Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 24(16)2023 Aug 17.
Article in English | MEDLINE | ID: mdl-37629079

ABSTRACT

Type 2 diabetes (T2D) is associated with increased risk of atherosclerotic vascular disease due to excessive vascular smooth muscle cell (VSMC) proliferation. Here, we investigated the role of mitochondrial dysfunction and Ca2+ levels in VSMC proliferation in T2D. VSMCs were isolated from normoglycemic and T2D-like mice induced by diet. The effects of mitochondrial Ca2+ uptake were studied using mice with selectively inhibited mitochondrial Ca2+/calmodulin-dependent kinase II (mtCaMKII) in VSMCs. Mitochondrial transition pore (mPTP) was blocked using ER-000444793. VSMCs from T2D compared to normoglycemic mice exhibited increased proliferation and baseline cytosolic Ca2+ levels ([Ca2+]cyto). T2D cells displayed lower endoplasmic reticulum Ca2+ levels, reduced mitochondrial Ca2+ entry, and increased Ca2+ leakage through the mPTP. Mitochondrial and cytosolic Ca2+ transients were diminished in T2D cells upon platelet-derived growth factor (PDGF) administration. Inhibiting mitochondrial Ca2+ uptake or the mPTP reduced VSMC proliferation in T2D, but had contrasting effects on [Ca2+]cyto. In T2D VSMCs, enhanced activation of Erk1/2 and its upstream regulators was observed, driven by elevated [Ca2+]cyto. Inhibiting mtCaMKII worsened the Ca2+ imbalance by blocking mitochondrial Ca2+ entry, leading to further increases in [Ca2+]cyto and Erk1/2 hyperactivation. Under these conditions, PDGF had no effect on VSMC proliferation. Inhibiting Ca2+-dependent signaling in the cytosol reduced excessive Erk1/2 activation and VSMC proliferation. Our findings suggest that altered Ca2+ handling drives enhanced VSMC proliferation in T2D, with mitochondrial dysfunction contributing to this process.


Subject(s)
Atherosclerosis , Diabetes Mellitus, Type 2 , Animals , Mice , Calcium , Platelet-Derived Growth Factor , Myocytes, Smooth Muscle , Cell Proliferation
2.
JACC Case Rep ; 9: 101533, 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36909267

ABSTRACT

Recognizing true from pseudo left ventricular aneurysm after myocardial infarction is paramount to guide clinical management and determine need for surgical urgency. We discuss a case of a postinfarction pseudoaneurysm that poses unique anatomic challenges and may hold a secret "DaVinci code" beyond current diagnostic criteria. (Level of Difficulty: Advanced.).

3.
bioRxiv ; 2023 Feb 16.
Article in English | MEDLINE | ID: mdl-36824758

ABSTRACT

Background: Type 2 diabetes (T2D) is associated with a strongly increased risk for restenosis after angioplasty driven by proliferation of vascular smooth muscle cells (VSMCs). Here, we sought to determine whether and how mitochondrial dysfunction in T2D drives VSMC proliferation with a focus on ROS and intracellular [Ca 2+ ] that both drive cell proliferation, occur in T2D and are regulated by mitochondrial activity. Methods: Using a diet-induced mouse model of T2D, the inhibition of the mitochondrial Ca 2+ /calmodulin-dependent kinase II (mtCaMKII), a regulator of Ca 2+ entry via the mitochondrial Ca 2+ uniporter selectively in VSMCs, we performed in vivo phenotyping after mechanical injury and established the mechanisms of excessive proliferation in cultured VSMCs. Results: In T2D, the inhibition of mtCaMKII reduced both neointima formation after mechanical injury and the proliferation of cultured VSMCs. VSMCs from T2D mice displayed accelerated proliferation, reduced mitochondrial Ca 2+ entry and membrane potential with elevated baseline [Ca 2+ ] cyto compared to cells from normoglycemic mice. Accelerated proliferation after PDGF treatment was driven by activation of Erk1/2 and its upstream regulators. Hyperactivation of Erk1/2 was Ca 2+ -dependent rather than mitochondrial ROS-driven Ca 2+ -dependent and included the activation of CaMKII in the cytosol. The inhibition of mtCaMKII exaggerated the Ca 2+ imbalance by lowering mitochondrial Ca 2+ entry and increasing baseline [Ca 2+ ] cyto , further enhancing baseline Erk1/2 activation. With inhibition of mtCaMKII, PDGF treatment had no additional effect on cell proliferation. Inhibition of activated CaMKII in the cytosol decreased excessive Erk1/2 activation and reduced VSMC proliferation. Conclusions: Collectively, our results provide evidence for the molecular mechanisms of enhanced VSMC proliferation after mechanical injury by mitochondrial Ca 2+ entry in T2D.

4.
Arterioscler Thromb Vasc Biol ; 39(6): 991-997, 2019 06.
Article in English | MEDLINE | ID: mdl-31070466

ABSTRACT

Mitochondria regulate major aspects of cell function by producing ATP, contributing to Ca2+ signaling, influencing redox potential, and controlling levels of reactive oxygen species. In this review, we will discuss recent findings that illustrate how mitochondrial respiration, Ca2+ handling, and production of reactive oxygen species affect vascular smooth muscle cell function during neointima formation. We will review mitochondrial fission/fusion as fundamental mechanisms for smooth muscle proliferation, migration, and metabolism and examine the role of mitochondrial mobility in cell migration. In addition, we will summarize novel aspects by which mitochondria regulate apoptosis.


Subject(s)
Mitochondria/metabolism , Muscle, Smooth, Vascular/metabolism , Neointima/metabolism , Reactive Oxygen Species/metabolism , Stress, Physiological/genetics , bcl-2 Homologous Antagonist-Killer Protein/genetics , Animals , Apoptosis/genetics , Cell Movement/genetics , Cell Proliferation , Cells, Cultured , Humans , Mitochondrial Dynamics/genetics , Signal Transduction/genetics
5.
Sci Signal ; 12(579)2019 04 30.
Article in English | MEDLINE | ID: mdl-31040260

ABSTRACT

The role of the mitochondrial Ca2+ uniporter (MCU) in physiologic cell proliferation remains to be defined. Here, we demonstrated that the MCU was required to match mitochondrial function to metabolic demands during the cell cycle. During the G1-S transition (the cycle phase with the highest mitochondrial ATP output), mitochondrial fusion, oxygen consumption, and Ca2+ uptake increased in wild-type cells but not in cells lacking MCU. In proliferating wild-type control cells, the addition of the growth factors promoted the activation of the Ca2+/calmodulin-dependent kinase II (CaMKII) and the phosphorylation of the mitochondrial fission factor Drp1 at Ser616 The lack of the MCU was associated with baseline activation of CaMKII, mitochondrial fragmentation due to increased Drp1 phosphorylation, and impaired mitochondrial respiration and glycolysis. The mitochondrial fission/fusion ratio and proliferation in MCU-deficient cells recovered after MCU restoration or inhibition of mitochondrial fragmentation or of CaMKII in the cytosol. Our data highlight a key function for the MCU in mitochondrial adaptation to the metabolic demands during cell cycle progression. Cytosolic CaMKII and the MCU participate in a regulatory circuit, whereby mitochondrial Ca2+ uptake affects cell proliferation through Drp1.


Subject(s)
Calcium Channels/metabolism , Cell Proliferation/physiology , G1 Phase Cell Cycle Checkpoints/physiology , Mitochondrial Dynamics/physiology , Myocytes, Smooth Muscle/metabolism , Animals , Calcium/metabolism , Calcium Channels/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Proliferation/genetics , Cells, Cultured , Dynamins/metabolism , Female , G1 Phase Cell Cycle Checkpoints/genetics , Male , Mice, Knockout , Mitochondrial Dynamics/genetics , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Phosphorylation
6.
Arterioscler Thromb Vasc Biol ; 38(6): 1333-1345, 2018 06.
Article in English | MEDLINE | ID: mdl-29599132

ABSTRACT

OBJECTIVE: The main objective of this study is to define the mechanisms by which mitochondria control vascular smooth muscle cell (VSMC) migration and impact neointimal hyperplasia. APPROACH AND RESULTS: The multifunctional CaMKII (Ca2+/calmodulin-dependent kinase II) in the mitochondrial matrix of VSMC drove a feed-forward circuit with the mitochondrial Ca2+ uniporter (MCU) to promote matrix Ca2+ influx. MCU was necessary for the activation of mitochondrial CaMKII (mtCaMKII), whereas mtCaMKII phosphorylated MCU at the regulatory site S92 that promotes Ca2+ entry. mtCaMKII was necessary and sufficient for platelet-derived growth factor-induced mitochondrial Ca2+ uptake. This effect was dependent on MCU. mtCaMKII and MCU inhibition abrogated VSMC migration and mitochondrial translocation to the leading edge. Overexpression of wild-type MCU, but not MCU S92A, mutant in MCU-/- VSMC rescued migration and mitochondrial mobility. Inhibition of microtubule, but not of actin assembly, blocked mitochondrial mobility. The outer mitochondrial membrane GTPase Miro-1 promotes mitochondrial mobility via microtubule transport but arrests it in subcellular domains of high Ca2+ concentrations. In Miro-1-/- VSMC, mitochondrial mobility and VSMC migration were abolished, and overexpression of mtCaMKII or a CaMKII inhibitory peptide in mitochondria (mtCaMKIIN) had no effect. Consistently, inhibition of mtCaMKII increased and prolonged cytosolic Ca2+ transients. mtCaMKII inhibition diminished phosphorylation of focal adhesion kinase and myosin light chain, leading to reduced focal adhesion turnover and cytoskeletal remodeling. In a transgenic model of selective mitochondrial CaMKII inhibition in VSMC, neointimal hyperplasia was significantly reduced after vascular injury. CONCLUSIONS: These findings identify mitochondrial CaMKII as a key regulator of mitochondrial Ca2+ uptake via MCU, thereby controlling mitochondrial translocation and VSMC migration after vascular injury.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium/metabolism , Carotid Artery Injuries/enzymology , Cell Movement , Mitochondria, Muscle/enzymology , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Neointima , Animals , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Carotid Artery Injuries/genetics , Carotid Artery Injuries/pathology , Cells, Cultured , Disease Models, Animal , Hyperplasia , Male , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria, Muscle/pathology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
7.
Vascul Pharmacol ; 60(2): 75-83, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24418021

ABSTRACT

Activation of the Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) and reactive oxygen species (ROS) promote neointimal hyperplasia after vascular injury. CaMKII can be directly activated by ROS through oxidation. In this study, we determined whether abolishing the oxidative activation site of CaMKII alters vascular smooth muscle cell (VCMC) proliferation, migration and apoptosis in vitro and neointimal formation in vivo. VSMC isolated from a knock-in mouse with oxidation-resistant CaMKIIδ (CaMKII M2V) displayed similar proliferation but decreased migration and apoptosis. Surprisingly, ROS production and expression of the NADPH oxidase subunits p47 and p22 were decreased in M2V VSMC, whereas superoxide dismutase 2 protein expression was upregulated. In vivo, after carotid artery ligation, no differences in neointimal size or remodeling were observed. In contrast to VSMC, CaMKII expression and autonomous activity were significantly higher in M2V compared to WT carotid arteries, suggesting that an autoregulatory mechanism determines CaMKII activity in vivo. Our findings demonstrate that preventing oxidative activation of CaMKII decreases migration and apoptosis in vitro and suggest that CaMKII regulates ROS production. Our study presents novel evidence that CaMKII expression in vivo is regulated by a negative feedback loop following oxidative activation.


Subject(s)
Apoptosis/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Muscle, Smooth, Vascular/metabolism , Reactive Oxygen Species/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Carotid Arteries/metabolism , Cell Proliferation , Cytochrome b Group/metabolism , Female , Gene Expression Regulation , Gene Knock-In Techniques , Male , Mice , Muscle, Smooth, Vascular/cytology , NADPH Oxidases/metabolism , Neointima/metabolism , Oxidation-Reduction , Superoxide Dismutase/metabolism
8.
J Virol ; 84(8): 4041-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20130047

ABSTRACT

Human alpha-defensins are evolutionarily conserved effectors of the innate immune response with broadly acting antibacterial activity. Their role in antiviral immunity is less well understood. We previously showed that these antimicrobial peptides are potent inhibitors of human adenovirus infection. Based on biochemical studies and indirect evidence from confocal microscopy, we proposed that defensins bind to and stabilize the virus capsid and neutralize infection by preventing the release of the endosomalytic protein VI. To determine whether defensin action also restricts exposure of the viral genome, we developed a system to evaluate adenovirus uncoating during cell entry by monitoring the exposure of BrdU-labeled viral genomes. This assay allowed us to determine the kinetics of uncoating of virus particles in single cells. Using this assay, we now provide direct evidence that human alpha-defensins block adenovirus infection by preventing uncoating during cell entry.


Subject(s)
Adenoviridae Infections/immunology , Adenoviruses, Human/immunology , Adenoviruses, Human/physiology , Virus Internalization , alpha-Defensins/immunology , Bromodeoxyuridine/metabolism , Capsid/metabolism , Cell Line , Humans , Microscopy, Confocal , Staining and Labeling/methods , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...