Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Vasc Res ; 61(3): 109-121, 2024.
Article in English | MEDLINE | ID: mdl-38615660

ABSTRACT

INTRODUCTION: Following our recent finding that Ucp2 knockout promotes ferroptosis, we aimed to examine whether UCP2 alleviates myocardial ischemia/reperfusion injury (MI/RI) by inhibiting ferroptosis. METHODS: The left anterior descending coronary arteries of wild-type and Ucp2-/- C57BL/6 mice were ligated for 30 min and reperfused for 2 h to establish an MI/RI model. The effects of UCP2 on ferroptosis and MI/RI were determined by echocardiography, 2,3,5-triphenylttrazolium chloride staining, hematoxylin-eosin staining, Masson's trichrome staining, Sirius red staining, and analysis of myocardial injury markers and ferroptosis indicators. Ferrostatin-1 (Fer-1) and erastin (Era) were used to investigate whether UCP2 alleviated MI/RI by inhibiting ferroptosis and the molecular mechanism. RESULTS: UCP2 was upregulated in the MI/RI model in WT mice. Deletion of Ucp2 exacerbated ferroptosis, altered the expression levels of multiple ferroptosis-related genes, and significantly exacerbated MI/RI. Knockout of Ucp2 promoted ferroptosis induced by Era and inhibited the antiferroptotic effects of Fer-1. Knockout of Ucp2 activated the p53/TfR1 pathway to exacerbate ferroptosis. CONCLUSION: Our results showed that UCP2 inhibited ferroptosis in MI/RI, which might be related to regulation of the p53/TfR1 pathway.


Subject(s)
Disease Models, Animal , Ferroptosis , Mice, Inbred C57BL , Mice, Knockout , Myocardial Reperfusion Injury , Myocytes, Cardiac , Uncoupling Protein 2 , Animals , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/genetics , Uncoupling Protein 2/metabolism , Uncoupling Protein 2/genetics , Uncoupling Protein 2/deficiency , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Male , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Mice
2.
Sci Rep ; 11(1): 15449, 2021 07 29.
Article in English | MEDLINE | ID: mdl-34326395

ABSTRACT

Endoplasmic reticulum (ER) stress and uncoupling protein-2 (UCP2) activation are opposing modulators of endothelial dysfunction in atherosclerosis. Exercise reduces atherosclerosis plaques and enhances endothelial function. Our aim was to understand how exercise affects ER stress and UCP2 activation, and how that relates to endothelial dysfunction in an atherosclerotic murine model. Wild type (C57BL/6, WT) and apolipoprotein-E-knockout (ApoEtm1Unc, ApoE KO) mice underwent treadmill exercise training (EX) or remained sedentary for 12 weeks. Acetylcholine (ACh)-induced endothelium-dependent vasodilation was determined in the presence of an eNOS inhibitor (L-NAME), UCP2 inhibitor (genipin), and ER stress inducer (tunicamycin). UCP2, ER stress markers and NLRP3 inflammasome signaling were quantified by western blotting. p67phox and superoxide were visualized using immunofluorescence and DHE staining. Nitric oxide (NO) was measured by nitrate/nitrite assay. ACh-induced vasodilation was attenuated in coronary arterioles of ApoE KO mice but improved in ApoE KO-EX mice. Treatment of coronary arterioles with L-NAME, tunicamycin, and genipin significantly attenuated ACh-induced vasodilation in all mice except for ApoE KO mice. Exercise reduced expression of ER stress proteins, TXNIP/NLRP3 inflammasome signaling cascades, and Bax expression in the heart of ApoE KO-EX mice. Further, exercise diminished superoxide production and NADPH oxidase p67phox expression in coronary arterioles while simultaneously increasing UCP2 expression and nitric oxide (NO) production in the heart of ApoE KO-EX mice. Routine exercise alleviates endothelial dysfunction in atherosclerotic coronary arterioles in an eNOS, UCP2, and ER stress signaling specific manner, and resulting in reduced TXNIP/NLRP3 inflammasome activity and oxidative stress.


Subject(s)
Atherosclerosis/metabolism , Atherosclerosis/therapy , Coronary Vessels/metabolism , Endoplasmic Reticulum Stress , Exercise Therapy/methods , Physical Conditioning, Animal/methods , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/therapy , Uncoupling Protein 2/deficiency , Acetylcholine/pharmacology , Animals , Arterioles/drug effects , Arterioles/metabolism , Arterioles/physiopathology , Atherosclerosis/genetics , Disease Models, Animal , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Exercise Test , Iridoids/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Oxidative Stress/drug effects , Oxidative Stress/genetics , Plaque, Atherosclerotic/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Uncoupling Protein 2/antagonists & inhibitors , Vasodilation/drug effects , Vasodilation/genetics
3.
Neurochem Res ; 46(6): 1359-1371, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33735403

