Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters

Publication year range
1.
Biochem J ; 478(3): 633-646, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33493298

ABSTRACT

Activation of AMP-activated protein kinase (AMPK) is considered a valid strategy for the treatment of type 2 diabetes. However, despite the importance of adipose tissue for whole-body energy homeostasis, the effect of AMPK activation in adipocytes has only been studied to a limited extent and mainly with the AMP-mimetic 5-aminoimidazole-4-carboxamide-1-ß-d-ribofuranoside (AICAR), which has limited specificity. The aim of this study was to evaluate the effect of the allosteric AMPK activators A-769662 and 991 on glucose uptake in adipocytes. For this purpose, primary rat or human adipocytes, and cultured 3T3-L1 adipocytes, were treated with either of the allosteric activators, or AICAR, and basal and insulin-stimulated glucose uptake was assessed. Additionally, the effect of AMPK activators on insulin-stimulated phosphorylation of Akt and Akt substrate of 160 kDa was assessed. Furthermore, primary adipocytes from ADaM site binding drug-resistant AMPKß1 S108A knock-in mice were employed to investigate the specificity of the drugs for the observed effects. Our results show that insulin-stimulated adipocyte glucose uptake was significantly reduced by A-769662 but not 991, yet neither activator had any clear effects on basal or insulin-stimulated Akt/AS160 signaling. The use of AMPKß1 S108A mutant-expressing adipocytes revealed that the observed inhibition of glucose uptake by A-769662 is most likely AMPK-independent, a finding which is supported by the rapid inhibitory effect A-769662 exerts on glucose uptake in 3T3-L1 adipocytes. These data suggest that AMPK activation per se does not inhibit glucose uptake in adipocytes and that the effects of AICAR and A-769662 are AMPK-independent.


Subject(s)
Adenylate Kinase/physiology , Adipocytes/drug effects , Biphenyl Compounds/pharmacology , Glucose/metabolism , Pyrones/pharmacology , Thiophenes/pharmacology , 3T3-L1 Cells , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adipocytes/metabolism , Allosteric Site , Amino Acid Substitution , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Benzimidazoles/pharmacology , Benzoates/pharmacology , Cells, Cultured , Enzyme Activation/drug effects , Female , Gene Knock-In Techniques , Humans , Insulin/pharmacology , Male , Mice , Mice, Inbred C57BL , Mutation, Missense , Phosphorylation , Protein Processing, Post-Translational , Rats , Rats, Sprague-Dawley , Ribonucleotides/pharmacology
2.
Am J Physiol Endocrinol Metab ; 319(3): E459-E471, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32663099

ABSTRACT

Insulin resistance in obesity and type 2 diabetes has been shown to be associated with decreased de novo fatty acid (FA) synthesis in adipose tissue. It is known that insulin can acutely stimulate FA synthesis in adipocytes; however, the mechanisms underlying this effect are unclear. The rate-limiting step in FA synthesis is catalyzed by acetyl-CoA carboxylase (ACC), known to be regulated through inhibitory phosphorylation at S79 by the AMP-activated protein kinase (AMPK). Previous results from our laboratory showed an inhibition of AMPK activity by insulin, which was accompanied by PKB-dependent phosphorylation of AMPK at S485. However, whether the S485 phosphorylation is required for insulin-induced inhibition of AMPK or other mechanisms underlie the reduced kinase activity is not known. To investigate this, primary rat adipocytes were transduced with a recombinant adenovirus encoding AMPK-WT or a nonphosphorylatable AMPK S485A mutant. AMPK activity measurements by Western blot analysis and in vitro kinase assay revealed that WT and S485A AMPK were inhibited to a similar degree by insulin, indicating that AMPK S485 phosphorylation is not required for insulin-induced AMPK inhibition. Further analysis suggested an involvement of decreased AMP-to-ATP ratios in the insulin-induced inhibition of AMPK activity, whereas a possible contribution of phosphodiesterases was excluded. Furthermore, we show that insulin-induced AMPK S485 phosphorylation also occurs in human adipocytes, suggesting it to be of an importance yet to be revealed. Altogether, this study increases our understanding of how insulin regulates AMPK activity, and with that, FA synthesis, in adipose tissue.


Subject(s)
AMP-Activated Protein Kinases/antagonists & inhibitors , Adipocytes/drug effects , Adipocytes/enzymology , Insulin/pharmacology , AMP-Activated Protein Kinases/genetics , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Adipocytes/metabolism , Animals , Energy Metabolism/drug effects , Fatty Acids/metabolism , Glycerol/metabolism , Mutation , Phosphoric Diester Hydrolases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley
3.
Am J Physiol Endocrinol Metab ; 315(5): E1075-E1085, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30253109

