regulatory protein causes congenial lipoid adrenal hyperplasia. Hum. Mol. Genet. 4, 2299–2305 (1995)PubMedCrossRef
79.
Zurück zum Zitat L.K. Christenson, J.F. Strauss 3rd, Steroidogenic acute regulatory protein (StAR) and the intramitochondrial translocation of cholesterol. Biochim. Biophys. Acta 1529, 175–187 (2000)PubMedCrossRef L.K. Christenson, J.F. Strauss 3rd, Steroidogenic acute regulatory protein (StAR) and the intramitochondrial translocation of cholesterol. Biochim. Biophys. Acta 1529, 175–187 (2000)PubMedCrossRef
80.
Zurück zum Zitat M.I. New, Inborn errors of adrenal steroidogenesis. Mol. Cell. Endocrinol. 211, 75–83 (2003)PubMedCrossRef M.I. New, Inborn errors of adrenal steroidogenesis. Mol. Cell. Endocrinol. 211, 75–83 (2003)PubMedCrossRef
81.
Zurück zum Zitat N. Krone, J. Grotzinger, P.M. Holterhus, W.G. Sippell, H.P. Schwarz, F.G. Riepe, Congenital adrenal hyperplasia due to 11-hydroxylase deficiency–insights from two novel CYP11B1 mutations (p. M92X, p.R453Q). Horm. Res. 72, 281–286 (2009)PubMedCrossRef N. Krone, J. Grotzinger, P.M. Holterhus, W.G. Sippell, H.P. Schwarz, F.G. Riepe, Congenital adrenal hyperplasia due to 11-hydroxylase deficiency–insights from two novel CYP11B1 mutations (p. M92X, p.R453Q). Horm. Res. 72, 281–286 (2009)PubMedCrossRef
82.
Zurück zum Zitat E.M. Brett, R.J. Auchus, Genetic forms of adrenal insufficiency. Endocr. Pract. 21, 395–399 (2015)PubMedCrossRef E.M. Brett, R.J. Auchus, Genetic forms of adrenal insufficiency. Endocr. Pract. 21, 395–399 (2015)PubMedCrossRef
83.
Zurück zum Zitat B.P. Hauffa, W.L. Miller, M.M. Grumbach, F.A. Conte, S.L. Kaplan, Congenital adrenal hyperplasia due to deficient cholesterol side-chain cleavage activity (20, 22-desmolase) in a patient treated for 18 years. Clin. Endocrinol. 23, 481–493 (1985)CrossRef B.P. Hauffa, W.L. Miller, M.M. Grumbach, F.A. Conte, S.L. Kaplan, Congenital adrenal hyperplasia due to deficient cholesterol side-chain cleavage activity (20, 22-desmolase) in a patient treated for 18 years. Clin. Endocrinol. 23, 481–493 (1985)CrossRef
84.
Zurück zum Zitat H.S. Bose, O.H. Pescovitz, W.L. Miller, Spontaneous feminization in a 46, XX female patient with congenital lipoid adrenal hyperplasia due to a homozygous frameshift mutation in the steroidogenic acute regulatory protein. J. Clin. Endocrinol. Metab. 82, 1511–1515 (1997)PubMed H.S. Bose, O.H. Pescovitz, W.L. Miller, Spontaneous feminization in a 46, XX female patient with congenital lipoid adrenal hyperplasia due to a homozygous frameshift mutation in the steroidogenic acute regulatory protein. J. Clin. Endocrinol. Metab. 82, 1511–1515 (1997)PubMed
85.
Zurück zum Zitat K. Fujieda, T. Tajima, J. Nakae, S. Sageshima, K. Tachibana, S. Suwa, T. Sugawara, J.F. Strauss 3rd, Spontaneous puberty in,46, XX subjects with congenital lipoid adrenal hyperplasia. Ovarian steroidogenesis is spared to some extent despite inactivating mutations in the steroidogenic acute regulatory protein (StAR) gene. J. Clin. Investig. 99, 1265–1271 (1997)PubMedPubMedCentralCrossRef K. Fujieda, T. Tajima, J. Nakae, S. Sageshima, K. Tachibana, S. Suwa, T. Sugawara, J.F. Strauss 3rd, Spontaneous puberty in,46, XX subjects with congenital lipoid adrenal hyperplasia. Ovarian steroidogenesis is spared to some extent despite inactivating mutations in the steroidogenic acute regulatory protein (StAR) gene. J. Clin. Investig. 99, 1265–1271 (1997)PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat M. Shima, A. Tanae, K. Miki, N. Katsumata, S. Matsumoto, S. Nakajima, T. Harada, T. Shinagawa, T. Tanaka, S. Okada, Mechanism for the development of ovarian cysts in patients with congenital lipoid adrenal hyperplasia. Eur. J. Endocrinol. 142, 274–279 (2000)PubMedCrossRef M. Shima, A. Tanae, K. Miki, N. Katsumata, S. Matsumoto, S. Nakajima, T. Harada, T. Shinagawa, T. Tanaka, S. Okada, Mechanism for the development of ovarian cysts in patients with congenital lipoid adrenal hyperplasia. Eur. J. Endocrinol. 142, 274–279 (2000)PubMedCrossRef
87.
Zurück zum Zitat J.F. Strauss 3rd, T. Kishida, L.K. Christenson, T. Fujimoto, H. Hiroi, START domain proteins and the intracellular trafficking of cholesterol in steroidogenic cells. Mol. Cell. Endocrinol. 202, 59–65 (2000)CrossRef J.F. Strauss 3rd, T. Kishida, L.K. Christenson, T. Fujimoto, H. Hiroi, START domain proteins and the intracellular trafficking of cholesterol in steroidogenic cells. Mol. Cell. Endocrinol. 202, 59–65 (2000)CrossRef
88.
Zurück zum Zitat T. Kishida, I. Kostetskii, Z. Zhang, F. Martinez, P. Liu, S.U. Walkley, N.K. Dwyer, E.J. Blanchette-Mackie, G.L. Radice, J.F. Strauss 3rd, Targeted mutation of the MLN64 START domain causes only modest alterations in cellular sterol metabolism. J. Biol. Chem. 279, 19276–19285 (2004)PubMedCrossRef T. Kishida, I. Kostetskii, Z. Zhang, F. Martinez, P. Liu, S.U. Walkley, N.K. Dwyer, E.J. Blanchette-Mackie, G.L. Radice, J.F. Strauss 3rd, Targeted mutation of the MLN64 START domain causes only modest alterations in cellular sterol metabolism. J. Biol. Chem. 279, 19276–19285 (2004)PubMedCrossRef
89.
Zurück zum Zitat S.R. King, A.A. Matassa, E.K. White, L.P. Walsh, Y. Jo, R.M. Rao, D.M. Stocco, M.E. Reyland, Oxysterols regulate expression of the steroidogenic acute regulatory protein. J. Mol. Endocrinol. 32, 507–517 (2004)PubMedCrossRef S.R. King, A.A. Matassa, E.K. White, L.P. Walsh, Y. Jo, R.M. Rao, D.M. Stocco, M.E. Reyland, Oxysterols regulate expression of the steroidogenic acute regulatory protein. J. Mol. Endocrinol. 32, 507–517 (2004)PubMedCrossRef
90.
Zurück zum Zitat P.R. Manna, Y. Jo, D.M. Stocco, Regulation of Leydig cell steroidogenesis by extracellular signal-regulated kinase 1/2: role of protein kinase A and protein kinase C signaling. J. Endocrinol. 193, 53–63 (2007)PubMedCrossRef P.R. Manna, Y. Jo, D.M. Stocco, Regulation of Leydig cell steroidogenesis by extracellular signal-regulated kinase 1/2: role of protein kinase A and protein kinase C signaling. J. Endocrinol. 193, 53–63 (2007)PubMedCrossRef
91.
Zurück zum Zitat H.S. Bose, R.M. Whittal, Y. Ran, M. Bose, B.Y. Baker, W.L. Miller, StAR-like activity and molten globule behavior of StARD6, a male germ-line protein. Biochemistry 47, 2277–2288 (2008)PubMedCrossRef H.S. Bose, R.M. Whittal, Y. Ran, M. Bose, B.Y. Baker, W.L. Miller, StAR-like activity and molten globule behavior of StARD6, a male germ-line protein. Biochemistry 47, 2277–2288 (2008)PubMedCrossRef
92.
Zurück zum Zitat M. Esparza-Perusquia, S. Olvera-Sanchez, O. Flores-Herrera, H. Flores-Herrera, A. Guevara-Flores, J.P. Pardo, M.T. Espinosa-Garcia, F. Martinez, Mitochondrial proteases act on STARD3 to activate progesterone synthesis in human syncytiotrophoblast. Biochim. Biophys. Acta 1850, 107–117 (2015)PubMedCrossRef M. Esparza-Perusquia, S. Olvera-Sanchez, O. Flores-Herrera, H. Flores-Herrera, A. Guevara-Flores, J.P. Pardo, M.T. Espinosa-Garcia, F. Martinez, Mitochondrial proteases act on STARD3 to activate progesterone synthesis in human syncytiotrophoblast. Biochim. Biophys. Acta 1850, 107–117 (2015)PubMedCrossRef
93.
Zurück zum Zitat K.M. Caron, S.C. Soo, K.L. Parker, Targeted disruption of StAR provides novel insights into congenital adrenal hyperplasia. Endocr. Res. 24, 827–834 (1998)PubMedCrossRef K.M. Caron, S.C. Soo, K.L. Parker, Targeted disruption of StAR provides novel insights into congenital adrenal hyperplasia. Endocr. Res. 24, 827–834 (1998)PubMedCrossRef
94.
Zurück zum Zitat S.E. Bulun, Z. Lin, G. Imir, S. Amin, M. Demura, B. Yilmaz, R. Martin, H. Utsunomiya, S. Thung, B. Gurates, M. Tamura, D. Langoi, S. Deb, Regulation of aromatase expression in estrogen-responsive breast and uterine disease: from bench to treatment. Pharmacol. Rev. 57, 359–383 (2005)PubMedCrossRef S.E. Bulun, Z. Lin, G. Imir, S. Amin, M. Demura, B. Yilmaz, R. Martin, H. Utsunomiya, S. Thung, B. Gurates, M. Tamura, D. Langoi, S. Deb, Regulation of aromatase expression in estrogen-responsive breast and uterine disease: from bench to treatment. Pharmacol. Rev. 57, 359–383 (2005)PubMedCrossRef
95.
Zurück zum Zitat S.E. Bulun, E.R. Simpson, Aromatase expression in women’s cancers. Adv. Exp. Med. Biol. 630, 112–132 (2008)PubMedCrossRef S.E. Bulun, E.R. Simpson, Aromatase expression in women’s cancers. Adv. Exp. Med. Biol. 630, 112–132 (2008)PubMedCrossRef
96.
Zurück zum Zitat S.E. Bulun, Z. Lin, H. Zhao, M. Lu, S. Amin, S. Reierstad, D. Chen, Regulation of aromatase expression in breast cancer tissue. Ann. N. Y. Acad. Sci. 1155, 121–131 (2009)PubMedCrossRef S.E. Bulun, Z. Lin, H. Zhao, M. Lu, S. Amin, S. Reierstad, D. Chen, Regulation of aromatase expression in breast cancer tissue. Ann. N. Y. Acad. Sci. 1155, 121–131 (2009)PubMedCrossRef
97.
Zurück zum Zitat K.R. Holloway, A. Barbieri, S. Malyarchuk, M. Saxena, A. Nedeljkovic-Kurepa, M. Cameron Mehl, A. Wang, X. Gu, K. Pruitt, SIRT1 positively regulates breast cancer associated human aromatase (CYP19A1) expression. Mol. Endocrinol. 27, 480–490 (2013)PubMedPubMedCentralCrossRef K.R. Holloway, A. Barbieri, S. Malyarchuk, M. Saxena, A. Nedeljkovic-Kurepa, M. Cameron Mehl, A. Wang, X. Gu, K. Pruitt, SIRT1 positively regulates breast cancer associated human aromatase (CYP19A1) expression. Mol. Endocrinol. 27, 480–490 (2013)PubMedPubMedCentralCrossRef
98.
Zurück zum Zitat E.R. Simpson, C. Clyne, G. Rubin, W.C. Boon, K. Robertson, K. Britt, C. Speed, M. Jones, Aromatase—a brief overview. Annu. Rev. Physiol. 64, 93–127 (2002)PubMedCrossRef E.R. Simpson, C. Clyne, G. Rubin, W.C. Boon, K. Robertson, K. Britt, C. Speed, M. Jones, Aromatase—a brief overview. Annu. Rev. Physiol. 64, 93–127 (2002)PubMedCrossRef
99.
Zurück zum Zitat S.E. Bulun, S. Yang, Z. Fang, B. Gurates, M. Tamura, S. Sebastian, Estrogen production and metabolism in endometriosis. Ann. N. Y. Acad. Sci. 955, 396–406 (2002)CrossRef S.E. Bulun, S. Yang, Z. Fang, B. Gurates, M. Tamura, S. Sebastian, Estrogen production and metabolism in endometriosis. Ann. N. Y. Acad. Sci. 955, 396–406 (2002)CrossRef
101.
Zurück zum Zitat R.L. Barbieri, S. Missmer, Endometriosis and infertility: a cause-effect relationship? Ann. N. Y. Acad. Sci. 955, 396–406 (2002)CrossRef R.L. Barbieri, S. Missmer, Endometriosis and infertility: a cause-effect relationship? Ann. N. Y. Acad. Sci. 955, 396–406 (2002)CrossRef
102.
Zurück zum Zitat D.W. Cramer, S.A. Missmer, The epidemiology of endometriosis. Ann. N. Y. Acad. Sci. 955, 396–406 (2002)CrossRef D.W. Cramer, S.A. Missmer, The epidemiology of endometriosis. Ann. N. Y. Acad. Sci. 955, 396–406 (2002)CrossRef
103.
Zurück zum Zitat R.B. Ness, D.W. Cramer, M.T. Goodman, S.K. Kjaer, K. Mallin, B.J. Mosgaard, D.M. Purdie, H.A. Risch, R. Vergona, A.H. Wu, Infertility, fertility drugs, and ovarian cancer: a pooled analysis of case-control studies. Am. J. Epidemiol. 155, 217–224 (2002)PubMedCrossRef R.B. Ness, D.W. Cramer, M.T. Goodman, S.K. Kjaer, K. Mallin, B.J. Mosgaard, D.M. Purdie, H.A. Risch, R. Vergona, A.H. Wu, Infertility, fertility drugs, and ovarian cancer: a pooled analysis of case-control studies. Am. J. Epidemiol. 155, 217–224 (2002)PubMedCrossRef
104.
Zurück zum Zitat J.J. Kim, T. Kurita, S.E. Bulun, Progesterone action in endometrial cancer, endometriosis, uterine fibroids, and breast cancer. Endocr. Rev. 34, 130–162 (2013)PubMedPubMedCentralCrossRef J.J. Kim, T. Kurita, S.E. Bulun, Progesterone action in endometrial cancer, endometriosis, uterine fibroids, and breast cancer. Endocr. Rev. 34, 130–162 (2013)PubMedPubMedCentralCrossRef
105.
Zurück zum Zitat S.J. Tsai, M.H. Wu, C.C. Lin, H.S. Sun, H.M. Chen, Regulation of steroidogenic acute regulatory protein expression and progesterone production in endometriotic stromal cells. J. Clin. Endocrinol. Metab. 86, 5765–5773 (2001)PubMedCrossRef S.J. Tsai, M.H. Wu, C.C. Lin, H.S. Sun, H.M. Chen, Regulation of steroidogenic acute regulatory protein expression and progesterone production in endometriotic stromal cells. J. Clin. Endocrinol. Metab. 86, 5765–5773 (2001)PubMedCrossRef
106.
Zurück zum Zitat S. Yang, Z. Fang, T. Suzuki, H. Sasano, J. Zhou, B. Gurates, M. Tamura, K. Ferrer, S. Bulun, Regulation of aromatase P450 expression in endometriotic and endometrial stromal cells by CCAAT/enhancer binding proteins (C/EBPs): decreased C/EBPbeta in endometriosis is associated with overexpression of aromatase. J. Clin. Endocrinol. Metab. 87, 2336–2345 (2002)PubMed S. Yang, Z. Fang, T. Suzuki, H. Sasano, J. Zhou, B. Gurates, M. Tamura, K. Ferrer, S. Bulun, Regulation of aromatase P450 expression in endometriotic and endometrial stromal cells by CCAAT/enhancer binding proteins (C/EBPs): decreased C/EBPbeta in endometriosis is associated with overexpression of aromatase. J. Clin. Endocrinol. Metab. 87, 2336–2345 (2002)PubMed
107.
Zurück zum Zitat E. Attar, S.E. Bulun, Aromatase and other steroidogenic genes in endometriosis: translational aspects. Hum. Reprod. Update 12, 49–56 (2006)PubMedCrossRef E. Attar, S.E. Bulun, Aromatase and other steroidogenic genes in endometriosis: translational aspects. Hum. Reprod. Update 12, 49–56 (2006)PubMedCrossRef
108.
Zurück zum Zitat W.D. Nes, Y.O. Lukyanenko, Z.H. Jia, S. Quideau, W.N. Howald, T.K. Pratum, R.R. West, J.C. Hutson, Identification of the lipophilic factor produced by macrophages that stimulates steroidogenesis. Endocrinology 141, 953–958 (2000)PubMed W.D. Nes, Y.O. Lukyanenko, Z.H. Jia, S. Quideau, W.N. Howald, T.K. Pratum, R.R. West, J.C. Hutson, Identification of the lipophilic factor produced by macrophages that stimulates steroidogenesis. Endocrinology 141, 953–958 (2000)PubMed
109.
Zurück zum Zitat Y.O. Lukyanenko, J.J. Chen, J.C. Hutson, Production of 25-hydroxycholesterol by testicular macrophages and its effects on Leydig cells. Biol. Reprod. 64, 790–796 (2001)PubMedCrossRef Y.O. Lukyanenko, J.J. Chen, J.C. Hutson, Production of 25-hydroxycholesterol by testicular macrophages and its effects on Leydig cells. Biol. Reprod. 64, 790–796 (2001)PubMedCrossRef
110.
Zurück zum Zitat J.A. Crow, K.L. Herring, S. Xie, A. Borazjani, P.M. Potter, M.K. Ross, Inhibition of carboxylesterase activity of THP1 monocytes/macrophages and recombinant human carboxylesterase 1 by oxysterols and fatty acids. Biochim. Biophys. Acta 1801, 31–41 (2010)PubMedPubMedCentralCrossRef J.A. Crow, K.L. Herring, S. Xie, A. Borazjani, P.M. Potter, M.K. Ross, Inhibition of carboxylesterase activity of THP1 monocytes/macrophages and recombinant human carboxylesterase 1 by oxysterols and fatty acids. Biochim. Biophys. Acta 1801, 31–41 (2010)PubMedPubMedCentralCrossRef
111.
Zurück zum Zitat E.S. Surrey, K.M. Silverberg, M.W. Surrey, W.B. Schoolcraft, Effect of prolonged gonadotropin-releasing hormone agonist therapy on the outcome of in vitro fertilization-embryo transfer in patients with endometriosis. Fertil. Steril. 78, 699–704 (2002)PubMedCrossRef E.S. Surrey, K.M. Silverberg, M.W. Surrey, W.B. Schoolcraft, Effect of prolonged gonadotropin-releasing hormone agonist therapy on the outcome of in vitro fertilization-embryo transfer in patients with endometriosis. Fertil. Steril. 78, 699–704 (2002)PubMedCrossRef
112.
Zurück zum Zitat V.M. Rice, Conventional medical therapies for endometriosis. Ann. N. Y. Acad. Sci. 955, 396–406 (2002)CrossRef V.M. Rice, Conventional medical therapies for endometriosis. Ann. N. Y. Acad. Sci. 955, 396–406 (2002)CrossRef
113.
Zurück zum Zitat R.K. Ailawadi, S. Jobanputra, M. Kataria, B. Gurates, S.E. Bulun, Treatment of endometriosis and chronic pelvic pain with letrozole and norethindrone acetate: a pilot study. Fertil. Steril. 81, 290–296 (2004)PubMedCrossRef R.K. Ailawadi, S. Jobanputra, M. Kataria, B. Gurates, S.E. Bulun, Treatment of endometriosis and chronic pelvic pain with letrozole and norethindrone acetate: a pilot study. Fertil. Steril. 81, 290–296 (2004)PubMedCrossRef
115.
Zurück zum Zitat K.R. Feingold, J.K. Shigenaga, M.R. Kazemi, C.M. McDonald, S.M. Patzek, A.S. Cross, A. Moser, C. Grunfeld, Mechanisms of triglyceride accumulation in activated macrophages. J. Leukoc. Biol. 92, 829–839 (2012)PubMedPubMedCentralCrossRef K.R. Feingold, J.K. Shigenaga, M.R. Kazemi, C.M. McDonald, S.M. Patzek, A.S. Cross, A. Moser, C. Grunfeld, Mechanisms of triglyceride accumulation in activated macrophages. J. Leukoc. Biol. 92, 829–839 (2012)PubMedPubMedCentralCrossRef
116.
Zurück zum Zitat C. Holm, T.G. Kirchgessner, K.L. Svenson, G. Fredrikson, S. Nilsson, C.G. Miller, J.E. Shively, C. Heinzmann, R.S. Sparkes, T. Mohandas et al., Hormone-sensitive lipase: sequence, expression, and chromosomal localization to 19 cent-q13.3. Science 241, 1503–1506 (1988)PubMedCrossRef C. Holm, T.G. Kirchgessner, K.L. Svenson, G. Fredrikson, S. Nilsson, C.G. Miller, J.E. Shively, C. Heinzmann, R.S. Sparkes, T. Mohandas et al., Hormone-sensitive lipase: sequence, expression, and chromosomal localization to 19 cent-q13.3. Science 241, 1503–1506 (1988)PubMedCrossRef
117.
Zurück zum Zitat H. Li, M. Brochu, S.P. Wang, L. Rochdi, M. Cote, G. Mitchell, N. Gallo-Payet, Hormone-sensitive lipase deficiency in mice causes lipid storage in the adrenal cortex and impaired corticosterone response to corticotropin stimulation. Endocrinology 143, 3333–3340 (2002)PubMedCrossRef H. Li, M. Brochu, S.P. Wang, L. Rochdi, M. Cote, G. Mitchell, N. Gallo-Payet, Hormone-sensitive lipase deficiency in mice causes lipid storage in the adrenal cortex and impaired corticosterone response to corticotropin stimulation. Endocrinology 143, 3333–3340 (2002)PubMedCrossRef
118.
Zurück zum Zitat S. Chung, S.P. Wang, L. Pan, G. Mitchell, J. Trasler, L. Hermo, Infertility and testicular defects in hormone-sensitive lipase-deficient mice. Endocrinology 142, 4272–4281 (2001)PubMedCrossRef S. Chung, S.P. Wang, L. Pan, G. Mitchell, J. Trasler, L. Hermo, Infertility and testicular defects in hormone-sensitive lipase-deficient mice. Endocrinology 142, 4272–4281 (2001)PubMedCrossRef
119.
Zurück zum Zitat S.P. Wang, S. Chung, K. Soni, H. Bourdages, L. Hermo, J. Trasler, G.A. Mitchell, Expression of human hormone-sensitive lipase (HSL) in postmeiotic germ cells confers normal fertility to HSL-deficient mice. Endocrinology 145, 5688–5693 (2004)PubMedCrossRef S.P. Wang, S. Chung, K. Soni, H. Bourdages, L. Hermo, J. Trasler, G.A. Mitchell, Expression of human hormone-sensitive lipase (HSL) in postmeiotic germ cells confers normal fertility to HSL-deficient mice. Endocrinology 145, 5688–5693 (2004)PubMedCrossRef
120.
Zurück zum Zitat T. Osterlund, Structure-function relationships of hormone-sensitive lipase. Eur. J. Biochem. 268, 1899–1907 (2001)PubMedCrossRef T. Osterlund, Structure-function relationships of hormone-sensitive lipase. Eur. J. Biochem. 268, 1899–1907 (2001)PubMedCrossRef
121.
Zurück zum Zitat C. Holm, Molecular mechanisms regulating hormone-sensitive lipase and lipolysis. Biochem. Soc. Trans. 31, 1120–1124 (2003)PubMedCrossRef C. Holm, Molecular mechanisms regulating hormone-sensitive lipase and lipolysis. Biochem. Soc. Trans. 31, 1120–1124 (2003)PubMedCrossRef
122.
Zurück zum Zitat C. Krintel, M. Morgelin, D.T. Logan, C. Holm, Phosphorylation of hormone-sensitive lipase by protein kinase A in vitro promotes an increase in its hydrophobic surface area. FEBS J. 276, 4752–4762 (2009)PubMedCrossRef C. Krintel, M. Morgelin, D.T. Logan, C. Holm, Phosphorylation of hormone-sensitive lipase by protein kinase A in vitro promotes an increase in its hydrophobic surface area. FEBS J. 276, 4752–4762 (2009)PubMedCrossRef
123.
Zurück zum Zitat W.J. Shen, S. Patel, V. Natu, R. Hong, J. Wang, S. Azhar, F.B. Kraemer, Interaction of hormone-sensitive lipase with steroidogenic acute regulatory protein: facilitation of cholesterol transfer in adrenal. J. Biol. Chem. 278, 43870–43876 (2003)PubMedCrossRef W.J. Shen, S. Patel, V. Natu, R. Hong, J. Wang, S. Azhar, F.B. Kraemer, Interaction of hormone-sensitive lipase with steroidogenic acute regulatory protein: facilitation of cholesterol transfer in adrenal. J. Biol. Chem. 278, 43870–43876 (2003)PubMedCrossRef
124.
Zurück zum Zitat R.M. Rao, Y. Jo, S. Leers-Sucheta, H.S. Bose, W.L. Miller, S. Azhar, D.M. Stocco, Differential regulation of steroid hormone biosynthesis in R2C and MA-10 Leydig tumor cells: role of SR-B1-mediated selective cholesteryl ester transport. Biol. Reprod. 68, 114–121 (2003)PubMedCrossRef R.M. Rao, Y. Jo, S. Leers-Sucheta, H.S. Bose, W.L. Miller, S. Azhar, D.M. Stocco, Differential regulation of steroid hormone biosynthesis in R2C and MA-10 Leydig tumor cells: role of SR-B1-mediated selective cholesteryl ester transport. Biol. Reprod. 68, 114–121 (2003)PubMedCrossRef
125.
Zurück zum Zitat S. Uda, S. Spolitu, F. Angius, M. Collu, S. Accossu, S. Banni, E. Murru, F. Sanna, B. Batetta, Role of HDL in cholesteryl ester metabolism of lipopolysaccharide-activated P388D1 macrophages. J. Lipid Res. 54, 3158–3169 (2013)PubMedPubMedCentralCrossRef S. Uda, S. Spolitu, F. Angius, M. Collu, S. Accossu, S. Banni, E. Murru, F. Sanna, B. Batetta, Role of HDL in cholesteryl ester metabolism of lipopolysaccharide-activated P388D1 macrophages. J. Lipid Res. 54, 3158–3169 (2013)PubMedPubMedCentralCrossRef
126.
Zurück zum Zitat J.T. Gwynne, J.F. Strauss 3rd, The role of lipoproteins in steroidogenesis and cholesterol metabolism in steroidogenic glands. Endocr. Rev. 3, 299–329 (1982)PubMedCrossRef J.T. Gwynne, J.F. Strauss 3rd, The role of lipoproteins in steroidogenesis and cholesterol metabolism in steroidogenic glands. Endocr. Rev. 3, 299–329 (1982)PubMedCrossRef
127.
Zurück zum Zitat M.S. Brown, J.L. Goldstein, A receptor-mediated pathway for cholesterol homeostasis. Science 232, 34–47 (1986)PubMedCrossRef M.S. Brown, J.L. Goldstein, A receptor-mediated pathway for cholesterol homeostasis. Science 232, 34–47 (1986)PubMedCrossRef
128.
Zurück zum Zitat E.J. Blanchette-Mackie, Intracellular cholesterol trafficking: role of the NPC1 protein. Biochim. Biophys. Acta 1486, 171–183 (2000)PubMedCrossRef E.J. Blanchette-Mackie, Intracellular cholesterol trafficking: role of the NPC1 protein. Biochim. Biophys. Acta 1486, 171–183 (2000)PubMedCrossRef
129.
Zurück zum Zitat H. Watari, E.J. Blanchette-Mackie, N.K. Dwyer, G. Sun, J.M. Glick, S. Patel, E.B. Neufeld, P.G. Pentchev, J.F. Strauss 3rd, NPC1-containing compartment of human granulosa-lutein cells: a role in the intracellular trafficking of cholesterol supporting steroidogenesis. Exp. Cell Res. 255, 56–66 (2000)PubMedCrossRef H. Watari, E.J. Blanchette-Mackie, N.K. Dwyer, G. Sun, J.M. Glick, S. Patel, E.B. Neufeld, P.G. Pentchev, J.F. Strauss 3rd, NPC1-containing compartment of human granulosa-lutein cells: a role in the intracellular trafficking of cholesterol supporting steroidogenesis. Exp. Cell Res. 255, 56–66 (2000)PubMedCrossRef
130.
Zurück zum Zitat N.Y. Gevry, B.D. Murphy, The role and regulation of the Niemann-Pick C1 gene in adrenal steroidogenesis. Endocr. Res. 28, 403–412 (2002)PubMedCrossRef N.Y. Gevry, B.D. Murphy, The role and regulation of the Niemann-Pick C1 gene in adrenal steroidogenesis. Endocr. Res. 28, 403–412 (2002)PubMedCrossRef
131.
Zurück zum Zitat E. Rigamonti, L. Helin, S. Lestavel, A.L. Mutka, M. Lepore, C. Fontaine, M.A. Bouhlel, S. Bultel, J.C. Fruchart, E. Ikonen, V. Clavey, B. Staels, G. Chinetti-Gbaguidi, Liver X receptor activation controls intracellular cholesterol trafficking and esterification in human macrophages. Circ. Res. 97, 682–689 (2005)PubMedCrossRef E. Rigamonti, L. Helin, S. Lestavel, A.L. Mutka, M. Lepore, C. Fontaine, M.A. Bouhlel, S. Bultel, J.C. Fruchart, E. Ikonen, V. Clavey, B. Staels, G. Chinetti-Gbaguidi, Liver X receptor activation controls intracellular cholesterol trafficking and esterification in human macrophages. Circ. Res. 97, 682–689 (2005)PubMedCrossRef
132.
Zurück zum Zitat K.M. Robertson, G.U. Schuster, K.R. Steffensen, O. Hovatta, S. Meaney, K. Hultenby, L.C. Johansson, K. Svechnikov, O. Soder, J.A. Gustafsson, The liver X receptor-{beta} is essential for maintaining cholesterol homeostasis in the testis. Endocrinology 146, 2519–2530 (2005)PubMedCrossRef K.M. Robertson, G.U. Schuster, K.R. Steffensen, O. Hovatta, S. Meaney, K. Hultenby, L.C. Johansson, K. Svechnikov, O. Soder, J.A. Gustafsson, The liver X receptor-{beta} is essential for maintaining cholesterol homeostasis in the testis. Endocrinology 146, 2519–2530 (2005)PubMedCrossRef
133.
Zurück zum Zitat D.H. Volle, K. Mouzat, R. Duggavathi, B. Siddeek, P. Dechelotte, B. Sion, G. Veyssiere, M. Benahmed, J.M. Lobaccaro, Multiple roles of the nuclear receptors for oxysterols liver X receptor to maintain male fertility. Mol. Endocrinol. 21, 1014–1027 (2007)PubMedCrossRef D.H. Volle, K. Mouzat, R. Duggavathi, B. Siddeek, P. Dechelotte, B. Sion, G. Veyssiere, M. Benahmed, J.M. Lobaccaro, Multiple roles of the nuclear receptors for oxysterols liver X receptor to maintain male fertility. Mol. Endocrinol. 21, 1014–1027 (2007)PubMedCrossRef
134.
Zurück zum Zitat M. Clagett-Dame, H.F. DeLuca, The role of vitamin A in mammalian reproduction and embryonic development. Annu. Rev. Nutr. 22, 347–381 (2002)PubMedCrossRef M. Clagett-Dame, H.F. DeLuca, The role of vitamin A in mammalian reproduction and embryonic development. Annu. Rev. Nutr. 22, 347–381 (2002)PubMedCrossRef
135.
Zurück zum Zitat A. Molotkov, N. Molotkova, G. Duester, Retinoic acid guides eye morphogenetic movements via paracrine signaling but is unnecessary for retinal dorsoventral patterning. Development 133, 1901–1910 (2006)PubMedPubMedCentralCrossRef A. Molotkov, N. Molotkova, G. Duester, Retinoic acid guides eye morphogenetic movements via paracrine signaling but is unnecessary for retinal dorsoventral patterning. Development 133, 1901–1910 (2006)PubMedPubMedCentralCrossRef
136.
Zurück zum Zitat A.W. See, M. Clagett-Dame, The temporal requirement for vitamin A in the developing eye: mechanism of action in optic fissure closure and new roles for the vitamin in regulating cell proliferation and adhesion in the embryonic retina. Dev. Biol. 325, 94–105 (2009)PubMedCrossRef A.W. See, M. Clagett-Dame, The temporal requirement for vitamin A in the developing eye: mechanism of action in optic fissure closure and new roles for the vitamin in regulating cell proliferation and adhesion in the embryonic retina. Dev. Biol. 325, 94–105 (2009)PubMedCrossRef
138.
139.
Zurück zum Zitat A. Slominski, B. Zbytek, G. Nikolakis, P.R. Manna, C. Skobowiat, M. Zmijewski, W. Li, Z. Janjetovic, A. Postlethwaite, C.C. Zouboulis, R.C. Tuckey, Steroidogenesis in the skin: implications for local immune functions. J. Steroid Biochem. Mol. Biol. 137, 107–123 (2010)CrossRef A. Slominski, B. Zbytek, G. Nikolakis, P.R. Manna, C. Skobowiat, M. Zmijewski, W. Li, Z. Janjetovic, A. Postlethwaite, C.C. Zouboulis, R.C. Tuckey, Steroidogenesis in the skin: implications for local immune functions. J. Steroid Biochem. Mol. Biol. 137, 107–123 (2010)CrossRef
140.
Zurück zum Zitat D. Bonhomme, A.M. Minni, S. Alfos, P. Roux, E. Richard, P. Higueret, M.P. Moisan, V. Pallet, K. Touyarot, Vitamin A status regulates glucocorticoid availability in Wistar rats: consequences on cognitive functions and hippocampal neurogenesis? Front. Behav. Neurosci. 8, 1–13 (2014)CrossRef D. Bonhomme, A.M. Minni, S. Alfos, P. Roux, E. Richard, P. Higueret, M.P. Moisan, V. Pallet, K. Touyarot, Vitamin A status regulates glucocorticoid availability in Wistar rats: consequences on cognitive functions and hippocampal neurogenesis? Front. Behav. Neurosci. 8, 1–13 (2014)CrossRef
141.
Zurück zum Zitat N. Srivastava, ATP binding cassette transporter A1–key roles in cellular lipid transport and atherosclerosis. Mol. Cell. Biochem. 237, 155–164 (2002)PubMedCrossRef N. Srivastava, ATP binding cassette transporter A1–key roles in cellular lipid transport and atherosclerosis. Mol. Cell. Biochem. 237, 155–164 (2002)PubMedCrossRef
142.
Zurück zum Zitat G. Chinetti-Gbaguidi, E. Rigamonti, L. Helin, A.L. Mutka, M. Lepore, J.C. Fruchart, V. Clavey, E. Ikonen, S. Lestavel, B. Staels, Peroxisome proliferator-activated receptor alpha controls cellular cholesterol trafficking in macrophages. J. Lipid Res. 46, 2717–2725 (2005)PubMedCrossRef G. Chinetti-Gbaguidi, E. Rigamonti, L. Helin, A.L. Mutka, M. Lepore, J.C. Fruchart, V. Clavey, E. Ikonen, S. Lestavel, B. Staels, Peroxisome proliferator-activated receptor alpha controls cellular cholesterol trafficking in macrophages. J. Lipid Res. 46, 2717–2725 (2005)PubMedCrossRef
143.
Zurück zum Zitat J.F. Oram, A.M. Vaughan, ATP-Binding cassette cholesterol transporters and cardiovascular disease. Circ. Res. 99, 1031–1043 (2006)PubMedCrossRef J.F. Oram, A.M. Vaughan, ATP-Binding cassette cholesterol transporters and cardiovascular disease. Circ. Res. 99, 1031–1043 (2006)PubMedCrossRef
144.
Zurück zum Zitat S. Bultel, L. Helin, V. Clavey, G. Chinetti-Gbaguidi, E. Rigamonti, M. Colin, J.C. Fruchart, B. Staels, S. Lestavel, Liver X receptor activation induces the uptake of cholesteryl esters from high density lipoproteins in primary human macrophages. Arterioscler. Thromb. Vasc. Biol. 28, 2288–2295 (2008)PubMedCrossRef S. Bultel, L. Helin, V. Clavey, G. Chinetti-Gbaguidi, E. Rigamonti, M. Colin, J.C. Fruchart, B. Staels, S. Lestavel, Liver X receptor activation induces the uptake of cholesteryl esters from high density lipoproteins in primary human macrophages. Arterioscler. Thromb. Vasc. Biol. 28, 2288–2295 (2008)PubMedCrossRef
145.
Zurück zum Zitat Y. Yuan, P. Li, J. Ye, Lipid homeostasis and the formation of macrophage-derived foam cells in atherosclerosis. Protein Cell 3, 173–181 (2012)PubMedCrossRef Y. Yuan, P. Li, J. Ye, Lipid homeostasis and the formation of macrophage-derived foam cells in atherosclerosis. Protein Cell 3, 173–181 (2012)PubMedCrossRef
146.
Zurück zum Zitat M. Bodzioch, E. Orso, J. Klucken, T. Langmann, A. Bottcher, W. Diederich, W. Drobnik, S. Barlage, C. Buchler, M. Porsch-Ozcurumez, W.E. Kaminski, H.W. Hahmann, K. Oette, G. Rothe, C. Aslanidis, K.J. Lackner, G. Schmitz, The gene encoding ATP-binding cassette transporter 1 is mutated in Tangier disease. Nat. Genet. 22, 347–351 (1999)PubMedCrossRef M. Bodzioch, E. Orso, J. Klucken, T. Langmann, A. Bottcher, W. Diederich, W. Drobnik, S. Barlage, C. Buchler, M. Porsch-Ozcurumez, W.E. Kaminski, H.W. Hahmann, K. Oette, G. Rothe, C. Aslanidis, K.J. Lackner, G. Schmitz, The gene encoding ATP-binding cassette transporter 1 is mutated in Tangier disease. Nat. Genet. 22, 347–351 (1999)PubMedCrossRef
147.
Zurück zum Zitat F. Tazoe, H. Yagyu, H. Okazaki, M. Igarashi, K. Eto, S. Nagashima, T. Inaba, H. Shimano, J. Osuga, S. Ishibashi, Induction of ABCA1 by overexpression of hormone-sensitive lipase in macrophages. Biochem. Biophys. Res. Commun. 376, 111–115 (2008)PubMedCrossRef F. Tazoe, H. Yagyu, H. Okazaki, M. Igarashi, K. Eto, S. Nagashima, T. Inaba, H. Shimano, J. Osuga, S. Ishibashi, Induction of ABCA1 by overexpression of hormone-sensitive lipase in macrophages. Biochem. Biophys. Res. Commun. 376, 111–115 (2008)PubMedCrossRef
148.
Zurück zum Zitat Y. Ning, Q. Bai, H. Lu, X. Li, W.M. Pandak, F. Zhao, S. Chen, S. Ren, L. Yin, Overexpression of mitochondrial cholesterol delivery protein, StAR, decreases intracellular lipids and inflammatory factors secretion in macrophages. Atherosclerosis 204, 0114–0120 (2009)CrossRef Y. Ning, Q. Bai, H. Lu, X. Li, W.M. Pandak, F. Zhao, S. Chen, S. Ren, L. Yin, Overexpression of mitochondrial cholesterol delivery protein, StAR, decreases intracellular lipids and inflammatory factors secretion in macrophages. Atherosclerosis 204, 0114–0120 (2009)CrossRef
149.
Zurück zum Zitat J.M. Taylor, F. Borthwick, C. Bartholomew, A. Graham, Overexpression of steroidogenic acute regulatory protein increases macrophage cholesterol efflux to apolipoprotein AI. Cardiovasc. Res. 86, 526–534 (2010)PubMedCrossRef J.M. Taylor, F. Borthwick, C. Bartholomew, A. Graham, Overexpression of steroidogenic acute regulatory protein increases macrophage cholesterol efflux to apolipoprotein AI. Cardiovasc. Res. 86, 526–534 (2010)PubMedCrossRef
150.
Zurück zum Zitat A. Nohara, J. Kobayashi, H. Mabuchi, Retinoid X receptor heterodimer variants and cardiovascular risk factors. J. Atheroscler. Thromb. 16, 303–318 (2009)PubMedCrossRef A. Nohara, J. Kobayashi, H. Mabuchi, Retinoid X receptor heterodimer variants and cardiovascular risk factors. J. Atheroscler. Thromb. 16, 303–318 (2009)PubMedCrossRef
151.
Zurück zum Zitat M. Hozoji-Inada, Y. Munehira, K. Nagao, N. Kioka, K. Ueda, Liver X receptor beta (LXRbeta) interacts directly with ATP-binding cassette A1 (ABCA1) to promote high density lipoprotein formation during acute cholesterol accumulation. J. Biol. Chem. 286, 20117–20124 (2011)PubMedPubMedCentralCrossRef M. Hozoji-Inada, Y. Munehira, K. Nagao, N. Kioka, K. Ueda, Liver X receptor beta (LXRbeta) interacts directly with ATP-binding cassette A1 (ABCA1) to promote high density lipoprotein formation during acute cholesterol accumulation. J. Biol. Chem. 286, 20117–20124 (2011)PubMedPubMedCentralCrossRef
152.
Zurück zum Zitat M. Ayaori, E. Yakushiji, M. Ogura, K. Nakaya, T. Hisada, H. Uto-Kondo, S. Takiguchi, Y. Terao, M. Sasaki, T. Komatsu, M. Iizuka, M. Yogo, Y. Uehara, H. Kagechika, T. Nakanishi, K. Ikewaki, Retinoic acid receptor agonists regulate expression of ATP-binding cassette transporter G1 in macrophages. Biochim. Biophys. Acta 1821, 561–572 (2012)PubMedCrossRef M. Ayaori, E. Yakushiji, M. Ogura, K. Nakaya, T. Hisada, H. Uto-Kondo, S. Takiguchi, Y. Terao, M. Sasaki, T. Komatsu, M. Iizuka, M. Yogo, Y. Uehara, H. Kagechika, T. Nakanishi, K. Ikewaki, Retinoic acid receptor agonists regulate expression of ATP-binding cassette transporter G1 in macrophages. Biochim. Biophys. Acta 1821, 561–572 (2012)PubMedCrossRef
153.
Zurück zum Zitat P.R. Manna, S.R. Sennoune, R. Martinez-Zaguilan, A.T. Slominski, K. Pruitt, Regulation of retinoid mediated cholesterol efflux involves liver X receptor activation in mouse macrophages. Biochem. Biophys. Res. Commun. 464, 312–317 (2015)PubMedCrossRef P.R. Manna, S.R. Sennoune, R. Martinez-Zaguilan, A.T. Slominski, K. Pruitt, Regulation of retinoid mediated cholesterol efflux involves liver X receptor activation in mouse macrophages. Biochem. Biophys. Res. Commun. 464, 312–317 (2015)PubMedCrossRef
154.
Zurück zum Zitat G. Chinetti, J.C. Fruchart, B. Staels, Transcriptional regulation of macrophage cholesterol trafficking by PPARalpha and LXR. Biochem. Soc. Trans. 34, 1128–1131 (2006)PubMedCrossRef G. Chinetti, J.C. Fruchart, B. Staels, Transcriptional regulation of macrophage cholesterol trafficking by PPARalpha and LXR. Biochem. Soc. Trans. 34, 1128–1131 (2006)PubMedCrossRef
155.
Zurück zum Zitat N. Krone, N.A. Hanley, W. Arlt, Age-specific changes in sex steroid biosynthesis and sex development. Best. Pract. Res. Clin. Endocrinol. Metab. 21, 393–401 (2007)PubMedCrossRef N. Krone, N.A. Hanley, W. Arlt, Age-specific changes in sex steroid biosynthesis and sex development. Best. Pract. Res. Clin. Endocrinol. Metab. 21, 393–401 (2007)PubMedCrossRef
156.
157.
Zurück zum Zitat E. Makrantonaki, C.C. Zouboulis, Molecular mechanisms of skin aging: state of the art. Ann. N. Y. Acad. Sci. 1119, 40–50 (2007)PubMedCrossRef E. Makrantonaki, C.C. Zouboulis, Molecular mechanisms of skin aging: state of the art. Ann. N. Y. Acad. Sci. 1119, 40–50 (2007)PubMedCrossRef
158.
Zurück zum Zitat I. Huhtaniemi, A. Perheentupa, Diagnosis and therapy of male hormonal changes related to aging. Duodecim 125, 1099–1106 (2009)PubMed I. Huhtaniemi, A. Perheentupa, Diagnosis and therapy of male hormonal changes related to aging. Duodecim 125, 1099–1106 (2009)PubMed
159.
Zurück zum Zitat M.L. Traub, N. Santoro, Reproductive aging and its consequences for general health. Ann. N. Y. Acad. Sci. 1204, 179–187 (2010)PubMedCrossRef M.L. Traub, N. Santoro, Reproductive aging and its consequences for general health. Ann. N. Y. Acad. Sci. 1204, 179–187 (2010)PubMedCrossRef
160.
Zurück zum Zitat B. Manor, L.A. Lipsitz, Physiologic complexity and aging: implications for physical function and rehabilitation. Prog. Neuropsychopharmacol. Biol. Psychiatry 45, 287–293 (2013)PubMedPubMedCentralCrossRef B. Manor, L.A. Lipsitz, Physiologic complexity and aging: implications for physical function and rehabilitation. Prog. Neuropsychopharmacol. Biol. Psychiatry 45, 287–293 (2013)PubMedPubMedCentralCrossRef
161.
Zurück zum Zitat T. Hertoghe, The “multiple hormone deficiency” theory of aging: is human senescence caused mainly by multiple hormone deficiencies? Ann. N. Y. Acad. Sci. 1057, 448–465 (2005)PubMedCrossRef T. Hertoghe, The “multiple hormone deficiency” theory of aging: is human senescence caused mainly by multiple hormone deficiencies? Ann. N. Y. Acad. Sci. 1057, 448–465 (2005)PubMedCrossRef
162.
Zurück zum Zitat A. Clegg, J. Young, S. Iliffe, M.O. Rikkert, K. Rockwood, Frailty in elderly people. Lancet 381, 752–762 (2013)PubMedCrossRef A. Clegg, J. Young, S. Iliffe, M.O. Rikkert, K. Rockwood, Frailty in elderly people. Lancet 381, 752–762 (2013)PubMedCrossRef
163.
Zurück zum Zitat N.D. Shaw, S.S. Srouji, S.N. Histed, K.E. McCurnin, J.E. Hall, Aging attenuates the pituitary response to gonadotropin-releasing hormone. J. Clin. Endocrinol. Metab. 94, 3259–3264 (2009)PubMedPubMedCentralCrossRef N.D. Shaw, S.S. Srouji, S.N. Histed, K.E. McCurnin, J.E. Hall, Aging attenuates the pituitary response to gonadotropin-releasing hormone. J. Clin. Endocrinol. Metab. 94, 3259–3264 (2009)PubMedPubMedCentralCrossRef
164.
Zurück zum Zitat I. Huhtaniemi, G. Forti, Male late-onset hypogonadism: pathogenesis, diagnosis and treatment. Nat. Rev. Urol. 8, 335–344 (2011)PubMedCrossRef I. Huhtaniemi, G. Forti, Male late-onset hypogonadism: pathogenesis, diagnosis and treatment. Nat. Rev. Urol. 8, 335–344 (2011)PubMedCrossRef
165.
166.
Zurück zum Zitat J.A. Janovick, M.D. Stewart, D. Jacob, L.D. Martin, J.M. Deng, C.A. Stewart, Y. Wang, A. Cornea, L. Chavali, S. Lopez, S. Mitalipov, E. Kang, H.S. Lee, P.R. Manna, D.M. Stocco, R.R. Behringer, P.M. Conn, Restoration of testis function in hypogonadotropic hypogonadal mice harboring a misfolded GnRHR mutant by pharmacoperone drug therapy. Proc. Natl. Acad. Sci. 110, 21030–21035 (2013)PubMedPubMedCentralCrossRef J.A. Janovick, M.D. Stewart, D. Jacob, L.D. Martin, J.M. Deng, C.A. Stewart, Y. Wang, A. Cornea, L. Chavali, S. Lopez, S. Mitalipov, E. Kang, H.S. Lee, P.R. Manna, D.M. Stocco, R.R. Behringer, P.M. Conn, Restoration of testis function in hypogonadotropic hypogonadal mice harboring a misfolded GnRHR mutant by pharmacoperone drug therapy. Proc. Natl. Acad. Sci. 110, 21030–21035 (2013)PubMedPubMedCentralCrossRef
167.
Zurück zum Zitat C. Feart, V. Pallet, C. Boucheron, D. Higueret, S. Alfos, L. Letenneur, J.F. Dartigues, P. Higueret, Aging affects the retinoic acid and the triiodothyronine nuclear receptor mRNA expression in human peripheral blood mononuclear cells. Eur. J. Endocrinol. 152, 449–458 (2005)PubMedCrossRef C. Feart, V. Pallet, C. Boucheron, D. Higueret, S. Alfos, L. Letenneur, J.F. Dartigues, P. Higueret, Aging affects the retinoic acid and the triiodothyronine nuclear receptor mRNA expression in human peripheral blood mononuclear cells. Eur. J. Endocrinol. 152, 449–458 (2005)PubMedCrossRef
168.
Zurück zum Zitat K. Ono, M. Yamada, Vitamin A and Alzheimer’s disease. Geriatr. Gerontol. Int. 12, 180–188 (2012)PubMedCrossRef K. Ono, M. Yamada, Vitamin A and Alzheimer’s disease. Geriatr. Gerontol. Int. 12, 180–188 (2012)PubMedCrossRef
169.
Zurück zum Zitat M.V. Blagosklonny, J. Campisi, D.A. Sinclair, A. Bartke, M.A. Blasco, W.M. Bonner, V.A. Bohr, R.M. Brosh Jr, A. Brunet, R.A. Depinho, L.A. Donehower, C.E. Finch, T. Finkel, M. Gorospe, A.V. Gudkov, M.N. Hall, S. Hekimi, S.L. Helfand, J. Karlseder, C. Kenyon, G. Kroemer, V. Longo, A. Nussenzweig, H.D. Osiewacz, D.S. Peeper, T.A. Rando, K.L. Rudolph, P. Sassone-Corsi, M. Serrano, N.E. Sharpless, V.P. Skulachev, J.L. Tilly, J. Tower, E. Verdin, J. Vijg, Impact papers on aging in 2009. Aging 2, 111–121 (2010)PubMedPubMedCentral M.V. Blagosklonny, J. Campisi, D.A. Sinclair, A. Bartke, M.A. Blasco, W.M. Bonner, V.A. Bohr, R.M. Brosh Jr, A. Brunet, R.A. Depinho, L.A. Donehower, C.E. Finch, T. Finkel, M. Gorospe, A.V. Gudkov, M.N. Hall, S. Hekimi, S.L. Helfand, J. Karlseder, C. Kenyon, G. Kroemer, V. Longo, A. Nussenzweig, H.D. Osiewacz, D.S. Peeper, T.A. Rando, K.L. Rudolph, P. Sassone-Corsi, M. Serrano, N.E. Sharpless, V.P. Skulachev, J.L. Tilly, J. Tower, E. Verdin, J. Vijg, Impact papers on aging in 2009. Aging 2, 111–121 (2010)PubMedPubMedCentral
170.
Zurück zum Zitat M.L. Batrinos, The aging of the endocrine hypothalamus and its dependent endocrine glands. Hormones 11, 241–253 (2012)PubMedCrossRef M.L. Batrinos, The aging of the endocrine hypothalamus and its dependent endocrine glands. Hormones 11, 241–253 (2012)PubMedCrossRef
171.
172.
Zurück zum Zitat D. Ortuno-Sahagun, M. Pallas, A.E. Rojas-Mayorquin, Oxidative stress in aging: advances in proteomic approaches. Oxid. Med. Cell. Longev. 2014, 1–18 (2014)CrossRef D. Ortuno-Sahagun, M. Pallas, A.E. Rojas-Mayorquin, Oxidative stress in aging: advances in proteomic approaches. Oxid. Med. Cell. Longev. 2014, 1–18 (2014)CrossRef
173.
Zurück zum Zitat P.Y. Liu, P.Y. Takahashi, P.D. Roebuck, A. Iranmanesh, J.D. Veldhuis, Age-specific changes in the regulation of LH-dependent testosterone secretion: assessing responsiveness to varying endogenous gonadotropin output in normal men. Am. J. Physiol. Regul. Integr. Comp. Physiol. 289, R721–R728 (2005)PubMedCrossRef P.Y. Liu, P.Y. Takahashi, P.D. Roebuck, A. Iranmanesh, J.D. Veldhuis, Age-specific changes in the regulation of LH-dependent testosterone secretion: assessing responsiveness to varying endogenous gonadotropin output in normal men. Am. J. Physiol. Regul. Integr. Comp. Physiol. 289, R721–R728 (2005)PubMedCrossRef
174.
Zurück zum Zitat X. Wang, D.M. Stocco, The decline in testosterone biosynthesis during male aging: a consequence of multiple alterations. Mol. Cell. Endocrinol. 238, 1–7 (2005)PubMedCrossRef X. Wang, D.M. Stocco, The decline in testosterone biosynthesis during male aging: a consequence of multiple alterations. Mol. Cell. Endocrinol. 238, 1–7 (2005)PubMedCrossRef
175.
Zurück zum Zitat E. Makrantonaki, P. Schonknecht, A.M. Hossini, E. Kaiser, M.M. Katsouli, J. Adjaye, J. Schroder, C.C. Zouboulis, Skin and brain age together: the role of hormones in the ageing process. Exp. Gerontol. 45, 801–813 (2010)PubMedCrossRef E. Makrantonaki, P. Schonknecht, A.M. Hossini, E. Kaiser, M.M. Katsouli, J. Adjaye, J. Schroder, C.C. Zouboulis, Skin and brain age together: the role of hormones in the ageing process. Exp. Gerontol. 45, 801–813 (2010)PubMedCrossRef
176.
Zurück zum Zitat A.T. Slominski, M.A. Zmijewski, C. Skobowiat, B. Zbytek, R.M. Slominski, J.D. Steketee, Sensing the environment: regulation of local and global homeostasis by the skin’s neuroendocrine system. Adv. Anat. Embryol. Cell Biol. 212, 1–115 (2012)CrossRef A.T. Slominski, M.A. Zmijewski, C. Skobowiat, B. Zbytek, R.M. Slominski, J.D. Steketee, Sensing the environment: regulation of local and global homeostasis by the skin’s neuroendocrine system. Adv. Anat. Embryol. Cell Biol. 212, 1–115 (2012)CrossRef
177.
Zurück zum Zitat M.C. Beattie, H. Chen, J. Fan, V. Papadopoulos, P. Miller, B.R. Zirkin, Aging and luteinizing hormone effects on reactive oxygen species production and DNA damage in rat leydig cells. Biol. Reprod. 88, 1–7 (2013)CrossRef M.C. Beattie, H. Chen, J. Fan, V. Papadopoulos, P. Miller, B.R. Zirkin, Aging and luteinizing hormone effects on reactive oxygen species production and DNA damage in rat leydig cells. Biol. Reprod. 88, 1–7 (2013)CrossRef
178.
Zurück zum Zitat L. Zhou, M.C. Beattie, C.Y. Lin, J. Liu, K. Traore, V. Papadopoulos, B.R. Zirkin, H. Chen, Oxidative stress and phthalate-induced down-regulation of steroidogenesis in MA-10 Leydig cells. Reprod. Toxicol. 42, 95–101 (2013)PubMedCrossRef L. Zhou, M.C. Beattie, C.Y. Lin, J. Liu, K. Traore, V. Papadopoulos, B.R. Zirkin, H. Chen, Oxidative stress and phthalate-induced down-regulation of steroidogenesis in MA-10 Leydig cells. Reprod. Toxicol. 42, 95–101 (2013)PubMedCrossRef
179.
Zurück zum Zitat G. Vitale, S. Salvioli, C. Franceschi, Oxidative stress and the ageing endocrine system. Nat. Rev. Endocrinol. 9, 228–240 (2013)PubMedCrossRef G. Vitale, S. Salvioli, C. Franceschi, Oxidative stress and the ageing endocrine system. Nat. Rev. Endocrinol. 9, 228–240 (2013)PubMedCrossRef
180.
Zurück zum Zitat Y. Kong, S.E. Trabucco, H. Zhang, Oxidative stress, mitochondrial dysfunction and the mitochondria theory of aging. Interdiscip. Top. Gerontol. 39, 86–107 (2014)PubMedCrossRef Y. Kong, S.E. Trabucco, H. Zhang, Oxidative stress, mitochondrial dysfunction and the mitochondria theory of aging. Interdiscip. Top. Gerontol. 39, 86–107 (2014)PubMedCrossRef
182.
Zurück zum Zitat I. Rodrigues Siqueira, C. Fochesatto, I.L. da Silva Torres, C. Dalmaz, C. Alexandre Netto, Aging affects oxidative state in hippocampus, hypothalamus and adrenal glands of Wistar rats. Life Sci. 78, 271–278 (2005)PubMedCrossRef I. Rodrigues Siqueira, C. Fochesatto, I.L. da Silva Torres, C. Dalmaz, C. Alexandre Netto, Aging affects oxidative state in hippocampus, hypothalamus and adrenal glands of Wistar rats. Life Sci. 78, 271–278 (2005)PubMedCrossRef
183.
Zurück zum Zitat T. Diemer, J.A. Allen, K.H. Hales, D.B. Hales, Reactive oxygen disrupts mitochondria in MA-10 tumor Leydig cells and inhibits steroidogenic acute regulatory (StAR) protein and steroidogenesis. Endocrinology 144, 2882–2891 (2003)PubMedCrossRef T. Diemer, J.A. Allen, K.H. Hales, D.B. Hales, Reactive oxygen disrupts mitochondria in MA-10 tumor Leydig cells and inhibits steroidogenic acute regulatory (StAR) protein and steroidogenesis. Endocrinology 144, 2882–2891 (2003)PubMedCrossRef
184.
Zurück zum Zitat A.S. Midzak, H. Chen, V. Papadopoulos, B.R. Zirkin, Leydig cell aging and the mechanisms of reduced testosterone synthesis. Mol. Cell. Endocrinol. 299, 23–31 (2009)PubMedCrossRef A.S. Midzak, H. Chen, V. Papadopoulos, B.R. Zirkin, Leydig cell aging and the mechanisms of reduced testosterone synthesis. Mol. Cell. Endocrinol. 299, 23–31 (2009)PubMedCrossRef
185.
Zurück zum Zitat A. Slominski, J. Wortsman, Neuroendocrinology of the skin. Endocr. Rev. 21, 457–487 (2000)PubMed A. Slominski, J. Wortsman, Neuroendocrinology of the skin. Endocr. Rev. 21, 457–487 (2000)PubMed
186.
Zurück zum Zitat P.M. Elias, J.S. Wakefield, Therapeutic implications of a barrier-based pathogenesis of atopic dermatitis. Clin. Rev. Allergy Immunol. 41, 282–295 (2011)PubMedPubMedCentralCrossRef P.M. Elias, J.S. Wakefield, Therapeutic implications of a barrier-based pathogenesis of atopic dermatitis. Clin. Rev. Allergy Immunol. 41, 282–295 (2011)PubMedPubMedCentralCrossRef
188.
Zurück zum Zitat A. Slominski, J. Wortsman, D.J. Tobin, The cutaneous serotoninergic/melatoninergic system: securing a place under the sun. FASEB. J. 19, 176–194 (2005)PubMedCrossRef A. Slominski, J. Wortsman, D.J. Tobin, The cutaneous serotoninergic/melatoninergic system: securing a place under the sun. FASEB. J. 19, 176–194 (2005)PubMedCrossRef
189.
Zurück zum Zitat A. Slominski, B. Zbytek, A. Szczesniewski, I. Semak, J. Kaminski, T. Sweatman, J. Wortsman, CRH stimulation of corticosteroids production in melanocytes is mediated by ACTH. Am. J. Physiol. Endocrinol. Metab. 288, E701–E706 (2005)PubMedCrossRef A. Slominski, B. Zbytek, A. Szczesniewski, I. Semak, J. Kaminski, T. Sweatman, J. Wortsman, CRH stimulation of corticosteroids production in melanocytes is mediated by ACTH. Am. J. Physiol. Endocrinol. Metab. 288, E701–E706 (2005)PubMedCrossRef
190.
Zurück zum Zitat A.T. Slominski, M.A. Zmijewski, B. Zbytek, D.J. Tobin, T.C. Theoharides, J. Rivier, Key Role of CRF in the skin stress response system. Endocr. Rev. 34, 827–884 (2013)PubMedPubMedCentralCrossRef A.T. Slominski, M.A. Zmijewski, B. Zbytek, D.J. Tobin, T.C. Theoharides, J. Rivier, Key Role of CRF in the skin stress response system. Endocr. Rev. 34, 827–884 (2013)PubMedPubMedCentralCrossRef
191.
Zurück zum Zitat A.T. Slominski, M.A. Zmijewski, I. Semak, B. Zbytek, A. Pisarchik, W. Li, J. Zjawiony, R.C. Tuckey, Cytochromes P450 and skin cancer: role of local endocrine pathways. Anticancer. Agents Med. Chem. 14, 77–96 (2014)PubMedPubMedCentralCrossRef A.T. Slominski, M.A. Zmijewski, I. Semak, B. Zbytek, A. Pisarchik, W. Li, J. Zjawiony, R.C. Tuckey, Cytochromes P450 and skin cancer: role of local endocrine pathways. Anticancer. Agents Med. Chem. 14, 77–96 (2014)PubMedPubMedCentralCrossRef
193.
Zurück zum Zitat N. Cirillo, S.S. Prime, Keratinocytes synthesize and activate cortisol. J. Cell. Biochem. 112, 1499–1505 (2011)PubMedCrossRef N. Cirillo, S.S. Prime, Keratinocytes synthesize and activate cortisol. J. Cell. Biochem. 112, 1499–1505 (2011)PubMedCrossRef
194.
Zurück zum Zitat T. Inoue, Y. Miki, K. Abe, M. Hatori, M. Hosaka, Y. Kariya, S. Kakuo, T. Fujimura, A. Hachiya, S. Honma, S. Aiba, H. Sasano, Sex steroid synthesis in human skin in situ: the roles of aromatase and steroidogenic acute regulatory protein in the homeostasis of human skin. Mol. Cell. Endocrinol. 362, 19–28 (2012)PubMedCrossRef T. Inoue, Y. Miki, K. Abe, M. Hatori, M. Hosaka, Y. Kariya, S. Kakuo, T. Fujimura, A. Hachiya, S. Honma, S. Aiba, H. Sasano, Sex steroid synthesis in human skin in situ: the roles of aromatase and steroidogenic acute regulatory protein in the homeostasis of human skin. Mol. Cell. Endocrinol. 362, 19–28 (2012)PubMedCrossRef
195.
Zurück zum Zitat A. Slominski, J. Zjawiony, J. Wortsman, I. Semak, J. Stewart, A. Pisarchik, T. Sweatman, J. Marcos, C. Dunbar, C.R. Tuckey, A novel pathway for sequential transformation of 7-dehydrocholesterol and expression of the P450scc system in mammalian skin. Eur. J. Biochem. 271, 4178–4188 (2004)PubMedPubMedCentralCrossRef A. Slominski, J. Zjawiony, J. Wortsman, I. Semak, J. Stewart, A. Pisarchik, T. Sweatman, J. Marcos, C. Dunbar, C.R. Tuckey, A novel pathway for sequential transformation of 7-dehydrocholesterol and expression of the P450scc system in mammalian skin. Eur. J. Biochem. 271, 4178–4188 (2004)PubMedPubMedCentralCrossRef
196.
Zurück zum Zitat P.M. Elias, D. Crumrine, A. Paller, M. Rodriguez-Martin, M.L. Williams, Pathogenesis of the cutaneous phenotype in inherited disorders of cholesterol metabolism: therapeutic implications for topical treatment of these disorders. Dermatoendocrinol 3, 100–106 (2011)PubMedPubMedCentralCrossRef P.M. Elias, D. Crumrine, A. Paller, M. Rodriguez-Martin, M.L. Williams, Pathogenesis of the cutaneous phenotype in inherited disorders of cholesterol metabolism: therapeutic implications for topical treatment of these disorders. Dermatoendocrinol 3, 100–106 (2011)PubMedPubMedCentralCrossRef
197.
Zurück zum Zitat S. Suomela, O. Elomaa, T. Skoog, R. Ala-aho, L. Jeskanen, J. Parssinen, L. Latonen, R. Grenman, J. Kere, V.M. Kahari, U. Saarialho-Kere, CCHCR1 is up-regulated in skin cancer and associated with EGFR expression. PLoS ONE 4, e6030 (2009)PubMedPubMedCentralCrossRef S. Suomela, O. Elomaa, T. Skoog, R. Ala-aho, L. Jeskanen, J. Parssinen, L. Latonen, R. Grenman, J. Kere, V.M. Kahari, U. Saarialho-Kere, CCHCR1 is up-regulated in skin cancer and associated with EGFR expression. PLoS ONE 4, e6030 (2009)PubMedPubMedCentralCrossRef
198.
Zurück zum Zitat R.F. Hannen, A.E. Michael, A. Jaulim, R. Bhogal, J.M. Burrin, M.P. Philpott, Steroid synthesis by primary human keratinocytes; implications for skin disease. Biochem. Biophys. Res. Commun. 404, 62–67 (2011)PubMedCrossRef R.F. Hannen, A.E. Michael, A. Jaulim, R. Bhogal, J.M. Burrin, M.P. Philpott, Steroid synthesis by primary human keratinocytes; implications for skin disease. Biochem. Biophys. Res. Commun. 404, 62–67 (2011)PubMedCrossRef
199.
Zurück zum Zitat A.T. Slominski, P.R. Manna, R.C. Tuckey, Cutaneous glucocorticosteroidogenesis: securing local homeostasis and the skin integrity. Exp. Dermatol. 23, 369–374 (2014)PubMedPubMedCentralCrossRef A.T. Slominski, P.R. Manna, R.C. Tuckey, Cutaneous glucocorticosteroidogenesis: securing local homeostasis and the skin integrity. Exp. Dermatol. 23, 369–374 (2014)PubMedPubMedCentralCrossRef
200.
Zurück zum Zitat N. Vernet, C. Dennefeld, C. Rochette-Egly, M. Oulad-Abdelghani, P. Chambon, N.B. Ghyselinck, M. Mark, Retinoic acid metabolism and signaling pathways in the adult and developing mouse testis. Endocrinology 147, 96–110 (2006)PubMedCrossRef N. Vernet, C. Dennefeld, C. Rochette-Egly, M. Oulad-Abdelghani, P. Chambon, N.B. Ghyselinck, M. Mark, Retinoic acid metabolism and signaling pathways in the adult and developing mouse testis. Endocrinology 147, 96–110 (2006)PubMedCrossRef
202.
Zurück zum Zitat J. Gericke, J. Ittensohn, J. Mihaly, S. Alvarez, R. Alvarez, D. Torocsik, A.R. de Lera, R. Ruhl, Regulation of retinoid-mediated signaling involved in skin homeostasis by RAR and RXR agonists/antagonists in mouse skin. PLoS ONE 8, e62643 (2013)PubMedPubMedCentralCrossRef J. Gericke, J. Ittensohn, J. Mihaly, S. Alvarez, R. Alvarez, D. Torocsik, A.R. de Lera, R. Ruhl, Regulation of retinoid-mediated signaling involved in skin homeostasis by RAR and RXR agonists/antagonists in mouse skin. PLoS ONE 8, e62643 (2013)PubMedPubMedCentralCrossRef
203.
Zurück zum Zitat S. Mukherjee, A. Date, V. Patravale, H.C. Korting, A. Roeder, G. Weindl, Retinoids in the treatment of skin aging: an overview of clinical efficacy and safety. Clin. Interv. Aging 1, 327–348 (2006)PubMedPubMedCentralCrossRef S. Mukherjee, A. Date, V. Patravale, H.C. Korting, A. Roeder, G. Weindl, Retinoids in the treatment of skin aging: an overview of clinical efficacy and safety. Clin. Interv. Aging 1, 327–348 (2006)PubMedPubMedCentralCrossRef
204.
205.
Zurück zum Zitat P.L. So, M.A. Fujimoto, E.H. Epstein Jr, Pharmacologic retinoid signaling and physiologic retinoic acid receptor signaling inhibit basal cell carcinoma tumorigenesis. Mol. Cancer Ther. 7, 1275–1284 (2008)PubMedPubMedCentralCrossRef P.L. So, M.A. Fujimoto, E.H. Epstein Jr, Pharmacologic retinoid signaling and physiologic retinoic acid receptor signaling inhibit basal cell carcinoma tumorigenesis. Mol. Cancer Ther. 7, 1275–1284 (2008)PubMedPubMedCentralCrossRef
206.
Zurück zum Zitat J. Mihaly, A. Gamlieli, M. Worm, R. Ruhl, Decreased retinoid concentration and retinoid signalling pathways in human atopic dermatitis. Exp. Dermatol. 20, 326–330 (2011)PubMedCrossRef J. Mihaly, A. Gamlieli, M. Worm, R. Ruhl, Decreased retinoid concentration and retinoid signalling pathways in human atopic dermatitis. Exp. Dermatol. 20, 326–330 (2011)PubMedCrossRef
207.
208.
Zurück zum Zitat M. Mark, N.B. Ghyselinck, P. Chambon, Function of retinoid nuclear receptors: lessons from genetic and pharmacological dissections of the retinoic acid signaling pathway during mouse embryogenesis. Annu. Rev. Pharmacol. Toxicol. 46, 451–480 (2006)PubMedCrossRef M. Mark, N.B. Ghyselinck, P. Chambon, Function of retinoid nuclear receptors: lessons from genetic and pharmacological dissections of the retinoic acid signaling pathway during mouse embryogenesis. Annu. Rev. Pharmacol. Toxicol. 46, 451–480 (2006)PubMedCrossRef
209.
Zurück zum Zitat J.K. Wickenheisser, V.L. Nelson-DeGrave, K.L. Hendricks, R.S. Legro, J.F. Strauss 3rd, J.M. McAllister, Retinoids and retinol differentially regulate steroid biosynthesis in ovarian theca cells. isolated from normal cycling women and women with polycystic ovary syndrome. J. Clin. Endocrinol. Metab. 90, 4858–4865 (2005)PubMedCrossRef J.K. Wickenheisser, V.L. Nelson-DeGrave, K.L. Hendricks, R.S. Legro, J.F. Strauss 3rd, J.M. McAllister, Retinoids and retinol differentially regulate steroid biosynthesis in ovarian theca cells. isolated from normal cycling women and women with polycystic ovary syndrome. J. Clin. Endocrinol. Metab. 90, 4858–4865 (2005)PubMedCrossRef
210.
Zurück zum Zitat P. Tucci, E. Cione, G. Genchi, Retinoic acid-induced testosterone production and retinoylation reaction are concomitant and exhibit a positive correlation in Leydig (TM-3) cells. J. Bioenerg. Biomembr. 40, 111–115 (2008)PubMedCrossRef P. Tucci, E. Cione, G. Genchi, Retinoic acid-induced testosterone production and retinoylation reaction are concomitant and exhibit a positive correlation in Leydig (TM-3) cells. J. Bioenerg. Biomembr. 40, 111–115 (2008)PubMedCrossRef
211.
Zurück zum Zitat K. Itoh, Y. Hiromori, N. Kato, I. Yoshida, N. Itoh, M. Ike, H. Nagase, K. Tanaka, T. Nakanishi, Placental steroidogenesis in rats is independent of signaling pathways induced by retinoic acids. Gen. Comp. Endocrinol. 163, 285–291 (2009)PubMedCrossRef K. Itoh, Y. Hiromori, N. Kato, I. Yoshida, N. Itoh, M. Ike, H. Nagase, K. Tanaka, T. Nakanishi, Placental steroidogenesis in rats is independent of signaling pathways induced by retinoic acids. Gen. Comp. Endocrinol. 163, 285–291 (2009)PubMedCrossRef
212.
Zurück zum Zitat E. Munetsuna, Y. Hojo, M. Hattori, H. Ishii, S. Kawato, A. Ishida, S.A. Kominami, T. Yamazaki, Retinoic acid stimulates 17beta-estradiol and testosterone synthesis in rat hippocampal slice cultures. Endocrinology 150, 4260–4269 (2009)PubMedCrossRef E. Munetsuna, Y. Hojo, M. Hattori, H. Ishii, S. Kawato, A. Ishida, S.A. Kominami, T. Yamazaki, Retinoic acid stimulates 17beta-estradiol and testosterone synthesis in rat hippocampal slice cultures. Endocrinology 150, 4260–4269 (2009)PubMedCrossRef
213.
Zurück zum Zitat A. Kushida, H. Tamura, Retinoic acids induce neurosteroid biosynthesis in human glial GI-1 Cells via the induction of steroidogenic genes. J. Biochem. 146, 917–923 (2009)PubMedCrossRef A. Kushida, H. Tamura, Retinoic acids induce neurosteroid biosynthesis in human glial GI-1 Cells via the induction of steroidogenic genes. J. Biochem. 146, 917–923 (2009)PubMedCrossRef
Metadaten
Titel
Role of the steroidogenic acute regulatory protein in health and disease
verfasst von
Pulak R. Manna
Cloyce L. Stetson
Andrzej T. Slominski
Kevin Pruitt
Publikationsdatum
01.01.2016
Verlag
Springer US
Erschienen in
Endocrine / Ausgabe 1/2016
Print ISSN: 1355-008X
Elektronische ISSN: 1559-0100
DOI
https://doi.org/10.1007/s12020-015-0715-6

Weitere Artikel der Ausgabe 1/2016

Endocrine 1/2016 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.