ABSTRACT

Mitochondrial uncoupling protein 2 (UCP2) deficiency exacerbates brain damage following cerebral ischemia/reperfusion (I/R). The Nod-like receptor protein-3 (NLRP3) inflammasome also plays a vital role in cerebral I/R damage. However, the effect of UCP2 on NLRP3 inflammasome-mediated hyperglycemia and I/R damage is not clear. In the present study, UCP2-knockout (UCP2-/-) and wild-type (WT) mice were used to establish a model of middle cerebral artery occlusion (MCAO) and reperfusion under normo- and hyperglycemic conditions. HT22 cells were established as a model of oxygen-glucose deprivation and reoxygenation (OGD/R) with high glucose to mimic hyperglycemia and I/R in vitro. HT22 cells were treated with/without different concentrations of the UCP2-specific inhibitor genipin for different periods of time. The results showed that UCP2 deficiency significantly increased histopathological changes and apoptosis after cerebral I/R damage in hyperglycemic mice. Moreover, UCP2 deficiency enhanced NLRP3 inflammasome activation in neurons when cerebral I/R damage was exacerbated by hyperglycemia. Furthermore, UCP2 deficiency enhanced NLRP3 inflammasome activation and reactive oxygen species (ROS) production in HT22 cells under OGD/R and high-glucose conditions. UCP2 deficiency aggravated hyperglycemia-induced exacerbation of cerebral I/R damage. UCP2 deficiency also enhanced NLRP3 inflammasome activation and ROS production in neurons in vitro and in vivo. These findings suggest that UCP2 deficiency enhances NLRP3 inflammasome activation following hyperglycemia-induced exacerbation of cerebral I/R damage in vitro and in vivo. UCP2 may be a potential therapeutic target for hyperglycemia-induced exacerbation of cerebral I/R damage.


Subject(s)
Hyperglycemia/metabolism , Infarction, Middle Cerebral Artery/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Reperfusion Injury/metabolism , Uncoupling Protein 2/deficiency , Animals , Apoptosis/physiology , Brain/pathology , Cell Line , Female , Glucose/deficiency , Glucose/pharmacology , Hyperglycemia/pathology , Hypoxia/physiopathology , Infarction, Middle Cerebral Artery/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Reactive Oxygen Species/metabolism , Reperfusion Injury/pathology
4.
Int J Med Sci ; 17(17): 2869-2878, 2020.
Article in English | MEDLINE | ID: mdl-33162815

ABSTRACT

Objective: Uncoupling protein 2 (UCP2) is a member of inner mitochondrial membrane proteins and deletion of UCP2 exacerbates brain damage after cerebral ischemia/reperfusion (I/R). Nevertheless, its functional role during cerebral I/R is not entirely understood. The objective of present study was to explore the influence of UCP2 deletion on mitochondrial autophagy (mitophagy) and mitochondria-mediated cell death pathway after cerebral I/R. Methods: UCP2-/- and wildtype (WT) mice were subjected to 60 min middle cerebral artery occlusion (MCAO) and allowed reperfusion for 24 hours. Infarct volume and histological outcomes were assessed, reactive oxygen species (ROS) and autophagy markers were measured, and mitochondrial ultrastructure was examined. Results: Deletion of UCP2 enlarged infarct volume, increased numbers of necrotic and TUNEL positive cells, and significantly increased pro-apoptotic protein levels in UCP2-/- mice compared with WT mice subjected to the same duration of I/R. Further, deletion of UCP2 increased ROS production, elevated LC3, Beclin1 and PINK1, while it suppressed p62 compared with respective WT ischemic controls. Electron microscopic study demonstrated the number of autophagosomes was higher in the UCP2-/- group, compared with the WT group. Conclusions: It is concluded that deletion of UCP2 exacerbates cerebral I/R injury via reinforcing mitophagy and cellular apoptosis in mice.


Subject(s)
Brain Ischemia/complications , Brain/pathology , Infarction, Middle Cerebral Artery/complications , Reperfusion Injury/pathology , Uncoupling Protein 2/deficiency , Animals , Apoptosis , Autophagosomes/pathology , Brain/blood supply , Brain/cytology , Brain Ischemia/pathology , Disease Models, Animal , Female , Humans , Infarction, Middle Cerebral Artery/pathology , Male , Mice , Mice, Knockout , Mitochondria/pathology , Mitophagy , Oxidative Stress , Reactive Oxygen Species/metabolism , Reperfusion Injury/etiology , Uncoupling Protein 2/genetics
5.
Cell Death Dis ; 11(1): 26, 2020 01 13.
Article in English | MEDLINE | ID: mdl-31932578