ABSTRACT

Activation of AMP-activated protein kinase (AMPK) is considered an attractive strategy for the treatment of type 2 diabetes. Favorable metabolic effects of AMPK activation are mainly observed in skeletal muscle and liver tissue, whereas the effects in human adipose tissue are only poorly understood. Previous studies, which largely employed the AMPK activator 5-aminoimidazole-4-carboxamide-1-ß-d-ribofuranoside (AICAR), suggest an antilipolytic role of AMPK in adipocytes. The aim of this work was to reinvestigate the role of AMPK in the regulation of lipolysis, using the novel allosteric small-molecule AMPK activators A-769662 and 991, with a focus on human adipocytes. For this purpose, human primary subcutaneous adipocytes were treated with A-769662, 991, or AICAR, as a control, before being stimulated with isoproterenol. AMPK activity status, glycerol release, and the phosphorylation of hormone-sensitive lipase (HSL), a key regulator of lipolysis, were then monitored. Our results show that both A-769662 and 991 activated AMPK to a level that was similar to, or greater than, that induced by AICAR. In contrast to AICAR, which as expected was antilipolytic, neither A-769662 nor 991 affected lipolysis in human adipocytes, although 991 treatment led to altered HSL phosphorylation. Furthermore, we suggest that HSL Ser660 is an important regulator of lipolytic activity in human adipocytes. These data suggest that the antilipolytic effect observed with AICAR in previous studies is, at least to some extent, AMPK independent.


Subject(s)
Adenylate Kinase/metabolism , Adipocytes/drug effects , Adipose Tissue/drug effects , Catecholamines/pharmacology , Lipolysis/drug effects , Pyrones/pharmacology , Thiophenes/pharmacology , Adipocytes/metabolism , Adipose Tissue/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Biphenyl Compounds , Female , Humans , Lipolysis/physiology , Male , Mice , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Ribonucleotides/pharmacology , Sterol Esterase/metabolism
4.
Diabetologia ; 60(2): 314-323, 2017 02.
Article in English | MEDLINE | ID: mdl-27807598

ABSTRACT

AIMS/HYPOTHESIS: Salt-inducible kinases (SIKs) are related to the metabolic regulator AMP-activated protein kinase (AMPK). SIK2 is abundant in adipose tissue. The aims of this study were to investigate the expression of SIKs in relation to human obesity and insulin resistance, and to evaluate whether changes in the expression of SIKs might play a causal role in the development of disturbed glucose uptake in human adipocytes. METHODS: SIK mRNA and protein was determined in human adipose tissue or adipocytes, and correlated to clinical variables. SIK2 and SIK3 expression and phosphorylation were analysed in adipocytes treated with TNF-α. Glucose uptake, GLUT protein levels and localisation, phosphorylation of protein kinase B (PKB/Akt) and the SIK substrate histone deacetylase 4 (HDAC4) were analysed after the SIKs had been silenced using small interfering RNA (siRNA) or inhibited using a pan-SIK-inhibitor (HG-9-91-01). RESULTS: We demonstrate that SIK2 and SIK3 mRNA are downregulated in adipose tissue from obese individuals and that the expression is regulated by weight change. SIK2 is also negatively associated with in vivo insulin resistance (HOMA-IR), independently of BMI and age. Moreover, SIK2 protein levels and specific kinase activity display a negative correlation to BMI in human adipocytes. Furthermore, SIK2 and SIK3 are downregulated by TNF-α in adipocytes. Silencing or inhibiting SIK1-3 in adipocytes results in reduced phosphorylation of HDAC4 and PKB/Akt, less GLUT4 at the plasma membrane, and lower basal and insulin-stimulated glucose uptake in adipocytes. CONCLUSION/INTERPRETATION: This is the first study to describe the expression and function of SIKs in human adipocytes. Our data suggest that SIKs might be protective in the development of obesity-induced insulin resistance, with implications for future treatment strategies.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Glucose/metabolism , Insulin/metabolism , Obesity/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Adult , Aged , Animals , Blotting, Western , Female , Humans , Insulin Resistance/genetics , Insulin Resistance/physiology , Male , Mice , Middle Aged , Phosphorylation/drug effects , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/pharmacology
5.
J Cell Sci ; 128(3): 472-86, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25472719

ABSTRACT

Salt-inducible kinase 2 (SIK2) is an AMP-activated protein kinase (AMPK) related kinase abundantly expressed in adipose tissue. Our aim was to identify molecular targets and functions of SIK2 in adipocytes, and to address the role of PKA-mediated phosphorylation of SIK2 on Ser358. Modulation of SIK2 in adipocytes resulted in altered phosphorylation of CREB-regulated transcription co-activator 2 (CRTC2), CRTC3 and class IIa histone deacetylase 4 (HDAC4). Furthermore, CRTC2, CRTC3, HDAC4 and protein phosphatase 2A (PP2A) interacted with SIK2, and the binding of CRTCs and PP2A to wild-type but not Ser358Ala SIK2, was reduced by cAMP elevation. Silencing of SIK2 resulted in reduced GLUT4 (also known as SLC2A4) protein levels, whereas cells treated with CRTC2 or HDAC4 siRNA displayed increased levels of GLUT4. Overexpression or pharmacological inhibition of SIK2 resulted in increased and decreased glucose uptake, respectively. We also describe a SIK2­CRTC2­HDAC4 pathway and its regulation in human adipocytes, strengthening the physiological relevance of our findings. Collectively, we demonstrate that SIK2 acts directly on CRTC2, CRTC3 and HDAC4, and that the cAMP­PKA pathway reduces the interaction of SIK2 with CRTCs and PP2A. Downstream, SIK2 increases GLUT4 levels and glucose uptake in adipocytes.


