Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
Front Cell Dev Biol ; 10: 979330, 2022.
Article En | MEDLINE | ID: mdl-36072335

Beige adipocytes play key roles in organismal energy and metabolic balance. In this study, we assessed whether the supplementation of human white adipocytes, differentiated from human adipose tissue-derived stem cells, with nicotinamide riboside (NR), a potent NAD + precursor, can shift differentiation to beige adipocytes (beiging). NR induced mitochondrial biogenesis and the expression of beige markers (TBX1 and UCP1) in white adipocytes demonstrating that NR can declutch beiging. NR did not induce PARP activity but supported SIRT1 induction, which plays a key role in beiging. NR induced etomoxir-resistant respiration, suggesting increases in the oxidation of carbohydrates, carbohydrate breakdown products, or amino acids. Furthermore, NR boosted oligomycin-resistant respiration corresponding to uncoupled respiration. Enhanced etomoxir and oligomycin-resistant respiration were dependent on mitochondrial reactive-species production. Taken together, NR supplementation can induce beiging and uncoupled respiration, which are beneficial for combatting metabolic diseases.

2.
PLoS One ; 15(9): e0236081, 2020.
Article En | MEDLINE | ID: mdl-32960890

Type 2 diabetes mellitus (T2DM), one of the most common metabolic diseases, is characterized by insulin resistance and inadequate insulin secretion of ß cells. Glycogen phosphorylase (GP) is the key enzyme in glycogen breakdown, and contributes to hepatic glucose production during fasting or during insulin resistance. Pharmacological GP inhibitors are potential glucose lowering agents, which may be used in T2DM therapy. A natural product isolated from the cultured broth of the fungal strain No. 138354, called 2,3-bis(4-hydroxycinnamoyloxy)glutaric acid (FR258900), was discovered a decade ago. In vivo studies showed that FR258900 significantly reduced blood glucose levels in diabetic mice. We previously showed that GP inhibitors can potently enhance the function of ß cells. The purpose of this study was to assess whether an analogue of FR258900 can influence ß cell function. BF142 (Meso-Dimethyl 2,3-bis[(E)-3-(4-acetoxyphenyl)prop-2-enamido]butanedioate) treatment activated the glucose-stimulated insulin secretion pathway, as indicated by enhanced glycolysis, increased mitochondrial oxidation, significantly increased ATP production, and elevated calcium influx in MIN6 cells. Furthermore, BF142 induced mTORC1-specific phosphorylation of S6K, increased levels of PDX1 and insulin protein, and increased insulin secretion. Our data suggest that BF142 can influence ß cell function and can support the insulin producing ability of ß cells.


Cinnamates/pharmacology , Enzyme Inhibitors/pharmacology , Glutarates/pharmacology , Glycogen Phosphorylase/antagonists & inhibitors , Insulin Secretion/drug effects , Insulin-Secreting Cells/drug effects , Succinic Acid/pharmacology , Animals , Cell Line, Tumor , Cinnamates/chemistry , Enzyme Inhibitors/chemistry , Glucose/metabolism , Glutarates/chemistry , Glycogen Phosphorylase/metabolism , Glycolysis/drug effects , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Methylation , Mice , Succinic Acid/chemistry
3.
J Transl Med ; 17(1): 229, 2019 07 19.
Article En | MEDLINE | ID: mdl-31324258