ABSTRACT

Mitochondrial dysfunction leads to loss of renal function and structure; however, the precise mechanisms by which mitochondrial function can regulate renal fibrosis remain unclear. Proximal tubular cells (PTCs) prefer fatty acid oxidation as their energy source and dysregulation of lipid metabolism has been linked to tubulointerstitial fibrosis (TIF). Here, we demonstrated that mitochondrial uncoupling protein 2 (UCP2) regulates TIF through the stimulation of lipid deposition and extracellular matrix (ECM) accumulation. We show that UCP2 expression was increased in human biopsy sample and mouse kidney tissues with TIF. Moreover, UCP2-deficient mice displayed mitigated renal fibrosis in I/R-induced mouse model of TIF. Consistent with these results, UCP2 deficiency displayed reduced lipid deposition and ECM accumulation in vivo and in vitro. In UCP2-deficient PTCs, inhibition of TIF resulted from downregulation of hypoxia-inducible factor-1α (HIF-1α), a key regulator of lipid metabolism and ECM accumulation. Furthermore, we describe a molecular mechanism by which UCP2 regulates HIF-1α stabilization through regulation of mitochondrial respiration and tissue hypoxia during TIF. HIF-1α inhibition by siRNA suppressed lipid and ECM accumulation by restoration of PPARα and CPT1α, as well as suppression of fibronectin and collagen I expression in PTCs. In conclusion, our results suggest that UCP2 regulates TIF by inducing the HIF-1α stabilization pathway in tubular cells. These results identify UCP2 as a potential therapeutic target in treating chronic renal fibrosis.


Subject(s)
Hypoxia/metabolism , Ischemia/metabolism , Kidney Diseases/metabolism , Kidney/injuries , Lipid Metabolism , Uncoupling Protein 2/metabolism , Animals , Cell Respiration , Cells, Cultured , Extracellular Matrix/metabolism , Fibrosis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney/pathology , Kidney Diseases/pathology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Mice, Inbred C57BL , Mitochondria/metabolism , Oxygen/metabolism , Protective Agents/metabolism , Reperfusion Injury/pathology , Uncoupling Protein 2/deficiency
6.
Cardiovasc Res ; 115(7): 1217-1227, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30850841

ABSTRACT

AIMS: The role of uncoupling protein 2 (UCP2) in cardiac adaptation to pressure overload remains unclear. In a classical model of left ventricular pressure overload genetic deletion of UCP2 (UCP2-/-) protected against cardiac hypertrophy and failure. However, in UCP2-/- mice increased proliferation of pulmonary arterial smooth muscle cells induces mild pulmonary hypertension, right ventricular (RV) hypertrophy, and reduced cardiac output. This suggests a different role for UCP2 in RV and left ventricular adaptation to pressure overload. To clarify this situation in more detail UCP2-/- and wild-type mice were exposed to pulmonary arterial banding (PAB). METHODS AND RESULTS: Mice were analysed (haemodynamics, morphometry, and echocardiography) 3 weeks after PAB or sham surgery. Myocytes and non-myocytes were isolated and analysed separately. Cell shortening of myocytes and fura-2 loading of cardiomyocytes were used to characterize their function. Brd assay was performed to study fibroblast proliferation. Isolated mitochondria were analysed to investigate the role of UCP2 for reactive oxygen species (ROS) production. UCP2 mRNA was 2.7-fold stronger expressed in RV myocytes than in left ventricular myocytes and stronger expressed in non-myocytes compared with myocytes. Three weeks after PAB, cardiac output was reduced in wild type but preserved in UCP2-/- mice. UCP2-/- had increased RV wall thickness, but lower RV internal diameters and displayed a significant stronger fibrosis. Cardiac fibroblasts from UCP2-/- had reduced proliferation rates but higher collagen-1 expression. Myocytes isolated from mice after PAB banding showed preserved function that was further improved by UCP2-/-. Mitochondrial ROS production and respiration was similar between UCP2-/- or wild-type hearts. CONCLUSION: Despite a mild pulmonary hypertension in UCP2-/- mice, hearts from these mice are well preserved against additional pressure overload (severe pulmonary hypertension). This-at least in part-depends on different behaviour of non-myocytes (fibroblasts).


Subject(s)
Fibroblasts/metabolism , Gene Silencing , Heart Failure/prevention & control , Hypertension, Pulmonary/prevention & control , Myocytes, Cardiac/metabolism , Uncoupling Protein 2/deficiency , Ventricular Function, Right , Ventricular Remodeling , Animals , Cells, Cultured , Collagen Type I/metabolism , Disease Models, Animal , Fibroblasts/pathology , Fibrosis , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/physiopathology , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/genetics , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myocytes, Cardiac/pathology , Reactive Oxygen Species/metabolism , Uncoupling Protein 2/genetics , Ventricular Function, Left
7.
J Pathol ; 247(3): 392-405, 2019 03.
Article in English | MEDLINE | ID: mdl-30426490