Subject(s)
Glucose/metabolism , Histone Deacetylases/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , 3T3 Cells , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , Cell Line , Cyclic AMP/metabolism , Glucose Transporter Type 4/metabolism , HEK293 Cells , Histone Deacetylases/genetics , Humans , Mice , Mice, Knockout , Phosphorylation , Protein Binding , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA Interference , RNA, Small Interfering , Rats , Signal Transduction , Transcription Factors/genetics
6.
J Vasc Res ; 54(4): 235-245, 2017.
Article in English | MEDLINE | ID: mdl-28768281

ABSTRACT

BACKGROUND: Smooth muscle cells are important for atherosclerotic plaque stability. Their proper ability to communicate with the extracellular matrix is crucial for maintaining the correct tissue integrity. In this study, we have investigated the role of ß-sarcoglycan within the matrix-binding dystrophin-glycoprotein complex in the development of atherosclerosis. RESULTS: Atherosclerotic plaque development was significantly reduced in ApoE-deficient mice lacking ß-sarcoglycan, and their plaques contained an increase in differentiated smooth muscle cells. ApoE-deficient mice lacking ß-sarcoglycan showed a reduction in ovarian adipose tissue and adipocyte size, while the total weight of the animals was not significantly different. Western blot analysis of adipose tissues showed a decreased activation of protein kinase B, while that of AMP-activated kinase was increased in mice lacking ß-sarcoglycan. Analysis of plasma in ß-sarcoglycan-deficient mice revealed reduced levels of leptin, adiponectin, insulin, cholesterol, and triglycerides but increased levels of IL-6, IL-17, and TNF-α. CONCLUSIONS: Our results indicate that the dystrophin-glycoprotein complex and ß-sarcoglycan can affect the atherosclerotic process. Furthermore, the results show the effects of ß-sarcoglycan deficiency on adipose tissue and lipid metabolism, which may also have contributed to the atherosclerotic plaque reduction.


Subject(s)
Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Atherosclerosis/prevention & control , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Plaque, Atherosclerotic , Sarcoglycans/deficiency , AMP-Activated Protein Kinases/metabolism , Adipocytes/metabolism , Adipocytes/pathology , Adipokines/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Dystrophin-Associated Protein Complex/metabolism , Female , Genetic Predisposition to Disease , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Phenotype , Proto-Oncogene Proteins c-akt/metabolism , Sarcoglycans/genetics
7.
J Biol Chem ; 290(11): 6763-76, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25593322

ABSTRACT

Cyclic nucleotide phosphodiesterase 3A (PDE3) regulates cAMP-mediated signaling in the heart, and PDE3 inhibitors augment contractility in patients with heart failure. Studies in mice showed that PDE3A, not PDE3B, is the subfamily responsible for these inotropic effects and that murine PDE3A1 associates with sarcoplasmic reticulum Ca(2+) ATPase 2 (SERCA2), phospholamban (PLB), and AKAP18 in a multiprotein signalosome in human sarcoplasmic reticulum (SR). Immunohistochemical staining demonstrated that PDE3A co-localizes in Z-bands of human cardiac myocytes with desmin, SERCA2, PLB, and AKAP18. In human SR fractions, cAMP increased PLB phosphorylation and SERCA2 activity; this was potentiated by PDE3 inhibition but not by PDE4 inhibition. During gel filtration chromatography of solubilized SR membranes, PDE3 activity was recovered in distinct high molecular weight (HMW) and low molecular weight (LMW) peaks. HMW peaks contained PDE3A1 and PDE3A2, whereas LMW peaks contained PDE3A1, PDE3A2, and PDE3A3. Western blotting showed that endogenous HMW PDE3A1 was the principal PKA-phosphorylated isoform. Phosphorylation of endogenous PDE3A by rPKAc increased cAMP-hydrolytic activity, correlated with shift of PDE3A from LMW to HMW peaks, and increased co-immunoprecipitation of SERCA2, cav3, PKA regulatory subunit (PKARII), PP2A, and AKAP18 with PDE3A. In experiments with recombinant proteins, phosphorylation of recombinant human PDE3A isoforms by recombinant PKA catalytic subunit increased co-immunoprecipitation with rSERCA2 and rat rAKAP18 (recombinant AKAP18). Deletion of the recombinant human PDE3A1/PDE3A2 N terminus blocked interactions with recombinant SERCA2. Serine-to-alanine substitutions identified Ser-292/Ser-293, a site unique to human PDE3A1, as the principal site regulating its interaction with SERCA2. These results indicate that phosphorylation of human PDE3A1 at a PKA site in its unique N-terminal extension promotes its incorporation into SERCA2/AKAP18 signalosomes, where it regulates a discrete cAMP pool that controls contractility by modulating phosphorylation-dependent protein-protein interactions, PLB phosphorylation, and SERCA2 activity.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Myocardium/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , A Kinase Anchor Proteins/analysis , A Kinase Anchor Proteins/metabolism , Calcium/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/analysis , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/analysis , Humans , Myocardium/cytology , Myocardium/enzymology , Myocardium/ultrastructure , Phosphorylation , Protein Interaction Maps , Protein Isoforms/analysis , Protein Isoforms/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/analysis
8.
J Cell Sci ; 125(Pt 21): 5084-95, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22946044

