JME
HOME HELP FEEDBACK SUBSCRIPTIONS ARCHIVE SEARCH TABLE OF CONTENTS
 QUICK SEARCH:   [advanced]


     


Journal of Molecular Endocrinology (2009) 42 283-289    DOI: 10.1677/JME-08-0156
© 2009 Society for Endocrinology

This Article
Right arrow Abstract Freely available
Right arrow Full Text (PDF)
Right arrow All Versions of this Article:
JME-08-0156v1
42/4/283    most recent
Right arrow Alert me when this article is cited
Right arrow Alert me if a correction is posted
Services
Right arrow Similar articles in this journal
Right arrow Similar articles in PubMed
Right arrow Alert me to new issues of the journal
Right arrow Download to citation manager
Citing Articles
Right arrow Citing Articles via HighWire
Right arrow Citing Articles via Web of Science (4)
Right arrow Citing Articles via Google Scholar
Google Scholar
Right arrow Articles by Bánhegyi, G.
Right arrow Articles by Benedetti, A.
Right arrow Search for Related Content
PubMed
Right arrow PubMed Citation
Right arrow Articles by Bánhegyi, G.
Right arrow Articles by Benedetti, A.

Review

Hexose-6-phosphate dehydrogenase: linking endocrinology and metabolism in the endoplasmic reticulum

Gábor Bánhegyi1,2, Miklós Csala2 and Angelo Benedetti1

1 Dipartimento di Fisiopatologia, Medicina Sperimentale e Sanità Pubblica, Università di Siena, Via Aldo Moro, 53100 Siena, Italy2 Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University & MTA-SE Pathobiochemistry Research Group, 1444 Budapest, Hungary

(Correspondence should be addressed to A Benedetti; Email: benedetti{at}unisi.it)


    Abstract
 Top
 Abstract
 Introduction
 Prereceptorial glucocorticoid...
 HSD11B1 activity is determined...
 Intracellular glucose-6...
 Pathobiochemical aspects
 Declaration of interest
 Funding
 References
 
Hexose-6-phosphate dehydrogenase (H6PD) got into the focus of interest due to its role in the prereceptorial activation of glucocorticoids, which has been implicated in the pathomechanism of metabolic syndrome. Genetic observations, results gained in H6PD knockout mice, and studies on differentiating adipocytes demonstrated the importance of the enzyme in metabolic regulation. A nutrient-sensing function can be postulated for the enzyme, which links metabolism to endocrinology in the endoplasmic reticulum. This review provides an overview of the recent developments concerning the enzyme and its impact on various branches of the intermediary metabolism, which make it an important subject for the research on obesity, diabetes, and metabolic syndrome.


    Introduction
 Top
 Abstract
 Introduction
 Prereceptorial glucocorticoid...
 HSD11B1 activity is determined...
 Intracellular glucose-6...
 Pathobiochemical aspects
 Declaration of interest
 Funding
 References
 
The aim of the present review is to summarize the recently discovered roles of hexose-6-phosphate dehydrogenase (H6PD) in the intermediary metabolism, redox homeostasis, and signaling of the cell. Although the enzyme has been known for decades, it drew little attention until recent observations regarding its subcellular topology, tissue distribution, protein-protein interactions, and especially its role in prereceptorial glucocorticoid activation. The novel findings have revealed that H6PD activity is an important link between the intermediary metabolism and the autocrine/paracrine effects of glucocorticoid hormones, and hence this enzyme should be considered as an intracellular nutrient sensor.

The endoplasmic reticulum (ER) as a metabolic compartment is involved in almost all important pathways of the intermediary metabolism (Csala et al. 2006). Accumulating evidence suggests that the ER plays a prominent role in nutrient sensing. The cells (hepatocytes, adipocytes, pancreatic β-cells, etc.) can adapt their ER capabilities to cope with the alterations of metabolic demand. Extreme metabolic conditions (either over- or undernutrition) result in ER stress sensed fundamentally by the luminal machinery of protein folding. ER stress causes the accumulation of immature proteins and triggers the unfolded protein response (UPR) that finally leads to a variety of downstream effects, such as apoptosis, inflammation, and insulin resistance (Hotamisligil 2005, Yoshida 2007, Zhang & Kaufman 2008).

Recent observations have indicated that, besides the above signaling pathway, ER nutrient sensing can track alternative paths. Overfeeding stimulates intracellular activation of glucocorticoids in many cell types, and the concomitant autocrine and paracrine effects seem to be important elements of the pathomechanism of obesity, metabolic syndrome, and type 2 diabetes (Tomlinson et al. 2004, Morton & Seckl 2008). H6PD emerged as an indispensable component of the intracellular glucocorticoid activating system. The alterations of the cellular metabolic state can affect the system through the activity of H6PD that is tightly coupled to the local production of glucocorticoids from inactive precursors in the ER lumen. Therefore, H6PD can be regarded as a metabolic sensor in the ER, which connects intermediary metabolism to hormonal signaling. In accordance with the sensor function, H6PD seems to be a ubiquitous enzyme with a moderate inducibility implying that the substrate supply rather than enzyme level defines its activity. The wide distribution suggests that H6PD is also a housekeeping enzyme, playing a prosurvival role by maintaining the redox homeostasis of the ER/SR lumen. The enzymology and physiological functions of H6PD have been delineated by excellent recent reviews (Hewitt et al. 2005, White et al. 2007, 2008); therefore, the main properties of H6PD are only briefly summarized in Table 1.


View this table:
[in this window]
[in a new window]

 
Table 1 Biochemical properties of hexose-6-phosphate dehydrogenase (H6PD)

 

    Prereceptorial glucocorticoid activation in health and disease
 Top
 Abstract
 Introduction
 Prereceptorial glucocorticoid...
 HSD11B1 activity is determined...
 Intracellular glucose-6...
 Pathobiochemical aspects
 Declaration of interest
 Funding
 References
 
Glucocorticoid hormones form an important component of the homeostatic control mechanisms that, together with the immune system and autonomic nervous system, are critical in adapting to environmental challenges and thus maintain stability through change over time. Glucocorticoids regulate pathways leading to cellular proliferation, differentiation, or death, those induced (or repressed) in response to infection, tissue damage, and inflammation, pathways that maintain metabolic homeostasis and those that regulate mood and memory (Stahn et al. 2007). The relative expression levels of glucocorticoid receptors and β (Lewis-Tuffin & Cidlowski 2006) as well as the intracellular level of the hormone are the principal determinants of the glucocorticoid action. Since inactive glucocorticoids constitute a major fraction in the blood, the intracellular level of the hormonally active form critically depends upon the prereceptorial activation of glucocorticoids by 11β-hydroxysteroid dehydrogenase type 1 (HSD11B1) (Tomlinson et al. 2004, Morton & Seckl 2008). By converting intrinsically inert glucocorticoids (cortisone, 11-dehydrocorticosterone) into their active forms (cortisol, corticosterone), HSD11B1 increases glucocorticoid access to receptors. Alterations of HSD11B1 expression and activity have been implicated in the pathomechanism of hypertension, type 2 diabetes, atherosclerosis, metabolic syndrome, obesity, age-related cognitive dysfunction, osteoporosis, arthritis, and many other human diseases (Stimson & Walker 2007, Chapman & Seckl 2008, Pierotti et al. 2008).


    HSD11B1 activity is determined by the [NADPH]/[NADP+] ratio
 Top
 Abstract
 Introduction
 Prereceptorial glucocorticoid...
 HSD11B1 activity is determined...
 Intracellular glucose-6...
 Pathobiochemical aspects
 Declaration of interest
 Funding
 References
 
HSD11B1 enzyme is an integral membrane protein of the ER with luminal active site (Ozols 1995, Mziaut et al. 1999, Odermatt et al. 1999). It catalyzes the interconversion between cortisone and cortisol using NADPH or NADP+ coenzyme. Although the reaction is reversible in vitro, the enzyme acts as a reductase in vivo. Due to the relatively low free enthalpy change of the reaction, the enzyme activity greatly depends on the [NADPH]/[NADP+] ratio in the ER lumen (Atanasov et al. 2004, Banhegyi et al. 2004, Bujalska et al. 2005). Although the origin, the exact composition, and the redox state of luminal pyridine nucleotides are unknown, several indirect observations show that they are separated by the membrane barrier from the cytosolic pool (Czegle et al. 2006, Piccirella et al. 2006). The redox state of the cytosolic pyridine nucleotides is determined by several oxidoreductases. By contrast, H6PD seems to be the major - if not the only - enzyme responsible for NADP+ reduction in the ER lumen (Hewitt et al. 2005). Furthermore, according to the present knowledge, the substrate supply for H6PD is ensured by a sole ER membrane protein, the glucose-6-phosphate transporter (G6PT). Thus, in the ER of hepatocyte, adipocyte, and neutrophil granulocyte (and possibly a number of other cells), HSD11B1 can be considered as a component of a complex system, which also includes H6PD and G6PT (Fig. 1). The cooperation of the three proteins is ensured by sharing a common separate pyridine nucleotide pool.



View larger version (18K):
[in this window]
[in a new window]

 
Figure 1 Scheme of the G6PT-H6PD-HSD11B1 triad in the ER. Glucose-6-phosphate (G6P) is transported by G6P transporter (G6PT) to the ER lumen, where it is converted to phosphogluconate (6PG) by H6PD in a redox reaction generating NADPH. HSD11B1 utilizes NADPH as cofactor, allowing the conversion of cortisone to cortisol.

 
Theoretically, these circumstances can make the G6PT-H6PD-HSD11B1 system an excellent metabolic sensor of the ER. Minor changes in the [NADPH]/[NADP+] ratio in a small compartment can lead to significant alterations in glucocorticoid activation. In fact, an about tenfold excess of NADPH is required for the appropriate functioning of HSD11B1 as a reductase; a dramatic increase in the activity starts at 9:1 [NADPH]/[NADP+] ratio (Dzyakanchuk et al. 2008). It means that the in vivo set point of the NADPH-NADP+ redox couple is much more reduced in the ER lumen than in the cytosol (Díaz-Flores et al. 2006, Pollak et al. 2007, Panten & Rustenbeck 2008). H6PD is more resistant to feedback inhibition by NADPH than its cytosolic counterpart glucose-6-phosphate dehydrogenase (Oka et al. 1981), which allows the almost complete reduction of the ER luminal pyridine nucleotide pool. The redox state of luminal NADPH/NADP+ system is defined by cytosolic glucose-6-phosphate level with the transmission of G6PT and H6PD.