ABSTRACT

Acute kidney injury (AKI) is a public health concern, with high morbidity and mortality rates in hospitalized patients and because survivors have an increased risk of progression to chronic kidney disease. Mitochondrial damage is the critical driver of AKI-associated dysfunction and loss of tubular epithelial cells; however, the pathways that mediate these events are poorly defined. Here, in murine ischemia/reperfusion (I/R)-induced AKI, we determined that mitochondrial damage is associated with the level of renal uncoupling protein 2 (UCP2). In hypoxia-damaged proximal tubular cells, a disruption of mitochondrial dynamics demonstrated by mitochondrial fragmentation and disturbance between fusion and fission was clearly indicated. Ucp2-deficient mice (knockout mice) with I/R injury experienced more severe AKI and mitochondrial fragmentation than wild-type mice. Moreover, genetic or pharmacological treatment increased UCP2 expression, improved renal function, reduced tubular injury and limited mitochondrial fission. In cultured proximal tubular epithelial cells, hypoxia-induced mitochondrial fission was exacerbated in cells with UCP2 deletion, whereas an increase in UCP2 ameliorated the hypoxia-induced disturbance of the balance between mitochondrial fusion and fission. Furthermore, results following modulation of UCP2 suggested it has a role in preserving mitochondrial integrity by preventing loss of membrane potential and reducing subsequent mitophagy. Taken together, our results indicate that UCP2 is protective against AKI and suggest that enhancing UCP2 to improve mitochondrial dynamics has potential as a strategy for improving outcomes of renal injury. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Acute Kidney Injury/pathology , Mitochondria/physiology , Uncoupling Protein 2/physiology , Acute Kidney Injury/etiology , Acute Kidney Injury/metabolism , Acute Kidney Injury/prevention & control , Animals , Cell Hypoxia/physiology , Cells, Cultured , Kidney Tubules, Proximal/ultrastructure , Male , Membrane Potential, Mitochondrial/physiology , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron , Mitochondrial Dynamics/physiology , Mitophagy/physiology , Protein Kinases/physiology , Reperfusion Injury/complications , Uncoupling Protein 2/deficiency , Uncoupling Protein 2/genetics , Uncoupling Protein 2/metabolism , Up-Regulation/physiology
8.
Acta Physiol (Oxf) ; 223(4): e13058, 2018 08.
Article in English | MEDLINE | ID: mdl-29480974

ABSTRACT

AIM: Uncoupling protein-2 (UCP-2) can induce mitochondrial uncoupling in the diabetic kidney. Although mitochondrial uncoupling reduces oxidative stress originating from the mitochondria and can be regarded as a protective mechanism, the increased oxygen consumption occurring secondarily to increased mitochondria uncoupling, that is leak respiration, may contribute to kidney tissue hypoxia. Using UCP-2-/- mice, we tested the hypothesis that UCP-2-mediated leak respiration is important for the development of diabetes-induced intrarenal hypoxia and proteinuria. METHODS: Kidney function, in vivo oxygen metabolism, urinary protein leakage and mitochondrial function were determined in wild-type and UCP-2-/- mice during normoglycaemia and 2 weeks after diabetes induction. RESULTS: Diabetic wild-type mice displayed mitochondrial leak respiration, pronounced intrarenal hypoxia, proteinuria and increased urinary KIM-1 excretion. However, diabetic UCP-2-/- mice did not develop increased mitochondrial leak respiration and presented with normal intrarenal oxygen levels, urinary protein and KIM-1 excretion. CONCLUSION: Although functioning as an antioxidant system, mitochondria uncoupling is always in co-occurrence with increased oxygen consumption, that is leak respiration; a potentially detrimental side effect as it can result in kidney tissue hypoxia; an acknowledged unifying pathway to nephropathy. Indeed, this study demonstrates a novel mechanism in which UCP-2-mediated mitochondrial leak respiration is necessary for the development of diabetes-induced intrarenal tissue hypoxia and proteinuria.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 1/complications , Diabetic Nephropathies/prevention & control , Kidney/metabolism , Mitochondria/metabolism , Oxygen/metabolism , Proteinuria/prevention & control , Uncoupling Protein 2/deficiency , Animals , Cell Hypoxia , Cell Respiration , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Diabetic Nephropathies/etiology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Gene Deletion , Genetic Predisposition to Disease , Hepatitis A Virus Cellular Receptor 1/metabolism , Mice, Knockout , Oxidative Stress , Oxygen Consumption , Phenotype , Proteinuria/etiology , Proteinuria/genetics , Proteinuria/metabolism , Uncoupling Protein 2/genetics
9.
Mitochondrion ; 42: 50-53, 2018 09.
Article in English | MEDLINE | ID: mdl-29154852

ABSTRACT

Polycystic liver diseases (PCLDs) are autosomal dominant disorders. To date, 3 genes are known to be associated with the disease, SEC63 and PRKCSH and LRP5. Here, we report that mice deficient in the mitochondrial uncoupling protein 2 gene (Ucp2-/-) spontaneously developed PCLDs when they were over 12months old. Macroscopical observation, blood chemistry as well as histopathological analysis demonstrated the PCLDs found in Ucp2-/- mice were very similar to the findings in human PCLDs. This is the first report describing the gene encoding mitochondrial protein is causative for PCLDs. UCP2 may be a biomarker of the PCLDs in humans.