ABSTRACT

Specificity and versatility in cyclic AMP (cAMP) signalling are governed by the spatial localisation and temporal dynamics of the signal. Phosphodiesterases (PDEs) are important for shaping cAMP signals by hydrolyzing the nucleotide. In pancreatic ß-cells, glucose triggers sub-plasma-membrane cAMP oscillations, which are important for insulin secretion, but the mechanisms underlying the oscillations are poorly understood. Here, we investigated the role of different PDEs in the generation of cAMP oscillations by monitoring the concentration of cAMP in the sub-plasma-membrane space ([cAMP](pm)) with ratiometric evanescent wave microscopy in MIN6 cells or mouse pancreatic ß-cells expressing a fluorescent translocation biosensor. The general PDE inhibitor IBMX increased [cAMP](pm), and whereas oscillations were frequently observed at 50 µM IBMX, 300 µM-1 mM of the inhibitor caused a stable increase in [cAMP](pm). The [cAMP](pm) was nevertheless markedly suppressed by the adenylyl cyclase inhibitor 2',5'-dideoxyadenosine, indicating IBMX-insensitive cAMP degradation. Among IBMX-sensitive PDEs, PDE3 was most important for maintaining a low basal level of [cAMP](pm) in unstimulated cells. After glucose induction of [cAMP](pm) oscillations, inhibitors of PDE1, PDE3 and PDE4 inhibitors the average cAMP level, often without disturbing the [cAMP](pm) rhythmicity. Knockdown of the IBMX-insensitive PDE8B by shRNA in MIN6 cells increased the basal level of [cAMP](pm) and prevented the [cAMP](pm)-lowering effect of 2',5'-dideoxyadenosine after exposure to IBMX. Moreover, PDE8B-knockdown cells showed reduced glucose-induced [cAMP](pm) oscillations and loss of the normal pulsatile pattern of insulin secretion. It is concluded that [cAMP](pm) oscillations in ß-cells are caused by periodic variations in cAMP generation, and that several PDEs, including PDE1, PDE3 and the IBMX-insensitive PDE8B, are required for shaping the sub-membrane cAMP signals and pulsatile insulin release.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/physiology , Cell Membrane/metabolism , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/physiology , Cyclic Nucleotide Phosphodiesterases, Type 3/physiology , Insulin/metabolism , Animals , Cells, Cultured , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Female , Glucose/physiology , Insulin Secretion , Islets of Langerhans/enzymology , Islets of Langerhans/metabolism , Isoenzymes/physiology , Kinetics , Mice , Mice, Inbred C57BL , Periodicity , Phosphatidylinositol Phosphates/metabolism , Second Messenger Systems , Single-Cell Analysis
9.
Cell Tissue Res ; 352(3): 469-78, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23584706

ABSTRACT

Several studies have demonstrated a link between diabetes and the dysfunction of the inner ear. Few studies, however, have reported the signalling mechanisms involved in metabolic control in human inner ear cells. Knowledge of the expression and role of the insulin receptor and downstream signalling components in the inner ear is sparce. Our immunohistochemistry approach has shown that the insulin receptor, insulin receptor substrate 1 (IRS1), protein kinase B (PKB) and insulin-sensitive glucose transporter (GLUT4) are expressed in the sensory epithelium of the human saccule, which also exhibits expression of a calcium-sensitive cAMP/cGMP phosphodiesterase 1C (PDE1C) and the vasopressin type 2 receptor. IRS1 and PDE1C are selectively expressed in sensory epithelial hair cells, whereas the other components are expressed in sensory epithelial supporting cells or in both cell types, as judged from co-expression or non-co-expression with glial fibrillary acidic protein, a marker for supporting cells. Furthermore, IRS1 appears to be localized in association with sensory nerves, whereas GLUT4 is expressed in the peri-nuclear area of stromal cells, as is the case for aquaporin 2. Thus, the insulin receptor, insulin signalling components and selected cAMP signalling components are expressed in the human saccule. In addition to well-known mechanisms of diabetes complications, such as neuropathy and vascular lesions, the expression of these proteins in the saccule could have a role in the observed link between diabetes and balance/hearing disorders.


Subject(s)
Epithelium/metabolism , Insulin/metabolism , Saccule and Utricle/metabolism , Sensation , Signal Transduction , Aquaporin 2/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Glucose Transporter Type 4/metabolism , Hair Cells, Auditory/cytology , Hair Cells, Auditory/enzymology , Humans , Insulin Receptor Substrate Proteins/metabolism , Models, Biological , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/metabolism , Receptors, Vasopressin/metabolism , Saccule and Utricle/cytology , Stromal Cells/cytology , Stromal Cells/metabolism
10.
Acta Otolaryngol ; 143(2): 127-133, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36735299

ABSTRACT

BACKGROUND: Diabetes is associated with inner ear dysfunction. Furthermore, C57BL/6J mice fed high fat diet (HFD), a model for insulin resistance and diabetes, develop endolymphatic hydrops (EH). AIM: Evaluate if betahistine, spironolactone (aldosterone antagonist) and empagliflozin (sodium -glucose cotransporter2 inhibitor) can prevent EH induced by HFD and explore potential mechanisms. METHODS: C57BL/6J mice fed HFD were treated with respective drug. The size of the endolymphatic fluid compartment was measured using contrast enhanced MRI. Secondarily, mice treated with cilostamide, a phosphodiesterase3 inhibitor, to induce EH and HEI-OC1 auditory cells were used to study potential cellular mechanisms of betahistine. RESULTS: HFD-induced EH was prevented by betahistine but not by spironolactone and empagliflozin. Betahistine induced phosphorylation of protein kinaseA substrates but did not prevent cilostamide-induced EH. CONCLUSIONS: Betahistine prevents the development of EH in mice fed HFD, most likely not involving pathways downstream of phosphodiesterase3, an enzyme with implications for dysfunction in diabetes. The finding that spironolactone did not prevent HFD-induced EH suggests different mechanisms for EH induction/treatment since spironolactone prevents EH induced by vasopressin, as previously observed. SIGNIFICANCE: This further demonstrates that independent mechanisms can cause hydropic inner ear diseases which suggests different therapeutic approaches and emphazises the need for personalized medicine.


Subject(s)
Diabetes Mellitus , Endolymphatic Hydrops , Insulin Resistance , Animals , Mice , Betahistine/adverse effects , Spironolactone/pharmacology , Spironolactone/therapeutic use , Mice, Inbred C57BL , Endolymphatic Hydrops/drug therapy , Endolymphatic Hydrops/etiology , Endolymphatic Hydrops/prevention & control , Magnetic Resonance Imaging
11.
Biochem Biophys Res Commun ; 425(4): 812-7, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22892131

ABSTRACT

The incretin - glucose-dependent insulinotropic polypeptide (GIP) - and the pro-inflammatory cytokine osteopontin are known to have important roles in the regulation of adipose tissue functions. In this work we show that GIP stimulates lipogenesis and osteopontin expression in primary adipocytes. The GIP-induced increase in osteopontin expression was inhibited by the NFAT (the transcription factor nuclear factor of activated T-cells) inhibitor A-285222. Also, the NFAT kinase glycogen synthase kinase (GSK) 3 was upregulated by GIP. To test whether cAMP might be involved in GIP-mediated effects on osteopontin a number of strategies were used. Thus, the ß3-adrenergic receptor agonist CL316,243 stimulated osteopontin expression, an effects which was mimicked by OPC3911, a specific inhibitor of phosphodiesterase 3. Furthermore, treatment of phosphodiesterase 3B knock-out mice with CL316,243 resulted in a dramatic upregulation of osteopontin in adipose tissue which was not the case in wild-type mice. In summary, we delineate mechanisms by which GIP stimulates osteopontin in adipocytes. Given the established link between osteopontin and insulin resistance, our data suggest that GIP by stimulating osteopontin expression, also could promote insulin resistance in adipocytes.


Subject(s)
Adipocytes/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Gastric Inhibitory Polypeptide/physiology , Lipogenesis/physiology , NFATC Transcription Factors/physiology , Osteopontin/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Animals , Gastric Inhibitory Polypeptide/pharmacology , Insulin/metabolism , Lipogenesis/drug effects , Male , Mice , NFATC Transcription Factors/antagonists & inhibitors , Osteopontin/genetics , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley
12.
Front Physiol ; 13: 928964, 2022.
Article in English | MEDLINE | ID: mdl-35991175

ABSTRACT

AMP-activated protein kinase (AMPK) activation is considered a useful strategy for the treatment of type 2 diabetes (T2D). It is unclear whether the expression and/or activity of AMPK in adipocytes is dysregulated in obesity. Also, the expression/activity pattern of AMPKß isoforms, which are targets for AMPK activators, in adipocytes remains elusive. In this study we show that the two AMPKß isoforms make roughly equal contributions to AMPK activity in primary human and mouse adipocytes, whereas in cultured 3T3-L1 adipocytes of mouse origin and in primary rat adipocytes, ß1-associated activity clearly dominates. Additionally, we found that obesity is not associated with changes in AMPK subunit expression or kinase activity in adipocytes isolated from subcutaneous adipose tissue from individuals with various BMI.

13.
Acta Otolaryngol ; 142(1): 6-12, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34962430

ABSTRACT

BACKGROUND: The mechanisms of association between diabetes and inner ear dysfunction are unknown, although endolymphatic hydrops may be involved. We have previously shown that insulin signaling components are expressed in human saccule and that insulin signaling takes place in HEI-OC1 auditory cells. AIM: To explore Nedd4-2 as a target for insulin signaling. MATERIALS AND METHODS: Effects of insulin were analyzed using western blot and confocal microscopy in HEI-OC1 auditory cells. RESULTS: Insulin induced phosphorylation of Nedd4-2 and increased the amount of ENaC at the plasma membrane. Also, protein kinase B (PKB) and NDRG1, a substrate for SGK1 (serum and glucocorticoid stimulated kinase), were phosphorylated in response to insulin. The SGK1 inhibitor GSK650394 prevented insulin-induced phosphorylation of NRDG1, but not of PKB and Nedd4-2, whereas the phosphatidylinositol 3-kinase (PI3K) inhibitor wortmannin and the PKB inhibitor MK2206 inhibited phosphorylation of all components. Ceramides prevented insulin-induced phosphorylation of PKB and NDRG1, but not of Nedd4-2. The ceramide metabolite sphingosine 1-phosphate induced phosphorylation of Nedd4-2. CONCLUSIONS: Insulin induces phosphorylation of Nedd4-2, most likely involving PI3K/PKB signaling. Sphingosine 1-phosphate might protect Nedd4-2 against ceramide-induced insulin resistance. SIGNIFICANCE: Insulin-mediated regulation of Nedd4-2 might impact on inner ear sodium homeostasis with implications for diabetes-induced inner ear damage.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Epithelial Sodium Channels/metabolism , Insulin/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Repressor Proteins/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Animals , Cell Line , Ceramides/pharmacology , Ear, Inner/cytology , Phosphorylation
14.
Peptides ; 151: 170747, 2022 05.
Article in English | MEDLINE | ID: mdl-35065097

ABSTRACT

Impaired beta cell function and beta cell death are key features of type 2 diabetes (T2D). Cocaine- and amphetamine-regulated transcript (CART) is necessary for normal islet function in mice. CART increases glucose-stimulated insulin secretion in vivo in mice and in vitro in human islets and CART protects beta cells against glucotoxicity-induced cell death in vitro in rats. Furthermore, beta cell CART is upregulated in T2D patients and in diabetic rodent models as a consequence of hyperglycaemia. The aim of this study was to assess the impact of upregulated beta cell CART on islet hormone secretion and glucose homeostasis in a transgenic mouse model. To this end, mice with beta cell-specific overexpression of CART (CARTtg mice) were generated. CARTtg mice challenged by aging, high fat diet feeding or streptozotocin treatment were phenotyped with respect to in vivo and in vitro insulin and glucagon secretion, glucose homeostasis, and beta cell mass. In addition, the impact of adenoviral overexpression of CART on insulin secretion was studied in INS-1 832/13 cells. CARTtg mice had a normal metabolic phenotype under basal conditions. On the other hand, with age CARTtg mice displayed increased insulin secretion and improved glucose elimination, compared with age-matched WT mice. Furthermore, compared with WT controls, CARTtg mice had increased insulin secretion after feeding a high fat diet, as well as lower glucose levels and higher insulin secretion after streptozotocin treatment. Viral overexpression of CART in INS-1 832/13 cells resulted in increased glucose-stimulated insulin secretion. Together, these results imply that beta cell CART acts to increase insulin secretion when beta cell function is challenged. We propose that the increase in beta cell CART is part of a compensatory mechanisms trying to counteract the hyperglycaemia in T2D.


Subject(s)
Diabetes Mellitus, Type 2 , Hyperglycemia , Insulin Resistance , Insulin-Secreting Cells , Islets of Langerhans , Animals , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Glucose/metabolism , Humans , Hyperglycemia/genetics , Hyperglycemia/metabolism , Insulin/metabolism , Insulin Resistance/genetics , Insulin Secretion , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Mice , Nerve Tissue Proteins/genetics , Rats , Streptozocin
15.
Front Physiol ; 12: 740666, 2021.
Article in English | MEDLINE | ID: mdl-34630160

ABSTRACT

Lipid uptake can be facilitated via caveolae, specific plasma membrane invaginations abundantly expressed in adipocytes. The dynamin-related protein EH domain-containing 2 (EHD2) stabilizes caveolae at the cell surface. Here, we have examined the importance of EHD2 for lipid handling using primary adipocytes isolated from EHD2 knockout (Ehd2-/- ) C57BL6/N mice. Following high-fat diet (HFD) feeding, we found a clear impairment of epididymal, but not inguinal, adipose tissue expansion in Ehd2-/- compared with Ehd2+/+ (WT) mice. Cell size distribution analysis revealed that Ehd2-/- mice had a lower proportion of small adipocytes, and an accumulation of medium-sized adipocytes in both epididymal and inguinal adipose tissue. Further, PPARγ activity, FABP4 and caveolin-1 expression were decreased in adipocytes isolated from Ehd2-/- mice. Inguinal adipocytes isolated from Ehd2-/- mice displayed reduced lipolysis in response to beta adrenergic receptor agonist, which was associated with reduced phosphorylation of perilipin-1 and hormone sensitive lipase (HSL). This impairment could not be rescued using a cAMP analog, indicating that impaired lipolysis in Ehd2-/- primary adipocytes likely occurs at the level of, or downstream of, protein kinase A (PKA). Altogether, these findings pinpoint the importance of EHD2 for maintained intracellular lipid metabolism, and emphasize differences in mechanisms regulating lipid handling in various adipose-tissue depots.

16.
Biochem J ; 424(3): 399-410, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-19747167

ABSTRACT

In adipocytes, PDE3B (phosphodiesterase 3B) is an important regulatory effector in signalling pathways controlled by insulin and cAMP-increasing hormones. Stimulation of 3T3-L1 adipocytes with insulin or the beta3-adrenergic receptor agonist CL316243 (termed CL) indicated that insulin preferentially phosphorylated/activated PDE3B associated with internal membranes (endoplasmic reticulum/Golgi), whereas CL preferentially phosphorylated/activated PDE3B associated with caveolae. siRNA (small interfering RNA)-mediated KD (knockdown) of CAV-1 (caveolin-1) in 3T3-L1 adipocytes resulted in down-regulation of expression of membrane-associated PDE3B. Insulin-induced activation of PDE3B was reduced, whereas CL-mediated activation was almost totally abolished. Similar results were obtained in adipocytes from Cav-1-deficient mice. siRNA-mediated KD of CAV-1 in 3T3-L1 adipocytes also resulted in inhibition of CL-stimulated phosphorylation of HSL (hormone-sensitive lipase) and perilipin A, and of lipolysis. Superose 6 gel-filtration chromatography of solubilized membrane proteins from adipocytes stimulated with insulin or CL demonstrated the reversible assembly of distinct macromolecular complexes that contained 32P-phosphorylated PDE3B and signalling molecules thought to be involved in its activation. Insulin- and CL-induced macromolecular complexes were enriched in cholesterol, and contained certain common signalling proteins [14-3-3, PP2A (protein phosphatase 2A) and cav-1]. The complexes present in insulin-stimulated cells contained tyrosine-phosphorylated IRS-1 (insulin receptor substrate 1) and its downstream signalling proteins, whereas CL-activated complexes contained beta3-adrenergic receptor, PKA-RII [PKA (cAMP-dependent protein kinase)-regulatory subunit] and HSL. Insulin- and CL-mediated macromolecular complex formation was significantly inhibited by CAV-1 KD. These results suggest that cav-1 acts as a molecular chaperone or scaffolding molecule in cholesterol-rich lipid rafts that may be necessary for the proper stabilization and activation of PDE3B in response to CL and insulin.


Subject(s)
Adipocytes/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Dioxoles/pharmacology , Insulin/pharmacology , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Adrenergic beta-Agonists/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Blotting, Western , Caveolae/drug effects , Caveolae/metabolism , Caveolin 1/genetics , Caveolin 1/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/genetics , Endoplasmic Reticulum/enzymology , Enzyme Activation/drug effects , Gene Expression Regulation, Enzymologic , Golgi Apparatus/enzymology , Lipolysis/drug effects , Macromolecular Substances/metabolism , Mice , Phosphorylation/drug effects , Protein Transport/drug effects , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Substrate Specificity
17.
Adipocyte ; 9(1): 587-599, 2020 12.
Article in English | MEDLINE | ID: mdl-33016185

ABSTRACT

The aim of this study was to elucidate mechanisms whereby bile acids exert beneficial metabolic effects, using the Cyp8b1-/- mouse as model. These mice are unable to synthesize cholic acid, resulting in increased synthesis of chenodeoxycholic acid and enlarged bile acid pool. Cyp8b1-/- mice were found to be protected against high-fat diet induced obesity. Bomb calorimetry measurements showed increased faecal energy output in Cyp8b1-/ mice. Indirect calorimetry measurements demonstrated increased energy expenditure in Cyp8b1-/- mice. Meal tolerance tests revealed no differences in glucose disposal, but the insulin response was lower in Cyp8b1-/- mice. Intravenous glucose tolerance tests, as well as static incubations of isolated islets, showed no difference between the groups, whereas insulin tolerance tests demonstrated improved insulin sensitivity in Cyp8b1-/- mice. The genes encoding mitochondrial transcription factor A (TFAM) and type 2-iodothyronine deiodinase were upregulated in brown adipose tissue of Cyp8b1/- mice and Western blot analyses showed increased abundance of TFAM, and a trend towards increased abundance of UCP1. The upregulation of TFAM and UCP1 was accompanied by increased mitochondrial density, as shown by transmission electron microscopy. White adipocytes of Cyp8b1-/- mice exhibited increased responsiveness to both catecholamines and insulin in lipolysis experiments and increased insulin-stimulated lipogenesis. In conclusion, increased energy expenditure, mitochondrial density of brown adipocytes and faecal energy output may all contribute to the protection against diet-induced obesity of Cyp8b1-/- mice. Enhanced insulin sensitivity of Cyp8b1-/- mice is accompanied by increased hormonal responsiveness of white adipocytes.


Subject(s)
Adipose Tissue/metabolism , Diet, High-Fat , Energy Metabolism , Obesity/etiology , Obesity/metabolism , Steroid 12-alpha-Hydroxylase/deficiency , Adipocytes/metabolism , Adipose Tissue/pathology , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Disease Susceptibility , Glucose/metabolism , Insulin/metabolism , Insulin Resistance , Lipid Metabolism , Lipogenesis/genetics , Lipolysis/genetics , Mice , Mice, Knockout , Mitochondria/metabolism , Obesity/pathology
18.
Article in English | MEDLINE | ID: mdl-32238362

ABSTRACT

OBJECTIVE: The mechanisms underlying the association between diabetes and inner ear dysfunction are not known yet. The aim of the present study is to evaluate the impact of obesity/insulin resistance on inner ear fluid homeostasis in vivo, and to investigate whether the organ of Corti could be a target tissue for insulin signaling using auditory House Ear Institute-Organ of Corti 1 (HEI-OC1) cells as an in vitro model. METHODS: High fat diet (HFD) fed C57BL/6J mice were used as a model to study the impact of insulin resistance on the inner ear. In one study, 12 C57BL/6J mice were fed either control diet or HFD and the size of the inner ear endolymphatic fluid compartment (EFC) was measured after 30 days using MRI and gadolinium contrast as a read-out. In another study, the size of the inner ear EFC was evaluated in eight C57BL/6J mice both before and after HFD feeding, with the same techniques. HEI-OC1 auditory cells were used as a model to investigate insulin signaling in organ of Corti cells. RESULTS: HFD feeding induced an expansion of the EFC in C57BL/6J mice, a hallmark of inner ear dysfunction. Insulin also induced phosphorylation of protein kinase B (PKB/Akt) at Ser473, in a PI3-kinase-dependent manner. The phosphorylation of PKB was inhibited by isoproterenol and IBMX, a general phosphodiesterase (PDE) inhibitor. PDE1B, PDE4D and the insulin-sensitive PDE3B were found expressed and catalytically active in HEI-OC1 cells. Insulin decreased and AICAR, an activator of AMP-activated protein kinase, increased the phosphorylation at the inhibitory Ser79 of acetyl-CoA carboxylase, the rate-limiting enzyme in de novo lipogenesis. Furthermore, the activity of hormone-sensitive lipase, the rate-limiting enzyme in lipolysis, was detected in HEI-OC1 cells. CONCLUSIONS: The organ of Corti could be a target tissue for insulin action, and inner ear insulin resistance might contribute to the association between diabetes and inner ear dysfunction.


Subject(s)
Ear, Inner , Insulin Resistance , Animals , Cyclic Nucleotide Phosphodiesterases, Type 1 , Insulin , Mice , Mice, Inbred C57BL , Organ of Corti
19.
J Clin Invest ; 116(12): 3240-51, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17143332

ABSTRACT

Cyclic nucleotide phosphodiesterase 3B (PDE3B) has been suggested to be critical for mediating insulin/IGF-1 inhibition of cAMP signaling in adipocytes, liver, and pancreatic beta cells. In Pde3b-KO adipocytes we found decreased adipocyte size, unchanged insulin-stimulated phosphorylation of protein kinase B and activation of glucose uptake, enhanced catecholamine-stimulated lipolysis and insulin-stimulated lipogenesis, and blocked insulin inhibition of catecholamine-stimulated lipolysis. Glucose, alone or in combination with glucagon-like peptide-1, increased insulin secretion more in isolated pancreatic KO islets, although islet size and morphology and immunoreactive insulin and glucagon levels were unchanged. The beta(3)-adrenergic agonist CL 316,243 (CL) increased lipolysis and serum insulin more in KO mice, but blood glucose reduction was less in CL-treated KO mice. Insulin resistance was observed in KO mice, with liver an important site of alterations in insulin-sensitive glucose production. In KO mice, liver triglyceride and cAMP contents were increased, and the liver content and phosphorylation states of several insulin signaling, gluconeogenic, and inflammation- and stress-related components were altered. Thus, PDE3B may be important in regulating certain cAMP signaling pathways, including lipolysis, insulin-induced antilipolysis, and cAMP-mediated insulin secretion. Altered expression and/or regulation of PDE3B may contribute to metabolic dysregulation, including systemic insulin resistance.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/genetics , Energy Metabolism/genetics , Homeostasis/genetics , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Adiponectin/metabolism , Animals , Blotting, Western , Catecholamines/metabolism , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3 , Energy Metabolism/physiology , Female , Homeostasis/physiology , Immunohistochemistry , Insulin/metabolism , Insulin Resistance/genetics , Insulin Resistance/physiology , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Lipolysis/genetics , Lipolysis/physiology , Liver/metabolism , Male , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology , Time Factors , Triglycerides/metabolism
20.
J Vasc Res ; 46(1): 25-35, 2009.
Article in English | MEDLINE | ID: mdl-18515972

ABSTRACT

BACKGROUND/AIM: Augmented adrenergic control of total peripheral vascular resistance (TPVR) in spontaneously hypertensive rats (SHR) may result from deficiencies in the vasodilatory system(s). Here, we studied the effect of cyclic AMP (cAMP) on TPVR-baseline and adrenergic vasoconstriction in SHR and normotensive controls (WKY). METHODS: Blood pressure and cardiac output were monitored in anesthetized rats, and TPVR calculated. RESULTS: cAMP-analogue (8CPT-cAMP) and phosphodiesterase (PDE) 3 inhibitor (milrinone) reduced TPVR in both strains. G(i) inactivator (pertussis toxin) lowered TPVR but not in all SHR. DeltaTPVR induced by alpha(1)-adrenoceptor agonist (phenylephrine) was reduced by 8CPT-cAMP and milrinone in both strains. They also clearly reduced the response to endogenous noradrenaline release (tyramine) in SHR but had little effect in WKY. When pertussis toxin reduced baseline, it also eliminated the tyramine TPVR response. Propranolol did not change the effect of milrinone on the phenylephrine or tyramine response. Strain-related differences in aorta, femoral arteries or skeletal muscle PDE activity (total/PDE3/PDE4) were absent. CONCLUSIONS: cAMP signaling down-stream of cAMP was functional in SHR, and opposed alpha(1)-adrenoceptor vasoconstriction in both strains. G(i) activity greatly influenced the TPVR baseline and adrenergic TPVR responses, and its activity appeared increased in SHR. Therapeutics aiming to increase signaling through this pathway may turn out to be valuable in the treatment of hypertension.


Subject(s)
Cyclic AMP/physiology , Signal Transduction/physiology , Vascular Resistance/drug effects , Adenylate Cyclase Toxin/pharmacology , Animals , Aorta/metabolism , Blood Pressure/drug effects , Cardiac Output/drug effects , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Femoral Artery/metabolism , Hypertension/drug therapy , Hypertension/physiopathology , Male , Milrinone/pharmacology , Muscle, Striated/metabolism , Phenylephrine/pharmacology , Propranolol/pharmacology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Thionucleotides/pharmacology , Tyramine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL