Fig 4 - uploaded by Gabriele Toietta
Content may be subject to copyright.
Sirt-1 promotes CREB-dependent gene expression. ( A ) Forskolin-in- ducible physical association of CREB with Sirt-1 in PC12 cells. mycCREB or mycCREB- Δ LZ were transfected in PC12 cells and immunoprecipitated from protein lysates untreated or stimulated with Fsk for 30 min; the presence of Sirt-1 in immunocomplexes was veri fi ed by immunoblotting ( Upper ); anti- Ser133 CREB and anti-myc immunoblotting were used to con fi rm CREB phosphorylation by Fsk and assess the expression level of transfected CREB isoforms, respectively ( Lower ). ( Ba ) ChIP assays showing parallel interaction of CREB and Sirt-1 with the CRE-containing promoter regions of nNOS and PGC- 1 α in hippocampal neurons treated with NGF. Promoter ampli fi cation from total chromatin input is also reported as control. ( b ) CREB mediates Sirt-1 interaction with CRE-containing promoter regions. NGF-inducible binding of Sirt-1 to nNOS ( Upper ) and Sirt-1 promoter ( Lower ) are drastically reduced in hippocampal neurons lacking CREB. Binding of CREB to the same promoter regions con fi rms severe reduction of CREB binding in Cre-infected cells. rIgG (rabbit IgG) is a negative control for ChIP. Total chromatin input was equal throughout the lanes. ( C ) RT-PCR analysis showing reduced induction of nNOS and PGC-1 α mRNA by NGF in cortical neurons treated with the Sirt-1 inhibitor Nicotinamide (NAM). ( D ) ( a and b ) Representative RT-PCR analysis of PGC-1 α and nNOS mRNA expression in whole brains from WT and Sirt-1-de- fi cient mice under both AL and CR (6 mo) feeding. Each lane represents a pool of two mice. Actin was used as loading control. ( c ) Western blot analysis of whole brain protein homogenates from WT and Sirt-1 KO mice (fed AL) indicating reduced expression of nNOS but normal levels of immunoreactive CREB and total histone H4. Anti-AcH4K16 and antiactin immunostaining con fi rm, respectively, reduced deacetylase activity in the SirtKO sample and equal protein loading in the two lanes. 

Sirt-1 promotes CREB-dependent gene expression. ( A ) Forskolin-in- ducible physical association of CREB with Sirt-1 in PC12 cells. mycCREB or mycCREB- Δ LZ were transfected in PC12 cells and immunoprecipitated from protein lysates untreated or stimulated with Fsk for 30 min; the presence of Sirt-1 in immunocomplexes was veri fi ed by immunoblotting ( Upper ); anti- Ser133 CREB and anti-myc immunoblotting were used to con fi rm CREB phosphorylation by Fsk and assess the expression level of transfected CREB isoforms, respectively ( Lower ). ( Ba ) ChIP assays showing parallel interaction of CREB and Sirt-1 with the CRE-containing promoter regions of nNOS and PGC- 1 α in hippocampal neurons treated with NGF. Promoter ampli fi cation from total chromatin input is also reported as control. ( b ) CREB mediates Sirt-1 interaction with CRE-containing promoter regions. NGF-inducible binding of Sirt-1 to nNOS ( Upper ) and Sirt-1 promoter ( Lower ) are drastically reduced in hippocampal neurons lacking CREB. Binding of CREB to the same promoter regions con fi rms severe reduction of CREB binding in Cre-infected cells. rIgG (rabbit IgG) is a negative control for ChIP. Total chromatin input was equal throughout the lanes. ( C ) RT-PCR analysis showing reduced induction of nNOS and PGC-1 α mRNA by NGF in cortical neurons treated with the Sirt-1 inhibitor Nicotinamide (NAM). ( D ) ( a and b ) Representative RT-PCR analysis of PGC-1 α and nNOS mRNA expression in whole brains from WT and Sirt-1-de- fi cient mice under both AL and CR (6 mo) feeding. Each lane represents a pool of two mice. Actin was used as loading control. ( c ) Western blot analysis of whole brain protein homogenates from WT and Sirt-1 KO mice (fed AL) indicating reduced expression of nNOS but normal levels of immunoreactive CREB and total histone H4. Anti-AcH4K16 and antiactin immunostaining con fi rm, respectively, reduced deacetylase activity in the SirtKO sample and equal protein loading in the two lanes. 

Source publication
Article
Full-text available
Calorie restriction delays brain senescence and prevents neurodegeneration, but critical regulators of these beneficial responses other than the NAD(+)-dependent histone deacetylase Sirtuin-1 (Sirt-1) are unknown. We report that effects of calorie restriction on neuronal plasticity, memory and social behavior are abolished in mice lacking cAMP resp...

Contexts in source publication

Context 1
... tion of CREB protein, and the expression level of a number of mRNAs known to be regulated by CREB (23). CREB1 protein expression in control mice was not changed by the dietary regimen; instead, an increase in CREB phosphorylation on Serine 133 in the hippocampi of the CR group was revealed by phospho-speci fi c immunoblotting (Fig. 2 A ), suggesting that CR activates CREB in this brain area. Accordingly, mRNAs of “ canonical ” CREB targets peroxisome proliferator-activated receptor- γ coactivator-1 α ( PGC- 1 α ), nNOS , and phosphoenolpyruvate carboxykinase ( PEPCK ) were induced by CR in the cortex and hippocampus of CREB- pro fi cient mice (Fig. 2 B ). Of note, both PGC-1 α and NO, the product of NOS enzymes, are known to promote mitochondrial biogenesis and to participate in organismal response to CR (26). The three mRNAs were overall down-regulated in BCKO mice fed AL compared with controls, and their induction by CR, observed in control mice, was nearly abolished (Fig. 2 B ). Interestingly, the expression of other putative CREB targets like bcl-2 , NGF , and c-fos were unaffected by CR and CREB deletion (Fig. S2). Thus, collectively, these fi ndings demonstrate that nutrient availability selectively regulates CREB-dependent gene expression in the forebrain. We next asked how CR may affect CREB activity. Because Sirt- 1 is a metabolic sensor involved in several biological consequences of nutrient deprivation (27, 6), and animals lacking Sirt-1 in the brain show defective behavioral and hormonal responses to CR (7), we investigated the sirtuin as a potential CREB interactor. Sirt-1 mRNA was drastically reduced both in the cortex and hippocampus of BCKO mice, irrespective of the dietary regimen. Moreover, the moderate induction by CR that we observed in control brains was completely lost in the corresponding CREB- de fi cient tissues (Fig. 2 C ). Finally, acetylation of Histones H3 (AcK9) and H4 (AcK16), an inverse correlate of Sirt-1 activity (28), was abnormally high in BCKO hippocampal homogenates (Fig. 2 D ), con fi rming an overall reduction of Sirt-1 activity in this area of the brain. Transcriptional Regulation of Sirt-1 by CREB in Neurons. These observations suggested that Sirt-1 may represent a direct transcriptional target of CREB. Accordingly, CREB activators NGF and Forskolin (Fsk) (29) raised the level of immunoreactive Sirt-1 and of the corresponding mRNA in cultured primary cortical and hippocampal neurons (Fig. 3 A ). Bioinformatic analysis of the mouse Sirt-1 locus (NC_000076) revealed the presence of several putative cAMP Responsive Elements (CRE) both upstream and downstream of the transcription start site (TSS) (30), and an ∼ 300- bp segment encompassing two of those elements (TGACG at +1,998 and CGTCA at +2,012 from the annotated TSS) drove transcriptional response to Fsk and PKA in a standard luciferase reporter assay performed in PC12 pheochromocytoma cells (Fig. 3 B , a and b ). Moreover, ChIP from hippocampal neurons revealed that CREB binds the same genomic region in a fashion inducible by NGF (Fig. 3 Bc ). Finally, in primary neurons isolated from CREB loxP/loxP mice, deletion of CREB by adenoviral delivery of the Cre recombinase (Fig. 3 C , a and b ) led to a reduction of Sirt-1 immunoreactivity (Fig. 3 Cc ), and inhibited Sirt-1 mRNA induction by NGF and Fsk (Fig. 3 Cb ). Thus, we conclude that CREB directly regulates Sirt-1 mRNA and protein expression in neurons. Known transcriptional regulators of Sirt-1, such as p53 and Foxo3a (31), also interact with, and are either positively or negatively modulated by the deacetylase (32, 33); similarly, a myc-tagged form of human CREB1 and Sirt-1 could be easily coimmunopre- cipated in naive PC12 cells (Fig. 4 A ), indicating physical association between the two proteins; the association was rapidly induced by the PKA agonist Fsk, in parallel with phosphorylation of CREB on Serine 133. Short-time (30 min) stimulation with Fsk did not affect the total amount of cellular Sirt-1, but rather increased the stoichiometry of the binding to CREB, suggesting either an increased af fi nity between the two proteins, or the enhanced interaction with intermediate molecular partners co-recruited at target gene promoters. In keeping with the latter possibility, a mutant CREB that retains the phosphorylation site but is unable to bind DNA because of the deletion of the C-terminal transactivation domain ( Δ LZ CREB), displayed marginal physical interaction with Sirt-1, nor was the binding induced by Fsk (Fig. 4 A ). To con fi rm that SIRT1 and CREB colocalize on CREB- responsive chromatin regions in cortical neurons, cross-linked chromatin was immunoprecipitated with an anti-Sirt-1 antiserum and Sirt-1 binding to the regions of nNOS and PGC-1 α promoters surrounding the CRE elements determined by semiquantitative PCR. Sirt-1 was found to interact with both genes, in a fashion inducible by NGF and with a binding kinetic similar to that of CREB (Fig. 4 Ba ). Sirt-1 also bound the same Sirt-1 chromatin region surrounding the +1,998/+2,012 half-CRE sites that copre- cipitates with CREB, suggesting that Sirt-1 may self-regulate its CRE-dependent transactivation (Fig. 4 Bb ). Of note, Sirt-1 interaction with nNOS and Sirt-1 chromatin was indeed mediated by CREB, as revealed by its drastic reduction in CREB loxP/loxP neurons following the recombinase-mediated deletion of the factor (Fig. 4 Bb ) and by the failure of forced Sirt-1 overexpression to restore Sirt-1 chromatin binding in CREB-de fi cient cells (Fig. S3). To determine whether Sirt-1 modulates CREB transcriptional activity, cortical neurons were stimulated with NGF in the presence of sirtuin inhibitor Nicotinamide (34); induction of nNOS and PGC-1 α mRNA by NGF was blunted upon inhibition of Sirt-1 deacetylase activity (Fig. 4 C ). A similar result was obtained in naive PC12 cells by siRNA-mediated knock-down of Sirt-1 (Fig. S4 A ). In addition, Sirt-1 – de fi cient PC12 displayed impaired differentiation by NGF (as assessed by quanti fi cation of neurite outgrowth), a response largely dependent on CREB and nNOS in this cell model (35, 36) (Fig. S4 B ). In a complementary set of experiments, lentivirus-mediated overexpression of Sirt-1 increased the expression of nNOS and PGC-1 α mRNA in hippocampal neurons treated with NGF. However, Sirt-1 did not fully restore the expression of these genes in CREB-de fi cient cells, nor NGF protection from hydrogen peroxide-dependent cell death, which is largely lost in these cells (Fig. S5), was recovered upon transduction of the Sirt-1 cDNA. Collectively, the above data suggest that Sirt-1 and CREB are, at least to some extent, re- ciprocally dependent, and act in concert in the context of neurotrophin signaling. Unlike previously reported fi ndings (16), manipulations aimed at modulating Sirt-1 expression and activity in the above contexts did not affect the level of immunoreactive CREB1 (Fig. S4 Aa ). KO Mice. Finally, to verify whether Sirt-1 actually regulates CREB- dependent transcription in vivo and this is relevant for response to CR, we analyzed the expression of CREB targets nNOS and PGC- 1 α mRNAs in whole brains of Sirt-1 KO mice fed AL or CR for 25 – 28 wk (37). This analysis revealed that both genes are markedly hypoexpressed in Sirt-de fi cient mice compared with littermate controls under both dietary regimens (Fig. 4 D ), and their induction by CR, that was more pronounced for PGC-1 α , was also attenuated (Fig. 4 D , a and b ). The amount of immunoreactive CREB was comparable in brain homogenates of the two strains (Fig. 4 Dc ), nor CREB acetylation on lysine residues was affected by either Sirt-1 deletion or CR (Fig. S6), indicating that, at least in these experimental settings, Sirt-1 regulates CREB transcriptional activity independent of the acetylation and expression level of the factor. Taken together, the above fi ndings strongly suggest that Sirt-1 modulates the expression of CREB-dependent genes in mouse brain, and by extension, identify in the decrease of CREB- dependent transcription a molecular signature for a defective brain response to CR, shared by CREB and Sirt-1 mutant mice (7). The CREB transcription factor has been widely investigated as a metabolic sensor and regulator of glucose homeostasis in liver and fat tissue (15), and as a master switch of calcium and neuro- trophin-triggered transcriptional programs regulating neuronal differentiation, survival, and plasticity in the brain and peripheral nervous system (10, 11, 18). Evidence also exist for CREB roles in the control of appetite and food intake in the hypothalamus (38), but whether neuronal plasticity and high-order cognitive functions may be in fl uenced by nutrient cues and energy metabolism through CREB, an issue relevant to major diseases like AD and type 2 diabetes (3, 4), remains to be established. Our demonstra- tion of impaired electrophysiological, cognitive, and emotional response to CR in brain CREB KO mice, clearly suggests that this may indeed be the case. Interestingly, these differences emerge in the context of an overall comparable feeding behavior and metabolic response to CR between control and BCKO mice (Fig. 1 Ab ), indicating that potential effects of hypothalamic CREB signaling on energy balance and appetite regulation unlikely account for the observed phenotypes (38). Data reported in Fig. 2, showing CREB phosphorylation and transcriptional activation by CR, indicate that CREB is metaboli- cally regulated in the cortex and hippocampus, although the mechanism of such regulation needs to be further investigated. Because plasma from CR rodents has been previously shown to induce Sirt-1 expression in several organs/tissues, a humoral/hormonal mechanism for CREB activation could be envisaged (39); as an alternative, NO (40) or oxygen species (41) may mediate, cell-autonomously, this effect. Further research in this direction is warranted. Our ...
Context 2
... phosphorylation on Serine 133 in the hippocampi of the CR group was revealed by phospho-speci fi c immunoblotting (Fig. 2 A ), suggesting that CR activates CREB in this brain area. Accordingly, mRNAs of “ canonical ” CREB targets peroxisome proliferator-activated receptor- γ coactivator-1 α ( PGC- 1 α ), nNOS , and phosphoenolpyruvate carboxykinase ( PEPCK ) were induced by CR in the cortex and hippocampus of CREB- pro fi cient mice (Fig. 2 B ). Of note, both PGC-1 α and NO, the product of NOS enzymes, are known to promote mitochondrial biogenesis and to participate in organismal response to CR (26). The three mRNAs were overall down-regulated in BCKO mice fed AL compared with controls, and their induction by CR, observed in control mice, was nearly abolished (Fig. 2 B ). Interestingly, the expression of other putative CREB targets like bcl-2 , NGF , and c-fos were unaffected by CR and CREB deletion (Fig. S2). Thus, collectively, these fi ndings demonstrate that nutrient availability selectively regulates CREB-dependent gene expression in the forebrain. We next asked how CR may affect CREB activity. Because Sirt- 1 is a metabolic sensor involved in several biological consequences of nutrient deprivation (27, 6), and animals lacking Sirt-1 in the brain show defective behavioral and hormonal responses to CR (7), we investigated the sirtuin as a potential CREB interactor. Sirt-1 mRNA was drastically reduced both in the cortex and hippocampus of BCKO mice, irrespective of the dietary regimen. Moreover, the moderate induction by CR that we observed in control brains was completely lost in the corresponding CREB- de fi cient tissues (Fig. 2 C ). Finally, acetylation of Histones H3 (AcK9) and H4 (AcK16), an inverse correlate of Sirt-1 activity (28), was abnormally high in BCKO hippocampal homogenates (Fig. 2 D ), con fi rming an overall reduction of Sirt-1 activity in this area of the brain. Transcriptional Regulation of Sirt-1 by CREB in Neurons. These observations suggested that Sirt-1 may represent a direct transcriptional target of CREB. Accordingly, CREB activators NGF and Forskolin (Fsk) (29) raised the level of immunoreactive Sirt-1 and of the corresponding mRNA in cultured primary cortical and hippocampal neurons (Fig. 3 A ). Bioinformatic analysis of the mouse Sirt-1 locus (NC_000076) revealed the presence of several putative cAMP Responsive Elements (CRE) both upstream and downstream of the transcription start site (TSS) (30), and an ∼ 300- bp segment encompassing two of those elements (TGACG at +1,998 and CGTCA at +2,012 from the annotated TSS) drove transcriptional response to Fsk and PKA in a standard luciferase reporter assay performed in PC12 pheochromocytoma cells (Fig. 3 B , a and b ). Moreover, ChIP from hippocampal neurons revealed that CREB binds the same genomic region in a fashion inducible by NGF (Fig. 3 Bc ). Finally, in primary neurons isolated from CREB loxP/loxP mice, deletion of CREB by adenoviral delivery of the Cre recombinase (Fig. 3 C , a and b ) led to a reduction of Sirt-1 immunoreactivity (Fig. 3 Cc ), and inhibited Sirt-1 mRNA induction by NGF and Fsk (Fig. 3 Cb ). Thus, we conclude that CREB directly regulates Sirt-1 mRNA and protein expression in neurons. Known transcriptional regulators of Sirt-1, such as p53 and Foxo3a (31), also interact with, and are either positively or negatively modulated by the deacetylase (32, 33); similarly, a myc-tagged form of human CREB1 and Sirt-1 could be easily coimmunopre- cipated in naive PC12 cells (Fig. 4 A ), indicating physical association between the two proteins; the association was rapidly induced by the PKA agonist Fsk, in parallel with phosphorylation of CREB on Serine 133. Short-time (30 min) stimulation with Fsk did not affect the total amount of cellular Sirt-1, but rather increased the stoichiometry of the binding to CREB, suggesting either an increased af fi nity between the two proteins, or the enhanced interaction with intermediate molecular partners co-recruited at target gene promoters. In keeping with the latter possibility, a mutant CREB that retains the phosphorylation site but is unable to bind DNA because of the deletion of the C-terminal transactivation domain ( Δ LZ CREB), displayed marginal physical interaction with Sirt-1, nor was the binding induced by Fsk (Fig. 4 A ). To con fi rm that SIRT1 and CREB colocalize on CREB- responsive chromatin regions in cortical neurons, cross-linked chromatin was immunoprecipitated with an anti-Sirt-1 antiserum and Sirt-1 binding to the regions of nNOS and PGC-1 α promoters surrounding the CRE elements determined by semiquantitative PCR. Sirt-1 was found to interact with both genes, in a fashion inducible by NGF and with a binding kinetic similar to that of CREB (Fig. 4 Ba ). Sirt-1 also bound the same Sirt-1 chromatin region surrounding the +1,998/+2,012 half-CRE sites that copre- cipitates with CREB, suggesting that Sirt-1 may self-regulate its CRE-dependent transactivation (Fig. 4 Bb ). Of note, Sirt-1 interaction with nNOS and Sirt-1 chromatin was indeed mediated by CREB, as revealed by its drastic reduction in CREB loxP/loxP neurons following the recombinase-mediated deletion of the factor (Fig. 4 Bb ) and by the failure of forced Sirt-1 overexpression to restore Sirt-1 chromatin binding in CREB-de fi cient cells (Fig. S3). To determine whether Sirt-1 modulates CREB transcriptional activity, cortical neurons were stimulated with NGF in the presence of sirtuin inhibitor Nicotinamide (34); induction of nNOS and PGC-1 α mRNA by NGF was blunted upon inhibition of Sirt-1 deacetylase activity (Fig. 4 C ). A similar result was obtained in naive PC12 cells by siRNA-mediated knock-down of Sirt-1 (Fig. S4 A ). In addition, Sirt-1 – de fi cient PC12 displayed impaired differentiation by NGF (as assessed by quanti fi cation of neurite outgrowth), a response largely dependent on CREB and nNOS in this cell model (35, 36) (Fig. S4 B ). In a complementary set of experiments, lentivirus-mediated overexpression of Sirt-1 increased the expression of nNOS and PGC-1 α mRNA in hippocampal neurons treated with NGF. However, Sirt-1 did not fully restore the expression of these genes in CREB-de fi cient cells, nor NGF protection from hydrogen peroxide-dependent cell death, which is largely lost in these cells (Fig. S5), was recovered upon transduction of the Sirt-1 cDNA. Collectively, the above data suggest that Sirt-1 and CREB are, at least to some extent, re- ciprocally dependent, and act in concert in the context of neurotrophin signaling. Unlike previously reported fi ndings (16), manipulations aimed at modulating Sirt-1 expression and activity in the above contexts did not affect the level of immunoreactive CREB1 (Fig. S4 Aa ). KO Mice. Finally, to verify whether Sirt-1 actually regulates CREB- dependent transcription in vivo and this is relevant for response to CR, we analyzed the expression of CREB targets nNOS and PGC- 1 α mRNAs in whole brains of Sirt-1 KO mice fed AL or CR for 25 – 28 wk (37). This analysis revealed that both genes are markedly hypoexpressed in Sirt-de fi cient mice compared with littermate controls under both dietary regimens (Fig. 4 D ), and their induction by CR, that was more pronounced for PGC-1 α , was also attenuated (Fig. 4 D , a and b ). The amount of immunoreactive CREB was comparable in brain homogenates of the two strains (Fig. 4 Dc ), nor CREB acetylation on lysine residues was affected by either Sirt-1 deletion or CR (Fig. S6), indicating that, at least in these experimental settings, Sirt-1 regulates CREB transcriptional activity independent of the acetylation and expression level of the factor. Taken together, the above fi ndings strongly suggest that Sirt-1 modulates the expression of CREB-dependent genes in mouse brain, and by extension, identify in the decrease of CREB- dependent transcription a molecular signature for a defective brain response to CR, shared by CREB and Sirt-1 mutant mice (7). The CREB transcription factor has been widely investigated as a metabolic sensor and regulator of glucose homeostasis in liver and fat tissue (15), and as a master switch of calcium and neuro- trophin-triggered transcriptional programs regulating neuronal differentiation, survival, and plasticity in the brain and peripheral nervous system (10, 11, 18). Evidence also exist for CREB roles in the control of appetite and food intake in the hypothalamus (38), but whether neuronal plasticity and high-order cognitive functions may be in fl uenced by nutrient cues and energy metabolism through CREB, an issue relevant to major diseases like AD and type 2 diabetes (3, 4), remains to be established. Our demonstra- tion of impaired electrophysiological, cognitive, and emotional response to CR in brain CREB KO mice, clearly suggests that this may indeed be the case. Interestingly, these differences emerge in the context of an overall comparable feeding behavior and metabolic response to CR between control and BCKO mice (Fig. 1 Ab ), indicating that potential effects of hypothalamic CREB signaling on energy balance and appetite regulation unlikely account for the observed phenotypes (38). Data reported in Fig. 2, showing CREB phosphorylation and transcriptional activation by CR, indicate that CREB is metaboli- cally regulated in the cortex and hippocampus, although the mechanism of such regulation needs to be further investigated. Because plasma from CR rodents has been previously shown to induce Sirt-1 expression in several organs/tissues, a humoral/hormonal mechanism for CREB activation could be envisaged (39); as an alternative, NO (40) or oxygen species (41) may mediate, cell-autonomously, this effect. Further research in this direction is warranted. Our data identify in the complex interplay with the nutrient- sensitive histone deacetylase Sirt-1 a molecular connection between CREB and neuronal response to CR. Previous work from others has compellingly involved ...
Context 3
... mouse Sirt-1 locus (NC_000076) revealed the presence of several putative cAMP Responsive Elements (CRE) both upstream and downstream of the transcription start site (TSS) (30), and an ∼ 300- bp segment encompassing two of those elements (TGACG at +1,998 and CGTCA at +2,012 from the annotated TSS) drove transcriptional response to Fsk and PKA in a standard luciferase reporter assay performed in PC12 pheochromocytoma cells (Fig. 3 B , a and b ). Moreover, ChIP from hippocampal neurons revealed that CREB binds the same genomic region in a fashion inducible by NGF (Fig. 3 Bc ). Finally, in primary neurons isolated from CREB loxP/loxP mice, deletion of CREB by adenoviral delivery of the Cre recombinase (Fig. 3 C , a and b ) led to a reduction of Sirt-1 immunoreactivity (Fig. 3 Cc ), and inhibited Sirt-1 mRNA induction by NGF and Fsk (Fig. 3 Cb ). Thus, we conclude that CREB directly regulates Sirt-1 mRNA and protein expression in neurons. Known transcriptional regulators of Sirt-1, such as p53 and Foxo3a (31), also interact with, and are either positively or negatively modulated by the deacetylase (32, 33); similarly, a myc-tagged form of human CREB1 and Sirt-1 could be easily coimmunopre- cipated in naive PC12 cells (Fig. 4 A ), indicating physical association between the two proteins; the association was rapidly induced by the PKA agonist Fsk, in parallel with phosphorylation of CREB on Serine 133. Short-time (30 min) stimulation with Fsk did not affect the total amount of cellular Sirt-1, but rather increased the stoichiometry of the binding to CREB, suggesting either an increased af fi nity between the two proteins, or the enhanced interaction with intermediate molecular partners co-recruited at target gene promoters. In keeping with the latter possibility, a mutant CREB that retains the phosphorylation site but is unable to bind DNA because of the deletion of the C-terminal transactivation domain ( Δ LZ CREB), displayed marginal physical interaction with Sirt-1, nor was the binding induced by Fsk (Fig. 4 A ). To con fi rm that SIRT1 and CREB colocalize on CREB- responsive chromatin regions in cortical neurons, cross-linked chromatin was immunoprecipitated with an anti-Sirt-1 antiserum and Sirt-1 binding to the regions of nNOS and PGC-1 α promoters surrounding the CRE elements determined by semiquantitative PCR. Sirt-1 was found to interact with both genes, in a fashion inducible by NGF and with a binding kinetic similar to that of CREB (Fig. 4 Ba ). Sirt-1 also bound the same Sirt-1 chromatin region surrounding the +1,998/+2,012 half-CRE sites that copre- cipitates with CREB, suggesting that Sirt-1 may self-regulate its CRE-dependent transactivation (Fig. 4 Bb ). Of note, Sirt-1 interaction with nNOS and Sirt-1 chromatin was indeed mediated by CREB, as revealed by its drastic reduction in CREB loxP/loxP neurons following the recombinase-mediated deletion of the factor (Fig. 4 Bb ) and by the failure of forced Sirt-1 overexpression to restore Sirt-1 chromatin binding in CREB-de fi cient cells (Fig. S3). To determine whether Sirt-1 modulates CREB transcriptional activity, cortical neurons were stimulated with NGF in the presence of sirtuin inhibitor Nicotinamide (34); induction of nNOS and PGC-1 α mRNA by NGF was blunted upon inhibition of Sirt-1 deacetylase activity (Fig. 4 C ). A similar result was obtained in naive PC12 cells by siRNA-mediated knock-down of Sirt-1 (Fig. S4 A ). In addition, Sirt-1 – de fi cient PC12 displayed impaired differentiation by NGF (as assessed by quanti fi cation of neurite outgrowth), a response largely dependent on CREB and nNOS in this cell model (35, 36) (Fig. S4 B ). In a complementary set of experiments, lentivirus-mediated overexpression of Sirt-1 increased the expression of nNOS and PGC-1 α mRNA in hippocampal neurons treated with NGF. However, Sirt-1 did not fully restore the expression of these genes in CREB-de fi cient cells, nor NGF protection from hydrogen peroxide-dependent cell death, which is largely lost in these cells (Fig. S5), was recovered upon transduction of the Sirt-1 cDNA. Collectively, the above data suggest that Sirt-1 and CREB are, at least to some extent, re- ciprocally dependent, and act in concert in the context of neurotrophin signaling. Unlike previously reported fi ndings (16), manipulations aimed at modulating Sirt-1 expression and activity in the above contexts did not affect the level of immunoreactive CREB1 (Fig. S4 Aa ). KO Mice. Finally, to verify whether Sirt-1 actually regulates CREB- dependent transcription in vivo and this is relevant for response to CR, we analyzed the expression of CREB targets nNOS and PGC- 1 α mRNAs in whole brains of Sirt-1 KO mice fed AL or CR for 25 – 28 wk (37). This analysis revealed that both genes are markedly hypoexpressed in Sirt-de fi cient mice compared with littermate controls under both dietary regimens (Fig. 4 D ), and their induction by CR, that was more pronounced for PGC-1 α , was also attenuated (Fig. 4 D , a and b ). The amount of immunoreactive CREB was comparable in brain homogenates of the two strains (Fig. 4 Dc ), nor CREB acetylation on lysine residues was affected by either Sirt-1 deletion or CR (Fig. S6), indicating that, at least in these experimental settings, Sirt-1 regulates CREB transcriptional activity independent of the acetylation and expression level of the factor. Taken together, the above fi ndings strongly suggest that Sirt-1 modulates the expression of CREB-dependent genes in mouse brain, and by extension, identify in the decrease of CREB- dependent transcription a molecular signature for a defective brain response to CR, shared by CREB and Sirt-1 mutant mice (7). The CREB transcription factor has been widely investigated as a metabolic sensor and regulator of glucose homeostasis in liver and fat tissue (15), and as a master switch of calcium and neuro- trophin-triggered transcriptional programs regulating neuronal differentiation, survival, and plasticity in the brain and peripheral nervous system (10, 11, 18). Evidence also exist for CREB roles in the control of appetite and food intake in the hypothalamus (38), but whether neuronal plasticity and high-order cognitive functions may be in fl uenced by nutrient cues and energy metabolism through CREB, an issue relevant to major diseases like AD and type 2 diabetes (3, 4), remains to be established. Our demonstra- tion of impaired electrophysiological, cognitive, and emotional response to CR in brain CREB KO mice, clearly suggests that this may indeed be the case. Interestingly, these differences emerge in the context of an overall comparable feeding behavior and metabolic response to CR between control and BCKO mice (Fig. 1 Ab ), indicating that potential effects of hypothalamic CREB signaling on energy balance and appetite regulation unlikely account for the observed phenotypes (38). Data reported in Fig. 2, showing CREB phosphorylation and transcriptional activation by CR, indicate that CREB is metaboli- cally regulated in the cortex and hippocampus, although the mechanism of such regulation needs to be further investigated. Because plasma from CR rodents has been previously shown to induce Sirt-1 expression in several organs/tissues, a humoral/hormonal mechanism for CREB activation could be envisaged (39); as an alternative, NO (40) or oxygen species (41) may mediate, cell-autonomously, this effect. Further research in this direction is warranted. Our data identify in the complex interplay with the nutrient- sensitive histone deacetylase Sirt-1 a molecular connection between CREB and neuronal response to CR. Previous work from others has compellingly involved neuronal Sirt-1 in hormonal and metabolic adaptation to dietary restriction in mice (7). Results presented here clearly suggest that the two molecules are part of the same CR-sensitive signaling cascade. However, other genes potentially relevant to mitochondrial biogenesis and neuronal response to calorie restriction, namely nNOS and PGC-1 α (40), and some PGC-1 α targets — including CPT1 , CoxIV , and UCP-2 — are also critically down-regulated, together with Sirt-1, in the cortex and hippocampus of BCKO mice (Fig. 2 B and Fig. S2 b ). This fi nding, and evidence from Fig. S3 that overexpression of Sirt-1 does not rescue NGF signaling in CREB-de fi cient hippocampal neurons in vitro, suggests that up-regulation of Sirt-1 is not the only mechanism whereby CREB participates in neuronal response to nutrients. Instead, cooperation with CREB is likely critical for the action of neuronal Sirt-1 in CR. This view is supported by the unexpected fi nding that CREB-dependent genes involved in neuronal plasticity, survival, and stress resistance (42, 43), and induced by calorie restriction (Fig. 2), are markedly down-regulated in Sirt-1 – de fi cient cultured neurons (PC12) and in the brain of Sirt-1 KO mice. Because the latter strain is, like BCKO mice, impaired in brain response to reduced food intake (7), the above evidence further support the notion that CREB-dependent transcription has a pivotal role in the neuronal effects of calorie restriction, and identify in the CREB – Sirt-1 axis a major component of the nutrient sensitive molecular network that connects caloric intake and energy metabolism to brain health. Biochemical details of how Sirt-1 affects CREB activity remains to be clari fi ed. Unlike other transcription factors, CREB does not appear to be deacetylated by Sirt-1 (Fig. S6); moreover, we could not detect consistent effects of Sirt-1 on the expression of CREB (Fig. 4 Dc , and Figs. S4 Aa and S6), unlike that described by Gao et al. (16). Those authors, however, made their important observations on Sirt-1 Δ ex4 mice that, unlike Sirt-1KO mice used in our experiments, do express an inactive form of Sirt-1 potentially acting in a dominant negative fashion against other molecules (deacetylases?) capable of regulating CREB expression. On the other hand, Sirt-1 ...
Context 4
... and downstream of the transcription start site (TSS) (30), and an ∼ 300- bp segment encompassing two of those elements (TGACG at +1,998 and CGTCA at +2,012 from the annotated TSS) drove transcriptional response to Fsk and PKA in a standard luciferase reporter assay performed in PC12 pheochromocytoma cells (Fig. 3 B , a and b ). Moreover, ChIP from hippocampal neurons revealed that CREB binds the same genomic region in a fashion inducible by NGF (Fig. 3 Bc ). Finally, in primary neurons isolated from CREB loxP/loxP mice, deletion of CREB by adenoviral delivery of the Cre recombinase (Fig. 3 C , a and b ) led to a reduction of Sirt-1 immunoreactivity (Fig. 3 Cc ), and inhibited Sirt-1 mRNA induction by NGF and Fsk (Fig. 3 Cb ). Thus, we conclude that CREB directly regulates Sirt-1 mRNA and protein expression in neurons. Known transcriptional regulators of Sirt-1, such as p53 and Foxo3a (31), also interact with, and are either positively or negatively modulated by the deacetylase (32, 33); similarly, a myc-tagged form of human CREB1 and Sirt-1 could be easily coimmunopre- cipated in naive PC12 cells (Fig. 4 A ), indicating physical association between the two proteins; the association was rapidly induced by the PKA agonist Fsk, in parallel with phosphorylation of CREB on Serine 133. Short-time (30 min) stimulation with Fsk did not affect the total amount of cellular Sirt-1, but rather increased the stoichiometry of the binding to CREB, suggesting either an increased af fi nity between the two proteins, or the enhanced interaction with intermediate molecular partners co-recruited at target gene promoters. In keeping with the latter possibility, a mutant CREB that retains the phosphorylation site but is unable to bind DNA because of the deletion of the C-terminal transactivation domain ( Δ LZ CREB), displayed marginal physical interaction with Sirt-1, nor was the binding induced by Fsk (Fig. 4 A ). To con fi rm that SIRT1 and CREB colocalize on CREB- responsive chromatin regions in cortical neurons, cross-linked chromatin was immunoprecipitated with an anti-Sirt-1 antiserum and Sirt-1 binding to the regions of nNOS and PGC-1 α promoters surrounding the CRE elements determined by semiquantitative PCR. Sirt-1 was found to interact with both genes, in a fashion inducible by NGF and with a binding kinetic similar to that of CREB (Fig. 4 Ba ). Sirt-1 also bound the same Sirt-1 chromatin region surrounding the +1,998/+2,012 half-CRE sites that copre- cipitates with CREB, suggesting that Sirt-1 may self-regulate its CRE-dependent transactivation (Fig. 4 Bb ). Of note, Sirt-1 interaction with nNOS and Sirt-1 chromatin was indeed mediated by CREB, as revealed by its drastic reduction in CREB loxP/loxP neurons following the recombinase-mediated deletion of the factor (Fig. 4 Bb ) and by the failure of forced Sirt-1 overexpression to restore Sirt-1 chromatin binding in CREB-de fi cient cells (Fig. S3). To determine whether Sirt-1 modulates CREB transcriptional activity, cortical neurons were stimulated with NGF in the presence of sirtuin inhibitor Nicotinamide (34); induction of nNOS and PGC-1 α mRNA by NGF was blunted upon inhibition of Sirt-1 deacetylase activity (Fig. 4 C ). A similar result was obtained in naive PC12 cells by siRNA-mediated knock-down of Sirt-1 (Fig. S4 A ). In addition, Sirt-1 – de fi cient PC12 displayed impaired differentiation by NGF (as assessed by quanti fi cation of neurite outgrowth), a response largely dependent on CREB and nNOS in this cell model (35, 36) (Fig. S4 B ). In a complementary set of experiments, lentivirus-mediated overexpression of Sirt-1 increased the expression of nNOS and PGC-1 α mRNA in hippocampal neurons treated with NGF. However, Sirt-1 did not fully restore the expression of these genes in CREB-de fi cient cells, nor NGF protection from hydrogen peroxide-dependent cell death, which is largely lost in these cells (Fig. S5), was recovered upon transduction of the Sirt-1 cDNA. Collectively, the above data suggest that Sirt-1 and CREB are, at least to some extent, re- ciprocally dependent, and act in concert in the context of neurotrophin signaling. Unlike previously reported fi ndings (16), manipulations aimed at modulating Sirt-1 expression and activity in the above contexts did not affect the level of immunoreactive CREB1 (Fig. S4 Aa ). KO Mice. Finally, to verify whether Sirt-1 actually regulates CREB- dependent transcription in vivo and this is relevant for response to CR, we analyzed the expression of CREB targets nNOS and PGC- 1 α mRNAs in whole brains of Sirt-1 KO mice fed AL or CR for 25 – 28 wk (37). This analysis revealed that both genes are markedly hypoexpressed in Sirt-de fi cient mice compared with littermate controls under both dietary regimens (Fig. 4 D ), and their induction by CR, that was more pronounced for PGC-1 α , was also attenuated (Fig. 4 D , a and b ). The amount of immunoreactive CREB was comparable in brain homogenates of the two strains (Fig. 4 Dc ), nor CREB acetylation on lysine residues was affected by either Sirt-1 deletion or CR (Fig. S6), indicating that, at least in these experimental settings, Sirt-1 regulates CREB transcriptional activity independent of the acetylation and expression level of the factor. Taken together, the above fi ndings strongly suggest that Sirt-1 modulates the expression of CREB-dependent genes in mouse brain, and by extension, identify in the decrease of CREB- dependent transcription a molecular signature for a defective brain response to CR, shared by CREB and Sirt-1 mutant mice (7). The CREB transcription factor has been widely investigated as a metabolic sensor and regulator of glucose homeostasis in liver and fat tissue (15), and as a master switch of calcium and neuro- trophin-triggered transcriptional programs regulating neuronal differentiation, survival, and plasticity in the brain and peripheral nervous system (10, 11, 18). Evidence also exist for CREB roles in the control of appetite and food intake in the hypothalamus (38), but whether neuronal plasticity and high-order cognitive functions may be in fl uenced by nutrient cues and energy metabolism through CREB, an issue relevant to major diseases like AD and type 2 diabetes (3, 4), remains to be established. Our demonstra- tion of impaired electrophysiological, cognitive, and emotional response to CR in brain CREB KO mice, clearly suggests that this may indeed be the case. Interestingly, these differences emerge in the context of an overall comparable feeding behavior and metabolic response to CR between control and BCKO mice (Fig. 1 Ab ), indicating that potential effects of hypothalamic CREB signaling on energy balance and appetite regulation unlikely account for the observed phenotypes (38). Data reported in Fig. 2, showing CREB phosphorylation and transcriptional activation by CR, indicate that CREB is metaboli- cally regulated in the cortex and hippocampus, although the mechanism of such regulation needs to be further investigated. Because plasma from CR rodents has been previously shown to induce Sirt-1 expression in several organs/tissues, a humoral/hormonal mechanism for CREB activation could be envisaged (39); as an alternative, NO (40) or oxygen species (41) may mediate, cell-autonomously, this effect. Further research in this direction is warranted. Our data identify in the complex interplay with the nutrient- sensitive histone deacetylase Sirt-1 a molecular connection between CREB and neuronal response to CR. Previous work from others has compellingly involved neuronal Sirt-1 in hormonal and metabolic adaptation to dietary restriction in mice (7). Results presented here clearly suggest that the two molecules are part of the same CR-sensitive signaling cascade. However, other genes potentially relevant to mitochondrial biogenesis and neuronal response to calorie restriction, namely nNOS and PGC-1 α (40), and some PGC-1 α targets — including CPT1 , CoxIV , and UCP-2 — are also critically down-regulated, together with Sirt-1, in the cortex and hippocampus of BCKO mice (Fig. 2 B and Fig. S2 b ). This fi nding, and evidence from Fig. S3 that overexpression of Sirt-1 does not rescue NGF signaling in CREB-de fi cient hippocampal neurons in vitro, suggests that up-regulation of Sirt-1 is not the only mechanism whereby CREB participates in neuronal response to nutrients. Instead, cooperation with CREB is likely critical for the action of neuronal Sirt-1 in CR. This view is supported by the unexpected fi nding that CREB-dependent genes involved in neuronal plasticity, survival, and stress resistance (42, 43), and induced by calorie restriction (Fig. 2), are markedly down-regulated in Sirt-1 – de fi cient cultured neurons (PC12) and in the brain of Sirt-1 KO mice. Because the latter strain is, like BCKO mice, impaired in brain response to reduced food intake (7), the above evidence further support the notion that CREB-dependent transcription has a pivotal role in the neuronal effects of calorie restriction, and identify in the CREB – Sirt-1 axis a major component of the nutrient sensitive molecular network that connects caloric intake and energy metabolism to brain health. Biochemical details of how Sirt-1 affects CREB activity remains to be clari fi ed. Unlike other transcription factors, CREB does not appear to be deacetylated by Sirt-1 (Fig. S6); moreover, we could not detect consistent effects of Sirt-1 on the expression of CREB (Fig. 4 Dc , and Figs. S4 Aa and S6), unlike that described by Gao et al. (16). Those authors, however, made their important observations on Sirt-1 Δ ex4 mice that, unlike Sirt-1KO mice used in our experiments, do express an inactive form of Sirt-1 potentially acting in a dominant negative fashion against other molecules (deacetylases?) capable of regulating CREB expression. On the other hand, Sirt-1 dependent modulation of nutrient sensitive cofactors of CREB, like TORC/Crtc (38), has been described in the liver ...
Context 5
... for response to CR, we analyzed the expression of CREB targets nNOS and PGC- 1 α mRNAs in whole brains of Sirt-1 KO mice fed AL or CR for 25 – 28 wk (37). This analysis revealed that both genes are markedly hypoexpressed in Sirt-de fi cient mice compared with littermate controls under both dietary regimens (Fig. 4 D ), and their induction by CR, that was more pronounced for PGC-1 α , was also attenuated (Fig. 4 D , a and b ). The amount of immunoreactive CREB was comparable in brain homogenates of the two strains (Fig. 4 Dc ), nor CREB acetylation on lysine residues was affected by either Sirt-1 deletion or CR (Fig. S6), indicating that, at least in these experimental settings, Sirt-1 regulates CREB transcriptional activity independent of the acetylation and expression level of the factor. Taken together, the above fi ndings strongly suggest that Sirt-1 modulates the expression of CREB-dependent genes in mouse brain, and by extension, identify in the decrease of CREB- dependent transcription a molecular signature for a defective brain response to CR, shared by CREB and Sirt-1 mutant mice (7). The CREB transcription factor has been widely investigated as a metabolic sensor and regulator of glucose homeostasis in liver and fat tissue (15), and as a master switch of calcium and neuro- trophin-triggered transcriptional programs regulating neuronal differentiation, survival, and plasticity in the brain and peripheral nervous system (10, 11, 18). Evidence also exist for CREB roles in the control of appetite and food intake in the hypothalamus (38), but whether neuronal plasticity and high-order cognitive functions may be in fl uenced by nutrient cues and energy metabolism through CREB, an issue relevant to major diseases like AD and type 2 diabetes (3, 4), remains to be established. Our demonstra- tion of impaired electrophysiological, cognitive, and emotional response to CR in brain CREB KO mice, clearly suggests that this may indeed be the case. Interestingly, these differences emerge in the context of an overall comparable feeding behavior and metabolic response to CR between control and BCKO mice (Fig. 1 Ab ), indicating that potential effects of hypothalamic CREB signaling on energy balance and appetite regulation unlikely account for the observed phenotypes (38). Data reported in Fig. 2, showing CREB phosphorylation and transcriptional activation by CR, indicate that CREB is metaboli- cally regulated in the cortex and hippocampus, although the mechanism of such regulation needs to be further investigated. Because plasma from CR rodents has been previously shown to induce Sirt-1 expression in several organs/tissues, a humoral/hormonal mechanism for CREB activation could be envisaged (39); as an alternative, NO (40) or oxygen species (41) may mediate, cell-autonomously, this effect. Further research in this direction is warranted. Our data identify in the complex interplay with the nutrient- sensitive histone deacetylase Sirt-1 a molecular connection between CREB and neuronal response to CR. Previous work from others has compellingly involved neuronal Sirt-1 in hormonal and metabolic adaptation to dietary restriction in mice (7). Results presented here clearly suggest that the two molecules are part of the same CR-sensitive signaling cascade. However, other genes potentially relevant to mitochondrial biogenesis and neuronal response to calorie restriction, namely nNOS and PGC-1 α (40), and some PGC-1 α targets — including CPT1 , CoxIV , and UCP-2 — are also critically down-regulated, together with Sirt-1, in the cortex and hippocampus of BCKO mice (Fig. 2 B and Fig. S2 b ). This fi nding, and evidence from Fig. S3 that overexpression of Sirt-1 does not rescue NGF signaling in CREB-de fi cient hippocampal neurons in vitro, suggests that up-regulation of Sirt-1 is not the only mechanism whereby CREB participates in neuronal response to nutrients. Instead, cooperation with CREB is likely critical for the action of neuronal Sirt-1 in CR. This view is supported by the unexpected fi nding that CREB-dependent genes involved in neuronal plasticity, survival, and stress resistance (42, 43), and induced by calorie restriction (Fig. 2), are markedly down-regulated in Sirt-1 – de fi cient cultured neurons (PC12) and in the brain of Sirt-1 KO mice. Because the latter strain is, like BCKO mice, impaired in brain response to reduced food intake (7), the above evidence further support the notion that CREB-dependent transcription has a pivotal role in the neuronal effects of calorie restriction, and identify in the CREB – Sirt-1 axis a major component of the nutrient sensitive molecular network that connects caloric intake and energy metabolism to brain health. Biochemical details of how Sirt-1 affects CREB activity remains to be clari fi ed. Unlike other transcription factors, CREB does not appear to be deacetylated by Sirt-1 (Fig. S6); moreover, we could not detect consistent effects of Sirt-1 on the expression of CREB (Fig. 4 Dc , and Figs. S4 Aa and S6), unlike that described by Gao et al. (16). Those authors, however, made their important observations on Sirt-1 Δ ex4 mice that, unlike Sirt-1KO mice used in our experiments, do express an inactive form of Sirt-1 potentially acting in a dominant negative fashion against other molecules (deacetylases?) capable of regulating CREB expression. On the other hand, Sirt-1 dependent modulation of nutrient sensitive cofactors of CREB, like TORC/Crtc (38), has been described in the liver and may occur in neuronal cells. Alternatively, Sirt-1 may be recruited by CREB on target promoters and act either on histones (Fig. 2 D ) or, in trans , on other (maybe com- peting) transcription factors. In conclusion, CREB1, an effector of neurotrophins involved in several age-associated neurodegenerative diseases, mediates at least some brain responses to dietary restriction. This action involves the up-regulation of Sirt-1 that in turn bolsters the CREB- dependent expression of genes involved in neuronal metabolism, survival and plasticity (Fig. S7). Although molecular details on how Sirt-1 regulates gene transcription by CREB need to be further investigated, the CREB-Sirt-1 axis outlines a unique molecular network at the crossroad of energy metabolism, metabolic diseases, and brain ...
Context 6
... object and spatial memory. This indicator of neuronal plasticity declines in rodents with age, in a fashion that is attenuated by calorie restriction (24). To extend our analysis of CR effects on the brain of CREB-de fi cient mice, we tested LTP at CA3-CA1 synapses in hippocampal brain slices obtained from animals euthanized after 5 wk of either CR or AL feeding regimen. As previously shown (25), LTP was not affected by CREB status in the two AL-fed groups. Strikingly, however, LTP increased in control mice under CR [AL 136.6 ± 8.0% ( n = 10 slices) vs. CR 161.3 ± 8.2% ( n = 9 slices); P < 0.05] but not in the BCKO strain [AL 130.3 ± 8.6% ( n = 9 slices) vs. CR 121.0 ± 8.5% ( n = 10 slices), n.s.] (Fig. 1 C ), indicating that bene fi cial effect of CR on LTP is also abrogated by brain-speci fi c CREB deletion. The increased LTP induced by CR in control mice was independent on changes in basal synaptic transmission, because input/output curves obtained plotting fi eld excitatory postsynaptic potential (fEPSP) amplitudes vs. stimulus intensity in hippocampal brain slices of CR and AL animals were superimposable. Because CREB appeared to participate in brain response to CR, we assessed whether diet affected the total amount/phosphoryla- tion of CREB protein, and the expression level of a number of mRNAs known to be regulated by CREB (23). CREB1 protein expression in control mice was not changed by the dietary regimen; instead, an increase in CREB phosphorylation on Serine 133 in the hippocampi of the CR group was revealed by phospho-speci fi c immunoblotting (Fig. 2 A ), suggesting that CR activates CREB in this brain area. Accordingly, mRNAs of “ canonical ” CREB targets peroxisome proliferator-activated receptor- γ coactivator-1 α ( PGC- 1 α ), nNOS , and phosphoenolpyruvate carboxykinase ( PEPCK ) were induced by CR in the cortex and hippocampus of CREB- pro fi cient mice (Fig. 2 B ). Of note, both PGC-1 α and NO, the product of NOS enzymes, are known to promote mitochondrial biogenesis and to participate in organismal response to CR (26). The three mRNAs were overall down-regulated in BCKO mice fed AL compared with controls, and their induction by CR, observed in control mice, was nearly abolished (Fig. 2 B ). Interestingly, the expression of other putative CREB targets like bcl-2 , NGF , and c-fos were unaffected by CR and CREB deletion (Fig. S2). Thus, collectively, these fi ndings demonstrate that nutrient availability selectively regulates CREB-dependent gene expression in the forebrain. We next asked how CR may affect CREB activity. Because Sirt- 1 is a metabolic sensor involved in several biological consequences of nutrient deprivation (27, 6), and animals lacking Sirt-1 in the brain show defective behavioral and hormonal responses to CR (7), we investigated the sirtuin as a potential CREB interactor. Sirt-1 mRNA was drastically reduced both in the cortex and hippocampus of BCKO mice, irrespective of the dietary regimen. Moreover, the moderate induction by CR that we observed in control brains was completely lost in the corresponding CREB- de fi cient tissues (Fig. 2 C ). Finally, acetylation of Histones H3 (AcK9) and H4 (AcK16), an inverse correlate of Sirt-1 activity (28), was abnormally high in BCKO hippocampal homogenates (Fig. 2 D ), con fi rming an overall reduction of Sirt-1 activity in this area of the brain. Transcriptional Regulation of Sirt-1 by CREB in Neurons. These observations suggested that Sirt-1 may represent a direct transcriptional target of CREB. Accordingly, CREB activators NGF and Forskolin (Fsk) (29) raised the level of immunoreactive Sirt-1 and of the corresponding mRNA in cultured primary cortical and hippocampal neurons (Fig. 3 A ). Bioinformatic analysis of the mouse Sirt-1 locus (NC_000076) revealed the presence of several putative cAMP Responsive Elements (CRE) both upstream and downstream of the transcription start site (TSS) (30), and an ∼ 300- bp segment encompassing two of those elements (TGACG at +1,998 and CGTCA at +2,012 from the annotated TSS) drove transcriptional response to Fsk and PKA in a standard luciferase reporter assay performed in PC12 pheochromocytoma cells (Fig. 3 B , a and b ). Moreover, ChIP from hippocampal neurons revealed that CREB binds the same genomic region in a fashion inducible by NGF (Fig. 3 Bc ). Finally, in primary neurons isolated from CREB loxP/loxP mice, deletion of CREB by adenoviral delivery of the Cre recombinase (Fig. 3 C , a and b ) led to a reduction of Sirt-1 immunoreactivity (Fig. 3 Cc ), and inhibited Sirt-1 mRNA induction by NGF and Fsk (Fig. 3 Cb ). Thus, we conclude that CREB directly regulates Sirt-1 mRNA and protein expression in neurons. Known transcriptional regulators of Sirt-1, such as p53 and Foxo3a (31), also interact with, and are either positively or negatively modulated by the deacetylase (32, 33); similarly, a myc-tagged form of human CREB1 and Sirt-1 could be easily coimmunopre- cipated in naive PC12 cells (Fig. 4 A ), indicating physical association between the two proteins; the association was rapidly induced by the PKA agonist Fsk, in parallel with phosphorylation of CREB on Serine 133. Short-time (30 min) stimulation with Fsk did not affect the total amount of cellular Sirt-1, but rather increased the stoichiometry of the binding to CREB, suggesting either an increased af fi nity between the two proteins, or the enhanced interaction with intermediate molecular partners co-recruited at target gene promoters. In keeping with the latter possibility, a mutant CREB that retains the phosphorylation site but is unable to bind DNA because of the deletion of the C-terminal transactivation domain ( Δ LZ CREB), displayed marginal physical interaction with Sirt-1, nor was the binding induced by Fsk (Fig. 4 A ). To con fi rm that SIRT1 and CREB colocalize on CREB- responsive chromatin regions in cortical neurons, cross-linked chromatin was immunoprecipitated with an anti-Sirt-1 antiserum and Sirt-1 binding to the regions of nNOS and PGC-1 α promoters surrounding the CRE elements determined by semiquantitative PCR. Sirt-1 was found to interact with both genes, in a fashion inducible by NGF and with a binding kinetic similar to that of CREB (Fig. 4 Ba ). Sirt-1 also bound the same Sirt-1 chromatin region surrounding the +1,998/+2,012 half-CRE sites that copre- cipitates with CREB, suggesting that Sirt-1 may self-regulate its CRE-dependent transactivation (Fig. 4 Bb ). Of note, Sirt-1 interaction with nNOS and Sirt-1 chromatin was indeed mediated by CREB, as revealed by its drastic reduction in CREB loxP/loxP neurons following the recombinase-mediated deletion of the factor (Fig. 4 Bb ) and by the failure of forced Sirt-1 overexpression to restore Sirt-1 chromatin binding in CREB-de fi cient cells (Fig. S3). To determine whether Sirt-1 modulates CREB transcriptional activity, cortical neurons were stimulated with NGF in the presence of sirtuin inhibitor Nicotinamide (34); induction of nNOS and PGC-1 α mRNA by NGF was blunted upon inhibition of Sirt-1 deacetylase activity (Fig. 4 C ). A similar result was obtained in naive PC12 cells by siRNA-mediated knock-down of Sirt-1 (Fig. S4 A ). In addition, Sirt-1 – de fi cient PC12 displayed impaired differentiation by NGF (as assessed by quanti fi cation of neurite outgrowth), a response largely dependent on CREB and nNOS in this cell model (35, 36) (Fig. S4 B ). In a complementary set of experiments, lentivirus-mediated overexpression of Sirt-1 increased the expression of nNOS and PGC-1 α mRNA in hippocampal neurons treated with NGF. However, Sirt-1 did not fully restore the expression of these genes in CREB-de fi cient cells, nor NGF protection from hydrogen peroxide-dependent cell death, which is largely lost in these cells (Fig. S5), was recovered upon transduction of the Sirt-1 cDNA. Collectively, the above data suggest that Sirt-1 and CREB are, at least to some extent, re- ciprocally dependent, and act in concert in the context of neurotrophin signaling. Unlike previously reported fi ndings (16), manipulations aimed at modulating Sirt-1 expression and activity in the above contexts did not affect the level of immunoreactive CREB1 (Fig. S4 Aa ). KO Mice. Finally, to verify whether Sirt-1 actually regulates CREB- dependent transcription in vivo and this is relevant for response to CR, we analyzed the expression of CREB targets nNOS and PGC- 1 α mRNAs in whole brains of Sirt-1 KO mice fed AL or CR for 25 – 28 wk (37). This analysis revealed that both genes are markedly hypoexpressed in Sirt-de fi cient mice compared with littermate controls under both dietary regimens (Fig. 4 D ), and their induction by CR, that was more pronounced for PGC-1 α , was also attenuated (Fig. 4 D , a and b ). The amount of immunoreactive CREB was comparable in brain homogenates of the two strains (Fig. 4 Dc ), nor CREB acetylation on lysine residues was affected by either Sirt-1 deletion or CR (Fig. S6), indicating that, at least in these experimental settings, Sirt-1 regulates CREB transcriptional activity independent of the acetylation and expression level of the factor. Taken together, the above fi ndings strongly suggest that Sirt-1 modulates the expression of CREB-dependent genes in mouse brain, and by extension, identify in the decrease of CREB- dependent transcription a molecular signature for a defective brain response to CR, shared by CREB and Sirt-1 mutant mice (7). The CREB transcription factor has been widely investigated as a metabolic sensor and regulator of glucose homeostasis in liver and fat tissue (15), and as a master switch of calcium and neuro- trophin-triggered transcriptional programs regulating neuronal differentiation, survival, and plasticity in the brain and peripheral nervous system (10, 11, 18). Evidence also exist for CREB roles in the control of appetite and food intake in the hypothalamus (38), but whether neuronal plasticity and high-order cognitive functions ...
Context 7
... targets peroxisome proliferator-activated receptor- γ coactivator-1 α ( PGC- 1 α ), nNOS , and phosphoenolpyruvate carboxykinase ( PEPCK ) were induced by CR in the cortex and hippocampus of CREB- pro fi cient mice (Fig. 2 B ). Of note, both PGC-1 α and NO, the product of NOS enzymes, are known to promote mitochondrial biogenesis and to participate in organismal response to CR (26). The three mRNAs were overall down-regulated in BCKO mice fed AL compared with controls, and their induction by CR, observed in control mice, was nearly abolished (Fig. 2 B ). Interestingly, the expression of other putative CREB targets like bcl-2 , NGF , and c-fos were unaffected by CR and CREB deletion (Fig. S2). Thus, collectively, these fi ndings demonstrate that nutrient availability selectively regulates CREB-dependent gene expression in the forebrain. We next asked how CR may affect CREB activity. Because Sirt- 1 is a metabolic sensor involved in several biological consequences of nutrient deprivation (27, 6), and animals lacking Sirt-1 in the brain show defective behavioral and hormonal responses to CR (7), we investigated the sirtuin as a potential CREB interactor. Sirt-1 mRNA was drastically reduced both in the cortex and hippocampus of BCKO mice, irrespective of the dietary regimen. Moreover, the moderate induction by CR that we observed in control brains was completely lost in the corresponding CREB- de fi cient tissues (Fig. 2 C ). Finally, acetylation of Histones H3 (AcK9) and H4 (AcK16), an inverse correlate of Sirt-1 activity (28), was abnormally high in BCKO hippocampal homogenates (Fig. 2 D ), con fi rming an overall reduction of Sirt-1 activity in this area of the brain. Transcriptional Regulation of Sirt-1 by CREB in Neurons. These observations suggested that Sirt-1 may represent a direct transcriptional target of CREB. Accordingly, CREB activators NGF and Forskolin (Fsk) (29) raised the level of immunoreactive Sirt-1 and of the corresponding mRNA in cultured primary cortical and hippocampal neurons (Fig. 3 A ). Bioinformatic analysis of the mouse Sirt-1 locus (NC_000076) revealed the presence of several putative cAMP Responsive Elements (CRE) both upstream and downstream of the transcription start site (TSS) (30), and an ∼ 300- bp segment encompassing two of those elements (TGACG at +1,998 and CGTCA at +2,012 from the annotated TSS) drove transcriptional response to Fsk and PKA in a standard luciferase reporter assay performed in PC12 pheochromocytoma cells (Fig. 3 B , a and b ). Moreover, ChIP from hippocampal neurons revealed that CREB binds the same genomic region in a fashion inducible by NGF (Fig. 3 Bc ). Finally, in primary neurons isolated from CREB loxP/loxP mice, deletion of CREB by adenoviral delivery of the Cre recombinase (Fig. 3 C , a and b ) led to a reduction of Sirt-1 immunoreactivity (Fig. 3 Cc ), and inhibited Sirt-1 mRNA induction by NGF and Fsk (Fig. 3 Cb ). Thus, we conclude that CREB directly regulates Sirt-1 mRNA and protein expression in neurons. Known transcriptional regulators of Sirt-1, such as p53 and Foxo3a (31), also interact with, and are either positively or negatively modulated by the deacetylase (32, 33); similarly, a myc-tagged form of human CREB1 and Sirt-1 could be easily coimmunopre- cipated in naive PC12 cells (Fig. 4 A ), indicating physical association between the two proteins; the association was rapidly induced by the PKA agonist Fsk, in parallel with phosphorylation of CREB on Serine 133. Short-time (30 min) stimulation with Fsk did not affect the total amount of cellular Sirt-1, but rather increased the stoichiometry of the binding to CREB, suggesting either an increased af fi nity between the two proteins, or the enhanced interaction with intermediate molecular partners co-recruited at target gene promoters. In keeping with the latter possibility, a mutant CREB that retains the phosphorylation site but is unable to bind DNA because of the deletion of the C-terminal transactivation domain ( Δ LZ CREB), displayed marginal physical interaction with Sirt-1, nor was the binding induced by Fsk (Fig. 4 A ). To con fi rm that SIRT1 and CREB colocalize on CREB- responsive chromatin regions in cortical neurons, cross-linked chromatin was immunoprecipitated with an anti-Sirt-1 antiserum and Sirt-1 binding to the regions of nNOS and PGC-1 α promoters surrounding the CRE elements determined by semiquantitative PCR. Sirt-1 was found to interact with both genes, in a fashion inducible by NGF and with a binding kinetic similar to that of CREB (Fig. 4 Ba ). Sirt-1 also bound the same Sirt-1 chromatin region surrounding the +1,998/+2,012 half-CRE sites that copre- cipitates with CREB, suggesting that Sirt-1 may self-regulate its CRE-dependent transactivation (Fig. 4 Bb ). Of note, Sirt-1 interaction with nNOS and Sirt-1 chromatin was indeed mediated by CREB, as revealed by its drastic reduction in CREB loxP/loxP neurons following the recombinase-mediated deletion of the factor (Fig. 4 Bb ) and by the failure of forced Sirt-1 overexpression to restore Sirt-1 chromatin binding in CREB-de fi cient cells (Fig. S3). To determine whether Sirt-1 modulates CREB transcriptional activity, cortical neurons were stimulated with NGF in the presence of sirtuin inhibitor Nicotinamide (34); induction of nNOS and PGC-1 α mRNA by NGF was blunted upon inhibition of Sirt-1 deacetylase activity (Fig. 4 C ). A similar result was obtained in naive PC12 cells by siRNA-mediated knock-down of Sirt-1 (Fig. S4 A ). In addition, Sirt-1 – de fi cient PC12 displayed impaired differentiation by NGF (as assessed by quanti fi cation of neurite outgrowth), a response largely dependent on CREB and nNOS in this cell model (35, 36) (Fig. S4 B ). In a complementary set of experiments, lentivirus-mediated overexpression of Sirt-1 increased the expression of nNOS and PGC-1 α mRNA in hippocampal neurons treated with NGF. However, Sirt-1 did not fully restore the expression of these genes in CREB-de fi cient cells, nor NGF protection from hydrogen peroxide-dependent cell death, which is largely lost in these cells (Fig. S5), was recovered upon transduction of the Sirt-1 cDNA. Collectively, the above data suggest that Sirt-1 and CREB are, at least to some extent, re- ciprocally dependent, and act in concert in the context of neurotrophin signaling. Unlike previously reported fi ndings (16), manipulations aimed at modulating Sirt-1 expression and activity in the above contexts did not affect the level of immunoreactive CREB1 (Fig. S4 Aa ). KO Mice. Finally, to verify whether Sirt-1 actually regulates CREB- dependent transcription in vivo and this is relevant for response to CR, we analyzed the expression of CREB targets nNOS and PGC- 1 α mRNAs in whole brains of Sirt-1 KO mice fed AL or CR for 25 – 28 wk (37). This analysis revealed that both genes are markedly hypoexpressed in Sirt-de fi cient mice compared with littermate controls under both dietary regimens (Fig. 4 D ), and their induction by CR, that was more pronounced for PGC-1 α , was also attenuated (Fig. 4 D , a and b ). The amount of immunoreactive CREB was comparable in brain homogenates of the two strains (Fig. 4 Dc ), nor CREB acetylation on lysine residues was affected by either Sirt-1 deletion or CR (Fig. S6), indicating that, at least in these experimental settings, Sirt-1 regulates CREB transcriptional activity independent of the acetylation and expression level of the factor. Taken together, the above fi ndings strongly suggest that Sirt-1 modulates the expression of CREB-dependent genes in mouse brain, and by extension, identify in the decrease of CREB- dependent transcription a molecular signature for a defective brain response to CR, shared by CREB and Sirt-1 mutant mice (7). The CREB transcription factor has been widely investigated as a metabolic sensor and regulator of glucose homeostasis in liver and fat tissue (15), and as a master switch of calcium and neuro- trophin-triggered transcriptional programs regulating neuronal differentiation, survival, and plasticity in the brain and peripheral nervous system (10, 11, 18). Evidence also exist for CREB roles in the control of appetite and food intake in the hypothalamus (38), but whether neuronal plasticity and high-order cognitive functions may be in fl uenced by nutrient cues and energy metabolism through CREB, an issue relevant to major diseases like AD and type 2 diabetes (3, 4), remains to be established. Our demonstra- tion of impaired electrophysiological, cognitive, and emotional response to CR in brain CREB KO mice, clearly suggests that this may indeed be the case. Interestingly, these differences emerge in the context of an overall comparable feeding behavior and metabolic response to CR between control and BCKO mice (Fig. 1 Ab ), indicating that potential effects of hypothalamic CREB signaling on energy balance and appetite regulation unlikely account for the observed phenotypes (38). Data reported in Fig. 2, showing CREB phosphorylation and transcriptional activation by CR, indicate that CREB is metaboli- cally regulated in the cortex and hippocampus, although the mechanism of such regulation needs to be further investigated. Because plasma from CR rodents has been previously shown to induce Sirt-1 expression in several organs/tissues, a humoral/hormonal mechanism for CREB activation could be envisaged (39); as an alternative, NO (40) or oxygen species (41) may mediate, cell-autonomously, this effect. Further research in this direction is warranted. Our data identify in the complex interplay with the nutrient- sensitive histone deacetylase Sirt-1 a molecular connection between CREB and neuronal response to CR. Previous work from others has compellingly involved neuronal Sirt-1 in hormonal and metabolic adaptation to dietary restriction in mice (7). Results presented here clearly suggest that the two molecules are part of the same CR-sensitive signaling cascade. However, other genes ...
Context 8
... overall down-regulated in BCKO mice fed AL compared with controls, and their induction by CR, observed in control mice, was nearly abolished (Fig. 2 B ). Interestingly, the expression of other putative CREB targets like bcl-2 , NGF , and c-fos were unaffected by CR and CREB deletion (Fig. S2). Thus, collectively, these fi ndings demonstrate that nutrient availability selectively regulates CREB-dependent gene expression in the forebrain. We next asked how CR may affect CREB activity. Because Sirt- 1 is a metabolic sensor involved in several biological consequences of nutrient deprivation (27, 6), and animals lacking Sirt-1 in the brain show defective behavioral and hormonal responses to CR (7), we investigated the sirtuin as a potential CREB interactor. Sirt-1 mRNA was drastically reduced both in the cortex and hippocampus of BCKO mice, irrespective of the dietary regimen. Moreover, the moderate induction by CR that we observed in control brains was completely lost in the corresponding CREB- de fi cient tissues (Fig. 2 C ). Finally, acetylation of Histones H3 (AcK9) and H4 (AcK16), an inverse correlate of Sirt-1 activity (28), was abnormally high in BCKO hippocampal homogenates (Fig. 2 D ), con fi rming an overall reduction of Sirt-1 activity in this area of the brain. Transcriptional Regulation of Sirt-1 by CREB in Neurons. These observations suggested that Sirt-1 may represent a direct transcriptional target of CREB. Accordingly, CREB activators NGF and Forskolin (Fsk) (29) raised the level of immunoreactive Sirt-1 and of the corresponding mRNA in cultured primary cortical and hippocampal neurons (Fig. 3 A ). Bioinformatic analysis of the mouse Sirt-1 locus (NC_000076) revealed the presence of several putative cAMP Responsive Elements (CRE) both upstream and downstream of the transcription start site (TSS) (30), and an ∼ 300- bp segment encompassing two of those elements (TGACG at +1,998 and CGTCA at +2,012 from the annotated TSS) drove transcriptional response to Fsk and PKA in a standard luciferase reporter assay performed in PC12 pheochromocytoma cells (Fig. 3 B , a and b ). Moreover, ChIP from hippocampal neurons revealed that CREB binds the same genomic region in a fashion inducible by NGF (Fig. 3 Bc ). Finally, in primary neurons isolated from CREB loxP/loxP mice, deletion of CREB by adenoviral delivery of the Cre recombinase (Fig. 3 C , a and b ) led to a reduction of Sirt-1 immunoreactivity (Fig. 3 Cc ), and inhibited Sirt-1 mRNA induction by NGF and Fsk (Fig. 3 Cb ). Thus, we conclude that CREB directly regulates Sirt-1 mRNA and protein expression in neurons. Known transcriptional regulators of Sirt-1, such as p53 and Foxo3a (31), also interact with, and are either positively or negatively modulated by the deacetylase (32, 33); similarly, a myc-tagged form of human CREB1 and Sirt-1 could be easily coimmunopre- cipated in naive PC12 cells (Fig. 4 A ), indicating physical association between the two proteins; the association was rapidly induced by the PKA agonist Fsk, in parallel with phosphorylation of CREB on Serine 133. Short-time (30 min) stimulation with Fsk did not affect the total amount of cellular Sirt-1, but rather increased the stoichiometry of the binding to CREB, suggesting either an increased af fi nity between the two proteins, or the enhanced interaction with intermediate molecular partners co-recruited at target gene promoters. In keeping with the latter possibility, a mutant CREB that retains the phosphorylation site but is unable to bind DNA because of the deletion of the C-terminal transactivation domain ( Δ LZ CREB), displayed marginal physical interaction with Sirt-1, nor was the binding induced by Fsk (Fig. 4 A ). To con fi rm that SIRT1 and CREB colocalize on CREB- responsive chromatin regions in cortical neurons, cross-linked chromatin was immunoprecipitated with an anti-Sirt-1 antiserum and Sirt-1 binding to the regions of nNOS and PGC-1 α promoters surrounding the CRE elements determined by semiquantitative PCR. Sirt-1 was found to interact with both genes, in a fashion inducible by NGF and with a binding kinetic similar to that of CREB (Fig. 4 Ba ). Sirt-1 also bound the same Sirt-1 chromatin region surrounding the +1,998/+2,012 half-CRE sites that copre- cipitates with CREB, suggesting that Sirt-1 may self-regulate its CRE-dependent transactivation (Fig. 4 Bb ). Of note, Sirt-1 interaction with nNOS and Sirt-1 chromatin was indeed mediated by CREB, as revealed by its drastic reduction in CREB loxP/loxP neurons following the recombinase-mediated deletion of the factor (Fig. 4 Bb ) and by the failure of forced Sirt-1 overexpression to restore Sirt-1 chromatin binding in CREB-de fi cient cells (Fig. S3). To determine whether Sirt-1 modulates CREB transcriptional activity, cortical neurons were stimulated with NGF in the presence of sirtuin inhibitor Nicotinamide (34); induction of nNOS and PGC-1 α mRNA by NGF was blunted upon inhibition of Sirt-1 deacetylase activity (Fig. 4 C ). A similar result was obtained in naive PC12 cells by siRNA-mediated knock-down of Sirt-1 (Fig. S4 A ). In addition, Sirt-1 – de fi cient PC12 displayed impaired differentiation by NGF (as assessed by quanti fi cation of neurite outgrowth), a response largely dependent on CREB and nNOS in this cell model (35, 36) (Fig. S4 B ). In a complementary set of experiments, lentivirus-mediated overexpression of Sirt-1 increased the expression of nNOS and PGC-1 α mRNA in hippocampal neurons treated with NGF. However, Sirt-1 did not fully restore the expression of these genes in CREB-de fi cient cells, nor NGF protection from hydrogen peroxide-dependent cell death, which is largely lost in these cells (Fig. S5), was recovered upon transduction of the Sirt-1 cDNA. Collectively, the above data suggest that Sirt-1 and CREB are, at least to some extent, re- ciprocally dependent, and act in concert in the context of neurotrophin signaling. Unlike previously reported fi ndings (16), manipulations aimed at modulating Sirt-1 expression and activity in the above contexts did not affect the level of immunoreactive CREB1 (Fig. S4 Aa ). KO Mice. Finally, to verify whether Sirt-1 actually regulates CREB- dependent transcription in vivo and this is relevant for response to CR, we analyzed the expression of CREB targets nNOS and PGC- 1 α mRNAs in whole brains of Sirt-1 KO mice fed AL or CR for 25 – 28 wk (37). This analysis revealed that both genes are markedly hypoexpressed in Sirt-de fi cient mice compared with littermate controls under both dietary regimens (Fig. 4 D ), and their induction by CR, that was more pronounced for PGC-1 α , was also attenuated (Fig. 4 D , a and b ). The amount of immunoreactive CREB was comparable in brain homogenates of the two strains (Fig. 4 Dc ), nor CREB acetylation on lysine residues was affected by either Sirt-1 deletion or CR (Fig. S6), indicating that, at least in these experimental settings, Sirt-1 regulates CREB transcriptional activity independent of the acetylation and expression level of the factor. Taken together, the above fi ndings strongly suggest that Sirt-1 modulates the expression of CREB-dependent genes in mouse brain, and by extension, identify in the decrease of CREB- dependent transcription a molecular signature for a defective brain response to CR, shared by CREB and Sirt-1 mutant mice (7). The CREB transcription factor has been widely investigated as a metabolic sensor and regulator of glucose homeostasis in liver and fat tissue (15), and as a master switch of calcium and neuro- trophin-triggered transcriptional programs regulating neuronal differentiation, survival, and plasticity in the brain and peripheral nervous system (10, 11, 18). Evidence also exist for CREB roles in the control of appetite and food intake in the hypothalamus (38), but whether neuronal plasticity and high-order cognitive functions may be in fl uenced by nutrient cues and energy metabolism through CREB, an issue relevant to major diseases like AD and type 2 diabetes (3, 4), remains to be established. Our demonstra- tion of impaired electrophysiological, cognitive, and emotional response to CR in brain CREB KO mice, clearly suggests that this may indeed be the case. Interestingly, these differences emerge in the context of an overall comparable feeding behavior and metabolic response to CR between control and BCKO mice (Fig. 1 Ab ), indicating that potential effects of hypothalamic CREB signaling on energy balance and appetite regulation unlikely account for the observed phenotypes (38). Data reported in Fig. 2, showing CREB phosphorylation and transcriptional activation by CR, indicate that CREB is metaboli- cally regulated in the cortex and hippocampus, although the mechanism of such regulation needs to be further investigated. Because plasma from CR rodents has been previously shown to induce Sirt-1 expression in several organs/tissues, a humoral/hormonal mechanism for CREB activation could be envisaged (39); as an alternative, NO (40) or oxygen species (41) may mediate, cell-autonomously, this effect. Further research in this direction is warranted. Our data identify in the complex interplay with the nutrient- sensitive histone deacetylase Sirt-1 a molecular connection between CREB and neuronal response to CR. Previous work from others has compellingly involved neuronal Sirt-1 in hormonal and metabolic adaptation to dietary restriction in mice (7). Results presented here clearly suggest that the two molecules are part of the same CR-sensitive signaling cascade. However, other genes potentially relevant to mitochondrial biogenesis and neuronal response to calorie restriction, namely nNOS and PGC-1 α (40), and some PGC-1 α targets — including CPT1 , CoxIV , and UCP-2 — are also critically down-regulated, together with Sirt-1, in the cortex and hippocampus of BCKO mice (Fig. 2 B and Fig. S2 b ). This fi nding, and evidence from Fig. S3 that overexpression of Sirt-1 does not rescue NGF ...
Context 9
... in cultured primary cortical and hippocampal neurons (Fig. 3 A ). Bioinformatic analysis of the mouse Sirt-1 locus (NC_000076) revealed the presence of several putative cAMP Responsive Elements (CRE) both upstream and downstream of the transcription start site (TSS) (30), and an ∼ 300- bp segment encompassing two of those elements (TGACG at +1,998 and CGTCA at +2,012 from the annotated TSS) drove transcriptional response to Fsk and PKA in a standard luciferase reporter assay performed in PC12 pheochromocytoma cells (Fig. 3 B , a and b ). Moreover, ChIP from hippocampal neurons revealed that CREB binds the same genomic region in a fashion inducible by NGF (Fig. 3 Bc ). Finally, in primary neurons isolated from CREB loxP/loxP mice, deletion of CREB by adenoviral delivery of the Cre recombinase (Fig. 3 C , a and b ) led to a reduction of Sirt-1 immunoreactivity (Fig. 3 Cc ), and inhibited Sirt-1 mRNA induction by NGF and Fsk (Fig. 3 Cb ). Thus, we conclude that CREB directly regulates Sirt-1 mRNA and protein expression in neurons. Known transcriptional regulators of Sirt-1, such as p53 and Foxo3a (31), also interact with, and are either positively or negatively modulated by the deacetylase (32, 33); similarly, a myc-tagged form of human CREB1 and Sirt-1 could be easily coimmunopre- cipated in naive PC12 cells (Fig. 4 A ), indicating physical association between the two proteins; the association was rapidly induced by the PKA agonist Fsk, in parallel with phosphorylation of CREB on Serine 133. Short-time (30 min) stimulation with Fsk did not affect the total amount of cellular Sirt-1, but rather increased the stoichiometry of the binding to CREB, suggesting either an increased af fi nity between the two proteins, or the enhanced interaction with intermediate molecular partners co-recruited at target gene promoters. In keeping with the latter possibility, a mutant CREB that retains the phosphorylation site but is unable to bind DNA because of the deletion of the C-terminal transactivation domain ( Δ LZ CREB), displayed marginal physical interaction with Sirt-1, nor was the binding induced by Fsk (Fig. 4 A ). To con fi rm that SIRT1 and CREB colocalize on CREB- responsive chromatin regions in cortical neurons, cross-linked chromatin was immunoprecipitated with an anti-Sirt-1 antiserum and Sirt-1 binding to the regions of nNOS and PGC-1 α promoters surrounding the CRE elements determined by semiquantitative PCR. Sirt-1 was found to interact with both genes, in a fashion inducible by NGF and with a binding kinetic similar to that of CREB (Fig. 4 Ba ). Sirt-1 also bound the same Sirt-1 chromatin region surrounding the +1,998/+2,012 half-CRE sites that copre- cipitates with CREB, suggesting that Sirt-1 may self-regulate its CRE-dependent transactivation (Fig. 4 Bb ). Of note, Sirt-1 interaction with nNOS and Sirt-1 chromatin was indeed mediated by CREB, as revealed by its drastic reduction in CREB loxP/loxP neurons following the recombinase-mediated deletion of the factor (Fig. 4 Bb ) and by the failure of forced Sirt-1 overexpression to restore Sirt-1 chromatin binding in CREB-de fi cient cells (Fig. S3). To determine whether Sirt-1 modulates CREB transcriptional activity, cortical neurons were stimulated with NGF in the presence of sirtuin inhibitor Nicotinamide (34); induction of nNOS and PGC-1 α mRNA by NGF was blunted upon inhibition of Sirt-1 deacetylase activity (Fig. 4 C ). A similar result was obtained in naive PC12 cells by siRNA-mediated knock-down of Sirt-1 (Fig. S4 A ). In addition, Sirt-1 – de fi cient PC12 displayed impaired differentiation by NGF (as assessed by quanti fi cation of neurite outgrowth), a response largely dependent on CREB and nNOS in this cell model (35, 36) (Fig. S4 B ). In a complementary set of experiments, lentivirus-mediated overexpression of Sirt-1 increased the expression of nNOS and PGC-1 α mRNA in hippocampal neurons treated with NGF. However, Sirt-1 did not fully restore the expression of these genes in CREB-de fi cient cells, nor NGF protection from hydrogen peroxide-dependent cell death, which is largely lost in these cells (Fig. S5), was recovered upon transduction of the Sirt-1 cDNA. Collectively, the above data suggest that Sirt-1 and CREB are, at least to some extent, re- ciprocally dependent, and act in concert in the context of neurotrophin signaling. Unlike previously reported fi ndings (16), manipulations aimed at modulating Sirt-1 expression and activity in the above contexts did not affect the level of immunoreactive CREB1 (Fig. S4 Aa ). KO Mice. Finally, to verify whether Sirt-1 actually regulates CREB- dependent transcription in vivo and this is relevant for response to CR, we analyzed the expression of CREB targets nNOS and PGC- 1 α mRNAs in whole brains of Sirt-1 KO mice fed AL or CR for 25 – 28 wk (37). This analysis revealed that both genes are markedly hypoexpressed in Sirt-de fi cient mice compared with littermate controls under both dietary regimens (Fig. 4 D ), and their induction by CR, that was more pronounced for PGC-1 α , was also attenuated (Fig. 4 D , a and b ). The amount of immunoreactive CREB was comparable in brain homogenates of the two strains (Fig. 4 Dc ), nor CREB acetylation on lysine residues was affected by either Sirt-1 deletion or CR (Fig. S6), indicating that, at least in these experimental settings, Sirt-1 regulates CREB transcriptional activity independent of the acetylation and expression level of the factor. Taken together, the above fi ndings strongly suggest that Sirt-1 modulates the expression of CREB-dependent genes in mouse brain, and by extension, identify in the decrease of CREB- dependent transcription a molecular signature for a defective brain response to CR, shared by CREB and Sirt-1 mutant mice (7). The CREB transcription factor has been widely investigated as a metabolic sensor and regulator of glucose homeostasis in liver and fat tissue (15), and as a master switch of calcium and neuro- trophin-triggered transcriptional programs regulating neuronal differentiation, survival, and plasticity in the brain and peripheral nervous system (10, 11, 18). Evidence also exist for CREB roles in the control of appetite and food intake in the hypothalamus (38), but whether neuronal plasticity and high-order cognitive functions may be in fl uenced by nutrient cues and energy metabolism through CREB, an issue relevant to major diseases like AD and type 2 diabetes (3, 4), remains to be established. Our demonstra- tion of impaired electrophysiological, cognitive, and emotional response to CR in brain CREB KO mice, clearly suggests that this may indeed be the case. Interestingly, these differences emerge in the context of an overall comparable feeding behavior and metabolic response to CR between control and BCKO mice (Fig. 1 Ab ), indicating that potential effects of hypothalamic CREB signaling on energy balance and appetite regulation unlikely account for the observed phenotypes (38). Data reported in Fig. 2, showing CREB phosphorylation and transcriptional activation by CR, indicate that CREB is metaboli- cally regulated in the cortex and hippocampus, although the mechanism of such regulation needs to be further investigated. Because plasma from CR rodents has been previously shown to induce Sirt-1 expression in several organs/tissues, a humoral/hormonal mechanism for CREB activation could be envisaged (39); as an alternative, NO (40) or oxygen species (41) may mediate, cell-autonomously, this effect. Further research in this direction is warranted. Our data identify in the complex interplay with the nutrient- sensitive histone deacetylase Sirt-1 a molecular connection between CREB and neuronal response to CR. Previous work from others has compellingly involved neuronal Sirt-1 in hormonal and metabolic adaptation to dietary restriction in mice (7). Results presented here clearly suggest that the two molecules are part of the same CR-sensitive signaling cascade. However, other genes potentially relevant to mitochondrial biogenesis and neuronal response to calorie restriction, namely nNOS and PGC-1 α (40), and some PGC-1 α targets — including CPT1 , CoxIV , and UCP-2 — are also critically down-regulated, together with Sirt-1, in the cortex and hippocampus of BCKO mice (Fig. 2 B and Fig. S2 b ). This fi nding, and evidence from Fig. S3 that overexpression of Sirt-1 does not rescue NGF signaling in CREB-de fi cient hippocampal neurons in vitro, suggests that up-regulation of Sirt-1 is not the only mechanism whereby CREB participates in neuronal response to nutrients. Instead, cooperation with CREB is likely critical for the action of neuronal Sirt-1 in CR. This view is supported by the unexpected fi nding that CREB-dependent genes involved in neuronal plasticity, survival, and stress resistance (42, 43), and induced by calorie restriction (Fig. 2), are markedly down-regulated in Sirt-1 – de fi cient cultured neurons (PC12) and in the brain of Sirt-1 KO mice. Because the latter strain is, like BCKO mice, impaired in brain response to reduced food intake (7), the above evidence further support the notion that CREB-dependent transcription has a pivotal role in the neuronal effects of calorie restriction, and identify in the CREB – Sirt-1 axis a major component of the nutrient sensitive molecular network that connects caloric intake and energy metabolism to brain health. Biochemical details of how Sirt-1 affects CREB activity remains to be clari fi ed. Unlike other transcription factors, CREB does not appear to be deacetylated by Sirt-1 (Fig. S6); moreover, we could not detect consistent effects of Sirt-1 on the expression of CREB (Fig. 4 Dc , and Figs. S4 Aa and S6), unlike that described by Gao et al. (16). Those authors, however, made their important observations on Sirt-1 Δ ex4 mice that, unlike Sirt-1KO mice used in our experiments, do express an inactive form of Sirt-1 potentially acting in a dominant negative fashion against ...
Context 10
... fi c CREB deletion. The increased LTP induced by CR in control mice was independent on changes in basal synaptic transmission, because input/output curves obtained plotting fi eld excitatory postsynaptic potential (fEPSP) amplitudes vs. stimulus intensity in hippocampal brain slices of CR and AL animals were superimposable. Because CREB appeared to participate in brain response to CR, we assessed whether diet affected the total amount/phosphoryla- tion of CREB protein, and the expression level of a number of mRNAs known to be regulated by CREB (23). CREB1 protein expression in control mice was not changed by the dietary regimen; instead, an increase in CREB phosphorylation on Serine 133 in the hippocampi of the CR group was revealed by phospho-speci fi c immunoblotting (Fig. 2 A ), suggesting that CR activates CREB in this brain area. Accordingly, mRNAs of “ canonical ” CREB targets peroxisome proliferator-activated receptor- γ coactivator-1 α ( PGC- 1 α ), nNOS , and phosphoenolpyruvate carboxykinase ( PEPCK ) were induced by CR in the cortex and hippocampus of CREB- pro fi cient mice (Fig. 2 B ). Of note, both PGC-1 α and NO, the product of NOS enzymes, are known to promote mitochondrial biogenesis and to participate in organismal response to CR (26). The three mRNAs were overall down-regulated in BCKO mice fed AL compared with controls, and their induction by CR, observed in control mice, was nearly abolished (Fig. 2 B ). Interestingly, the expression of other putative CREB targets like bcl-2 , NGF , and c-fos were unaffected by CR and CREB deletion (Fig. S2). Thus, collectively, these fi ndings demonstrate that nutrient availability selectively regulates CREB-dependent gene expression in the forebrain. We next asked how CR may affect CREB activity. Because Sirt- 1 is a metabolic sensor involved in several biological consequences of nutrient deprivation (27, 6), and animals lacking Sirt-1 in the brain show defective behavioral and hormonal responses to CR (7), we investigated the sirtuin as a potential CREB interactor. Sirt-1 mRNA was drastically reduced both in the cortex and hippocampus of BCKO mice, irrespective of the dietary regimen. Moreover, the moderate induction by CR that we observed in control brains was completely lost in the corresponding CREB- de fi cient tissues (Fig. 2 C ). Finally, acetylation of Histones H3 (AcK9) and H4 (AcK16), an inverse correlate of Sirt-1 activity (28), was abnormally high in BCKO hippocampal homogenates (Fig. 2 D ), con fi rming an overall reduction of Sirt-1 activity in this area of the brain. Transcriptional Regulation of Sirt-1 by CREB in Neurons. These observations suggested that Sirt-1 may represent a direct transcriptional target of CREB. Accordingly, CREB activators NGF and Forskolin (Fsk) (29) raised the level of immunoreactive Sirt-1 and of the corresponding mRNA in cultured primary cortical and hippocampal neurons (Fig. 3 A ). Bioinformatic analysis of the mouse Sirt-1 locus (NC_000076) revealed the presence of several putative cAMP Responsive Elements (CRE) both upstream and downstream of the transcription start site (TSS) (30), and an ∼ 300- bp segment encompassing two of those elements (TGACG at +1,998 and CGTCA at +2,012 from the annotated TSS) drove transcriptional response to Fsk and PKA in a standard luciferase reporter assay performed in PC12 pheochromocytoma cells (Fig. 3 B , a and b ). Moreover, ChIP from hippocampal neurons revealed that CREB binds the same genomic region in a fashion inducible by NGF (Fig. 3 Bc ). Finally, in primary neurons isolated from CREB loxP/loxP mice, deletion of CREB by adenoviral delivery of the Cre recombinase (Fig. 3 C , a and b ) led to a reduction of Sirt-1 immunoreactivity (Fig. 3 Cc ), and inhibited Sirt-1 mRNA induction by NGF and Fsk (Fig. 3 Cb ). Thus, we conclude that CREB directly regulates Sirt-1 mRNA and protein expression in neurons. Known transcriptional regulators of Sirt-1, such as p53 and Foxo3a (31), also interact with, and are either positively or negatively modulated by the deacetylase (32, 33); similarly, a myc-tagged form of human CREB1 and Sirt-1 could be easily coimmunopre- cipated in naive PC12 cells (Fig. 4 A ), indicating physical association between the two proteins; the association was rapidly induced by the PKA agonist Fsk, in parallel with phosphorylation of CREB on Serine 133. Short-time (30 min) stimulation with Fsk did not affect the total amount of cellular Sirt-1, but rather increased the stoichiometry of the binding to CREB, suggesting either an increased af fi nity between the two proteins, or the enhanced interaction with intermediate molecular partners co-recruited at target gene promoters. In keeping with the latter possibility, a mutant CREB that retains the phosphorylation site but is unable to bind DNA because of the deletion of the C-terminal transactivation domain ( Δ LZ CREB), displayed marginal physical interaction with Sirt-1, nor was the binding induced by Fsk (Fig. 4 A ). To con fi rm that SIRT1 and CREB colocalize on CREB- responsive chromatin regions in cortical neurons, cross-linked chromatin was immunoprecipitated with an anti-Sirt-1 antiserum and Sirt-1 binding to the regions of nNOS and PGC-1 α promoters surrounding the CRE elements determined by semiquantitative PCR. Sirt-1 was found to interact with both genes, in a fashion inducible by NGF and with a binding kinetic similar to that of CREB (Fig. 4 Ba ). Sirt-1 also bound the same Sirt-1 chromatin region surrounding the +1,998/+2,012 half-CRE sites that copre- cipitates with CREB, suggesting that Sirt-1 may self-regulate its CRE-dependent transactivation (Fig. 4 Bb ). Of note, Sirt-1 interaction with nNOS and Sirt-1 chromatin was indeed mediated by CREB, as revealed by its drastic reduction in CREB loxP/loxP neurons following the recombinase-mediated deletion of the factor (Fig. 4 Bb ) and by the failure of forced Sirt-1 overexpression to restore Sirt-1 chromatin binding in CREB-de fi cient cells (Fig. S3). To determine whether Sirt-1 modulates CREB transcriptional activity, cortical neurons were stimulated with NGF in the presence of sirtuin inhibitor Nicotinamide (34); induction of nNOS and PGC-1 α mRNA by NGF was blunted upon inhibition of Sirt-1 deacetylase activity (Fig. 4 C ). A similar result was obtained in naive PC12 cells by siRNA-mediated knock-down of Sirt-1 (Fig. S4 A ). In addition, Sirt-1 – de fi cient PC12 displayed impaired differentiation by NGF (as assessed by quanti fi cation of neurite outgrowth), a response largely dependent on CREB and nNOS in this cell model (35, 36) (Fig. S4 B ). In a complementary set of experiments, lentivirus-mediated overexpression of Sirt-1 increased the expression of nNOS and PGC-1 α mRNA in hippocampal neurons treated with NGF. However, Sirt-1 did not fully restore the expression of these genes in CREB-de fi cient cells, nor NGF protection from hydrogen peroxide-dependent cell death, which is largely lost in these cells (Fig. S5), was recovered upon transduction of the Sirt-1 cDNA. Collectively, the above data suggest that Sirt-1 and CREB are, at least to some extent, re- ciprocally dependent, and act in concert in the context of neurotrophin signaling. Unlike previously reported fi ndings (16), manipulations aimed at modulating Sirt-1 expression and activity in the above contexts did not affect the level of immunoreactive CREB1 (Fig. S4 Aa ). KO Mice. Finally, to verify whether Sirt-1 actually regulates CREB- dependent transcription in vivo and this is relevant for response to CR, we analyzed the expression of CREB targets nNOS and PGC- 1 α mRNAs in whole brains of Sirt-1 KO mice fed AL or CR for 25 – 28 wk (37). This analysis revealed that both genes are markedly hypoexpressed in Sirt-de fi cient mice compared with littermate controls under both dietary regimens (Fig. 4 D ), and their induction by CR, that was more pronounced for PGC-1 α , was also attenuated (Fig. 4 D , a and b ). The amount of immunoreactive CREB was comparable in brain homogenates of the two strains (Fig. 4 Dc ), nor CREB acetylation on lysine residues was affected by either Sirt-1 deletion or CR (Fig. S6), indicating that, at least in these experimental settings, Sirt-1 regulates CREB transcriptional activity independent of the acetylation and expression level of the factor. Taken together, the above fi ndings strongly suggest that Sirt-1 modulates the expression of CREB-dependent genes in mouse brain, and by extension, identify in the decrease of CREB- dependent transcription a molecular signature for a defective brain response to CR, shared by CREB and Sirt-1 mutant mice (7). The CREB transcription factor has been widely investigated as a metabolic sensor and regulator of glucose homeostasis in liver and fat tissue (15), and as a master switch of calcium and neuro- trophin-triggered transcriptional programs regulating neuronal differentiation, survival, and plasticity in the brain and peripheral nervous system (10, 11, 18). Evidence also exist for CREB roles in the control of appetite and food intake in the hypothalamus (38), but whether neuronal plasticity and high-order cognitive functions may be in fl uenced by nutrient cues and energy metabolism through CREB, an issue relevant to major diseases like AD and type 2 diabetes (3, 4), remains to be established. Our demonstra- tion of impaired electrophysiological, cognitive, and emotional response to CR in brain CREB KO mice, clearly suggests that this may indeed be the case. Interestingly, these differences emerge in the context of an overall comparable feeding behavior and metabolic response to CR between control and BCKO mice (Fig. 1 Ab ), indicating that potential effects of hypothalamic CREB signaling on energy balance and appetite regulation unlikely account for the observed phenotypes (38). Data reported in Fig. 2, showing CREB phosphorylation and transcriptional activation by CR, indicate that CREB is metaboli- cally ...

Similar publications

Article
Full-text available
Maternal overnutrition and obesity during pregnancy can have long-term effects on offspring physiology and behaviour. These developmental programming effects may be mediated by fetal exposure to glucocorticoids, which is regulated in part by placental 11β-hydroxysteroid dehydrogenase (11β-HSD) type 1 and 2. We tested whether a maternal high-fat, hi...

Citations

... In addition, it has been identified that there are putative CREBbinding sites in the proximal promoter region of SIRT1, and it has been proven that SIRT1 promoter activity is induced by CREB; SIRT1 mRNA expression is also increased by the overexpression of CREB in HepG2 cells (Noriega et al., 2011). It has also been demonstrated that CREB directly mediates the transcription of SIRT1 by binding to SIRT1 chromatin and CREB deficiency markedly reduces the expression of SIRT1 (Fusco et al., 2012b). Moreover, SIRT1 and CREB may form a complex on the cAMPresponsive element, and SIRT1 may promote CREB transcriptional activity and the expression of target genes, creating a positive feedback loop (Fusco et al., 2012a;Fusco et al., 2012b). ...
... It has also been demonstrated that CREB directly mediates the transcription of SIRT1 by binding to SIRT1 chromatin and CREB deficiency markedly reduces the expression of SIRT1 (Fusco et al., 2012b). Moreover, SIRT1 and CREB may form a complex on the cAMPresponsive element, and SIRT1 may promote CREB transcriptional activity and the expression of target genes, creating a positive feedback loop (Fusco et al., 2012a;Fusco et al., 2012b). In this study, we have displayed that imperatorin enhances the phosphorylation of LKB1, AMPK, and CREB and subsequently elevates the activity of SIRT1. ...
Article
Full-text available
Background: Pulmonary fibrosis features in damaged pulmonary structure or over-produced extracellular matrix and impaired lung function, leading to respiratory failure and eventually death. Fibrotic lungs are characterized by the secretion of pro-fibrotic factors, transformation of fibroblasts to myofibroblasts, and accumulation of matrix proteins. Hypothesis/purpose: Imperatorin shows anti-inflammatory effects on alveolar macrophages against acute lung injury. We attempt to evaluate the properties of imperatorin on the basis of fibroblasts. Methods: In in vitro, zymosan was introduced to provoke pro-fibrotic responses in NIH/3T3 or MRC-5 pulmonary fibroblasts. Imperatorin was given for examining its effects against fibrosis. The mice were stimulated by bleomycin, and imperatorin was administered to evaluate the prophylactic potential in vivo. Results: The upregulated expression of connective tissue growth factor (CTGF), α-smooth muscle actin (α-SMA), and collagen protein due to zymosan introduction was decreased by imperatorin in fibroblasts. Zymosan induced the activity of transglutaminase 2 (TGase2) and lysyl oxidase (LOX), which was also inhibited by the administration of imperatorin. Imperatorin alone enhanced sirtuin 1 (SIRT1) activity and growth differentiation factor 15 (GDF15) secretion in fibroblasts via LKB1/AMPK/CREB pathways. In addition, GDF15 exerted a beneficial effect by reducing the protein expression of CTGF, α-SMA, and collagen and the activities of TGase and LOX. Moreover, orally administered imperatorin showed prophylactic effects on bleomycin-induced pulmonary fibrosis in mice. Conclusion: Imperatorin reduces fibrotic marker expression in fibroblasts and also increases GDF15 secretion via the LKB1/AMPK/CREB pathway, attenuating pro-fibrotic responses in vitro. Imperatorin also alleviates pulmonary fibrosis induced by bleomycin in vivo.
... Field recordings were performed on coronal slices containing the hippocampus as previously described (80,81). Briefly, animals were anesthetized by isoflurane inhalation (Esteve) and decapitated. ...
Article
Full-text available
Synaptic plasticity plays a crucial role in memory formation by regulating the communication between neurons. Although actin polymerization has been linked to synaptic plasticity and dendritic spine stability, the causal link between actin polymerization and memory encoding has not been identified yet. It is not clear whether actin polymerization and structural changes in dendritic spines are a driver or a consequence of learning and memory. Using an extrinsically disordered form of the protein kinase LIMK1, which rapidly and precisely acts on ADF/cofilin, a direct modifier of actin, we induced long-term enlargement of dendritic spines and enhancement of synaptic transmission in the hippocampus on command. The activation of extrinsically disordered LIMK1 in vivo improved memory encoding and slowed cognitive decline in aged mice exhibiting reduced cofilin phosphorylation. The engineered memory by an extrinsically disordered LIMK1 supports a direct causal link between actin-mediated synaptic transmission and memory.
... PKA activated the downstream SIRT1 and SIRT5, leading to improved mitochondrial function and metabolic homeostasis (Dehghan et al., 2019). Furthermore, CREB promotes SIRT1 transcription via binding to its promoter in neuronal cells (Fusco et al., 2012). CREB recruits SIRT-1 to DNA, which leads to the increased expression of CREB target genes, such as Peroxisome proliferator-activated receptor-γ coactivator 1-α (PGC1α) and neuronal NO synthase (Fusco et al., 2012). ...
... Furthermore, CREB promotes SIRT1 transcription via binding to its promoter in neuronal cells (Fusco et al., 2012). CREB recruits SIRT-1 to DNA, which leads to the increased expression of CREB target genes, such as Peroxisome proliferator-activated receptor-γ coactivator 1-α (PGC1α) and neuronal NO synthase (Fusco et al., 2012). The expression of SIRT1 was upregulated in Pkd1 mutant mouse renal epithelial cells and kidney tissues, and human ADPKD cells (Zhou et al., 2013;Warner et al., 2016). ...
Article
Full-text available
Autosomal dominant polycystic kidney disease (ADPKD), with an estimated genetic prevalence between 1:400 and 1:1,000 individuals, is the third most common cause of end stage kidney disease after diabetes mellitus and hypertension. Over the last 3 decades there has been great progress in understanding its pathogenesis. This allows the stratification of therapeutic targets into four levels, gene mutation and polycystin disruption, proximal mechanisms directly caused by disruption of polycystin function, downstream regulatory and signaling pathways, and non-specific pathophysiologic processes shared by many other diseases. Dysfunction of the polycystins, encoded by the PKD genes, is closely associated with disruption of calcium and upregulation of cyclic AMP and protein kinase A (PKA) signaling, affecting most downstream regulatory, signaling, and pathophysiologic pathways altered in this disease. Interventions acting on G protein coupled receptors to inhibit of 3′,5′-cyclic adenosine monophosphate (cAMP) production have been effective in preclinical trials and have led to the first approved treatment for ADPKD. However, completely blocking cAMP mediated PKA activation is not feasible and PKA activation independently from cAMP can also occur in ADPKD. Therefore, targeting the cAMP/PKA/CREB pathway beyond cAMP production makes sense. Redundancy of mechanisms, numerous positive and negative feedback loops, and possibly counteracting effects may limit the effectiveness of targeting downstream pathways. Nevertheless, interventions targeting important regulatory, signaling and pathophysiologic pathways downstream from cAMP/PKA activation may provide additive or synergistic value and build on a strategy that has already had success. The purpose of this manuscript is to review the role of cAMP and PKA signaling and their multiple downstream pathways as potential targets for emergent therapies for ADPKD.
... Because protein deacetylase sirtuin 1 (Sirt1) acts as a transcriptional activator of SERCA2a gene expression in high-glucose conditions (20) and Sirt1 is a direct transcriptional target of cAMP responsive element binding protein (CREB), which is activated by cAMP-PKA (21), we assessed whether PDE4 inhibitor restores SERCA2a expression through activation of cAMP-PKA-CREB-Sirt1 signal pathway. In line with previous research (21), forskolin, a PKA activator, could facilitate CREB phosphorylation on serine group, five random fields per heart. Boxed areas in the top row are enlarged in the middle row of panels. ...
... Moreover, resveratrol activates Sirt1 in vivo due to its effect on cAMP signaling by directly inhibiting PDE4 (4). It also has been reported that cAMP increased the protein level of sirtuins and Sirt1 is a direct transcriptional target of CREB, which is activated by cAMP-PKA in the nucleus (21,31). Given that PDE4D5 appears to form puncta in nucleus and plays a critical role in the functional nuclear cAMP-PKA signaling (32,33) and the expression of cardiac PDE4D5 was increased in human failing hearts with diabetes and HFD mice, we demonstrated that PDE4D5 overexpression reduced CREB phosphorylation, leading to decreased expression of Sirt1 and SERCA2a. ...
Article
Patients with type 2 diabetes have a substantial risk of developing cardiovascular disease. Phosphodiesterase 4 (PDE4) dysregulation is of pathophysiological importance in metabolic disorders. To determine the role of PDE4 in diabetic cardiac dysfunction, mice fed with high-fat diet (HFD) were treated by pharmacological inhibition of PDE4 or cardiac specific knocking down of PDE4D. Mice on HFD developed diabetes and cardiac dysfunction with increased cardiac PDE4D5 expression. PDE4 inhibitor roflumilast can reverse hyperglycemia and cardiac dysfunction, accompanied by the decrease of PDE4D expression and increase of muscle-specific microRNA miR-1 level in hearts. Either cardiac specific PDE4D knockdown or miR-1 overexpression significantly reversed cardiac dysfunction in HFD-mice, despite persistence of hyperglycemia. Gain- and loss-of-function studies of PDE4D in cardiomyocytes implicated that inhibition of insulin-induced PDE4D protected cardiac hypertrophy by preserving miR-1 expression in cardiomyocytes through promoting cAMP-CREB-Sirt1 signaling-induced SERCA2a expression. We further revealed that insulin also induced PDE4D expression in cardiac fibroblasts, which causes cardiac fibrosis through TGF-β1 signaling-mediated miR-1 reduction. Importantly, the expression of PDE4D5 was increased in human failing hearts with diabetes. These studies elucidate a novel mechanism by which hyperinsulinemia-induced cardiac PDE4D expression contributes to diabetic cardiac remodeling through reducing the expression of miR-1 and upregulation of miR-1 target hypertrophy and fibrosis-associated genes. Our study suggests a therapeutic potential of PDE4 inhibitor roflumilast in preventing or treating cardiac dysfunction in diabetes in addition to lowering glucose.
... Thus, CR-induced levels of ghrelin contribute to enhanced cognitive function. CR also confers neuroprotective effects mediated through nutrient-sensing pathways (such as the SIRT1/mTOR signaling pathways (Ma et al., 2015;Quintas et al., 2012;Yang et al., 2014), CREB/SIRT1 signaling pathways (Fusco et al., 2012), FGF21-induced AMPK/mTOR pathway (Ruhlmann et al., 2016), AMPK signaling pathway (Ma et al., 2017), and insulin-PI3K/Akt signaling pathway ). ...
Article
Alzheimer's disease (AD) is one of the fastest growing cognitive decline-related neurological diseases. To date, effective curative strategies have remained elusive. A growing body of evidence indicates that dietary patterns have significant effects on cognitive function and the risk of developing AD. Previous studies on the association between diet and AD risk have mainly focused on individual food components and specific nutrients, and the mechanisms responsible for the beneficial effects of dietary patterns on AD are not well understood. This article provides a comprehensive overview of the effects of dietary patterns, including the Mediterranean diet (MedDiet), dietary approaches to stop hypertension (DASH) diet, Mediterranean-DASH diet intervention for neurological delay (MIND), ketogenic diet, caloric restriction, intermittent fasting, methionine restriction, and low-protein and high-carbohydrate diet, on cognitive impairment and summarizes the underlying mechanisms by which dietary patterns attenuate cognitive impairment, especially highlighting the modulation of dietary patterns on cognitive impairment through gut microbiota. Furthermore, considering the variability in individual metabolic responses to dietary intake, we put forward a framework to develop personalized dietary patterns for people with cognitive disorders or AD based on individual gut microbiome compositions.
... A beneficial effect of elevating SIRT1 levels is also in line with previous reports that SIRT1 expression is decreased in MJD models [11,48], which we also confirmed in our MJD zebrafish and stable HEK293 cell models. This decreased level of SIRT1 expression may be explained by previous findings that polyQ-expanded ataxin-3 and huntingtin proteins have been shown to interact with cAMP response element binding (CREB) protein [42], a transcription factor known to regulate levels of SIRT1 [17,30]. Interestingly, valproate treatment has been shown to activate the CREB pathway [24,40], which may explain the increase in SIRT1 levels produced by valproate treatment here. ...
Article
Full-text available
Machado-Joseph disease (MJD, also known as spinocerebellar ataxia type 3) is a fatal neurodegenerative disease that impairs control and coordination of movement. Here we tested whether treatment with the histone deacetylase inhibitor sodium valproate (valproate) prevented a movement phenotype that develops in larvae of a transgenic zebrafish model of the disease. We found that treatment with valproate improved the swimming of the MJD zebrafish, affected levels of acetylated histones 3 and 4, but also increased expression of polyglutamine expanded human ataxin-3. Proteomic analysis of protein lysates generated from the treated and untreated MJD zebrafish also predicted that valproate treatment had activated the sirtuin longevity signaling pathway and this was confirmed by findings of increased SIRT1 protein levels and sirtuin activity in valproate treated MJD zebrafish and HEK293 cells expressing ataxin-3 84Q, respectively. Treatment with resveratrol (another compound known to activate the sirtuin pathway), also improved swimming in the MJD zebrafish. Co-treatment with valproate alongside EX527, a SIRT1 activity inhibitor, prevented induction of autophagy by valproate and the beneficial effects of valproate on the movement in the MJD zebrafish, supporting that they were both dependent on sirtuin activity. These findings provide the first evidence of sodium valproate inducing activation of the sirtuin pathway. Further, they indicate that drugs that target the sirtuin pathway, including sodium valproate and resveratrol, warrant further investigation for the treatment of MJD and related neurodegenerative diseases. Graphical abstract
... It has been found that a deficiency in the CREB pathway in mature neurons reduces the response of mice toward 30% CR, which is in stark contrast to wildtype mice, in which cognitive functions and electrophysiological parameters were enhanced after CR. However, CREB can directly activate Sirtuin1 in the mouse hippocampus (Fusco et al., 2012). In neurodegenerative diseases, Sirtuin1 can potentiate CREBtarget of rapamycin complex 1 transcriptional activity to improve disease symptoms (Jeong et al., 2011). ...
Article
Full-text available
Aging and neurodegenerative diseases are frequently associated with the disruption of the extracellular microenvironment, which includes mesenchyme and body fluid components. Caloric restriction (CR) has been recognized as a lifestyle intervention that can improve long-term health. In addition to preventing metabolic disorders, CR has been shown to improve brain health owing to its enhancing effect on cognitive functions or retarding effect on the progression of neurodegenerative diseases. This article summarizes current findings regarding the neuroprotective effects of CR, which include the modulation of metabolism, autophagy, oxidative stress, and neuroinflammation. This review may offer future perspectives for brain aging interventions.
... 47,48 Glucose can also stimulate the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway, 49,50 which enhances the transcription of NOS1 gene via the CREB transcription factor. [51][52][53] Additionally, both the PI3K/Akt 54, 55 and cAMP/PKA 40,48,56 pathways play important roles in the regulation of the phosphorylation of NOS1. Therefore, we speculate that in diabetes, the glucose load transported from tubular fluid into the macula densa cells via SGLT1 activates the cAMP /PKA and PI3K/Akt signaling pathways, which increases the expression of NOS1 via CREB and upregulates the activity of NOS1 by phosphorylation at Ser1417 ( Figure 5). ...
Article
Full-text available
An increase of glomerular filtration rate (GFR) is a common observation in early diabetes and is considered a key risk factor for subsequent kidney injury. However, the mechanisms underlying diabetic hyperfiltration have not been fully clarified. Here, we tested the hypothesis that macula densa neuronal nitric oxide synthase (NOS1) is upregulated via sodium glucose cotransporter type 1 (SGLT1) in diabetes, which then inhibits tubuloglomerular feedback (TGF) promoting glomerular hyperfiltration. Therefore, we examined changes in cortical NOS1 expression and phosphorylation, nitric oxide production in the macula densa, TGF response, and GFR during the early stage of insulin-deficient (Akita) diabetes in wild-type and macula densa-specific NOS1 knockout mice. A set of sophisticated techniques including microperfusion of juxtaglomerular apparatus in vitro, micropuncture of kidney tubules in vivo, and clearance kinetics of plasma fluorescent-sinistrin were employed. Complementary studies tested the role of SGLT1 in SGLT1 knockout mice and explored NOS1 expression and phosphorylation in kidney biopsies of cadaveric donors. Diabetic mice had upregulated macula densa NOS1, inhibited TGF and elevated GFR. Macula densa-selective NOS1 knockout attenuated the diabetes-induced TGF inhibition and GFR elevation. Additionally, deletion of SGLT1 prevented the upregulation of macula densa NOS1 and attenuated inhibition of TGF in diabetic mice. Furthermore, the expression and phosphorylation levels of NOS1 were increased in cadaveric kidneys of diabetics and positively correlated with blood glucose as well as estimated GFR in the donors. Thus, our findings demonstrate that the macula densa SGLT1-NOS1-TGF pathway plays a crucial role in the control of GFR in diabetes.
... On the other hand, leptin also causes oxidative stress and most recent studies rather reported an increase in leptin concentration in depressed patients [53,54]. Despite leptin being reduced by CR, both putatively stimulate neuroplasticity via similar pathways [55,56]. In rat models, CR but also leptin injections were shown to directly enhance neurogenesis and cell survival in the hippocampus, a well-established core region of structural and functional impairment in depression [57,58]. ...
Article
Full-text available
Obesity is highly prevalent, causing substantial cardiovascular and mental health morbidity. Women show increased risk for mental health disorders, that is multiplied in obesity and related to cellular and psychological stress that can be targeted by non-pharmacological interventions. A total of 43 women underwent two weeks of caloric restriction, half of which also received 7 h of individualized clinical psychological intervention including psychoeducation, mindfulness, and heart-rate-variability biofeedback. Effects on body mass index (BMI), fatty liver index (FLI), bioimpedance measures, serum parameters, perceived stress (PSS), burn-out susceptibility (burn out diagnostic inventory) and dimensional psychiatric symptom load (brief symptom inventory, BSI) were analyzed with linear mixed effects models. Caloric restriction led to a reduction in BMI, body fat and FLI, decreased serum concentrations of leptin, PSS score, BSI dimensions and global severity index (all p ≤ 0.0001, withstanding Bonferroni–Holm correction). Benefits of add-on biofeedback were observed for BMI reduction (p = 0.041). Caloric restriction was effective in ameliorating both psychological wellbeing and metabolic functions following a BMI reduction. Biofeedback boosted effects on BMI reduction and the combinative therapy may be protective against common progression to mental health and cardiovascular disorders in overweight women while comparing favorably to pharmacological interventions in terms of side-effects and acceptability.
... On the other hand, CR has anti-inflammatory properties such as pro-inflammatory mediator suppressors (i.e. NF-κB, IL-1β, IL-6, TNF, cyclooxygenase 2, and inducible nitric oxide synthase) delaying brain senescence and preventing neurodegeneration [178][179][180]. CR also can suppress oxidative stress and damage. ...
Article
Adequate nutrition is vital for immune homeostasis. However, the incidence of obesity is increasing worldwide due to the adoption of the Western diet and a sedentary lifestyle. Obesity is associated with chronic inflammation which alters the function of adipose tissue, liver, pancreas, and the nervous system. Inflammation is related to cellular senescence, distinguished by irreversible cell cycle arrest. Senescent cells secrete the senescence-associated secretory phenotype (SASP) which contains pro-inflammatory factors. Targeting processes in senescence might have a salutary approach to obesity. The present review highlights the impact of an unhealthy diet on tissues affected by obesity, and the mechanisms that promote the consequent inflammation and senescence.