Subject(s)
Cysts/genetics , Liver Diseases/genetics , Uncoupling Protein 2/deficiency , Animals , Blood Chemical Analysis , Disease Models, Animal , Female , Histocytochemistry , Liver/pathology , Male , Mice, Inbred C57BL
10.
Sci Transl Med ; 9(418)2017 Nov 29.
Article in English | MEDLINE | ID: mdl-29187642

ABSTRACT

Limb remote ischemic preconditioning (RIPC) is an effective means of protection against ischemia/reperfusion (IR)-induced injury to multiple organs. Many studies are focused on identifying endocrine mechanisms that underlie the cross-talk between muscle and RIPC-mediated organ protection. We report that RIPC releases irisin, a myokine derived from the extracellular portion of fibronectin domain-containing 5 protein (FNDC5) in skeletal muscle, to protect against injury to the lung. Human patients with neonatal respiratory distress syndrome show reduced concentrations of irisin in the serum and increased irisin concentrations in the bronchoalveolar lavage fluid, suggesting transfer of irisin from circulation to the lung under physiologic stress. In mice, application of brief periods of ischemia preconditioning stimulates release of irisin into circulation and transfer of irisin to the lung subjected to IR injury. Irisin, via lipid raft-mediated endocytosis, enters alveolar cells and targets mitochondria. Interaction between irisin and mitochondrial uncoupling protein 2 (UCP2) allows for prevention of IR-induced oxidative stress and preservation of mitochondrial function. Animal model studies show that intravenous administration of exogenous irisin protects against IR-induced injury to the lung via improvement of mitochondrial function, whereas in UCP2-deficient mice or in the presence of a UCP2 inhibitor, the protective effect of irisin is compromised. These results demonstrate that irisin is a myokine that facilitates RIPC-mediated lung protection. Targeting the action of irisin in mitochondria presents a potential therapeutic intervention for pulmonary IR injury.


Subject(s)
Fibronectins/blood , Fibronectins/metabolism , Mitochondria/metabolism , Reperfusion Injury/blood , Reperfusion Injury/metabolism , Animals , Animals, Newborn , Female , Humans , Male , Mice , Oxidative Stress/physiology , Uncoupling Protein 2/antagonists & inhibitors , Uncoupling Protein 2/deficiency , Uncoupling Protein 2/metabolism
11.
J Am Heart Assoc ; 6(10)2017 Oct 12.
Article in English | MEDLINE | ID: mdl-29025747

ABSTRACT

BACKGROUND: Intracoronary stent restenosis, characterized by excessive smooth muscle cell (SMC) proliferation and myointimal hyperplasia, remains a clinical challenge. Mitochondrial membrane potential has been linked to the proliferative rate of SMCs. This study aimed to screen a critical gene regulating mitochondrial potential and to confirm its effects on myointimal formation in preclinical animal models. METHODS AND RESULTS: We performed transcriptome screening for genes differentially expressed in ligated versus unligated mouse carotid arteries. We observed that uncoupling protein 2 gene (Ucp2) mRNA, encoding UCP2, was transiently upregulated during the first 3 days after ligation and then significantly downregulated from day 7 through day 21, during which time neointima formed remarkably. The UCP2 protein level also declined after day 7 of ligation. In ligated carotid arteries, Ucp2-/- mice, compared with wild-type littermates, exhibited accelerated myointimal formation, which was associated with increased superoxide production and can be attenuated by treatment with antioxidant 4-hydroxy-2,2,6,6-tetramethyl-piperidinoxyl (TEMPOL). Knockdown of UCP2 enhanced human aortic SMC migration and proliferation that can also be attenuated by TEMPOL, whereas UCP2 overexpression inhibited SMC migration and proliferation, along with decreased activity of nuclear factor-κB. Moreover, nuclear factor-κB inhibitor attenuated UCP2 knockdown-enhanced SMC proliferation. Adenovirus-mediated overexpression of UCP2 inhibited myointimal formation in balloon-injured carotid arteries of rats and rabbits and in-stent stenosis of porcine coronary arteries. Moreover, UCP2 overexpression also suppressed neointimal hyperplasia in cultured human saphenous vein ex vivo. CONCLUSIONS: UCP2 inhibits myointimal hyperplasia after vascular injury, probably through suppressing nuclear factor-κB-dependent SMC proliferation and migration, rendering UCP2 a potential therapeutic target against restenosis.


Subject(s)
Carotid Artery Injuries/prevention & control , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima , Uncoupling Protein 2/metabolism , Animals , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Injuries/genetics , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Cell Movement , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Genetic Predisposition to Disease , Humans , Hyperplasia , Male , Membrane Potential, Mitochondrial , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/pathology , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , NF-kappa B/metabolism , Phenotype , Rabbits , Rats, Sprague-Dawley , Swine , Swine, Miniature , Time Factors , Tissue Culture Techniques , Uncoupling Protein 2/deficiency , Uncoupling Protein 2/genetics
12.
PLoS One ; 12(7): e0179743, 2017.
Article in English | MEDLINE | ID: mdl-28683125