BACKGROUND: Chemotherapy-induced left ventricular dysfunction represents a major clinical problem, which is often only recognised at an advanced stage, when supportive therapy is ineffective. Although an early heart failure treatment could positively influence the health status and clinical outcome, there is still no evidence of routine prophylactic cardioprotection for the majority of patients without previous cardiovascular history awaiting potentially cardiotoxic chemotherapy. In this study, we set out to investigate whether a prophylactic cardioprotective therapy relative to a conventionally scheduled heart failure treatment is more effective in preventing cardiotoxicity in a rodent model of doxorubicin (DOX)-induced cardiomyopathy. METHODS: Male Wistar rats (n = 7-11 per group) were divided into 4 subgroups, namely negative controls receiving intravenous saline (CON), positive controls receiving intravenous DOX (6 cycles; D-CON), and DOX-treated animals receiving either prophylactic (PRE, started 1 week before DOX) or conventionally applied (POST, started 1 month after DOX) combined heart failure therapy of oral bisoprolol, perindopril and eplerenone. Blood pressure, heart rate, body weight and echocardiographic parameters were monitored in vivo, whereas myocardial fibrosis, capillarisation, ultrastructure, myofilament function, apoptosis, oxidative stress and mitochondrial biogenesis were studied in vitro. RESULTS: The survival rate in the PRE group was significantly improved compared to D-CON (p = 0.0207). DOX increased the heart rate of the animals (p = 0.0193), while the blood pressure (p ≤ 0.0105) and heart rate (p = 0.0029) were significantly reduced in the PRE group compared to D-CON and POST. The ejection fraction remained preserved in the PRE group compared to D-CON or POST (p ≤ 0.0237), while none of the treatments could prevent the DOX-induced increase in the isovolumetric relaxation time. DOX decreased the rate of the actin-myosin cross-bridge cycle, irrespective of any treatment applied (p ≤ 0.0433). The myocardium of the D-CON and POST animals displayed pronounced ultrastructural damage, which was not apparent in the PRE group (p ≤ 0.033). While the DOX-induced apoptotic activity could be reduced in both the PRE and POST groups (p ≤ 0.0433), no treatment was able to prevent fibrotic remodelling or the disturbed mitochondrial biogenesis. CONCLUSION: For attenuating DOX-induced adverse myocardial effects, prophylactic cardioprotection has many advantages compared to a late-applied treatment.


Cardiomyopathies/chemically induced , Cardiomyopathies/therapy , Doxorubicin/adverse effects , Heart Failure/therapy , Animals , Apoptosis , Cardiomyopathies/diagnostic imaging , Caspase 3/metabolism , Disease Models, Animal , Echocardiography , Fibrosis , Heart Failure/diagnostic imaging , Male , Myocardium/pathology , Myocardium/ultrastructure , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats, Wistar , Survival Analysis
4.
Biochem Pharmacol ; 167: 76-85, 2019 09.
Article En | MEDLINE | ID: mdl-31251940

Mitochondrial biogenesis is a key feature of energy expenditure and organismal energy balance. Genetic deletion of PARP1 or PARP2 was shown to induce mitochondrial biogenesis and energy expenditure. In line with that, PARP inhibitors were shown to induce energy expenditure in skeletal muscle. We aimed to investigate whether pharmacological inhibition of PARPs induces brown or beige adipocyte differentiation. SVF fraction of human pericardial adipose tissue was isolated and human adipose-derived mesenchymal stem cells (hADMSCs) were differentiated to white and beige adipocytes. A subset of hADMSCs were differentiated to white adipocytes in the presence of Olaparib, a potent PARP inhibitor currently in clinical use, to induce browning. Olaparib induced morphological changes (smaller lipid droplets) in white adipocytes that is a feature of brown/beige adipocytes. Furthermore, Olaparib induced mitochondrial biogenesis in white adipocytes and enhanced UCP1 expression. We showed that Olaparib treatment inhibited nuclear and cytosolic PAR formation, induced NAD+/NADH ratio and consequently boosted SIRT1 and AMPK activity and the downstream transcriptional program leading to increases in OXPHOS. Olaparib treatment did not induce the expression of beige adipocyte markers in white adipocytes, suggesting the formation of brown or brown-like adipocytes. PARP1, PARP2 and tankyrases are key players in the formation of white adipose tissue. Hereby, we show that PARP inhibition induces the transdifferentiation of white adipocytes to brown-like adipocytes suggesting that PARP activity could be a determinant of the differentiation of these adipocyte lineages.


Adipocytes, Brown/metabolism , Adipocytes, White/metabolism , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Adipocytes, Brown/drug effects , Adipocytes, White/drug effects , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Humans
5.
PLoS One ; 13(1): e0187789, 2018.
Article En | MEDLINE | ID: mdl-29293500

Poly(ADP-ribose) polymerase (PARP)10 is a PARP family member that performs mono-ADP-ribosylation of target proteins. Recent studies have linked PARP10 to metabolic processes and metabolic regulators that prompted us to assess whether PARP10 influences mitochondrial oxidative metabolism. The depletion of PARP10 by specific shRNAs increased mitochondrial oxidative capacity in cellular models of breast, cervical, colorectal and exocrine pancreas cancer. Upon silencing of PARP10, mitochondrial superoxide production decreased in line with increased expression of antioxidant genes pointing out lower oxidative stress upon PARP10 silencing. Improved mitochondrial oxidative capacity coincided with increased AMPK activation. The silencing of PARP10 in MCF7 and CaCo2 cells decreased the proliferation rate that correlated with increased expression of anti-Warburg enzymes (Foxo1, PGC-1α, IDH2 and fumarase). By analyzing an online database we showed that lower PARP10 expression increases survival in gastric cancer. Furthermore, PARP10 expression decreased upon fasting, a condition that is characterized by increases in mitochondrial biogenesis. Finally, lower PARP10 expression is associated with increased fatty acid oxidation.


Mitochondria/physiology , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins/metabolism , Adenylate Kinase/metabolism , Animals , Blotting, Western , Cell Line , Cell Proliferation/physiology , Electrophoresis, Polyacrylamide Gel , Gene Silencing , Humans , Male , Mice, Inbred C57BL , Oxidation-Reduction , Oxidative Stress , Oxygen Consumption , Poly(ADP-ribose) Polymerases/genetics , Proto-Oncogene Proteins/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
6.
Br J Pharmacol ; 175(2): 301-319, 2018 01.
Article En | MEDLINE | ID: mdl-28409826

BACKGROUND AND PURPOSE: Glycogen phosphorylase (GP) is the key enzyme for glycogen degradation. GP inhibitors (GPi-s) are glucose lowering agents that cause the accumulation of glucose in the liver as glycogen. Glycogen metabolism has implications in beta cell function. Glycogen degradation can maintain cellular glucose levels, which feeds into catabolism to maintain insulin secretion, and elevated glycogen degradation levels contribute to glucotoxicity. The purpose of this study was to assess whether influencing glycogen metabolism in beta cells by GPi-s affects the function of these cells. EXPERIMENTAL APPROACH: The effects of structurally different GPi-s were investigated on MIN6 insulinoma cells and in a mouse model of diabetes. KEY RESULTS: GPi treatment increased glycogen content and, consequently, the surface area of glycogen in MIN6 cells. Furthermore, GPi treatment induced insulin receptor ß (InsRß), Akt and p70S6K phosphorylation, as well as pancreatic and duodenal homeobox 1(PDX1) and insulin expression. In line with these findings, GPi-s enhanced non-stimulated and glucose-stimulated insulin secretion in MIN6 cells. The InsRß was shown to co-localize with glycogen particles as confirmed by in silico screening, where components of InsR signalling were identified as glycogen-bound proteins. GPi-s also activated the pathway of insulin secretion, indicated by enhanced glycolysis, mitochondrial oxidation and calcium signalling. Finally, GPi-s increased the size of islets of Langerhans and improved glucose-induced insulin release in mice. CONCLUSION AND IMPLICATIONS: These data suggest that GPi-s also target beta cells and can be repurposed as agents to preserve beta cell function or even ameliorate beta cell dysfunction in different forms of diabetes. LINKED ARTICLES: This article is part of a themed section on Inventing New Therapies Without Reinventing the Wheel: The Power of Drug Repurposing. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.2/issuetoc.


Glycogen Phosphorylase/antagonists & inhibitors , Insulin-Secreting Cells/drug effects , Animals , Calcium Signaling/drug effects , Cells, Cultured , Glycogen/metabolism , Glycolysis/drug effects , Insulin/metabolism , Islets of Langerhans/drug effects , Male , Mice , Mitochondria/metabolism , Receptor, Insulin/metabolism
7.
PLoS One ; 11(6): e0157644, 2016.
Article En | MEDLINE | ID: mdl-27322180

Beige adipocytes are special cells situated in the white adipose tissue. Beige adipocytes, lacking thermogenic cues, morphologically look quite similar to regular white adipocytes, but with a markedly different response to adrenalin. White adipocytes respond to adrenergic stimuli by enhancing lipolysis, while in beige adipocytes adrenalin induces mitochondrial biogenesis too. A key step in the differentiation and function of beige adipocytes is the deacetylation of peroxisome proliferator-activated receptor (PPARγ) by SIRT1 and the consequent mitochondrial biogenesis. AMP-activated protein kinase (AMPK) is an upstream activator of SIRT1, therefore we set out to investigate the role of AMPK in beige adipocyte differentiation using human adipose-derived mesenchymal stem cells (hADMSCs) from pericardial adipose tissue. hADMSCs were differentiated to white and beige adipocytes and the differentiation medium of the white adipocytes was supplemented with 100 µM [(2R,3S,4R,5R)-5-(4-Carbamoyl-5-aminoimidazol-1-yl)-3,4-dihydroxyoxolan-2-yl]methyl dihydrogen phosphate (AICAR), a known activator of AMPK. The activation of AMPK with AICAR led to the appearance of beige-like morphological properties in differentiated white adipocytes. Namely, smaller lipid droplets appeared in AICAR-treated white adipocytes in a similar fashion as in beige cells. Moreover, in AICAR-treated white adipocytes the mitochondrial network was more fused than in white adipocytes; a fused mitochondrial system was characteristic to beige adipocytes. Despite the morphological similarities between AICAR-treated white adipocytes and beige cells, functionally AICAR-treated white adipocytes were similar to white adipocytes. We were unable to detect increases in basal or cAMP-induced oxygen consumption rate (a marker of mitochondrial biogenesis) when comparing control and AICAR-treated white adipocytes. Similarly, markers of beige adipocytes such as TBX1, UCP1, CIDEA, PRDM16 and TMEM26 remained the same when comparing control and AICAR-treated white adipocytes. Our data point out that in human pericardial hADMSCs the role of AMPK activation in controlling beige differentiation is restricted to morphological features, but not to actual metabolic changes.


AMP-Activated Protein Kinases/metabolism , Adipocytes, Beige/cytology , Adipocytes, White/enzymology , Adipose Tissue, White/cytology , Aminoimidazole Carboxamide/analogs & derivatives , Pericardium/cytology , Ribonucleotides/pharmacology , Stem Cells/enzymology , Adipocytes, Beige/drug effects , Adipocytes, Beige/enzymology , Aminoimidazole Carboxamide/pharmacology , Cell Shape/drug effects , Enzyme Activation/drug effects , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Dynamics/drug effects , Phenotype , Stem Cells/cytology , Stem Cells/drug effects
8.
Indian J Med Res ; 143(2): 220-6, 2016 Feb.
Article En | MEDLINE | ID: mdl-27121520

BACKGROUND & OBJECTIVES: Ischaemic stroke is a life burdening disease for which carotid endarterectomy (CEA) is considered a gold standard intervention. Pro-inflammatory markers like matrix metalloproteinases (MMPs) and their inhibitors (TIMPs) and S-100 Beta (S100B) may have a role in the early inflammation and cognitive decline following CEA. This study was aimed to describe the perioperative time courses and correlations between of MMP-9, TIMP-1 and S100B following CEA. METHODS: Fifty four patients scheduled for CEA were enrolled. Blood samples were collected at four time points, T 1 : preoperative, T 2 : 60 min after cross-clamp release, T 3 : first postoperative morning, T 4 : third postoperative morning. Twenty atherosclerotic patients were included as controls. Plasma MMP-9, TIMP-1 and S100B levels were estimated by ELISA. RESULTS: TIMP-1 was decreased significantly in the CEA group (P<0.01). Plasma MMP-9 was elevated and remained elevated from T 1-4 in the CEA group (P<0.05) with a marked elevation in T 3 compared to T 1 (P<0.05). MMP-9/TIMP-1 was elevated in the CEA group and increased further by T 2 and T 3 (P<0.05). S100B was elevated on T 2 and decreased on T 3-4 compared to T 1 . INTERPRETATION & CONCLUSIONS: Our study provides information on the dynamic changes of MMP-9-TIMP-1 system and S100B in the perioperative period. Preoperative reduction of TIMP-1 might be predictive for shunt requirement but future studies are required for verification.


Endarterectomy, Carotid/adverse effects , Matrix Metalloproteinase 9/blood , S100 Calcium Binding Protein beta Subunit/blood , Stroke/surgery , Tissue Inhibitor of Metalloproteinase-1/blood , Aged , Biomarkers/blood , Carotid Arteries/physiopathology , Carotid Arteries/surgery , Female , Humans , Inflammation/blood , Inflammation/physiopathology , Male , Middle Aged , Perioperative Period , Stroke/blood , Stroke/physiopathology
9.
PLoS One ; 11(2): e0150232, 2016.
Article En | MEDLINE | ID: mdl-26919657

Cancer cells are characterized by metabolic alterations, namely, depressed mitochondrial oxidation, enhanced glycolysis and pentose phosphate shunt flux to support rapid cell growth, which is called the Warburg effect. In our study we assessed the metabolic consequences of a joint treatment of MCF-7 breast cancer cells with AICAR, an inducer of AMP-activated kinase (AMPK) jointly with methotrexate (MTX), a folate-analog antimetabolite that blunts de novo nucleotide synthesis. MCF7 cells, a model of breast cancer cells, were resistant to the individual application of AICAR or MTX, however combined treatment of AICAR and MTX reduced cell proliferation. Prolonged joint application of AICAR and MTX induced AMPK and consequently enhanced mitochondrial oxidation and reduced the rate of glycolysis. These metabolic changes suggest an anti-Warburg rearrangement of metabolism that led to the block of the G1/S and the G2/M transition slowing down cell cycle. The slowdown of cell proliferation was abolished when mitotropic transcription factors, PGC-1α, PGC-1ß or FOXO1 were silenced. In human breast cancers higher expression of AMPKα and FOXO1 extended survival. AICAR and MTX exerts similar additive antiproliferative effect on other breast cancer cell lines, such as SKBR and 4T1 cells, too. Our data not only underline the importance of Warburg metabolism in breast cancer cells but nominate the AICAR+MTX combination as a potential cytostatic regime blunting Warburg metabolism. Furthermore, we suggest the targeting of AMPK and FOXO1 to combat breast cancer.


AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/pathology , Cytostatic Agents/pharmacology , Energy Metabolism/drug effects , Forkhead Transcription Factors/metabolism , Methotrexate/pharmacology , Neoplasm Proteins/metabolism , Ribonucleotides/pharmacology , Aminoimidazole Carboxamide/administration & dosage , Aminoimidazole Carboxamide/pharmacology , Antimetabolites, Antineoplastic/pharmacology , Bone Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Cell Cycle/drug effects , Cell Division/drug effects , Cell Line, Tumor , Cytostatic Agents/administration & dosage , Drug Screening Assays, Antitumor , Drug Synergism , Enzyme Induction/drug effects , Female , Forkhead Box Protein O1 , Gene Expression Regulation, Neoplastic , Glycolysis/drug effects , Humans , Lactates/metabolism , MCF-7 Cells , Membrane Potential, Mitochondrial/drug effects , Methotrexate/administration & dosage , Molecular Targeted Therapy , Osteosarcoma/pathology , RNA Interference , Ribonucleotides/administration & dosage , Transcription Factors/antagonists & inhibitors
10.
Trends Endocrinol Metab ; 26(2): 75-83, 2015 Feb.
Article En | MEDLINE | ID: mdl-25497347

Mitochondria are essential in cellular stress responses. Mitochondrial output to environmental stress is a major factor in metabolic adaptation and is regulated by a complex network of energy and nutrient sensing proteins. Activation of poly(ADP-ribose) polymerases (PARPs) has been known to impair mitochondrial function; however, our view of PARP-mediated mitochondrial dysfunction and injury has only recently fundamentally evolved. In this review, we examine our current understanding of PARP-elicited mitochondrial damage, PARP-mediated signal transduction pathways, transcription factors that interact with PARPs and govern mitochondrial biogenesis, as well as mitochondrial diseases that are mediated by PARPs. With PARP activation emerging as a common underlying mechanism in numerous pathologies, a better understanding the role of various PARPs in mitochondrial regulation may help open new therapeutic avenues.


Mitochondria/physiology , Poly(ADP-ribose) Polymerases/physiology , Animals , Humans , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Poly(ADP-ribose) Polymerases/genetics , Signal Transduction/genetics , Transcription, Genetic
11.
Biochim Biophys Acta ; 1842(4): 594-602, 2014 Apr.
Article En | MEDLINE | ID: mdl-24365238

Poly(ADP-ribose) polymerase-2 (PARP-2) is acknowledged as a DNA repair enzyme. However, recent investigations have attributed unique roles to PARP-2 in metabolic regulation in the liver. We assessed changes in hepatic lipid homeostasis upon the deletion of PARP-2 and found that cholesterol levels were higher in PARP-2(-/-) mice as compared to wild-type littermates. To uncover the molecular background, we analyzed changes in steady-state mRNA levels upon the knockdown of PARP-2 in HepG2 cells and in murine liver that revealed higher expression of sterol-regulatory element binding protein (SREBP)-1 dependent genes. We demonstrated that PARP-2 is a suppressor of the SREBP1 promoter, and the suppression of the SREBP1 gene depends on the enzymatic activation of PARP-2. Consequently, the knockdown of PARP-2 enhances SREBP1 expression that in turn induces the genes driven by SREBP1 culminating in higher hepatic cholesterol content. We did not detect hypercholesterolemia, higher fecal cholesterol content or increase in serum LDL, although serum HDL levels decreased in the PARP-2(-/-) mice. In cells and mice where PARP-2 was deleted we observed decreased ABCA1 mRNA and protein expression that is probably linked to lower HDL levels. In our current study we show that PARP-2 impacts on hepatic and systemic cholesterol homeostasis. Furthermore, the depletion of PARP-2 leads to lower HDL levels which represent a risk factor to cardiovascular diseases.


Cholesterol/metabolism , Lipoproteins, HDL/blood , Liver/metabolism , Poly(ADP-ribose) Polymerases/physiology , Animals , Hep G2 Cells , Humans , Male , Mice , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/physiology
12.
PLoS One ; 8(7): e69420, 2013.
Article En | MEDLINE | ID: mdl-23936011

Glycogen phosphorylase (GP) catalyzes the breakdown of glycogen and largely contributes to hepatic glucose production making GP inhibition an attractive target to modulate glucose levels in diabetes. Hereby we present the metabolic effects of a novel, potent, glucose-based GP inhibitor (KB228) tested in vitro and in vivo under normoglycemic and diabetic conditions. KB228 administration enhanced glucose sensitivity in chow-fed and obese, diabetic mice that was a result of higher hepatic glucose uptake. Besides improved glucose sensitivity, we have observed further unexpected metabolic rearrangements. KB228 administration increased oxygen consumption that was probably due to the overexpression of uncoupling protein-2 (UCP2) that was observed in animal and cellular models. Furthermore, KB228 treatment induced mammalian target of rapamycin complex 2 (mTORC2) in mice. Our data demonstrate that glucose based GP inhibitors are capable of reducing glucose levels in mice under normo and hyperglycemic conditions. Moreover, these GP inhibitors induce accommodation in addition to GP inhibition--such as enhanced mitochondrial oxidation and mTORC2 signaling--to cope with the glucose influx and increased glycogen deposition in the cells, however the molecular mechanism of accommodation is unexplored.


Diabetes Mellitus, Experimental/drug therapy , Enzyme Inhibitors/pharmacology , Glucose/pharmacology , Glycogen Phosphorylase/antagonists & inhibitors , Liver Glycogen/metabolism , Urea/pharmacology , Animals , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/physiopathology , Enzyme Inhibitors/chemical synthesis , Gene Expression/drug effects , Glucose/analogs & derivatives , Glucose/chemical synthesis , Glucose Tolerance Test , Glycogen Phosphorylase/metabolism , Ion Channels/agonists , Ion Channels/genetics , Ion Channels/metabolism , Liver/drug effects , Liver/metabolism , Liver Glycogen/antagonists & inhibitors , Male , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Obese , Mitochondrial Proteins/agonists , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Multiprotein Complexes/agonists , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Oxygen Consumption/drug effects , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Uncoupling Protein 2 , Urea/analogs & derivatives , Urea/chemical synthesis
13.
Cell Mol Life Sci ; 69(24): 4079-92, 2012 Dec.
Article En | MEDLINE | ID: mdl-22581363

Poly(ADP-ribose) polymerase (PARP)-2 is a nuclear enzyme that belongs to the PARP family and PARP-2 is responsible for 5-15 % of total cellular PARP activity. PARP-2 was originally described in connection to DNA repair and in physiological and pathophysiological processes associated with genome maintenance (e.g., centromere and telomere protection, spermiogenesis, thymopoiesis, azoospermia, and tumorigenesis). Recent reports have identified important rearrangements in gene expression upon the knockout of PARP-2. Such rearrangements heavily impact inflammation and metabolism. Metabolic effects are mediated through modifying PPARγ and SIRT1 function. Altered gene expression gives rise to a complex phenotype characterized primarily by enhanced mitochondrial activity that results both in beneficial (loss of fat, enhanced insulin sensitivity) and in disadvantageous (pancreatic beta cell hypofunction upon high fat feeding) consequences. Enhanced mitochondrial biogenesis provides protection in oxidative stress-related diseases. Hereby, we review the recent developments in PARP-2 research with special attention to the involvement of PARP-2 in transcriptional and metabolic regulation.


DNA Repair , Poly(ADP-ribose) Polymerases/physiology , Transcription, Genetic , Animals , Chromatin Assembly and Disassembly , Gene Expression Regulation , Genomic Instability , Humans , Mice , Models, Genetic , Oxidative Stress/genetics , Poly(ADP-ribose) Polymerases/chemistry , Poly(ADP-ribose) Polymerases/genetics , Protein Structure, Tertiary , Sirtuin 1/metabolism , Sirtuin 1/physiology , Spermatogenesis/genetics
...