    Intracellular glucose-6-phosphate concentration as a function of nutrient load
 Top
 Abstract
 Introduction
 Prereceptorial glucocorticoid...
 HSD11B1 activity is determined...
 Intracellular glucose-6...
 Pathobiochemical aspects
 Declaration of interest
 Funding
 References
 
Intracellular glucose-6-phosphate concentration is a function of blood glucose and insulin levels in insulin-sensitive non-gluconeogenic tissues such as adipose tissue and skeletal muscle (Denton et al. 1966, Sekar et al. 1998). Insulin-dependent translocation of GLUT4 glucose transporter (SLC2A4) into the plasma membrane enhances glucose uptake, which results in elevated intracellular glucose-6-phosphate level. Accumulated glucose-6-phosphate fuels the G6PT-H6PD-HSD11B1 triad, which finally leads to increased prereceptorial activation of glucocorticoids (Fig. 2). In gluconeogenic tissues, intracellular glucose-6-phosphate concentration is less fluctuating; however, it can be elevated by intense gluconeogenesis in starvation (Hems & Brosnan 1971).



View larger version (43K):
[in this window]
[in a new window]

 
Figure 2 The role of the G6PT-H6PD-HSD11B1 triad in nutrient sensing of insulin-sensitive cells. Insulin stimulates the translocation of GLUT4 into the plasma membrane and hence enhances glucose uptake in muscle and adipose tissue, which results in elevated intracellular glucose-6-phosphate (G6P) level. On the other hand, high level of free fatty acids - according to Randle's ‘glucose-fatty acid cycle’ - inhibits glucose catabolism at several key steps, most importantly at the level of the pyruvate dehydrogenase (PDH) reaction. As a consequence, glucose-6-phosphate accumulates within the cell. Accumulation of glucose-6-phosphate, in turn, fuels the G6PT-H6PD-HSD11B1 system, which leads to increased prereceptorial activation of glucocorticoids. Thus, oversupply of reducing equivalents - in the form of either carbohydrates or lipids - leads to the enhancement of glucocorticoid activation. Increasing concentrations are indicated by red arrows, and resulting enzyme inhibitions are also indicated in red. Abbreviations: FFA, free fatty acid; F6P, fructose-6-phosphate; F1,6bP, fructose-1,6-bisphosphate; Glc, glucose; G6P, glucose-6-phosphate; GR, glucocorticoid receptor; HK, hexokinase; IR, insulin receptor; MT, mitochondrion; PDH, pyruvate dehydrogenase; PFK, phosphofructokinase; Pyr, pyruvate and OAA, oxaloacetate.

 
The system is also suitable for the indirect sensing of fatty acid levels. According to Randle's hypothesis (glucose-fatty acid cycle), free fatty acids compete with glucose for substrate oxidation. Increased free fatty acid levels lead to elevated mitochondrial [acetyl-CoA]/[CoA] and [NADH]/[NAD+] ratios that inhibit pyruvate dehydrogenase activity and lead to an increase in citrate levels, which, in turn, decreases phosphofructokinase activity and hence induces an increase in glucose-6-phosphate concentration due to the inhibition of glycolysis (Randle et al. 1963, Randle 1998; Fig. 2).

In conclusion, overfeeding either by carbohydrates or lipids results in elevated glucose-6-phosphate level in adipose tissue and skeletal muscle. These conditions, in fact, are accompanied by enhanced glucocorticoid activation. In humans, ‘whole-body’ generation of cortisol by HSD11B1 increases within 2-3 h after consumption of a mixed meal (Basu et al. 2006). Similarly, hyperinsulinemia and elevated free fatty acid level induce an acute increase in HSD11B1 activity in adipose tissue in humans (Wake et al. 2006). A very recent study has shown that luminal NADPH concentration in the ER is highly sensitive to extracellular glucose levels in HEK-293 cells expressing HSD11B1. Lowering glucose in the culture medium dose dependently decreased the reductase activity of HSD11B1 and diminished the cortisol/cortisone ratio. Coexpression with H6PD potentiated the reductase activity of HSD11B1 at high glucose concentration (Dzyakanchuk et al. 2008). However, the same study points out that other cell types (H4IIE liver cells and 3T3-L1 adipocytes) are less sensitive to the changes in extracellular glucose concentration. The glucose-6-phosphate availability in the ER lumen changes dramatically in two subtypes of the glycogen storage disease type 1 (GSD1). The observation that G6PT deficiency (GSD1b) significantly reduces while glucose-6-phosphatase deficiency (GSD1a) remarkably increases HSD11B1 activity strongly supports our hypothesis and proves the participation of G6PT in glucose (or glucose-6-phosphate) sensing (Walker et al. 2007). A complex disorder of nutrient sensing and metabolic regulation was observed in H6PD knockout mice: fasting hypoglycemia, low hepatic glycogen content, increased sensitivity to insulin, and decreased negative feedback on the hypothalamic-pituitary-adrenal axis (Lavery et al. 2006, 2007, Rogoff et al. 2007). These findings raise the important possibility that the G6PT-H6PD-HSD11B1 system plays a key role in the dynamic modifications of the acute metabolic response to feeding. The acute responsiveness suggests that the activity rather than the expression of H6PD is important in the coupling between metabolism and hormonal response.

The functioning of the system has been evidenced also in specialized endocrine cells involved in the regulation of whole-body metabolism. Local activation of glucocorticoids is required for the inhibition of glucagon secretion in the -cells of pancreatic islets (Swali et al. 2008). The effect of inactive glucocorticoid precursors is abolished in H6PD-null mice or when the HSD11B1 activity is reduced with specific inhibitors (Lavery et al. 2006).


    Pathobiochemical aspects
 Top
 Abstract
 Introduction
 Prereceptorial glucocorticoid...
 HSD11B1 activity is determined...
 Intracellular glucose-6...
 Pathobiochemical aspects
 Declaration of interest
 Funding
 References
 
Beyond the physiological sensor mechanism, the G6PT-H6PD-HSD11B1 triad can participate in the pathomechanism of gluco-, lipo-, and glucolipotoxicity. Glucose and fatty acids activate the UPR and induce ER stress by an unknown mechanism. It can be supposed that the excess of reducing equivalents leads to a redox imbalance in the ER lumen with a concomitant ER stress (Banhegyi et al. 2007). On the other hand, pharmacological or genetic manipulation of the G6PT-H6PD-HSD11B1 triad leads to ER stress, UPR, and/or apoptosis in various cell types (Leuzzi et al. 2003, Belkaid et al. 2006, Kardon et al. 2008, Lavery et al. 2008) because of a deficiency in the production of reducing equivalents. The ER stress response and its effectors are activated to protect the cells from apoptosis; however, the activation of these processes under conditions of long-term elevation of free fatty acids and glucose can lead to cellular dysfunction and ultimately apoptosis. Pancreatic β-cells are especially sensitive to glucolipotoxicity, perhaps not independently from their metabolic sensor function; high capacity of nutrient sensing is necessarily accompanied by a reduced protection against nutrient toxicity (Wang et al. 2005, Chang-Chen et al. 2008, Hou et al. 2008). However, saturated fatty acids disrupt ER homeostasis and induce ER stress and apoptosis also in liver cells (Wei et al. 2006).

An additional interesting point relates to the involvement of H6PD in the formation of atherogenic hydroxysterols. Actually it has been observed that 7-ketosterols (Hult et al. 2004, Schweizer et al. 2004) and 7-keto neurosteroids (Nashev et al. 2007) are substrates and competitive inhibitors of HSD11B1. Potentially, the redox balance of oxysterols, determined by H6PD/HSD11B1, may be crucial in modulating glucocorticoid and oxysterol effects (Balazs et al. 2008, Wamil et al. 2008).

Taking it all-round, high nutrient (electron donor) intake in the form of either carbohydrates or lipids can lead to enhanced glucocorticoid activation, especially in insulin-sensitive, non-gluconeogenic tissues. It has been known for a long while that glucocorticoids induce a state of insulin resistance by directly inhibiting the translocation of the GLUT4 glucose transporters to the plasma membrane and by decreased sensitivity of glycogen synthesis and glucose oxidation to insulin in skeletal muscle (Rizza et al. 1982, Dimitriadis et al. 1997). Moreover, elevated local activation of glucocorticoids stimulates preadipocyte differentiation, increases the expression of lipoprotein lipase, glycerol production, and triglyceride synthesis in visceral adipose tissue and stimulates hepatic gluconeogenesis, thereby contributing to the development of the metabolic syndrome (Bujalska et al. 2008, Tomlinson et al. 2008).

In conclusion, overnutrition (excess of reducing power) causes the elevation of intracellular glucose-6-phosphate level, which activates H6PD via G6PT. The generated and maintained high [NADPH]/[NADP+] ratio in the ER lumen supports glucocorticoid activation by HSD11B1. High local glucocorticoid levels counter-regulate insulin action and promote nutrient storage, hence producing the most characteristic features of the metabolic syndrome. The role of HSD11B1 in metabolism-dependent autocrine and paracrine effects is supported by growing evidence; however, the mechanism may be more general in endocrinology. Although the subcellular localization of steroid oxidoreductases has not been systematically investigated, the different preferences of the isoenzymes to reductase or dehydrogenase activity - e.g. in case of 17β-hydroxysteroid dehydrogenase family (Luu-The 2001) - suggest that their active sites might be localized in different compartments of the cell and that the reductase activity might be due to colocalization and functional cooperation with H6PD.


    Declaration of interest
 Top
 Abstract
 Introduction
 Prereceptorial glucocorticoid...
 HSD11B1 activity is determined...
 Intracellular glucose-6...
 Pathobiochemical aspects
 Declaration of interest
 Funding
 References
 
The authors have nothing to disclose. The authors declare that there is no conflict of interest that would prejudice the impartiality of this scientific work.


    Funding
 Top
 Abstract
 Introduction
 Prereceptorial glucocorticoid...
 HSD11B1 activity is determined...
 Intracellular glucose-6...
 Pathobiochemical aspects
 Declaration of interest
 Funding
 References
 
This research did not receive any specific grant from any funding agency in the public, commercial or not-for-profit sector.


    Acknowledgements
 
Thanks are due to the János Bolyai Research Scholarship of the Hungarian Academy of Sciences.


    References
 Top
 Abstract
 Introduction
 Prereceptorial glucocorticoid...
 HSD11B1 activity is determined...
 Intracellular glucose-6...
 Pathobiochemical aspects
 Declaration of interest
 Funding
 References
 
Atanasov AG, Nashev LG, Schweizer RA, Frick C & Odermatt A 2004 Hexose-6-phosphate dehydrogenase determines the reaction direction of 11β-hydroxysteroid dehydrogenase type 1 as an oxoreductase. FEBS Letters 571 129-133.[CrossRef][Web of Science][Medline]

Atanasov AG, Nashev LG, Gelman L, Legeza B, Sack R, Portmann R & Odermatt A 2008 Direct protein-protein interaction of 11β-hydroxysteroid dehydrogenase type 1 and hexose-6-phosphate dehydrogenase in the endoplasmic reticulum lumen. Biochimica et Biophysica Acta 1783 1536-1543.[Medline]

Balazs Z, Nashev LG, Chandsawangbhuwana C, Baker ME & Odermatt A 2008 Hexose-6-phosphate dehydrogenase modulates the effect of inhibitors and alternative substrates of 11β-hydroxysteroid dehydrogenase 1. Molecular and Cellular Endocrinology (doi: 10.1016/j.mce.2008.10.021)

Banhegyi G, Benedetti A, Fulceri R & Senesi S 2004 Cooperativity between 11β-hydroxysteroid dehydrogenase type 1 and hexose-6-phosphate dehydrogenase in the lumen of the endoplasmic reticulum. Journal of Biological Chemistry 279 27017-27021.[Abstract/Free Full Text]

Banhegyi G, Benedetti A, Csala M & Mandl J 2007 Stress on redox. FEBS Letters 581 3634-3640.[CrossRef][Web of Science][Medline]

Basu R, Singh R, Basu A, Johnson CM & Rizza RA 2006 Effect of nutrient ingestion on total-body and splanchnic cortisol production in humans. Diabetes 55 667-674.[Abstract/Free Full Text]

Belkaid A, Copland IB, Massillon D & Annabi B 2006 Silencing of the human microsomal glucose-6-phosphate translocase induces glioma cell death: potential new anticancer target for curcumin. FEBS Letters 580 3746-3752.[CrossRef][Web of Science][Medline]

Beutler E & Morrison M 1967 Localization and characteristics of hexose 6-phosphate dehydrogenase (glucose dehydrogenase). Journal of Biological Chemistry 242 5289-5293.[Abstract/Free Full Text]

Bujalska IJ, Draper N, Michailidou Z, Tomlinson JW, White PC, Chapman KE, Walker EA & Stewart PM 2005 Hexose-6-phosphate dehydrogenase confers oxo-reductase activity upon 11β-hydroxysteroid dehydrogenase type 1. Journal of Molecular Endocrinology 34 675-684.[Abstract/Free Full Text]

Bujalska IJ, Hewitt KN, Hauton D, Lavery GG, Tomlinson JW, Walker EA & Stewart PM 2008 Lack of hexose-6-phosphate dehydrogenase impairs lipid mobilization from mouse adipose tissue. Endocrinology 149 2584-2591.[Abstract/Free Full Text]

Chang-Chen KJ, Mullur R & Bernal-Mizrachi E 2008 β-Cell failure as a complication of diabetes. Reviews in Endocrine and Metabolic Disorders 9 329-343.[CrossRef]

Chapman KE & Seckl JR 2008 11β-HSD1, inflammation, metabolic disease and age-related cognitive (dys)function. Neurochemical Research 33 624-636.[CrossRef][Web of Science][Medline]

Clarke JL & Mason PJ 2003 Murine hexose-6-phosphate dehydrogenase: a bifunctional enzyme with broad substrate specificity and 6-phosphogluconolactonase activity. Archives of Biochemistry and Biophysics 415 229-234.[CrossRef][Web of Science][Medline]

Collard F, Collet JF, Gerin I, Veiga-da-Cunha M & Van Schaftingen E 1999 Identification of the cDNA encoding human 6-phosphogluconolactonase, the enzyme catalyzing the second step of the pentose phosphate pathway(1). FEBS Letters 459 223-226.[CrossRef][Web of Science][Medline]

Csala M, Banhegyi G & Benedetti A 2006 Endoplasmic reticulum: a metabolic compartment. FEBS Letters 580 2160-2165.[CrossRef][Web of Science][Medline]

Czegle I, Piccirella S, Senesi S, Csala M, Mandl J, Banhegyi G, Fulceri R & Benedetti A 2006 Cooperativity between 11β-hydroxysteroid dehydrogenase type 1 and hexose-6-phosphate dehydrogenase is based on a common pyridine nucleotide pool in the lumen of the endoplasmic reticulum. Molecular and Cellular Endocrinology 248 24-25.[CrossRef][Web of Science][Medline]

Denton RM, Yorke RE & Randle PJ 1966 Measurement of concentrations of metabolites in adipose tissue and effects of insulin, alloxan-diabetes and adrenaline. Biochemical Journal 100 407-419.[Web of Science][Medline]

Díaz-Flores M, Ibanez-Hernandez MA, Galvan RE, Gutierrez M, Duran-Reyes G, Medina-Navarro R, Pascoe-Lira D, Ortega-Camarillo C, Vilar-Rojas C, Cruz M et al. 2006 Glucose-6-phosphate dehydrogenase activity and NADPH/NADP+ ratio in liver and pancreas are dependent on the severity of hyperglycemia in rat. Life Sciences 78 2601-2607.[CrossRef][Web of Science][Medline]

Dimitriadis G, Leighton B, Parry-Billings M, Sasson S, Young M, Krause U, Bevan S, Piva T, Wegener G & Newsholme EA 1997 Effects of glucocorticoid excess on the sensitivity of glucose transport and metabolism to insulin in rat skeletal muscle. Biochemical Journal 321 707-712.[Web of Science][Medline]

Dzyakanchuk AA, Balazs Z, Nashev LG, Amrein KE & Odermatt A 2008 11β-Hydroxysteroid dehydrogenase 1 reductase activity is dependent on a high ratio of NADPH/NADP(+) and is stimulated by extracellular glucose. Molecular and Cellular Endocrinology (doi: 10.1016/jmce.2008.08.009)

Hems DA & Brosnan JT 1971 Effects of metabolic acidosis and starvation on the content of intermediary metabolites in rat kidney. Biochemical Journal 123 391-397.[Web of Science][Medline]

Hewitt KN, Walker EA & Stewart PM 2005 Minireview: hexose-6-phosphate dehydrogenase and redox control of 11β-hydroxysteroid dehydrogenase type 1 activity. Endocrinology 146 2539-2543.[CrossRef][Web of Science][Medline]

Hotamisligil GS 2005 Role of endoplasmic reticulum stress and c-Jun NH2-terminal kinase pathways in inflammation and origin of obesity and diabetes. Diabetes 54 S73-S78.[Abstract/Free Full Text]

Hou ZQ, Li HL, Gao L, Pan L, Zhao JJ & Li GW 2008 Involvement of chronic stresses in rat islet and INS-1 cell glucotoxicity induced by intermittent high glucose. Molecular and Cellular Endocrinology 291 71-78.[CrossRef][Web of Science][Medline]

Hult M, Elleby B, Shafqat N, Svensson S, Rane A, Jornvall H, Abrahmsen L & Oppermann U 2004 Human and rodent type 1 11β-hydroxysteroid dehydrogenases are 7β-hydroxycholesterol dehydrogenases involved in oxysterol metabolism. Cellular and Molecular Life Sciences 61 992-999.[CrossRef][Web of Science][Medline]

Kardon T, Senesi S, Marcolongo P, Legeza B, Banhegyi G, Mandl J, Fulceri R & Benedetti A 2008 Maintenance of luminal NADPH in the endoplasmic reticulum promotes the survival of human neutrophil granulocytes. FEBS Letters 582 1809-1815.[CrossRef][Web of Science][Medline]

Lavery GG, Walker EA, Draper N, Jeyasuria P, Marcos J, Shackleton CH, Parker KL, White PC & Stewart PM 2006 Hexose-6-phosphate dehydrogenase knock-out mice lack 11β-hydroxysteroid dehydrogenase type 1-mediated glucocorticoid generation. Journal of Biological Chemistry 281 6546-6551.[Abstract/Free Full Text]

Lavery GG, Hauton D, Hewitt KN, Brice SM, Sherlock M, Walker EA & Stewart PM 2007 Hypoglycemia with enhanced hepatic glycogen synthesis in recombinant mice lacking hexose-6-phosphate dehydrogenase. Endocrinology 148 6100-6106.[Abstract/Free Full Text]

Lavery GG, Walker EA, Turan N, Rogoff D, Ryder JW, Shelton JM, Richardson JA, Falciani F, White PC, Stewart PM et al. 2008 Deletion of hexose-6-phosphate dehydrogenase activates the unfolded protein response pathway and induces skeletal myopathy. Journal of Biological Chemistry 283 8453-8461.[Abstract/Free Full Text]

Leuzzi R, Banhegyi G, Kardon T, Marcolongo P, Capecchi PL, Burger HJ, Benedetti A & Fulceri R 2003 Inhibition of microsomal glucose-6-phosphate transport in human neutrophils results in apoptosis: a potential explanation for neutrophil dysfunction in glycogen storage disease type 1b. Blood 101 2381-2387.[Abstract/Free Full Text]

Lewis-Tuffin LJ & Cidlowski JA 2006 The physiology of human glucocorticoid receptor β (hGRβ) and glucocorticoid resistance. Annals of the New York Academy of Sciences 1069 1-9.[CrossRef][Web of Science][Medline]

Luu-The V 2001 Analysis and characteristics of multiple types of human 17β-hydroxysteroid dehydrogenase. Journal of Steroid Biochemistry and Molecular Biology 76 143-151.[CrossRef][Web of Science][Medline]

Marcolongo P, Piccirella S, Senesi S, Wunderlich L, Gerin I, Mandl J, Fulceri R, Banhegyi G & Benedetti A 2007 The glucose-6-phosphate transporter-hexose-6-phosphate dehydrogenase-11β-hydroxysteroid dehydrogenase type 1 system of the adipose tissue. Endocrinology 148 2487-2495.[Abstract/Free Full Text]

Mason PJ, Stevens D, Diez A, Knight SW, Scopes DA & Vulliamy TJ 1999 Human hexose-6-phosphate dehydrogenase (glucose 1-dehydrogenase) encoded at 1p36: coding sequence and expression. Blood Cells, Molecules & Diseases 25 30-37.[CrossRef][Web of Science][Medline]

Morton NM & Seckl JR 2008 11β-Hydroxysteroid dehydrogenase type 1 and obesity. Frontiers of Hormone Research 36 146-164.[Web of Science][Medline]

Mziaut H, Korza G, Hand AR, Gerard C & Ozols J 1999 Targeting proteins to the lumen of endoplasmic reticulum using N-terminal domains of 11β-hydroxysteroid dehydrogenase and the 50-kDa esterase. Journal of Biological Chemistry 274 14122-14129.[Abstract/Free Full Text]

Nashev LG, Chandsawangbhuwana C, Balazs Z, Atanasov AG, Dick B, Frey FJ, Baker ME & Odermatt A 2007 Hexose-6-phosphate dehydrogenase modulates 11β-hydroxysteroid dehydrogenase type 1-dependent metabolism of 7-keto- and 7β-hydroxy-neurosteroids. PLoS ONE 2 e561[CrossRef][Medline]

Odermatt A, Arnold P, Stauffer A, Frey BM & Frey FJ 1999 The N-terminal anchor sequences of 11β-hydroxysteroid dehydrogenases determine their orientation in the endoplasmic reticulum membrane. Journal of Biological Chemistry 274 28762-28770.[Abstract/Free Full Text]

Oka K, Takahashi T & Hori SH 1981 Differential effects of the NADPH/NADP+ ratio on the activities of hexose-6-phosphate dehydrogenase and glucose-6-phosphate dehydrogenase. Biochimica et Biophysica Acta 662 318-325.[Medline]

Ozols J 1993 Isolation and the complete amino acid sequence of lumenal endoplasmic reticulum glucose-6-phosphate dehydrogenase. PNAS 90 5302-5306.[Abstract/Free Full Text]

Ozols J 1995 Lumenal orientation and post-translational modifications of the liver microsomal 11β-hydroxysteroid dehydrogenase. Journal of Biological Chemistry 270 2305-2312.[Abstract/Free Full Text]

Panten U & Rustenbeck I 2008 Fuel-induced amplification of insulin secretion in mouse pancreatic islets exposed to a high sulfonylurea concentration: role of the NADPH/NADP+ ratio. Diabetologia 51 101-109.[Medline]

Piccirella S, Czegle I, Lizak B, Margittai E, Senesi S, Papp E, Csala M, Fulceri R, Csermely P, Mandl J et al. 2006 Uncoupled redox systems in the lumen of the endoplasmic reticulum. Pyridine nucleotides stay reduced in an oxidative environment. Journal of Biological Chemistry 281 4671-4677.[Abstract/Free Full Text]

Pierotti S, Gandini L, Lenzi A & Isidori AM 2008 Pre-receptorial regulation of steroid hormones in bone cells: insights on glucocorticoid-induced osteoporosis. Journal of Steroid Biochemistry and Molecular Biology 108 292-299.[CrossRef][Web of Science][Medline]

Pollak N, Dolle C & Ziegler M 2007 The power to reduce: pyridine nucleotides - small molecules with a multitude of functions. Biochemical Journal 402 205-218.[CrossRef][Web of Science][Medline]

Randle PJ 1998 Regulatory interactions between lipids and carbohydrates: the glucose fatty acid cycle after 35 years. Diabetes/Metabolism Reviews 14 263-283.[CrossRef][Web of Science][Medline]

Randle PJ, Garland PB, Hales CN & Newsholme EA 1963 The glucose fatty-acid cycle. Its role in insulin sensitivity and the metabolic disturbances of diabetes mellitus. Lancet 1 785-789.[Web of Science][Medline]

Rizza RA, Mandarino LJ & Gerich JE 1982 Cortisol-induced insulin resistance in man: impaired suppression of glucose production and stimulation of glucose utilization due to a postreceptor detect of insulin action. Journal of Clinical Endocrinology and Metabolism 54 131-138.[Abstract/Free Full Text]

Rogoff D, Ryder JW, Black K, Yan Z, Burgess SC, McMillan DR & White PC 2007 Abnormalities of glucose homeostasis and the hypothalamic-pituitary-adrenal axis in mice lacking hexose-6-phosphate dehydrogenase. Endocrinology 148 5072-5080.[Abstract/Free Full Text]

van Schaftingen E & Gerin I 2002 The glucose-6-phosphatase system. Biochemical Journal 362 513-532.[CrossRef][Web of Science][Medline]

Schweizer RA, Zurcher M, Balazs Z, Dick B & Odermatt A 2004 Rapid hepatic metabolism of 7-ketocholesterol by 11β-hydroxysteroid dehydrogenase type 1: species-specific differences between the rat, human, and hamster enzyme. Journal of Biological Chemistry 279 18415-18424.[Abstract/Free Full Text]

Sekar N, Qian S & Shechter Y 1998 Vanadate elevates lipogenicity of starved rat adipose tissue: mechanism of action. Endocrinology 139 2514-2518.[Abstract/Free Full Text]

Stahn C, Lowenberg M, Hommes DW & Buttgereit F 2007 Molecular mechanisms of glucocorticoid action and selective glucocorticoid receptor agonists. Molecular and Cellular Endocrinology 275 71-78.[CrossRef][Web of Science][Medline]

Stimson RH & Walker BR 2007 Glucocorticoids and 11β-hydroxysteroid dehydrogenase type 1 in obesity and the metabolic syndrome. Minerva Endocrinologica 32 141-159.[Medline]

Swali A, Walker EA, Lavery GG, Tomlinson JW & Stewart PM 2008 11β-Hydroxysteroid dehydrogenase type 1 regulates insulin and glucagon secretion in pancreatic islets. Diabetologia 51 2003-2011.[Medline]

Tomlinson JW, Walker EA, Bujalska IJ, Draper N, Lavery GG, Cooper MS, Hewison M & Stewart PM 2004 11β-Hydroxysteroid dehydrogenase type 1: a tissue-specific regulator of glucocorticoid response. Endocrine Reviews 25 831-866.[Abstract/Free Full Text]

Tomlinson JW, Finney J, Hughes BA, Hughes SV & Stewart PM 2008 Reduced glucocorticoid production rate, decreased 5alpha-reductase activity, and adipose tissue insulin sensitization after weight loss. Diabetes 57 1536-1543.[Abstract/Free Full Text]

Wake DJ, Homer NZ, Andrew R & Walker BR 2006 Acute in vivo regulation of 11β-hydroxysteroid dehydrogenase type 1 activity by insulin and intralipid infusions in humans. Journal of Clinical Endocrinology and Metabolism 91 4682-4688.[Abstract/Free Full Text]

Walker EA, Ahmed A, Lavery GG, Tomlinson JW, Kim SY, Cooper MS, Ride JP, Hughes BA, Shackleton CH, McKiernan P et al. 2007 11β-Hydroxysteroid dehydrogenase type 1 regulation by intracellular glucose 6-phosphate provides evidence for a novel link between glucose metabolism and hypothalamo-pituitary-adrenal axis function. Journal of Biological Chemistry 282 27030-27036.[Abstract/Free Full Text]

Wamil M, Andrew R, Chapman KE, Street J, Morton NM & Seckl JR 2008 7-Oxysterols modulate glucocorticoid activity in adipocytes through competition for 11β-hydroxysteroid dehydrogenase type 1. Endocrinology 149 5905-5918.

Wang H, Kouri G & Wollheim CB 2005 ER stress and SREBP-1 activation are implicated in β-cell glucolipotoxicity. Journal of Cell Science 118 3905-3915.[Abstract/Free Full Text]

Wei Y, Wang D, Topczewski F & Pagliassotti MJ 2006 Saturated fatty acids induce endoplasmic reticulum stress and apoptosis independently of ceramide in liver cells. American Journal of Physiology 291 E275-E281.[Web of Science]

White PC, Rogoff D, McMillan DR & Lavery GG 2007 Hexose 6-phosphate dehydrogenase (H6PD) and corticosteroid metabolism. Molecular and Cellular Endocrinology 265-266 89-92.

White PC, Rogoff D & McMillan DR 2008 Physiological roles of 11β-hydroxysteroid dehydrogenase type 1 and hexose-6-phosphate dehydrogenase. Current Opinion in Pediatrics 20 453-457.[CrossRef][Web of Science][Medline]

Yoshida H 2007 ER stress and diseases. FEBS Journal 274 630-658.[CrossRef][Web of Science][Medline]

Zhang K & Kaufman RJ 2008 From endoplasmic-reticulum stress to the inflammatory response. Nature 454 455-462.[CrossRef][Web of Science][Medline]

Received in final form 24 November 2008
Accepted 5 December 2008
Made available online as an Accepted Preprint 5 December 2008




This article has been cited by other articles:


Home page
EndocrinologyHome page
G.-X. Hu, Q.-Q. Lian, B.-B. Chen, P. V. Prasad, N. Kumar, Z.-Q. Zheng, and R.-S. Ge
7{alpha}-Hydroxytestosterone Affects 11{beta}-Hydroxysteroid Dehydrogenase 1 Direction in Rat Leydig Cells
Endocrinology, February 1, 2010; 151(2): 748 - 754.
[Abstract] [Full Text] [PDF]


This Article
Right arrow Abstract Freely available
Right arrow Full Text (PDF)
Right arrow All Versions of this Article:
JME-08-0156v1
42/4/283    most recent
Right arrow Alert me when this article is cited
Right arrow Alert me if a correction is posted
Services
Right arrow Similar articles in this journal
Right arrow Similar articles in PubMed
Right arrow Alert me to new issues of the journal
Right arrow Download to citation manager
Citing Articles
Right arrow Citing Articles via HighWire
Right arrow Citing Articles via Web of Science (4)
Right arrow Citing Articles via Google Scholar
Google Scholar
Right arrow Articles by Bánhegyi, G.
Right arrow Articles by Benedetti, A.
Right arrow Search for Related Content
PubMed
Right arrow PubMed Citation
Right arrow Articles by Bánhegyi, G.
Right arrow Articles by Benedetti, A.


HOME HELP FEEDBACK SUBSCRIPTIONS ARCHIVE SEARCH TABLE OF CONTENTS