ABSTRACT

UCP-2 shows an important role in modulating of mitochondrial membrane potential and cell apoptosis. Whether or not UCP-2 could been a critical factor in preventing AAA formation is not known. We report that UCP-2 protein and mRNA expression were significantly higher in Ang-Ⅱ-induced AAA of mice. The incident rate of AAA in UCP-2-/-ApoE-/- mice after Ang-Ⅱtreatment was higher than the rate in the UCP-2+/+ApoE-/- mice. The abdominal aorta from UCP-2-/-ApoE-/- mice showed the medial hypertrophy, fragmentation of elastic lamellas and depletion of α-SMA. The NADPH oxidase activity and level of MDA was significantly higher in UCP-2-/-ApoE-/- mice than UCP-2+/+ApoE-/- or WT mice. Besides, the SOD activity is increased in UCP-2+/+ApoE-/- mice as compared with WT mice, whereas deficiency of UCP-2 decreased the increasing SOD activity in Ang-Ⅱ treated ApoE-/- mice. UCP-2 knockout up-regulated the MMP2 and MMP9 expression in aortic aneurysm. Ang-Ⅱ induced apoptosis of VSMCs was increased in UCP-2-/-ApoE-/- mice. And the expression of eNOS in vascular tissue from UCP-2-/-ApoE-/- mice is lower than WT and UCP-2+/+ApoE-/- mice. This study provides a mechanism by which UCP-2, via anti-oxidants and anti-apoptosis, participates in the preventing of AAA formation.


Subject(s)
Angiotensin II/pharmacology , Aorta, Abdominal/drug effects , Aortic Aneurysm, Abdominal/genetics , Apolipoproteins E/genetics , Uncoupling Protein 2/genetics , Actins/genetics , Actins/metabolism , Animals , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Apolipoproteins E/deficiency , Apoptosis/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Expression Regulation , Malondialdehyde/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Uncoupling Protein 2/deficiency
13.
Mitochondrion ; 34: 135-140, 2017 05.
Article in English | MEDLINE | ID: mdl-28351676

ABSTRACT

Mitochondria are fascinating organelles involved in various cellular-metabolic activities that are integral for mammalian development. Although they perform diverse, yet interconnected functions, mitochondria are remarkably regulated by complex signaling networks. Therefore, it is not surprising that mitochondrial dysfunction is involved in plethora of diseases, including neurodegenerative and metabolic disorders. One of the many factors that lead to mitochondrial-associated metabolic diseases is the uncoupling protein-2, a family of mitochondrial anion proteins present in the inner mitochondrial membrane. Since their discovery, uncoupling proteins have attracted considerable attention due to their involvement in mitochondrial-mediated oxidative stress and energy metabolism. This review attempts to provide a summary of recent developments in the field of uncoupling protein 2 relating to mitochondrial associated metabolic diseases.


Subject(s)
Metabolic Diseases/physiopathology , Uncoupling Protein 2/deficiency , Uncoupling Protein 2/metabolism , Animals , Energy Metabolism , Humans
14.
Biochimie ; 134: 118-126, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28130074

ABSTRACT

Uncoupling protein 2 (UCP2) was discovered in 1997 and classified as an uncoupling protein largely based on its homology of sequence with UCP1. Since its discovery, the uncoupling function of UCP2 has been questioned and there is yet no consensus on the true function of this protein. UCP2 was first proposed to be a reactive oxygen species (ROS) regulator and an insulin secretion modulator. More recently, it was demonstrated as a regulator of the mitochondrial fatty acid oxidation, which prompted us to investigate its role in the metabolic and thermogenic functions of brown adipose tissue. We first investigated the role of UCP2 in affecting the glycolysis capacity by evaluating the extracellular flux in cells lacking UCP2. We thereafter investigated the role of UCP2 in BAT thermogenesis with positron emission tomography using the metabolic tracers [11C]-acetate (metabolic activity), 2-deoxy-2-[18F]-fluoro-d-glucose (18FDG, glucose uptake) and 14(R,S)-[18F]fluoro-6-thia-heptadecanoic acid [18FTHA, non-esterified fatty acid (NEFA) uptake]. The effect of the ß3-adrenoreceptor (ADRB3) selective agonist, CL316,243 (CL), on BAT 18FDG and 18FTHA uptakes, as well as 11C-acetate activity was assessed in UCP2KO and UCP2WT mice exposed at room temperature or adapted to cold. Our results suggest that despite the fact that UCP2 does not have the uncoupling potential of UCP1, its contribution to BAT thermogenesis and to the adaptation to cold exposure appears crucial. Notably, we found that the absence of UCP2 promoted a shift toward glucose utilization and increased glycolytic capacity in BAT, which conferred a better oxidative/thermogenic activity/capacity following an acute adrenergic stimulation. However, following cold exposure, a context of high-energy demand, BAT of UCP2KO mice failed to adapt and thermogenesis was impaired. We conclude that UCP2 regulates BAT thermogenesis by favouring the utilization of NEFA, a process required for the adaptation to cold.


Subject(s)
Adipose Tissue, Brown/metabolism , Glucose/metabolism , Glycolysis/genetics , Mitochondria/metabolism , Thermogenesis/genetics , Uncoupling Protein 2/genetics , Acetic Acid/metabolism , Adipose Tissue, Brown/drug effects , Adrenergic beta-3 Receptor Agonists/pharmacology , Animals , Cold Temperature , Dioxoles/pharmacology , Fatty Acids/metabolism , Fluorodeoxyglucose F18/metabolism , Gene Expression Regulation , Glucose/pharmacology , Glycolysis/drug effects , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Positron-Emission Tomography , Radiopharmaceuticals/metabolism , Receptors, Adrenergic, beta-3/genetics , Receptors, Adrenergic, beta-3/metabolism , Signal Transduction , Thermogenesis/drug effects , Uncoupling Protein 2/deficiency
15.
Neurochem Res ; 42(1): 108-114, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27401256

ABSTRACT

The mitochondrial aspartate/glutamate transporter Aralar/AGC1/Slc25a12 is critically involved in brain aspartate synthesis, and AGC1 deficiency results in a drastic fall of brain aspartate levels in humans and mice. It has recently been described that the uncoupling protein UCP2 transports four carbon metabolites including aspartate. Since UCP2 is expressed in several brain cell types and AGC1 is mainly neuronal, we set to test whether UCP2 could be a mitochondrial aspartate carrier in the brain glial compartment. The study of the cerebral metabolism of (1-13C)-glucose in vivo in wild type and UCP2-knockout mice showed no differences in C3 or C2 labeling of aspartate, suggesting that UCP2 does not function as a mitochondrial aspartate carrier in brain. However, surprisingly, a clear decrease (of about 30-35 %) in the fractional enrichment of glutamate, glutamine and GABA was observed in the brains of UCP2-KO mice which was not associated with differences in either glucose or lactate enrichments. The results suggest that the dilution in the labeling of glutamate and its downstream metabolites could originate from the uptake of an unlabeled substrate that could not leave the matrix via UCP2 becoming trapped in the matrix. Understanding the nature of the unlabeled substrate and its precursor(s) as alternative substrates to glucose is of interest in the context of neurological diseases associated with UCP2.


Subject(s)
Cerebral Cortex/metabolism , Glucose/metabolism , Uncoupling Protein 2/physiology , Animals , Carbon Isotopes/metabolism , Glutamic Acid/metabolism , Glutamine/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Uncoupling Protein 2/deficiency , gamma-Aminobutyric Acid/metabolism
16.
Hepatobiliary Pancreat Dis Int ; 15(6): 647-654, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27919855

ABSTRACT

BACKGROUND: Uncoupling protein 2 (UCP2) has been suggested to inhibit mitochondrial production of reactive oxygen species (ROS) by decreasing the mitochondrial membrane potential. Experimental acute pancreatitis is associated with increased UCP2 expression, whereas UCP2 deficiency retards regeneration of aged mice from acute pancreatitis. Here, we have addressed biological and molecular functions of UCP2 in pancreatic stellate cells (PSCs), which are involved in pancreatic wound repair and fibrogenesis. METHODS: PSCs were isolated from 12 months old (aged) UCP2-/- mice and animals of the wild-type (WT) strain C57BL/6. Proliferation and cell death were assessed by employing trypan blue staining and a 5-bromo-2'-deoxyuridine incorporation assay. Intracellular fat droplets were visualized by oil red O staining. Levels of mRNA were determined by RT-PCR, while protein expression was analyzed by immunoblotting and immunofluorescence analysis. Intracellular ROS levels were measured with 2', 7'-dichlorofluorescin diacetate. Expression of senescence-associated beta-galactosidase (SA beta-Gal) was used as a surrogate marker of cellular senescence. RESULTS: PSCs derived from UCP2-/- mice proliferated at a lower rate than cells from WT mice. In agreement with this observation, the UCP2 inhibitor genipin displayed dose-dependent inhibitory effects on WT PSC growth. Interestingly, ROS levels in PSCs did not differ between the two strains, and PSCs derived from UCP2-/- mice did not senesce faster than those from corresponding WT cells. PSCs from UCP2-/- mice and WT animals were also indistinguishable with respect to the activation-dependent loss of intracellular fat droplets, expression of the activation marker alpha-smooth muscle actin, type I collagen and the autocrine/paracrine mediators interleukin-6 and transforming growth factor-beta1. CONCLUSIONS: A reduced proliferative capacity of PSC from aged UCP2-/- mice may contribute to the retarded regeneration after acute pancreatitis. Apart from their slower growth, PSC of UCP2-/- mice displayed no functional abnormalities. The antifibrotic potential of UCP2 inhibitors deserves further attention.


Subject(s)
Cell Proliferation , Pancreatic Stellate Cells/metabolism , Uncoupling Protein 2/metabolism , Animals , Biomarkers/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Gene Expression Profiling , Gene Expression Regulation , Genotype , Iridoids/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/pathology , Phenotype , Reactive Oxygen Species/metabolism , Signal Transduction , Uncoupling Protein 2/antagonists & inhibitors , Uncoupling Protein 2/deficiency , Uncoupling Protein 2/genetics
17.
Exp Hematol ; 44(11): 1085-1091.e2, 2016 11.
Article in English | MEDLINE | ID: mdl-27544660

ABSTRACT

Progress of age-related hematopoietic diseases such as myelodysplastic syndrome has previously been linked to enhanced levels of reactive oxygen species (ROS). Uncoupling protein 2 (UCP2) was found to reduce mitochondrial ROS production through uncoupling of the respiratory chain. The impact of UCP2 loss and elevated ROS on hematopoiesis during aging has not yet been investigated. In this study, UCP2 knockout mice were analyzed at aging stages of 3, 12, and 24 months with respect to oxidative and energy status of bone marrow cells. Further, the cellular bone marrow subpopulation composition was characterized, as were the differential blood counts at all time points. UCP2 knockout mice revealed enhanced levels of mitochondrial superoxide in elderly animals. Following oxidative stress, adenosine triphosphate (ATP) levels decreased more in the knockout mice than in the wild type. Investigation of bone marrow and blood counts of the knockout mice revealed an enhanced amount of monocytes and neutrophils, as well as a decreased amount of B cells and impaired erythropoiesis throughout aging. In summary, UCP2 induces protective effects on ROS and ATP levels during aging. Additionally, the results suggest an imbalance in hematopoiesis because of the lack of UCP2.


Subject(s)
Aging/blood , Aging/genetics , B-Lymphocytes , Leukocyte Count , Neutrophils , Uncoupling Protein 2/deficiency , Adenosine Triphosphate/metabolism , Aging/metabolism , Animals , B-Lymphocytes/metabolism , Biomarkers , Bone Marrow Cells/metabolism , Erythropoiesis/genetics , Immunophenotyping , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Neutrophils/metabolism , Oxidative Stress , Phenotype
18.
Mitochondrion ; 30: 42-50, 2016 09.
Article in English | MEDLINE | ID: mdl-27364833

ABSTRACT

Uncoupling protein (UCP) 2 is a mitochondrial transporter protein that plays various roles in cellular metabolism, including the glucose and lipid metabolism. Polymorphisms in UCP2 are associated with longevity in humans. In line with this, mice carrying the UCP2 transgene under the control of hypocretin promoter were reported to have an extended lifespan, while, conversely, mice deficient in Ucp2 demonstrated a significantly shorter lifespan. In this study, we examined the phenotype of aging in a large colony of Ucp2-deficient (Ucp2(-/-)) mice on the molecular level. We have found that the significantly shorter lives of Ucp2(-/-) mice is the result of an accelerated aging process throughout their entire lifespan. Thus, Ucp2(-/-) mice not only earlier gained sexual maturity, but also earlier progressed into an aging phenotype, reflected by a decrease in body weight, increased neutrophil numbers, and earlier emergence of spontaneous ulcerative dermatitis. Intriguingly, on the molecular level this acceleration in aging predominantly driven by increased levels of circulating IGF-1 in Ucp2(-/-) mice, hinting at a crosstalk between UCP2 and the classical Insulin/IGF-1 signaling aging pathway.


Subject(s)
Aging , Uncoupling Protein 2/metabolism , Animals , Cross-Sectional Studies , Female , Insulin-Like Growth Factor I/analysis , Longitudinal Studies , Male , Mice , Mice, Knockout , Serum/chemistry , Uncoupling Protein 2/deficiency
19.
Nucl Med Biol ; 43(10): 587-92, 2016 10.
Article in English | MEDLINE | ID: mdl-27451963

ABSTRACT

INTRODUCTION: Compounds that modulate cancer cell glucose metabolism could open new opportunities for antitumor therapy and for monitoring response using (18)F-FDG PET. Genipin, a natural dietary compound that blocks uncoupling protein 2 (UCP2)-mediated mitochondrial proton leakage, is a potential anticancer agent. We investigated the effect of genipin on glucose metabolism and the mitochondrial function of cancer cells. METHODS: Breast and colon cancer cells were assessed for effects of genipin on (18)F-FDG uptake. T47D breast cancer cells were further evaluated for time-dependent and dose-dependent effects on (18)F-FDG uptake, lactate release, oxygen consumption rate (OCR), reactive oxygen species (ROS) production, and mitochondrial membrane potential. The effects of UCP2 knockdown were evaluated using specific siRNA. RESULTS: Cancer cells displayed significant reductions in (18)F-FDG uptake by genipin. T47D cells showed the greatest reduction to 32.6±1.0% of controls by 250µM genipin. The effect occurred rapidly, reaching a plateau by 1h that lasted up to 24h. The effect was dose-dependent with a half-inhibitory concentration of 60.8µM. An accompanying decrease in lactate release was consistent with reduced glycolytic flux. OCR was significantly decreased by genipin to 82.2±11.4% of controls, and ROS generation was increased to 156.7±16.0%. These effects were largely reproduced by UCP2 knockdown with specific siRNA. CONCLUSIONS: Genipin decreased cancer cell (18)F-FDG uptake by reducing both glycolytic flux and mitochondrial oxidative respiration. This effect appeared to occur by blocking the ability of UCP2 to dissipate energy and restrict ROS production through proton leakage.


Subject(s)
Fluorodeoxyglucose F18/metabolism , Glucose/metabolism , Iridoids/pharmacology , Uncoupling Protein 2/metabolism , Biological Transport/drug effects , Breast Neoplasms/pathology , Cell Line, Tumor , Colonic Neoplasms/pathology , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Lactic Acid/biosynthesis , Matrix Metalloproteinases/metabolism , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Time Factors , Uncoupling Protein 2/deficiency , Uncoupling Protein 2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL