301
|
Abstract
The circulation of the lung is unique both in volume and function. For example, it is the only organ with two circulations: the pulmonary circulation, the main function of which is gas exchange, and the bronchial circulation, a systemic vascular supply that provides oxygenated blood to the walls of the conducting airways, pulmonary arteries and veins. The pulmonary circulation accommodates the entire cardiac output, maintaining high blood flow at low intravascular arterial pressure. As compared with the systemic circulation, pulmonary arteries have thinner walls with much less vascular smooth muscle and a relative lack of basal tone. Factors controlling pulmonary blood flow include vascular structure, gravity, mechanical effects of breathing, and the influence of neural and humoral factors. Pulmonary vascular tone is also altered by hypoxia, which causes pulmonary vasoconstriction. If the hypoxic stimulus persists for a prolonged period, contraction is accompanied by remodeling of the vasculature, resulting in pulmonary hypertension. In addition, genetic and environmental factors can also confer susceptibility to development of pulmonary hypertension. Under normal conditions, the endothelium forms a tight barrier, actively regulating interstitial fluid homeostasis. Infection and inflammation compromise normal barrier homeostasis, resulting in increased permeability and edema formation. This article focuses on reviewing the basics of the lung circulation (pulmonary and bronchial), normal development and transition at birth and vasoregulation. Mechanisms contributing to pathological conditions in the pulmonary circulation, in particular when barrier function is disrupted and during development of pulmonary hypertension, will also be discussed.
Collapse
Affiliation(s)
- Karthik Suresh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Larissa A. Shimoda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
302
|
Torres G, Morales PE, García-Miguel M, Norambuena-Soto I, Cartes-Saavedra B, Vidal-Peña G, Moncada-Ruff D, Sanhueza-Olivares F, San Martín A, Chiong M. Glucagon-like peptide-1 inhibits vascular smooth muscle cell dedifferentiation through mitochondrial dynamics regulation. Biochem Pharmacol 2016; 104:52-61. [PMID: 26807480 PMCID: PMC4775317 DOI: 10.1016/j.bcp.2016.01.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 01/20/2016] [Indexed: 11/29/2022]
Abstract
Glucagon-like peptide-1 (GLP-1) is a neuroendocrine hormone produced by gastrointestinal tract in response to food ingestion. GLP-1 plays a very important role in the glucose homeostasis by stimulating glucose-dependent insulin secretion, inhibiting glucagon secretion, inhibiting gastric emptying, reducing appetite and food intake. Because of these actions, the GLP-1 peptide-mimetic exenatide is one of the most promising new medicines for the treatment of type 2 diabetes. In vivo treatments with GLP-1 or exenatide prevent neo-intima layer formation in response to endothelial damage and atherosclerotic lesion formation in aortic tissue. Whether GLP-1 modulates vascular smooth muscle cell (VSMC) migration and proliferation by controlling mitochondrial dynamics is unknown. In this report, we showed that GLP-1 increased mitochondrial fusion and activity in a PKA-dependent manner in the VSMC cell line A7r5. GLP-1 induced a Ser-637 phosphorylation in the mitochondrial fission protein Drp1, and decreased Drp1 mitochondrial localization. GLP-1 inhibited PDGF-BB-induced VSMC migration and proliferation, actions inhibited by overexpressing wild type Drp1 and mimicked by the Drp1 inhibitor Mdivi-1 and by overexpressing dominant negative Drp1. These results show that GLP-1 stimulates mitochondrial fusion, increases mitochondrial activity and decreases PDGF-BB-induced VSMC dedifferentiation by a PKA/Drp1 signaling pathway. Our data suggest that GLP-1 inhibits vascular remodeling through a mitochondrial dynamics-dependent mechanism.
Collapse
Affiliation(s)
- Gloria Torres
- Advanced Center for Chronic Diseases (ACCDiS), Centro Estudios Moleculares de la Célula (CEMC), Departamento Bioquímica y Biología Molecular, Facultad Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile; Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Pablo E Morales
- Advanced Center for Chronic Diseases (ACCDiS), Centro Estudios Moleculares de la Célula (CEMC), Departamento Bioquímica y Biología Molecular, Facultad Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Marina García-Miguel
- Advanced Center for Chronic Diseases (ACCDiS), Centro Estudios Moleculares de la Célula (CEMC), Departamento Bioquímica y Biología Molecular, Facultad Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Ignacio Norambuena-Soto
- Advanced Center for Chronic Diseases (ACCDiS), Centro Estudios Moleculares de la Célula (CEMC), Departamento Bioquímica y Biología Molecular, Facultad Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Benjamín Cartes-Saavedra
- Advanced Center for Chronic Diseases (ACCDiS), Centro Estudios Moleculares de la Célula (CEMC), Departamento Bioquímica y Biología Molecular, Facultad Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Gonzalo Vidal-Peña
- Advanced Center for Chronic Diseases (ACCDiS), Centro Estudios Moleculares de la Célula (CEMC), Departamento Bioquímica y Biología Molecular, Facultad Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - David Moncada-Ruff
- Advanced Center for Chronic Diseases (ACCDiS), Centro Estudios Moleculares de la Célula (CEMC), Departamento Bioquímica y Biología Molecular, Facultad Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Fernanda Sanhueza-Olivares
- Advanced Center for Chronic Diseases (ACCDiS), Centro Estudios Moleculares de la Célula (CEMC), Departamento Bioquímica y Biología Molecular, Facultad Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Alejandra San Martín
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Mario Chiong
- Advanced Center for Chronic Diseases (ACCDiS), Centro Estudios Moleculares de la Célula (CEMC), Departamento Bioquímica y Biología Molecular, Facultad Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
303
|
Abstract
Mitochondria are a distinguishing feature of eukaryotic cells. Best known for their critical function in energy production via oxidative phosphorylation (OXPHOS), mitochondria are essential for nutrient and oxygen sensing and for the regulation of critical cellular processes, including cell death and inflammation. Such diverse functional roles for organelles that were once thought to be simple may be attributed to their distinct heteroplasmic genome, exclusive maternal lineage of inheritance, and ability to generate signals to communicate with other cellular organelles. Mitochondria are now thought of as one of the cell's most sophisticated and dynamic responsive sensing systems. Specific signatures of mitochondrial dysfunction that are associated with disease pathogenesis and/or progression are becoming increasingly important. In particular, the centrality of mitochondria in the pathological processes and clinical phenotypes associated with a range of lung diseases is emerging. Understanding the molecular mechanisms regulating the mitochondrial processes of lung cells will help to better define phenotypes and clinical manifestations associated with respiratory disease and to identify potential diagnostic and therapeutic targets.
Collapse
|
304
|
Maimaitijiang A, Zhuang X, Jiang X, Li Y. Dynamin-related protein inhibitor downregulates reactive oxygen species levels to indirectly suppress high glucose-induced hyperproliferation of vascular smooth muscle cells. Biochem Biophys Res Commun 2016; 471:474-8. [PMID: 26903301 DOI: 10.1016/j.bbrc.2016.02.051] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 02/14/2016] [Indexed: 02/01/2023]
Abstract
Hyperproliferation of vascular smooth muscle cells is a pathogenic mechanism common in diabetic vascular complications and is a putatively important therapeutic target. This study investigated multiple levels of biology, including cellular and organellar changes, as well as perturbations in protein synthesis and morphology. Quantitative and qualitative analysis was utilized to assess the effect of mitochondrial dynamic changes and reactive oxygen species(ROS) levels on high-glucose-induced hyperproliferation of vascular smooth muscle cells. The data demonstrated that the mitochondrial fission inhibitor Mdivi-1 and downregulation of ROS levels both effectively inhibited the high-glucose-induced hyperproliferation of vascular smooth muscle cells. Downregulation of ROS levels played a more direct role and ROS levels were also regulated by mitochondrial dynamics. Increased ROS levels induced excessive mitochondrial fission through dynamin-related protein (Drp 1), while Mdivi-1 suppressed the sensitivity of Drp1 to ROS levels, thus inhibiting excessive mitochondrial fission under high-glucose conditions. This study is the first to propose that mitochondrial dynamic changes and ROS levels interact with each other and regulate high-glucose-induced hyperproliferation of vascular smooth muscle cells. This finding provides novel ideas in understanding the pathogenesis of diabetic vascular remodeling and intervention.
Collapse
Affiliation(s)
| | - Xinyu Zhuang
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, PR China
| | - Xiaofei Jiang
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, PR China
| | - Yong Li
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, PR China.
| |
Collapse
|
305
|
Walters JW, Amos D, Ray K, Santanam N. Mitochondrial redox status as a target for cardiovascular disease. Curr Opin Pharmacol 2016; 27:50-5. [PMID: 26894468 DOI: 10.1016/j.coph.2016.01.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 01/25/2016] [Accepted: 01/29/2016] [Indexed: 02/07/2023]
Abstract
Mitochondria are major players in cellular energetics, oxidative stress and programmed cell death. Mitochondrial dynamics regulate and integrate these functions. Mitochondrial dysfunction is involved in cardiac hypertrophy, hypertension and myocardial ischemia/reperfusion injury. Reactive oxygen species generation is modulated by the fusion-fission pathway as well as key proteins such as sirtuins that act as metabolic sensors of cellular energetics. Mitochondrial redox status has thus become a good target for therapy against cardiovascular diseases. Recently, there is an influx of studies garnered towards assessing the beneficial effects of mitochondrial targeted antioxidants, drugs modulating the fusion-fission proteins, sirtuins, and other mitochondrial processes as potential cardio-protecting agents.
Collapse
Affiliation(s)
- James W Walters
- School of Arts & Sciences, Bluefield State College, Basic Science Building B213, 219 Rock Street, Bluefield, WV 24701, USA
| | - Deborah Amos
- Department of Pharmacology, Physiology & Toxicology, Joan C Edwards School of Medicine, Marshall University, One John Marshall Dr, Huntington, WV 25755, USA
| | - Kristeena Ray
- Department of Pharmacology, Physiology & Toxicology, Joan C Edwards School of Medicine, Marshall University, One John Marshall Dr, Huntington, WV 25755, USA
| | - Nalini Santanam
- Department of Pharmacology, Physiology & Toxicology, Joan C Edwards School of Medicine, Marshall University, One John Marshall Dr, Huntington, WV 25755, USA.
| |
Collapse
|
306
|
Altered Mitochondrial Dynamics Contributes to Propofol-induced Cell Death in Human Stem Cell-derived Neurons. Anesthesiology 2016; 123:1067-83. [PMID: 26352374 DOI: 10.1097/aln.0000000000000857] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Studies in developing animals have shown that anesthetic agents can lead to neuronal cell death and learning disabilities when administered early in life. Development of human embryonic stem cell-derived neurons has provided a valuable tool for understanding the effects of anesthetics on developing human neurons. Unbalanced mitochondrial fusion and fission lead to various pathological conditions including neurodegeneration. The aim of this study was to dissect the role of mitochondrial dynamics in propofol-induced neurotoxicity. METHODS Terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate in situ nick-end labeling staining was used to assess cell death in human embryonic stem cell-derived neurons. Mitochondrial fission was assessed using TOM20 staining and electron microscopy. Expression of mitochondrial fission-related proteins was assessed by Western blot, and confocal microscopy was used to assess opening time of the mitochondrial permeability transition pore (mPTP). RESULTS Exposure to 6 h of 20 μg/ml propofol increased cell death from 3.18 ± 0.17% in the control-treated group to 9.6 ± 0.95% and led to detrimental increases in mitochondrial fission (n = 5 coverslips per group) accompanied by increased expression of activated dynamin-related protein 1 and cyclin-dependent kinase 1, key proteins responsible for mitochondrial fission. Propofol exposure also induced earlier opening of the mPTP from 118.9 ± 3.1 s in the control-treated group to 73.3 ± 1.6 s. Pretreatment of the cells with mdivi-1, a mitochondrial fission blocker rescued the propofol-induced toxicity, mitochondrial fission, and mPTP opening time (n = 75 cells per group). Inhibiting cyclin-dependent kinase 1 attenuated the increase in cell death and fission and the increase in expression of activated dynamin-related protein 1. CONCLUSION These data demonstrate for the first time that propofol-induced neurotoxicity occurs through a mitochondrial fission/mPTP-mediated pathway.
Collapse
|
307
|
Mitofusin 2 Downregulation Triggers Pulmonary Artery Smooth Muscle Cell Proliferation and Apoptosis Imbalance in Rats With Hypoxic Pulmonary Hypertension Via the PI3K/Akt and Mitochondrial Apoptosis Pathways. J Cardiovasc Pharmacol 2016; 67:164-74. [DOI: 10.1097/fjc.0000000000000333] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
308
|
Vásquez-Trincado C, García-Carvajal I, Pennanen C, Parra V, Hill JA, Rothermel BA, Lavandero S. Mitochondrial dynamics, mitophagy and cardiovascular disease. J Physiol 2016; 594:509-25. [PMID: 26537557 DOI: 10.1113/jp271301] [Citation(s) in RCA: 459] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 08/30/2015] [Indexed: 12/14/2022] Open
Abstract
Cardiac hypertrophy is often initiated as an adaptive response to haemodynamic stress or myocardial injury, and allows the heart to meet an increased demand for oxygen. Although initially beneficial, hypertrophy can ultimately contribute to the progression of cardiac disease, leading to an increase in interstitial fibrosis and a decrease in ventricular function. Metabolic changes have emerged as key mechanisms involved in the development and progression of pathological remodelling. As the myocardium is a highly oxidative tissue, mitochondria play a central role in maintaining optimal performance of the heart. 'Mitochondrial dynamics', the processes of mitochondrial fusion, fission, biogenesis and mitophagy that determine mitochondrial morphology, quality and abundance have recently been implicated in cardiovascular disease. Studies link mitochondrial dynamics to the balance between energy demand and nutrient supply, suggesting that changes in mitochondrial morphology may act as a mechanism for bioenergetic adaptation during cardiac pathological remodelling. Another critical function of mitochondrial dynamics is the removal of damaged and dysfunctional mitochondria through mitophagy, which is dependent on the fission/fusion cycle. In this article, we discuss the latest findings regarding the impact of mitochondrial dynamics and mitophagy on the development and progression of cardiovascular pathologies, including diabetic cardiomyopathy, atherosclerosis, damage from ischaemia-reperfusion, cardiac hypertrophy and decompensated heart failure. We will address the ability of mitochondrial fusion and fission to impact all cell types within the myocardium, including cardiac myocytes, cardiac fibroblasts and vascular smooth muscle cells. Finally, we will discuss how these findings can be applied to improve the treatment and prevention of cardiovascular diseases.
Collapse
Affiliation(s)
- César Vásquez-Trincado
- Advanced Centre for Chronic Disease (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Centre for Molecular Studies of the Cell, Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile
| | - Ivonne García-Carvajal
- Advanced Centre for Chronic Disease (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Centre for Molecular Studies of the Cell, Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile
| | - Christian Pennanen
- Advanced Centre for Chronic Disease (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Centre for Molecular Studies of the Cell, Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile
| | - Valentina Parra
- Advanced Centre for Chronic Disease (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Centre for Molecular Studies of the Cell, Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Centre, Dallas, TX, USA
| | - Joseph A Hill
- Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Centre, Dallas, TX, USA.,Department of Molecular Biology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
| | - Beverly A Rothermel
- Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Centre, Dallas, TX, USA.,Department of Molecular Biology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
| | - Sergio Lavandero
- Advanced Centre for Chronic Disease (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Centre for Molecular Studies of the Cell, Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Centre, Dallas, TX, USA
| |
Collapse
|
309
|
Bryant AJ, Robinson LJ, Moore CS, Blackwell TR, Gladson S, Penner NL, Burman A, McClellan LJ, Polosukhin VV, Tanjore H, McConaha ME, Gleaves LA, Talati MA, Hemnes AR, Fessel JP, Lawson WE, Blackwell TS, West JD. Expression of mutant bone morphogenetic protein receptor II worsens pulmonary hypertension secondary to pulmonary fibrosis. Pulm Circ 2015; 5:681-90. [PMID: 26697175 DOI: 10.1086/683811] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Pulmonary fibrosis is often complicated by pulmonary hypertension (PH), and previous studies have shown a potential link between bone morphogenetic protein receptor II (BMPR2) and PH secondary to pulmonary fibrosis. We exposed transgenic mice expressing mutant BMPR2 and control mice to repetitive intraperitoneal injections of bleomycin for 4 weeks. The duration of transgene activation was too short for mutant BMPR2 mice to develop spontaneous PH. Mutant BMPR2 mice had increased right ventricular systolic pressure compared to control mice, without differences in pulmonary fibrosis. We found increased hypoxia-inducible factor (HIF)1-α stabilization in lungs of mutant-BMPR2-expressing mice compared to controls following bleomycin treatment. In addition, expression of the hypoxia response element protein connective tissue growth factor was increased in transgenic mice as well as in a human pulmonary microvascular endothelial cell line expressing mutant BMPR2. In mouse pulmonary vascular endothelial cells, mutant BMPR2 expression resulted in increased HIF1-α and reactive oxygen species production following exposure to hypoxia, both of which were attenuated with the antioxidant TEMPOL. These data suggest that expression of mutant BMPR2 worsens secondary PH through increased HIF activity in vascular endothelium. This pathway could be therapeutically targeted in patients with PH secondary to pulmonary fibrosis.
Collapse
Affiliation(s)
- Andrew J Bryant
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA ; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Linda J Robinson
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Christy S Moore
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Thomas R Blackwell
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Santhi Gladson
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Niki L Penner
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Ankita Burman
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Lucas J McClellan
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Vasiliy V Polosukhin
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Harikrishna Tanjore
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Melinda E McConaha
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Linda A Gleaves
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Megha A Talati
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Anna R Hemnes
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Joshua P Fessel
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - William E Lawson
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA ; Department of Veterans Affairs Medical Center, Nashville, Tennessee, USA
| | - Timothy S Blackwell
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA ; Department of Cell and Developmental Biology and Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - James D West
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
310
|
Flicker-assisted localization microscopy reveals altered mitochondrial architecture in hypertension. Sci Rep 2015; 5:16875. [PMID: 26593883 PMCID: PMC4655370 DOI: 10.1038/srep16875] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 10/21/2015] [Indexed: 12/18/2022] Open
Abstract
Mitochondrial morphology is central to normal physiology and disease development. However, in many live cells and tissues, complex mitochondrial structures exist and morphology has been difficult to quantify. We have measured the shape of electrically-discrete mitochondria, imaging them individually to restore detail hidden in clusters and demarcate functional boundaries. Stochastic “flickers” of mitochondrial membrane potential were visualized with a rapidly-partitioning fluorophore and the pixel-by-pixel covariance of spatio-temporal fluorescence changes analyzed. This Flicker-assisted Localization Microscopy (FaLM) requires only an epifluorescence microscope and sensitive camera. In vascular myocytes, the apparent variation in mitochondrial size was partly explained by densely-packed small mitochondria. In normotensive animals, mitochondria were small spheres or rods. In hypertension, mitochondria were larger, occupied more of the cell volume and were more densely clustered. FaLM provides a convenient tool for increased discrimination of mitochondrial architecture and has revealed mitochondrial alterations that may contribute to hypertension.
Collapse
|
311
|
Chen X, Talati M, Fessel JP, Hemnes AR, Gladson S, French J, Shay S, Trammell A, Phillips JA, Hamid R, Cogan JD, Dawson EP, Womble KE, Hedges LK, Martinez EG, Wheeler LA, Loyd JE, Majka SJ, West J, Austin ED. Estrogen Metabolite 16α-Hydroxyestrone Exacerbates Bone Morphogenetic Protein Receptor Type II-Associated Pulmonary Arterial Hypertension Through MicroRNA-29-Mediated Modulation of Cellular Metabolism. Circulation 2015; 133:82-97. [PMID: 26487756 DOI: 10.1161/circulationaha.115.016133] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 10/02/2015] [Indexed: 12/25/2022]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a proliferative disease of the pulmonary vasculature that preferentially affects women. Estrogens such as the metabolite 16α-hydroxyestrone (16αOHE) may contribute to PAH pathogenesis, and alterations in cellular energy metabolism associate with PAH. We hypothesized that 16αOHE promotes heritable PAH (HPAH) via microRNA-29 (miR-29) family upregulation and that antagonism of miR-29 would attenuate pulmonary hypertension in transgenic mouse models of Bmpr2 mutation. METHODS AND RESULTS MicroRNA array profiling of human lung tissue found elevation of microRNAs associated with energy metabolism, including the miR-29 family, among HPAH patients. miR-29 expression was 2-fold higher in Bmpr2 mutant mice lungs at baseline compared with controls and 4 to 8-fold higher in Bmpr2 mice exposed to 16αOHE 1.25 μg/h for 4 weeks. Blot analyses of Bmpr2 mouse lung protein showed significant reductions in peroxisome proliferator-activated receptor-γ and CD36 in those mice exposed to 16αOHE and protein derived from HPAH lungs compared with controls. Bmpr2 mice treated with anti-miR-29 (20-mg/kg injections for 6 weeks) had improvements in hemodynamic profile, histology, and markers of dysregulated energy metabolism compared with controls. Pulmonary artery smooth muscle cells derived from Bmpr2 murine lungs demonstrated mitochondrial abnormalities, which improved with anti-miR-29 transfection in vitro; endothelial-like cells derived from HPAH patient induced pluripotent stem cell lines were similar and improved with anti-miR-29 treatment. CONCLUSIONS 16αOHE promotes the development of HPAH via upregulation of miR-29, which alters molecular and functional indexes of energy metabolism. Antagonism of miR-29 improves in vivo and in vitro features of HPAH and reveals a possible novel therapeutic target.
Collapse
Affiliation(s)
- Xinping Chen
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Megha Talati
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Joshua P Fessel
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Anna R Hemnes
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Santhi Gladson
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Jaketa French
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Sheila Shay
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Aaron Trammell
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - John A Phillips
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Rizwan Hamid
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Joy D Cogan
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Elliott P Dawson
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Kristie E Womble
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Lora K Hedges
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Elizabeth G Martinez
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Lisa A Wheeler
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - James E Loyd
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Susan J Majka
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - James West
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Eric D Austin
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.).
| |
Collapse
|
312
|
Yamamori T, Ike S, Bo T, Sasagawa T, Sakai Y, Suzuki M, Yamamoto K, Nagane M, Yasui H, Inanami O. Inhibition of the mitochondrial fission protein dynamin-related protein 1 (Drp1) impairs mitochondrial fission and mitotic catastrophe after x-irradiation. Mol Biol Cell 2015; 26:4607-17. [PMID: 26466676 PMCID: PMC4678018 DOI: 10.1091/mbc.e15-03-0181] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 10/07/2015] [Indexed: 01/07/2023] Open
Abstract
The role of mitochondrial dynamics in cellular responses to ionizing radiation (IR) is still largely unknown. This study demonstrates that IR triggers Drp1-dependent mitochondrial fission and that Drp1 inhibition attenuates radiation-induced mitotic catastrophe, suggesting that Drp1 is involved in determining the fate of cells after irradiation. Accumulating evidence suggests that mitochondrial dynamics is crucial for the maintenance of cellular quality control and function in response to various stresses. However, the role of mitochondrial dynamics in cellular responses to ionizing radiation (IR) is still largely unknown. In this study, we provide evidence that IR triggers mitochondrial fission mediated by the mitochondrial fission protein dynamin-related protein 1 (Drp1). We also show IR-induced mitotic catastrophe (MC), which is a type of cell death associated with defective mitosis, and aberrant centrosome amplification in mouse embryonic fibroblasts (MEFs). These are attenuated by genetic or pharmacological inhibition of Drp1. Whereas radiation-induced aberrant centrosome amplification and MC are suppressed by the inhibition of Plk1 and CDK2 in wild-type MEFs, the inhibition of these kinases is ineffective in Drp1-deficient MEFs. Furthermore, the cyclin B1 level after irradiation is significantly higher throughout the time course in Drp1-deficient MEFs than in wild-type MEFs, implying that Drp1 is involved in the regulation of cyclin B1 level. These findings strongly suggest that Drp1 plays an important role in determining the fate of cells after irradiation via the regulation of mitochondrial dynamics.
Collapse
Affiliation(s)
- Tohru Yamamori
- Laboratory of Radiation Biology, Department of Environmental Veterinary Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Satoshi Ike
- Laboratory of Radiation Biology, Department of Environmental Veterinary Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Tomoki Bo
- Laboratory of Radiation Biology, Department of Environmental Veterinary Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Tomoya Sasagawa
- Laboratory of Radiation Biology, Department of Environmental Veterinary Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Yuri Sakai
- Laboratory of Radiation Biology, Department of Environmental Veterinary Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Motofumi Suzuki
- Laboratory of Radiation Biology, Department of Environmental Veterinary Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Kumiko Yamamoto
- Laboratory of Radiation Biology, Department of Environmental Veterinary Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Masaki Nagane
- Laboratory of Radiation Biology, Department of Environmental Veterinary Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Hironobu Yasui
- Laboratory of Radiation Biology, Department of Environmental Veterinary Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Osamu Inanami
- Laboratory of Radiation Biology, Department of Environmental Veterinary Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| |
Collapse
|
313
|
Novak EA, Mollen KP. Mitochondrial dysfunction in inflammatory bowel disease. Front Cell Dev Biol 2015; 3:62. [PMID: 26484345 PMCID: PMC4589667 DOI: 10.3389/fcell.2015.00062] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 09/14/2015] [Indexed: 12/12/2022] Open
Abstract
Inflammatory Bowel Disease (IBD) represents a group of idiopathic disorders characterized by chronic or recurring inflammation of the gastrointestinal tract. While the exact etiology of disease is unknown, IBD is recognized to be a complex, multifactorial disease that results from an intricate interplay of genetic predisposition, an altered immune response, changes in the intestinal microbiota, and environmental factors. Together, these contribute to a destruction of the intestinal epithelial barrier, increased gut permeability, and an influx of immune cells. Given that most cellular functions as well as maintenance of the epithelial barrier is energy-dependent, it is logical to assume that mitochondrial dysfunction may play a key role in both the onset and recurrence of disease. Indeed several studies have demonstrated evidence of mitochondrial stress and alterations in mitochondrial function within the intestinal epithelium of patients with IBD and mice undergoing experimental colitis. Although the hallmarks of mitochondrial dysfunction, including oxidative stress and impaired ATP production are known to be evident in the intestines of patients with IBD, it is as yet unclear whether these processes occur as a cause of consequence of disease. We provide a current review of mitochondrial function in the setting of intestinal inflammation during IBD.
Collapse
Affiliation(s)
- Elizabeth A Novak
- Department of Surgery, University of Pittsburgh School of Medicine Pittsburgh, PA, USA
| | - Kevin P Mollen
- Department of Surgery, University of Pittsburgh School of Medicine Pittsburgh, PA, USA
| |
Collapse
|
314
|
Dunham-Snary KJ, Hong ZG, Xiong PY, Del Paggio JC, Herr JE, Johri AM, Archer SL. A mitochondrial redox oxygen sensor in the pulmonary vasculature and ductus arteriosus. Pflugers Arch 2015; 468:43-58. [PMID: 26395471 DOI: 10.1007/s00424-015-1736-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 09/09/2015] [Accepted: 09/15/2015] [Indexed: 12/18/2022]
Abstract
The mammalian homeostatic oxygen sensing system (HOSS) initiates changes in vascular tone, respiration, and neurosecretion that optimize oxygen uptake and tissue oxygen delivery within seconds of detecting altered environmental or arterial PO2. The HOSS includes carotid body type 1 cells, adrenomedullary cells, neuroepithelial bodies, and smooth muscle cells (SMCs) in pulmonary arteries (PAs), ductus arteriosus (DA), and fetoplacental arteries. Hypoxic pulmonary vasoconstriction (HPV) optimizes ventilation-perfusion matching. In utero, HPV diverts placentally oxygenated blood from the non-ventilated lung through the DA. At birth, increased alveolar and arterial oxygen tension dilates the pulmonary vasculature and constricts the DA, respectively, thereby transitioning the newborn to an air-breathing organism. Though modulated by endothelial-derived relaxing and constricting factors, O2 sensing is intrinsic to PASMCs and DASMCs. Within the SMC's dynamic mitochondrial network, changes in PO2 alter the reduction-oxidation state of redox couples (NAD(+)/NADH, NADP(+)/NADPH) and the production of reactive oxygen species, ROS (e.g., H2O2), by complexes I and III of the electron transport chain (ETC). ROS and redox couples regulate ion channels, transporters, and enzymes, changing intracellular calcium [Ca(2+)]i and calcium sensitivity and eliciting homeostatic responses to hypoxia. In PASMCs, hypoxia inhibits ROS production and reduces redox couples, thereby inhibiting O2-sensitive voltage-gated potassium (Kv) channels, depolarizing the plasma membrane, activating voltage-gated calcium channels (CaL), increasing [Ca(2+)]i, and causing vasoconstriction. In DASMCs, elevated PO2 causes mitochondrial fission, increasing ETC complex I activity and ROS production. The DASMC's downstream response to elevated PO2 (Kv channel inhibition, CaL activation, increased [Ca(2+)]i, and rho kinase activation) is similar to the PASMC's hypoxic response. Impaired O2 sensing contributes to human diseases, including pulmonary arterial hypertension and patent DA.
Collapse
Affiliation(s)
- Kimberly J Dunham-Snary
- Department of Medicine, Queen's University, Etherington Hall, Room 3041, 94 Stuart St, Kingston, ON, K7L 3N6, Canada
| | - Zhigang G Hong
- Department of Medicine, Queen's University, Etherington Hall, Room 3041, 94 Stuart St, Kingston, ON, K7L 3N6, Canada
| | - Ping Y Xiong
- Department of Medicine, Queen's University, Etherington Hall, Room 3041, 94 Stuart St, Kingston, ON, K7L 3N6, Canada
| | - Joseph C Del Paggio
- Department of Medicine, Queen's University, Etherington Hall, Room 3041, 94 Stuart St, Kingston, ON, K7L 3N6, Canada
| | - Julia E Herr
- Department of Medicine, Queen's University, Etherington Hall, Room 3041, 94 Stuart St, Kingston, ON, K7L 3N6, Canada
| | - Amer M Johri
- Department of Medicine, Queen's University, Etherington Hall, Room 3041, 94 Stuart St, Kingston, ON, K7L 3N6, Canada
| | - Stephen L Archer
- Department of Medicine, Queen's University, Etherington Hall, Room 3041, 94 Stuart St, Kingston, ON, K7L 3N6, Canada.
| |
Collapse
|
315
|
Ong SB, Kalkhoran SB, Cabrera-Fuentes HA, Hausenloy DJ. Mitochondrial fusion and fission proteins as novel therapeutic targets for treating cardiovascular disease. Eur J Pharmacol 2015; 763:104-14. [PMID: 25987420 PMCID: PMC4784719 DOI: 10.1016/j.ejphar.2015.04.056] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Revised: 03/28/2015] [Accepted: 04/09/2015] [Indexed: 12/11/2022]
Abstract
The past decade has witnessed a number of exciting developments in the field of mitochondrial dynamics - a phenomenon in which changes in mitochondrial shape and movement impact on cellular physiology and pathology. By undergoing fusion and fission, mitochondria are able to change their morphology between elongated interconnected networks and discrete fragmented structures, respectively. The cardiac mitochondria, in particular, have garnered much interest due to their unique spatial arrangement in the adult cardiomyocyte, and the multiple roles they play in cell death and survival. In this article, we review the role of the mitochondrial fusion and fission proteins as novel therapeutic targets for treating cardiovascular disease.
Collapse
Affiliation(s)
- Sang-Bing Ong
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore; National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore; Department of Clinical Sciences, Faculty of Biosciences and Medical Engineering, Universiti Teknologi Malaysia, Johor Bahru, Malaysia
| | | | - Hector A Cabrera-Fuentes
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore; National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore; Institute of Biochemistry, Medical School, Justus-Liebig University, Giessen, Germany; Department of Microbiology, Kazan Federal University, Kazan, Russian Federation
| | - Derek J Hausenloy
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore; National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore; The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, UK; The National Institute of Health Research University College London Hospitals Biomedical Research Centre, UK.
| |
Collapse
|
316
|
Kim KY, Perkins GA, Shim MS, Bushong E, Alcasid N, Ju S, Ellisman MH, Weinreb RN, Ju WK. DRP1 inhibition rescues retinal ganglion cells and their axons by preserving mitochondrial integrity in a mouse model of glaucoma. Cell Death Dis 2015; 6:e1839. [PMID: 26247724 PMCID: PMC4558491 DOI: 10.1038/cddis.2015.180] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 05/13/2015] [Accepted: 05/18/2015] [Indexed: 11/09/2022]
Abstract
Glaucoma is the leading cause of irreversible blindness and is characterized by slow and progressive degeneration of the optic nerve head axons and retinal ganglion cell (RGC), leading to loss of visual function. Although oxidative stress and/or alteration of mitochondrial (mt) dynamics induced by elevated intraocular pressure (IOP) are associated with this neurodegenerative disease, the mechanisms that regulate mt dysfunction-mediated glaucomatous neurodegeneration are poorly understood. Using a mouse model of glaucoma, DBA/2J (D2), which spontaneously develops elevated IOP, as well as an in vitro RGC culture system, we show here that oxidative stress, as evidenced by increasing superoxide dismutase 2 (SOD2) and mt transcription factor A (Tfam) protein expression, triggers mt fission and loss by increasing dynamin-related protein 1 (DRP1) in the retina of glaucomatous D2 mice as well as in cultured RGCs exposed to elevated hydrostatic pressure in vitro. DRP1 inhibition by overexpressing DRP1 K38A mutant blocks mt fission and triggers a subsequent reduction of oxidative stress, as evidenced by decreasing SOD2 and Tfam protein expression. DRP1 inhibition promotes RGC survival by increasing phosphorylation of Bad at serine 112 in the retina and preserves RGC axons by maintaining mt integrity in the glial lamina of glaucomatous D2 mice. These findings demonstrate an important vicious cycle involved in glaucomatous neurodegeneration that starts with elevated IOP producing oxidative stress; the oxidative stress then leads to mt fission and a specific form of mt dysfunction that generates further oxidative stress, thus perpetuating the cycle. Our findings suggest that DRP1 is a potential therapeutic target for ameliorating oxidative stress-mediated mt fission and dysfunction in RGC and its axons during glaucomatous neurodegeneration. Thus, DRP1 inhibition may provide a new therapeutic strategy for protecting both RGCs and their axons in glaucoma and other optic neuropathies.
Collapse
Affiliation(s)
- K-Y Kim
- Department of Neuroscience, Center for Research in Biological Systems, National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA
| | - G A Perkins
- Department of Neuroscience, Center for Research in Biological Systems, National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA
| | - M S Shim
- Laboratory for Optic Nerve Biology, Department of Ophthalmology, Hamilton Glaucoma Center, University of California, San Diego, La Jolla, CA, USA
| | - E Bushong
- Department of Neuroscience, Center for Research in Biological Systems, National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA
| | - N Alcasid
- Department of Neuroscience, Center for Research in Biological Systems, National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA
| | - S Ju
- Department of Neuroscience, Center for Research in Biological Systems, National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA
| | - M H Ellisman
- Department of Neuroscience, Center for Research in Biological Systems, National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA
| | - R N Weinreb
- Laboratory for Optic Nerve Biology, Department of Ophthalmology, Hamilton Glaucoma Center, University of California, San Diego, La Jolla, CA, USA
| | - W-K Ju
- Laboratory for Optic Nerve Biology, Department of Ophthalmology, Hamilton Glaucoma Center, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
317
|
Kornfeld OS, Hwang S, Disatnik MH, Chen CH, Qvit N, Mochly-Rosen D. Mitochondrial reactive oxygen species at the heart of the matter: new therapeutic approaches for cardiovascular diseases. Circ Res 2015; 116:1783-99. [PMID: 25999419 DOI: 10.1161/circresaha.116.305432] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Reactive oxygen species (ROS) have been implicated in a variety of age-related diseases, including multiple cardiovascular disorders. However, translation of ROS scavengers (antioxidants) into the clinic has not been successful. These antioxidants grossly reduce total levels of cellular ROS including ROS that participate in physiological signaling. In this review, we challenge the traditional antioxidant therapeutic approach that targets ROS directly with novel approaches that improve mitochondrial functions to more effectively treat cardiovascular diseases.
Collapse
Affiliation(s)
- Opher S Kornfeld
- From the Department of Chemical and Systems Biology, Stanford University School of Medicine, CA
| | - Sunhee Hwang
- From the Department of Chemical and Systems Biology, Stanford University School of Medicine, CA
| | - Marie-Hélène Disatnik
- From the Department of Chemical and Systems Biology, Stanford University School of Medicine, CA
| | - Che-Hong Chen
- From the Department of Chemical and Systems Biology, Stanford University School of Medicine, CA
| | - Nir Qvit
- From the Department of Chemical and Systems Biology, Stanford University School of Medicine, CA
| | - Daria Mochly-Rosen
- From the Department of Chemical and Systems Biology, Stanford University School of Medicine, CA.
| |
Collapse
|
318
|
Ryan JJ, Archer SL. Emerging concepts in the molecular basis of pulmonary arterial hypertension: part I: metabolic plasticity and mitochondrial dynamics in the pulmonary circulation and right ventricle in pulmonary arterial hypertension. Circulation 2015; 131:1691-702. [PMID: 25964279 DOI: 10.1161/circulationaha.114.006979] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- John J Ryan
- From Division of Cardiovascular Medicine, Department of Medicine, University of Utah, Salt Lake City (J.J.R.); and Department of Medicine, Queen's University, Kingston, ON, Canada (S.L.A.)
| | - Stephen L Archer
- From Division of Cardiovascular Medicine, Department of Medicine, University of Utah, Salt Lake City (J.J.R.); and Department of Medicine, Queen's University, Kingston, ON, Canada (S.L.A.).
| |
Collapse
|
319
|
López-Crisosto C, Bravo-Sagua R, Rodriguez-Peña M, Mera C, Castro PF, Quest AFG, Rothermel BA, Cifuentes M, Lavandero S. ER-to-mitochondria miscommunication and metabolic diseases. Biochim Biophys Acta Mol Basis Dis 2015; 1852:2096-105. [PMID: 26171812 DOI: 10.1016/j.bbadis.2015.07.011] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Revised: 07/09/2015] [Accepted: 07/10/2015] [Indexed: 12/17/2022]
Abstract
Eukaryotic cells contain a variety of subcellular organelles, each of which performs unique tasks. Thus follows that in order to coordinate these different intracellular functions, a highly dynamic system of communication must exist between the various compartments. Direct endoplasmic reticulum (ER)-mitochondria communication is facilitated by the physical interaction of their membranes in dedicated structural domains known as mitochondria-associated membranes (MAMs), which facilitate calcium (Ca(2+)) and lipid transfer between organelles and also act as platforms for signaling. Numerous studies have demonstrated the importance of MAM in ensuring correct function of both organelles, and recently MAMs have been implicated in the genesis of various human diseases. Here, we review the salient structural features of interorganellar communication via MAM and discuss the most common experimental techniques employed to assess functionality of these domains. Finally, we will highlight the contribution of MAM to a variety of cellular functions and consider the potential role of MAM in the genesis of metabolic diseases. In doing so, the importance for cell functions of maintaining appropriate communication between ER and mitochondria will be emphasized.
Collapse
Affiliation(s)
- Camila López-Crisosto
- Advanced Center for Chronic Diseases (ACCDiS), Center for Molecular Studies of the Cell (CEMC), Faculty of Chemical & Pharmaceutical Sciences, Faculty of Medicine, University of Chile, Santiago 8380492, Chile
| | - Roberto Bravo-Sagua
- Advanced Center for Chronic Diseases (ACCDiS), Center for Molecular Studies of the Cell (CEMC), Faculty of Chemical & Pharmaceutical Sciences, Faculty of Medicine, University of Chile, Santiago 8380492, Chile
| | - Marcelo Rodriguez-Peña
- Advanced Center for Chronic Diseases (ACCDiS), Center for Molecular Studies of the Cell (CEMC), Faculty of Chemical & Pharmaceutical Sciences, Faculty of Medicine, University of Chile, Santiago 8380492, Chile
| | - Claudia Mera
- Advanced Center for Chronic Diseases (ACCDiS), Center for Molecular Studies of the Cell (CEMC), Faculty of Chemical & Pharmaceutical Sciences, Faculty of Medicine, University of Chile, Santiago 8380492, Chile
| | - Pablo F Castro
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Pontifical Catholic University of Chile, Santiago 8380492, Chile
| | - Andrew F G Quest
- Advanced Center for Chronic Diseases (ACCDiS), Center for Molecular Studies of the Cell (CEMC), Faculty of Chemical & Pharmaceutical Sciences, Faculty of Medicine, University of Chile, Santiago 8380492, Chile
| | - Beverly A Rothermel
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75235, USA; Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Mariana Cifuentes
- Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago 7830490, Chile
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Center for Molecular Studies of the Cell (CEMC), Faculty of Chemical & Pharmaceutical Sciences, Faculty of Medicine, University of Chile, Santiago 8380492, Chile; Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75235, USA; Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA.
| |
Collapse
|
320
|
Serum-glucocorticoid regulated kinase 1 regulates macrophage recruitment and activation contributing to monocrotaline-induced pulmonary arterial hypertension. Cardiovasc Toxicol 2015; 14:368-78. [PMID: 24825325 DOI: 10.1007/s12012-014-9260-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Sustained inflammation is associated with pulmonary vascular remodeling and arterial hypertension (PAH). Serum-glucocorticoid regulated kinase 1 (SGK1) has been shown to participate in vascular remodeling, but its role in inflammation-associated PAH remains unknown. In this study, the importance of SGK1 expression and activation was investigated on monocrotaline (MCT)-induced PAH, an inflammation-associated experimental model of PAH used in mice and rats. The expression of SGK1 in the lungs of rats with MCT-induced PAH was significantly increased. Furthermore, SGK1 knockout mice were resistant to MCT-induced PAH and showed less elevation of right ventricular systolic pressure and right ventricular hypertrophy and showed reduced pulmonary vascular remodeling in response to MCT injection. Administering the SGK1 inhibitor, EMD638683, to rats also prevented the development of MCT-induced PAH. The expression of SGK1 was shown to take place primarily in alveolar macrophages. EMD638683 treatment suppressed macrophage infiltration and inhibited the proliferation of pulmonary arterial smooth muscle cells (PASMCs) in the lungs of rats with MCT-induced PAH. Co-culture of bone marrow-derived macrophages (BMDMs) from wild-type (WT) mice promoted proliferation of PASMC in vitro, whereas BMDMs from either SGK1 knockout mice or WT mice with EMD638683 treatment failed to induce this response. Collectively, the present results demonstrated that SGK1 is important to the regulation of macrophage activation that contributes to the development of PAH; thus, SGK1 may be a potential therapeutic target for the treatment of PAH.
Collapse
|
321
|
Talati M, Hemnes A. Fatty acid metabolism in pulmonary arterial hypertension: role in right ventricular dysfunction and hypertrophy. Pulm Circ 2015; 5:269-78. [PMID: 26064451 DOI: 10.1086/681227] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 12/30/2014] [Indexed: 12/21/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a complex, multifactorial disease in which an increase in pulmonary vascular resistance leads to increased afterload on the right ventricle (RV), causing right heart failure and death. Our understanding of the pathophysiology of RV dysfunction in PAH is limited but is constantly improving. Increasing evidence suggests that in PAH RV dysfunction is associated with various components of metabolic syndrome, such as insulin resistance, hyperglycemia, and dyslipidemia. The relationship between RV dysfunction and fatty acid/glucose metabolites is multifaceted, and in PAH it is characterized by a shift in utilization of energy sources toward increased glucose utilization and reduced fatty acid consumption. RV dysfunction may be caused by maladaptive fatty acid metabolism resulting from an increase in fatty acid uptake by fatty acid transporter molecule CD36 and an imbalance between glucose and fatty acid oxidation in mitochondria. This leads to lipid accumulation in the form of triglycerides, diacylglycerol, and ceramides in the cytoplasm, hallmarks of lipotoxicity. Current interventions in animal models focus on improving RV dysfunction through altering fatty acid oxidation rates and limiting lipid accumulation, but more specific and effective therapies may be available in the coming years based on current research. In conclusion, a deeper understanding of the complex mechanisms of the metabolic remodeling of the RV will aid in the development of targeted treatments for RV failure in PAH.
Collapse
Affiliation(s)
- Megha Talati
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Anna Hemnes
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
322
|
Huetsch J, Shimoda LA. Na(+)/H(+) exchange and hypoxic pulmonary hypertension. Pulm Circ 2015; 5:228-43. [PMID: 26064449 DOI: 10.1086/680213] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 07/22/2014] [Indexed: 12/24/2022] Open
Abstract
Intracellular pH (pHi) homeostasis is key to the functioning of vascular smooth muscle cells, including pulmonary artery smooth muscle cells (PASMCs). Sodium-hydrogen exchange (NHE) is an important contributor to pHi control in PASMCs. In this review, we examine the role of NHE in PASMC function, in both physiologic and pathologic conditions. In particular, we focus on the contribution of NHE to the PASMC response to hypoxia, considering both acute hypoxic pulmonary vasoconstriction and the development of pulmonary vascular remodeling and pulmonary hypertension in response to chronic hypoxia. Hypoxic pulmonary hypertension remains a disease with limited therapeutic options. Thus, this review explores past efforts at disrupting NHE signaling and discusses the therapeutic potential that such efforts may have in the field of pulmonary hypertension.
Collapse
Affiliation(s)
- John Huetsch
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21224, USA
| | - Larissa A Shimoda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21224, USA
| |
Collapse
|
323
|
Li D, Li X, Guan Y, Guo X. Mitofusin-2-mediated tethering of mitochondria and endoplasmic reticulum promotes cell cycle arrest of vascular smooth muscle cells in G0/G1 phase. Acta Biochim Biophys Sin (Shanghai) 2015; 47:441-50. [PMID: 25926139 DOI: 10.1093/abbs/gmv035] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 02/26/2015] [Indexed: 11/13/2022] Open
Abstract
Mitofusin-2 (Mfn-2) is a hyperplasia suppressor. Changes in Mfn-2 expression are thought to reflect mitochondrial remodeling during cell proliferation. However, it is unclear how the participation of Mfn-2 in mitochondrial remodeling prevents cellular proliferation. Here we show that arresting vascular smooth muscle cells (VSMCs) in the G0/G1 phase by serum starvation up-regulates Mfn-2 expression and causes mitochondria to assemble into a tubular network and to attach to the endoplasmic reticulum (ER). In the S phase, short rod-shaped mitochondrial structures that were dissociated from the ER were observed. Levels of glucose, ATP, l-amino acid, and NADP(+) did not vary throughout the cell cycle. However, NAD(+) level was lower and NADH level was higher in the G0/G1 phase than in the S phase. Mitochondrial membrane potential was lower in the S phase than in the G0/G1 phase. Infecting VSMCs with an adenovirus encoding full-length Mfn-2 increased NADH level and reduced NAD(+) level, while infecting the cells with an adenovirus that silences the p21(ras) signature motif produced opposite effects. These results suggest that Mfn-2 up-regulation causes mitochondrial fusion into tubular networks and attachment to the ER, which in turn halts proliferation of VSMCs.
Collapse
Affiliation(s)
- Dan Li
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaolan Li
- Molecular Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yang Guan
- Department of Ultrastructural Pathology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaomei Guo
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
324
|
Zhang L, Ji L, Tang X, Chen X, Li Z, Mi X, Yang L. Inhibition to DRP1 translocation can mitigate p38 MAPK-signaling pathway activation in GMC induced by hyperglycemia. Ren Fail 2015; 37:903-10. [PMID: 25857570 DOI: 10.3109/0886022x.2015.1034607] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Diabetic nephropathy (DN) is a serious complication of diabetes with a poorly defined etiology and limited treatment options. Early intervention is a key to preventing the progression of DN. Dynamin-related protein 1 (DRP1) regulates mitochondrial morphology by promoting its fission and is involved in the pathogenesis of numerous diseases. Furthermore, DRP1 is also closely associated with the development of diabetes, but its functional role in DN remains unknown. This study investigated the effect of DRP1 on early stage of DN. DRP1 expression has increased significantly in glomerular mesangial cell (GMC), which is cultivated in high glucose (HG). Ultra-microstructural changes of nephrons, expression of collagen IV and phosph-p38, ROS production, and mitochondrial function were evaluated and, at the same time, were compared with glomerular mesangial cell (GMC) cultured in normal-glucose (NG), mannitol, and a medium with mitochondrial division inhibitor 1 (Midivi-1). Endogenous DRP1 expression increased in DN. Compared to the control groups ofNG and mannitol, overexpression of DRP1 destroyed pathological changes typical of the GMC, like accumulation of extracellular matrix, and an increase in mitochondria division. In addition, Overexpression of DRP1 promoted the activation of p38, the accumulation of ROS, mitochondrial dysfunction, and the synthesis of collagen IV, and all these changes are suppressed by Midivi-1. This study demonstrates that DRP1 overexpression can accelerate pathological changes in the GMC cultured in HG. Further studies are needed to clarify the underlying mechanism of this destructive function.
Collapse
Affiliation(s)
- LieMei Zhang
- a Division of Nephrology , West China Hospital of Sichuan University , Chengdu , Sichuan , China
| | | | | | | | | | | | | |
Collapse
|
325
|
Ryan JJ, Huston J, Kutty S, Hatton ND, Bowman L, Tian L, Herr JE, Johri AM, Archer SL. Right ventricular adaptation and failure in pulmonary arterial hypertension. Can J Cardiol 2015; 31:391-406. [PMID: 25840092 PMCID: PMC4385216 DOI: 10.1016/j.cjca.2015.01.023] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 01/19/2015] [Accepted: 01/19/2015] [Indexed: 01/22/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is an obstructive pulmonary vasculopathy, characterized by excess proliferation, apoptosis resistance, inflammation, fibrosis, and vasoconstriction. Although PAH therapies target some of these vascular abnormalities (primarily vasoconstriction), most do not directly benefit the right ventricle (RV). This is suboptimal because a patient's functional state and prognosis are largely determined by the success of the adaptation of the RV to the increased afterload. The RV initially hypertrophies but might ultimately decompensate, becoming dilated, hypokinetic, and fibrotic. A number of pathophysiologic abnormalities have been identified in the PAH RV, including: ischemia and hibernation (partially reflecting RV capillary rarefaction), autonomic activation (due to G protein receptor kinase 2-mediated downregulation and desensitization of β-adrenergic receptors), mitochondrial-metabolic abnormalities (notably increased uncoupled glycolysis and glutaminolysis), and fibrosis. Many RV abnormalities are detectable using molecular imaging and might serve as biomarkers. Some molecular pathways, such as those regulating angiogenesis, metabolism, and mitochondrial dynamics, are similarly deranged in the RV and pulmonary vasculature, offering the possibility of therapies that treat the RV and pulmonary circulation. An important paradigm in PAH is that the RV and pulmonary circulation constitute a unified cardiopulmonary unit. Clinical trials of PAH pharmacotherapies should assess both components of the cardiopulmonary unit.
Collapse
Affiliation(s)
- John J Ryan
- Division of Cardiovascular Medicine, Department of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Jessica Huston
- Department of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Shelby Kutty
- Pediatric Cardiology, University of Nebraska Medical Center, Children's Hospital and Medical Center, Omaha, Nebraska, USA
| | - Nathan D Hatton
- Division of Pulmonary Medicine, Department of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Lindsay Bowman
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Lian Tian
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Julia E Herr
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Amer M Johri
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Stephen L Archer
- Department of Medicine, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
326
|
Haslip M, Dostanic I, Huang Y, Zhang Y, Russell KS, Jurczak MJ, Mannam P, Giordano F, Erzurum SC, Lee PJ. Endothelial uncoupling protein 2 regulates mitophagy and pulmonary hypertension during intermittent hypoxia. Arterioscler Thromb Vasc Biol 2015; 35:1166-78. [PMID: 25814675 DOI: 10.1161/atvbaha.114.304865] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 03/10/2015] [Indexed: 01/06/2023]
Abstract
OBJECTIVES Pulmonary hypertension (PH) is a process of lung vascular remodeling, which can lead to right heart dysfunction and significant morbidity. The underlying mechanisms leading to PH are not well understood, and therapies are limited. Using intermittent hypoxia (IH) as a model of oxidant-induced PH, we identified an important role for endothelial cell mitophagy via mitochondrial uncoupling protein 2 (Ucp2) in the development of IH-induced PH. APPROACH AND RESULTS Ucp2 endothelial knockout (VE-KO) and Ucp2 Flox (Flox) mice were subjected to 5 weeks of IH. Ucp2 VE-KO mice exhibited higher right ventricular systolic pressure and worse right heart hypertrophy, as measured by increased right ventricle weight/left ventricle plus septal weight (RV/LV+S) ratio, at baseline and after IH. These changes were accompanied by increased mitophagy. Primary mouse lung endothelial cells transfected with Ucp2 siRNA and subjected to cyclic exposures to CoCl2 (chemical hypoxia) showed increased mitophagy, as measured by PTEN-induced putative kinase 1 and LC3BII/I ratios, decreased mitochondrial biogenesis, and increased apoptosis. Similar results were obtained in primary lung endothelial cells isolated from VE-KO mice. Moreover, silencing PTEN-induced putative kinase 1 in the endothelium of Ucp2 knockout mice, using endothelial-targeted lentiviral silencing RNA in vivo, prevented IH-induced PH. Human pulmonary artery endothelial cells from people with PH demonstrated changes similar to Ucp2-silenced mouse lung endothelial cells. CONCLUSIONS The loss of endothelial Ucp2 leads to excessive PTEN-induced putative kinase 1-induced mitophagy, inadequate mitochondrial biosynthesis, and increased apoptosis in endothelium. An endothelial Ucp2-PTEN-induced putative kinase 1 axis may be effective therapeutic targets in PH.
Collapse
Affiliation(s)
- Maria Haslip
- From the Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine (M.H., I.D., Y.Z., P.M., P.J.L.), Section of Cardiovascular Disease (Y.H., K.S.R, F.G.), and Section of Endocrinology and Metabolism (M.J.J.), Yale University School of Medicine, New Haven, CT; and Department of Pathobiology, Lerner Research Institute and Respiratory Institute, Cleveland Clinic, OH (S.C.E.)
| | - Iva Dostanic
- From the Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine (M.H., I.D., Y.Z., P.M., P.J.L.), Section of Cardiovascular Disease (Y.H., K.S.R, F.G.), and Section of Endocrinology and Metabolism (M.J.J.), Yale University School of Medicine, New Haven, CT; and Department of Pathobiology, Lerner Research Institute and Respiratory Institute, Cleveland Clinic, OH (S.C.E.)
| | - Yan Huang
- From the Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine (M.H., I.D., Y.Z., P.M., P.J.L.), Section of Cardiovascular Disease (Y.H., K.S.R, F.G.), and Section of Endocrinology and Metabolism (M.J.J.), Yale University School of Medicine, New Haven, CT; and Department of Pathobiology, Lerner Research Institute and Respiratory Institute, Cleveland Clinic, OH (S.C.E.)
| | - Yi Zhang
- From the Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine (M.H., I.D., Y.Z., P.M., P.J.L.), Section of Cardiovascular Disease (Y.H., K.S.R, F.G.), and Section of Endocrinology and Metabolism (M.J.J.), Yale University School of Medicine, New Haven, CT; and Department of Pathobiology, Lerner Research Institute and Respiratory Institute, Cleveland Clinic, OH (S.C.E.)
| | - Kerry S Russell
- From the Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine (M.H., I.D., Y.Z., P.M., P.J.L.), Section of Cardiovascular Disease (Y.H., K.S.R, F.G.), and Section of Endocrinology and Metabolism (M.J.J.), Yale University School of Medicine, New Haven, CT; and Department of Pathobiology, Lerner Research Institute and Respiratory Institute, Cleveland Clinic, OH (S.C.E.)
| | - Michael J Jurczak
- From the Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine (M.H., I.D., Y.Z., P.M., P.J.L.), Section of Cardiovascular Disease (Y.H., K.S.R, F.G.), and Section of Endocrinology and Metabolism (M.J.J.), Yale University School of Medicine, New Haven, CT; and Department of Pathobiology, Lerner Research Institute and Respiratory Institute, Cleveland Clinic, OH (S.C.E.)
| | - Praveen Mannam
- From the Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine (M.H., I.D., Y.Z., P.M., P.J.L.), Section of Cardiovascular Disease (Y.H., K.S.R, F.G.), and Section of Endocrinology and Metabolism (M.J.J.), Yale University School of Medicine, New Haven, CT; and Department of Pathobiology, Lerner Research Institute and Respiratory Institute, Cleveland Clinic, OH (S.C.E.)
| | - Frank Giordano
- From the Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine (M.H., I.D., Y.Z., P.M., P.J.L.), Section of Cardiovascular Disease (Y.H., K.S.R, F.G.), and Section of Endocrinology and Metabolism (M.J.J.), Yale University School of Medicine, New Haven, CT; and Department of Pathobiology, Lerner Research Institute and Respiratory Institute, Cleveland Clinic, OH (S.C.E.)
| | - Serpil C Erzurum
- From the Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine (M.H., I.D., Y.Z., P.M., P.J.L.), Section of Cardiovascular Disease (Y.H., K.S.R, F.G.), and Section of Endocrinology and Metabolism (M.J.J.), Yale University School of Medicine, New Haven, CT; and Department of Pathobiology, Lerner Research Institute and Respiratory Institute, Cleveland Clinic, OH (S.C.E.)
| | - Patty J Lee
- From the Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine (M.H., I.D., Y.Z., P.M., P.J.L.), Section of Cardiovascular Disease (Y.H., K.S.R, F.G.), and Section of Endocrinology and Metabolism (M.J.J.), Yale University School of Medicine, New Haven, CT; and Department of Pathobiology, Lerner Research Institute and Respiratory Institute, Cleveland Clinic, OH (S.C.E.).
| |
Collapse
|
327
|
Inhibition of the mitochondrial fission protein dynamin-related protein 1 improves survival in a murine cardiac arrest model. Crit Care Med 2015; 43:e38-47. [PMID: 25599491 DOI: 10.1097/ccm.0000000000000817] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
OBJECTIVES Survival following sudden cardiac arrest is poor despite advances in cardiopulmonary resuscitation and the use of therapeutic hypothermia. Dynamin-related protein 1, a regulator of mitochondrial fission, is an important determinant of reactive oxygen species generation, myocardial necrosis, and left ventricular function following ischemia/reperfusion injury, but its role in cardiac arrest is unknown. We hypothesized that dynamin-related protein 1 inhibition would improve survival, cardiac hemodynamics, and mitochondrial function in an in vivo model of cardiac arrest. DESIGN Laboratory investigation. SETTING University laboratory. INTERVENTIONS Anesthetized and ventilated adult female C57BL/6 wild-type mice underwent an 8-minute KCl-induced cardiac arrest followed by 90 seconds of cardiopulmonary resuscitation. Mice were then blindly randomized to a single IV injection of Mdivi-1 (0.24 mg/kg), a small molecule dynamin-related protein 1 inhibitor or vehicle (dimethyl sulfoxide). MEASUREMENTS AND MAIN RESULTS Following resuscitation from cardiac arrest, mitochondrial fission was evidenced by dynamin-related protein 1 translocation to the mitochondrial membrane and a decrease in mitochondrial size. Mitochondrial fission was associated with increased lactate and evidence of oxidative damage. Mdivi-1 administration during cardiopulmonary resuscitation inhibited dynamin-related protein 1 activation, preserved mitochondrial morphology, and decreased oxidative damage. Mdivi-1 also reduced the time to return of spontaneous circulation (116 ± 4 vs 143 ± 7 s; p < 0.001) during cardiopulmonary resuscitation and enhanced myocardial performance post-return of spontaneous circulation. These improvements were associated with significant increases in survival (65% vs 33%) and improved neurological scores up to 72 hours post cardiac arrest. CONCLUSIONS Post-cardiac arrest inhibition of dynamin-related protein 1 improves time to return of spontaneous circulation and myocardial hemodynamics, resulting in improved survival and neurological outcomes in a murine model of cardiac arrest. Pharmacological targeting of mitochondrial fission may be a promising therapy for cardiac arrest.
Collapse
|
328
|
Ryan J, Dasgupta A, Huston J, Chen KH, Archer SL. Mitochondrial dynamics in pulmonary arterial hypertension. J Mol Med (Berl) 2015; 93:229-42. [PMID: 25672499 DOI: 10.1007/s00109-015-1263-5] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 01/27/2015] [Accepted: 02/02/2015] [Indexed: 12/31/2022]
Abstract
Pulmonary arterial hypertension (PAH) is an idiopathic cardiopulmonary disease characterized by obstruction of small pulmonary arteries. Vascular obstruction is the consequence of excessive proliferation and apoptosis resistance of vascular cells, as well as inflammation, thrombosis, and vasoconstriction. Vascular obstruction increases the afterload faced by the right ventricle (RV), leading to RV failure. The proliferative, obstructive vasculopathy of PAH shares several mitochondrial abnormalities with cancer, notably a shift to aerobic glycolysis and mitochondrial fragmentation. Mitochondria in the pulmonary artery smooth muscle cell (PASMC) normally serve as oxygen sensors. In PAH, acquired mitochondrial abnormalities, including epigenetic silencing of superoxide dismutase (SOD2), disrupt oxygen sensing creating a pseudo-hypoxic environment characterized by normoxic activation of hypoxia-inducible factor-1α (HIF-1α). The resulting metabolic shift to aerobic glycolysis (the Warburg phenomenon) reflects inhibition of pyruvate dehydrogenase by pyruvate dehydrogenase kinases. In addition, altered mitochondrial dynamics result in mitochondrial fragmentation. The molecular basis of this structural change includes upregulation and activation of fission mediators, notably dynamin-related protein 1 (DRP-1), and downregulation of fusion mediators, especially mitofusin-2 (MFN2). These pathogenic mitochondrial abnormalities offer new therapeutic targets. Inhibition of mitotic fission or enhancement of fusion in PAH PASMC slows cell proliferation, causes cell cycle arrest, and induces apoptosis. DRP-1 inhibition or MFN2 gene therapy can regress PAH in experimental models of PAH. This review focuses on the etiology of mitochondrial fragmentation in PAH and explores the therapeutic implications of mitochondrial dynamics in the pulmonary vasculature and RV.
Collapse
Affiliation(s)
- John Ryan
- Division of Cardiovascular Medicine, Department of Medicine, University of Utah, Salt Lake City, UT, USA
| | | | | | | | | |
Collapse
|
329
|
Sharp WW, Archer SL. Mitochondrial dynamics in cardiovascular disease: fission and fusion foretell form and function. J Mol Med (Berl) 2015; 93:225-8. [PMID: 25669447 DOI: 10.1007/s00109-015-1258-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 12/03/2014] [Accepted: 01/26/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Willard W Sharp
- Division of Emergency Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | | |
Collapse
|
330
|
Sharp WW. Dynamin-related protein 1 as a therapeutic target in cardiac arrest. J Mol Med (Berl) 2015; 93:243-52. [PMID: 25659608 DOI: 10.1007/s00109-015-1257-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 01/13/2015] [Accepted: 01/26/2015] [Indexed: 12/23/2022]
Abstract
Despite improvements in cardiopulmonary resuscitation (CPR) quality, defibrillation technologies, and implementation of therapeutic hypothermia, less than 10 % of out-of-hospital cardiac arrest (OHCA) victims survive to hospital discharge. New resuscitation therapies have been slow to develop, in part, because the pathophysiologic mechanisms critical for resuscitation are not understood. During cardiac arrest, systemic cessation of blood flow results in whole body ischemia. CPR and the restoration of spontaneous circulation (ROSC), both result in immediate reperfusion injury of the heart that is characterized by severe contractile dysfunction. Unlike diseases of localized ischemia/reperfusion (IR) injury (myocardial infarction and stroke), global IR injury of organs results in profound organ dysfunction with far shorter ischemic times. The two most commonly injured organs following cardiac arrest resuscitation, the heart and brain, are critically dependent on mitochondrial function. New insights into mitochondrial dynamics and the role of the mitochondrial fission protein Dynamin-related protein 1 (Drp1) in apoptosis have made targeting these mechanisms attractive for IR therapy. In animal models, inhibiting Drp1 following IR injury or cardiac arrest confers protection to both the heart and brain. In this review, the relationship of the major mitochondrial fission protein Drp1 to ischemic changes in the heart and its targeting as a new therapeutic target following cardiac arrest are discussed.
Collapse
Affiliation(s)
- Willard W Sharp
- Section of Emergency Medicine, Department of Medicine, University of Chicago, 5841 S. Maryland Ave, MC 5068, Chicago, IL, 60637, USA,
| |
Collapse
|
331
|
Emerging therapies and future directions in pulmonary arterial hypertension. Can J Cardiol 2015; 31:489-501. [PMID: 25840098 DOI: 10.1016/j.cjca.2015.01.028] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 01/26/2015] [Accepted: 01/26/2015] [Indexed: 11/21/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a complex obliterative vascular disease. It remains deadly despite an explosion of basic research over the past 20 years that identified myriads of potential therapeutic targets, few of which have been translated into early phase trials. Despite the agreement over the past decade that its pathogenesis is based on an antiapoptotic and proproliferative environment within the pulmonary arterial wall, and not vasoconstriction, all the currently approved therapies were developed and tested in PAH because of their vasodilatory properties. Numerous potential therapies identified in preclinical research fail to be translated in clinical research. Here we discuss 7 concepts that might help address the "translational gap" in PAH. These include: a need to approach the "pulmonary arteries-right ventricle unit" comprehensively and develop right ventricle-specific therapies for heart failure; the metabolic and inflammatory theories of PAH that put many "diverse" abnormalities under 1 mechanistic roof, allowing the identification of more effective targets and biomarkers; the realization that PAH might be a systemic disease with primary abnormalities in extrapulmonary tissues including the right ventricle, skeletal muscle, immune system, and perhaps bone marrow, shifting our focus toward more systemic targets; the realization that many heritable components of PAH have an epigenetic basis that can be therapeutically targeted; and novel approaches like cell therapy or devices that can potentially improve access to transplanted organs. This progress marks the entrance into a new and exciting stage in our understanding and ability to fight this mysterious deadly disease.
Collapse
|
332
|
Wang L, Yu T, Lee H, O'Brien DK, Sesaki H, Yoon Y. Decreasing mitochondrial fission diminishes vascular smooth muscle cell migration and ameliorates intimal hyperplasia. Cardiovasc Res 2015; 106:272-83. [PMID: 25587046 DOI: 10.1093/cvr/cvv005] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 12/31/2014] [Indexed: 12/13/2022] Open
Abstract
AIMS Vascular smooth muscle cell (VSMC) migration in response to arterial wall injury is a critical process in the development of intimal hyperplasia. Cell migration is an energy-demanding process that is predicted to require mitochondrial function. Mitochondria are morphologically dynamic, undergoing continuous shape change through fission and fusion. However, the role of mitochondrial morphology in VSMC migration is not well understood. The aim of the study is to understand how mitochondrial fission contributes to VSMC migration and provides its in vivo relevance in the mouse model of intimal hyperplasia. METHODS AND RESULTS In primary mouse VSMCs, the chemoattractant PDGF induced mitochondrial shortening through the mitochondrial fission protein dynamin-like protein 1 (DLP1)/Drp1. Perturbation of mitochondrial fission by expressing the dominant-negative mutant DLP1-K38A or by DLP1 silencing greatly decreased PDGF-induced lamellipodia formation and VSMC migration, indicating that mitochondrial fission is an important process in VSMC migration. PDGF induced an augmentation of mitochondrial energetics as well as ROS production, both of which were found to be necessary for VSMC migration. Mechanistically, the inhibition of mitochondrial fission induced an increase of mitochondrial inner membrane proton leak in VSMCs, abrogating the PDGF-induced energetic enhancement and an ROS increase. In an in vivo model of intimal hyperplasia, transgenic mice expressing DLP1-K38A displayed markedly reduced ROS levels and neointima formation in response to femoral artery wire injury. CONCLUSIONS Mitochondrial fission is an integral process in cell migration, and controlling mitochondrial fission can limit VSMC migration and the pathological intimal hyperplasia by altering mitochondrial energetics and ROS levels.
Collapse
Affiliation(s)
- Li Wang
- Department of Physiology, Medical College of Georgia, Georgia Regents University, 1120 Fifteenth Street, Augusta, GA 30912-3000, USA
| | - Tianzheng Yu
- Department of Physiology, Medical College of Georgia, Georgia Regents University, 1120 Fifteenth Street, Augusta, GA 30912-3000, USA
| | - Hakjoo Lee
- Department of Physiology, Medical College of Georgia, Georgia Regents University, 1120 Fifteenth Street, Augusta, GA 30912-3000, USA
| | - Dawn K O'Brien
- Department of Physiology, Medical College of Georgia, Georgia Regents University, 1120 Fifteenth Street, Augusta, GA 30912-3000, USA
| | - Hiromi Sesaki
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yisang Yoon
- Department of Physiology, Medical College of Georgia, Georgia Regents University, 1120 Fifteenth Street, Augusta, GA 30912-3000, USA
| |
Collapse
|
333
|
Ikeda Y, Shirakabe A, Brady C, Zablocki D, Ohishi M, Sadoshima J. Molecular mechanisms mediating mitochondrial dynamics and mitophagy and their functional roles in the cardiovascular system. J Mol Cell Cardiol 2015; 78:116-22. [PMID: 25305175 PMCID: PMC4268018 DOI: 10.1016/j.yjmcc.2014.09.019] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 09/12/2014] [Accepted: 09/16/2014] [Indexed: 02/05/2023]
Abstract
Mitochondria are essential organelles that produce the cellular energy source, ATP. Dysfunctional mitochondria are involved in the pathophysiology of heart disease, which is associated with reduced levels of ATP and excessive production of reactive oxygen species. Mitochondria are dynamic organelles that change their morphology through fission and fusion in order to maintain their function. Fusion connects neighboring depolarized mitochondria and mixes their contents to maintain membrane potential. In contrast, fission segregates damaged mitochondria from intact ones, where the damaged part of mitochondria is subjected to mitophagy whereas the intact part to fusion. It is generally believed that mitochondrial fusion is beneficial for the heart, especially under stress conditions, because it consolidates the mitochondria's ability to supply energy. However, both excessive fusion and insufficient fission disrupt the mitochondrial quality control mechanism and potentiate cell death. In this review, we discuss the role of mitochondrial dynamics and mitophagy in the heart and the cardiomyocytes therein, with a focus on their roles in cardiovascular disease. This article is part of a Special Issue entitled "Mitochondria: From Basic Mitochondrial Biology to Cardiovascular Disease".
Collapse
Affiliation(s)
- Yoshiyuki Ikeda
- Department of Cell Biology & Molecular Medicine, NJ Medical School, Rutgers University, USA; Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan
| | - Akihiro Shirakabe
- Department of Cell Biology & Molecular Medicine, NJ Medical School, Rutgers University, USA
| | - Christopher Brady
- Department of Cell Biology & Molecular Medicine, NJ Medical School, Rutgers University, USA
| | - Daniela Zablocki
- Department of Cell Biology & Molecular Medicine, NJ Medical School, Rutgers University, USA
| | - Mitsuru Ohishi
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan
| | - Junichi Sadoshima
- Department of Cell Biology & Molecular Medicine, NJ Medical School, Rutgers University, USA.
| |
Collapse
|
334
|
Chiong M, Cartes-Saavedra B, Norambuena-Soto I, Mondaca-Ruff D, Morales PE, García-Miguel M, Mellado R. Mitochondrial metabolism and the control of vascular smooth muscle cell proliferation. Front Cell Dev Biol 2014; 2:72. [PMID: 25566542 PMCID: PMC4266092 DOI: 10.3389/fcell.2014.00072] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 11/28/2014] [Indexed: 12/12/2022] Open
Abstract
Differentiation and dedifferentiation of vascular smooth muscle cells (VSMCs) are essential processes of vascular development. VSMC have biosynthetic, proliferative, and contractile roles in the vessel wall. Alterations in the differentiated state of the VSMC play a critical role in the pathogenesis of a variety of cardiovascular diseases, including atherosclerosis, hypertension, and vascular stenosis. This review provides an overview of the current state of knowledge of molecular mechanisms involved in the control of VSMC proliferation, with particular focus on mitochondrial metabolism. Mitochondrial activity can be controlled by regulating mitochondrial dynamics, i.e., mitochondrial fusion and fission, and by regulating mitochondrial calcium handling through the interaction with the endoplasmic reticulum (ER). Alterations in both VSMC proliferation and mitochondrial function can be triggered by dysregulation of mitofusin-2, a small GTPase associated with mitochondrial fusion and mitochondrial–ER interaction. Several lines of evidence highlight the relevance of mitochondrial metabolism in the control of VSMC proliferation, indicating a new area to be explored in the treatment of vascular diseases.
Collapse
Affiliation(s)
- Mario Chiong
- Faculty of Chemical and Pharmaceutical Sciences, Advanced Center for Chronic Diseases, University of Chile Santiago, Chile
| | - Benjamín Cartes-Saavedra
- Faculty of Chemical and Pharmaceutical Sciences, Advanced Center for Chronic Diseases, University of Chile Santiago, Chile
| | - Ignacio Norambuena-Soto
- Faculty of Chemical and Pharmaceutical Sciences, Advanced Center for Chronic Diseases, University of Chile Santiago, Chile
| | - David Mondaca-Ruff
- Faculty of Chemical and Pharmaceutical Sciences, Advanced Center for Chronic Diseases, University of Chile Santiago, Chile
| | - Pablo E Morales
- Faculty of Chemical and Pharmaceutical Sciences, Advanced Center for Chronic Diseases, University of Chile Santiago, Chile
| | - Marina García-Miguel
- Faculty of Chemical and Pharmaceutical Sciences, Advanced Center for Chronic Diseases, University of Chile Santiago, Chile
| | - Rosemarie Mellado
- Faculty of Chemistry, Pontifical Catholic University of Chile Santiago, Chile
| |
Collapse
|
335
|
Hall AR, Burke N, Dongworth RK, Hausenloy DJ. Mitochondrial fusion and fission proteins: novel therapeutic targets for combating cardiovascular disease. Br J Pharmacol 2014; 171:1890-906. [PMID: 24328763 PMCID: PMC3976611 DOI: 10.1111/bph.12516] [Citation(s) in RCA: 207] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 10/21/2013] [Accepted: 10/28/2013] [Indexed: 12/18/2022] Open
Abstract
Mitochondria are no longer considered to be solely the static powerhouses of the cell. While they are undoubtedly essential to sustaining life and meeting the energy requirements of the cell through oxidative phosphorylation, they are now regarded as highly dynamic organelles with multiple functions, playing key roles in cell survival and death. In this review, we discuss the emerging role of mitochondrial fusion and fission proteins, as novel therapeutic targets for treating a wide range of cardiovascular diseases.
Collapse
Affiliation(s)
- A R Hall
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, NIHR University College London Hospitals Biomedical Research Centre, University College London Hospital & Medical School, London, UK
| | | | | | | |
Collapse
|
336
|
Paulin R, Dromparis P, Sutendra G, Gurtu V, Zervopoulos S, Bowers L, Haromy A, Webster L, Provencher S, Bonnet S, Michelakis ED. Sirtuin 3 deficiency is associated with inhibited mitochondrial function and pulmonary arterial hypertension in rodents and humans. Cell Metab 2014; 20:827-839. [PMID: 25284742 DOI: 10.1016/j.cmet.2014.08.011] [Citation(s) in RCA: 161] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 07/02/2014] [Accepted: 08/18/2014] [Indexed: 12/19/2022]
Abstract
Suppression of mitochondrial function promoting proliferation and apoptosis suppression has been described in the pulmonary arteries and extrapulmonary tissues in pulmonary arterial hypertension (PAH), but the cause of this metabolic remodeling is unknown. Mice lacking sirtuin 3 (SIRT3), a mitochondrial deacetylase, have increased acetylation and inhibition of many mitochondrial enzymes and complexes, suppressing mitochondrial function. Sirt3KO mice develop spontaneous PAH, exhibiting previously described molecular features of PAH pulmonary artery smooth muscle cells (PASMC). In human PAH PASMC and rats with PAH, SIRT3 is downregulated, and its normalization with adenovirus gene therapy reverses the disease phenotype. A loss-of-function SIRT3 polymorphism, linked to metabolic syndrome, is associated with PAH in an unbiased cohort of 162 patients and controls. If confirmed in large patient cohorts, these findings may facilitate biomarker and therapeutic discovery programs in PAH.
Collapse
Affiliation(s)
- Roxane Paulin
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2B7, Canada
| | - Peter Dromparis
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2B7, Canada
| | - Gopinath Sutendra
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2B7, Canada
| | - Vikram Gurtu
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2B7, Canada
| | | | - Lyndsay Bowers
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2B7, Canada
| | - Alois Haromy
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2B7, Canada
| | - Linda Webster
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2B7, Canada
| | - Steeve Provencher
- Department of Medicine, Laval University, IUCPQ Research Centre, Pulmonary Hypertension Research Group, Quebec, QC G1V 4G5, Canada
| | - Sebastien Bonnet
- Department of Medicine, Laval University, IUCPQ Research Centre, Pulmonary Hypertension Research Group, Quebec, QC G1V 4G5, Canada
| | | |
Collapse
|
337
|
PINK1 signalling in cancer biology. Biochim Biophys Acta Rev Cancer 2014; 1846:590-8. [PMID: 25450579 DOI: 10.1016/j.bbcan.2014.10.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 09/29/2014] [Accepted: 10/20/2014] [Indexed: 02/08/2023]
Abstract
PTEN-induced kinase 1 (PINK1) was identified initially in cancer cells as a gene up-regulated by overexpression of the major tumor suppressor, PTEN. Loss-of-function mutations in PINK1 were discovered subsequently to cause autosomal recessive Parkinson's disease. Substantial work during the past decade has revealed that PINK1 regulates several primary cellular processes of significance in cancer cell biology, including cell survival, stress resistance, mitochondrial homeostasis and the cell cycle. Mechanistically, PINK1 has been shown to interact on a number of levels with the pivotal oncogenic PI3-kinase/Akt/mTOR signalling axis and to control critical mitochondrial and metabolic functions that regulate cancer survival, growth, stress resistance and the cell cycle. A cytoprotective and chemoresistant function for PINK1 has been highlighted by some studies, supporting PINK1 as a target in cancer therapeutics. This article reviews the function of PINK1 in cancer cell biology, with an emphasis on the mechanisms by which PINK1 interacts with PI3-kinase/Akt signalling, mitochondrial homeostasis, and the potential context-dependent pro- and anti-tumorigenic functions of PINK1.
Collapse
|
338
|
Mitochondria: roles in pulmonary hypertension. Int J Biochem Cell Biol 2014; 55:93-7. [PMID: 25149415 DOI: 10.1016/j.biocel.2014.08.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 08/13/2014] [Indexed: 11/22/2022]
Abstract
Mitochondria are essential cell organelles responsible for ATP production in the presence of oxygen. In the pulmonary vasculature, mitochondria contribute to physiological intracellular signalling pathways through production of reactive oxygen species and play the role of oxygen sensors that coordinate hypoxic pulmonary vasoconstriction. Mitochondria also play a pathophysiological role in pulmonary hypertension (PH). This disease is characterized by increased pulmonary arterial pressure and remodelling of pulmonary arteries, leading to increased pulmonary vascular resistance, hypertrophy of the right ventricle, right heart failure and ultimately death. Mitochondrial alterations have been evidenced in PH in pulmonary arteries and in the right ventricle, in particular a chronic shift in energy production from mitochondrial oxidative phosphorylation to glycolysis. This shift, initially described in cancer cells, may play a central role in PH pathogenesis. Further studies of these metabolic mitochondrial alterations in PH may therefore open new therapeutic perspectives in this disease.
Collapse
|
339
|
Affiliation(s)
- Roxane Paulin
- From the Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | | |
Collapse
|
340
|
Wan YY, Zhang JF, Yang ZJ, Jiang LP, Wei YF, Lai QN, Wang JB, Xin HB, Han XJ. Involvement of Drp1 in hypoxia-induced migration of human glioblastoma U251 cells. Oncol Rep 2014; 32:619-26. [PMID: 24899388 DOI: 10.3892/or.2014.3235] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Accepted: 05/21/2014] [Indexed: 11/06/2022] Open
Abstract
Glioblastoma is one of the most aggressive brain tumors with high morbidity and mortality. Hypoxia is often the common characteristic of tumor microenvironment, and hypoxia-inducible factor-1α (HIF-1α) is an essential factor regulating the migratory activity of cancer cells including glioblastoma. Recently, mitochondrial dynamics was found to be involved in the aggression of cancer cells. However, whether dynamin-related protein 1 (Drp1) contributes to the migration of human glioblastoma cells under hypoxia remains unknown. In the present study, hypoxia was found to upregulate the transcription and expression of Drp1, and stimulated mitochondrial fission in glioblastoma U251 cells. Inhibition of HIF-1α with echinomycin blocked hypoxia‑induced expression of Drp1. Notably, Drp1 inhibitor Mdivi-1 efficiently attenuated hypoxia-induced mitochondrial fission and migration of U251 cells. In addition, three U251 stable cell lines expressing GFP, GFP-Drp1 and dominant negative GFP-Drp1‑K38A were established to examine the direct role of Drp1 in hypoxia-induced migration. MTT assay showed that there was no significant difference in proliferation of three cell lines. Compared with the GFP cell line, exogenously expressed GFP-Drp1-K38A inhibited hypoxia-induced migration of U251 cells, while stable expression of GFP-Drp1 enhanced the migration of U251 cells under hypoxia. Therefore, this study indicates the involvement of Drp1 in hypoxia-induced migration of human glioblastoma U251 cells, and suggests Drp1 to be a potential therapeutic target to suppress the aggression of glioblastoma in the future.
Collapse
Affiliation(s)
- Yu-Ying Wan
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Jian-Feng Zhang
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Zhang-Jian Yang
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Li-Ping Jiang
- Department of Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang, Jiangxi, P.R. China
| | - Yong-Fang Wei
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Qi-Nan Lai
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Jian-Bin Wang
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Hong-Bo Xin
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Xiao-Jian Han
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, P.R. China
| |
Collapse
|
341
|
Zhang X, Li ZL, Crane JA, Jordan KL, Pawar AS, Textor SC, Lerman A, Lerman LO. Valsartan regulates myocardial autophagy and mitochondrial turnover in experimental hypertension. Hypertension 2014; 64:87-93. [PMID: 24752430 DOI: 10.1161/hypertensionaha.113.02151] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Renovascular hypertension alters cardiac structure and function. Autophagy is activated during left ventricular hypertrophy and linked to adverse cardiac function. The angiotensin II receptor blocker, valsartan, lowers blood pressure and is cardioprotective, but whether it modulates autophagy in the myocardium is unclear. We hypothesized that valsartan would alleviate autophagy and improve left ventricular myocardial mitochondrial turnover in swine renovascular hypertension. Domestic pigs were randomized to control, unilateral renovascular hypertension, and renovascular hypertension treated with valsartan (320 mg/d) or conventional triple therapy (reserpine+hydralazine+hydrochlorothiazide) for 4 weeks after 6 weeks of renovascular hypertension (n=7 each group). Left ventricular remodeling, function, and myocardial oxygenation and microcirculation were assessed by multidetector computer tomography, blood oxygen level-dependent MRI, and microcomputer tomography. Myocardial autophagy, markers for mitochondrial degradation and biogenesis, and mitochondrial respiratory-chain proteins were examined ex vivo. Renovascular hypertension induced left ventricular hypertrophy and myocardial hypoxia, enhanced cellular autophagy and mitochondrial degradation, and suppressed mitochondrial biogenesis. Valsartan and triple therapy similarly decreased blood pressure, but valsartan solely alleviated left ventricular hypertrophy, ameliorated myocardial autophagy and mitophagy, and increased mitochondrial biogenesis. In contrast, triple therapy only slightly attenuated autophagy and preserved mitochondrial proteins, but elicited no improvement in mitophagy. These data suggest a novel potential role of valsartan in modulating myocardial autophagy and mitochondrial turnover in renovascular hypertension-induced hypertensive heart disease, which may possibly bolster cardiac repair via a blood pressure-independent manner.
Collapse
Affiliation(s)
- Xin Zhang
- From the Division of Nephrology and Hypertension (X.Z., Z.-L.L., J.A.C., K.L.J., A.S.P., S.C.T., L.O.L.) and Division of Cardiovascular Diseases (A.L., L.O.L.), Mayo Clinic, Rochester, MN; and Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China (Z.-L.L.)
| | - Zi-Lun Li
- From the Division of Nephrology and Hypertension (X.Z., Z.-L.L., J.A.C., K.L.J., A.S.P., S.C.T., L.O.L.) and Division of Cardiovascular Diseases (A.L., L.O.L.), Mayo Clinic, Rochester, MN; and Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China (Z.-L.L.)
| | - John A Crane
- From the Division of Nephrology and Hypertension (X.Z., Z.-L.L., J.A.C., K.L.J., A.S.P., S.C.T., L.O.L.) and Division of Cardiovascular Diseases (A.L., L.O.L.), Mayo Clinic, Rochester, MN; and Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China (Z.-L.L.)
| | - Kyra L Jordan
- From the Division of Nephrology and Hypertension (X.Z., Z.-L.L., J.A.C., K.L.J., A.S.P., S.C.T., L.O.L.) and Division of Cardiovascular Diseases (A.L., L.O.L.), Mayo Clinic, Rochester, MN; and Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China (Z.-L.L.)
| | - Aditya S Pawar
- From the Division of Nephrology and Hypertension (X.Z., Z.-L.L., J.A.C., K.L.J., A.S.P., S.C.T., L.O.L.) and Division of Cardiovascular Diseases (A.L., L.O.L.), Mayo Clinic, Rochester, MN; and Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China (Z.-L.L.)
| | - Stephen C Textor
- From the Division of Nephrology and Hypertension (X.Z., Z.-L.L., J.A.C., K.L.J., A.S.P., S.C.T., L.O.L.) and Division of Cardiovascular Diseases (A.L., L.O.L.), Mayo Clinic, Rochester, MN; and Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China (Z.-L.L.)
| | - Amir Lerman
- From the Division of Nephrology and Hypertension (X.Z., Z.-L.L., J.A.C., K.L.J., A.S.P., S.C.T., L.O.L.) and Division of Cardiovascular Diseases (A.L., L.O.L.), Mayo Clinic, Rochester, MN; and Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China (Z.-L.L.)
| | - Lilach O Lerman
- From the Division of Nephrology and Hypertension (X.Z., Z.-L.L., J.A.C., K.L.J., A.S.P., S.C.T., L.O.L.) and Division of Cardiovascular Diseases (A.L., L.O.L.), Mayo Clinic, Rochester, MN; and Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China (Z.-L.L.).
| |
Collapse
|
342
|
Wang Y, Tabas I. Emerging roles of mitochondria ROS in atherosclerotic lesions: causation or association? J Atheroscler Thromb 2014; 21:381-90. [PMID: 24717761 DOI: 10.5551/jat.23929] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Mitochondrial-derived reactive oxygen species (mtROS) is one of the major sources of cellular ROS, and excessive mtROS is associated with atherosclerosis progression in both human and mouse models. This review aims to summarize the most recent studies showing the existence, the causes and pathological consequences of excessive mtROS in atherosclerosis. Despite numerous association and causation studies demonstrating the importance of mtROS in atherosclerosis progression, the failure of antioxidant therapy in human randomized clinical trials demands more definitive, cell-type specific investigations. Better mechanistic understanding of mtROS in atherosclerosis may lead to more effective therapeutic strategies.
Collapse
|
343
|
Abstract
Pulmonary arterial hypertension (PAH) is a vascular remodeling disease of the lungs resulting in heart failure and premature death. Although, until recently, it was thought that PAH pathology is restricted to pulmonary arteries, several extrapulmonary organs are also affected. The realization that these tissues share a common metabolic abnormality (i.e., suppression of mitochondrial glucose oxidation and increased glycolysis) is important for our understanding of PAH, if not a paradigm shift. Here, we discuss an emerging metabolic theory, which proposes that PAH should be viewed as a syndrome involving many organs sharing a mitochondrial abnormality and explains many PAH features and provides novel biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Gopinath Sutendra
- Department of Medicine, University of Alberta, 2C2 Walter Mackenzie Centre, 8440 112 Street Northwest, Edmonton, AB T6G 2P4, Canada
| | - Evangelos D Michelakis
- Department of Medicine, University of Alberta, 2C2 Walter Mackenzie Centre, 8440 112 Street Northwest, Edmonton, AB T6G 2P4, Canada.
| |
Collapse
|
344
|
Alexandrou AT, Li JJ. Cell cycle regulators guide mitochondrial activity in radiation-induced adaptive response. Antioxid Redox Signal 2014; 20:1463-80. [PMID: 24180340 PMCID: PMC3936506 DOI: 10.1089/ars.2013.5684] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
SIGNIFICANCE There are accruing concerns on potential genotoxic agents present in the environment including low-dose ionizing radiation (LDIR) that naturally exists on earth's surface and atmosphere and is frequently used in medical diagnosis and nuclear industry. Although its long-term health risk is being evaluated and remains controversial, LDIR is shown to induce temporary but significant adaptive responses in mammalian cells and animals. The mechanisms guiding the mitochondrial function in LDIR-induced adaptive response represent a unique communication between DNA damage and cellular metabolism. Elucidation of the LDIR-regulated mitochondrial activity may reveal new mechanisms adjusting cellular function to cope with hazardous environmental stress. RECENT ADVANCES Key cell cycle regulators, including Cyclin D1/CDK4 and Cyclin B1/cyclin-dependent kinase 1 (CDK1) complexes, are actively involved in the regulation of mitochondrial functions via phosphorylation of their mitochondrial targets. Accumulating new evidence supports a concept that the Cyclin B1/CDK1 complex acts as a mediator in the cross talk between radiation-induced DNA damage and mitochondrial functions to coordinate cellular responses to low-level genotoxic stresses. CRITICAL ISSUES The LDIR-mediated mitochondrial activity via Cyclin B1/CDK1 regulation is an irreplaceable network that is able to harmonize vital cellular functions with adjusted mitochondrial metabolism to enhance cellular homeostasis. FUTURE DIRECTIONS Further investigation of the coordinative mechanism that regulates mitochondrial activities in sublethal stress conditions, including LDIR, will reveal new insights of how cells cope with genotoxic injury and will be vital for future targeted therapeutic interventions that reduce environmental injury and cancer risk.
Collapse
Affiliation(s)
- Aris T Alexandrou
- Department of Radiation Oncology, NCI-Designated Comprehensive Cancer Center, University of California at Davis , Sacramento, California
| | | |
Collapse
|
345
|
Tuder RM, Archer SL, Dorfmüller P, Erzurum SC, Guignabert C, Michelakis E, Rabinovitch M, Schermuly R, Stenmark KR, Morrell NW. Relevant issues in the pathology and pathobiology of pulmonary hypertension. J Am Coll Cardiol 2014; 62:D4-12. [PMID: 24355640 DOI: 10.1016/j.jacc.2013.10.025] [Citation(s) in RCA: 422] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 10/22/2013] [Indexed: 11/18/2022]
Abstract
Knowledge of the pathobiology of pulmonary hypertension (PH) continues to accelerate. However, fundamental gaps remain in our understanding of the underlying pathological changes in pulmonary arteries and veins in the different forms of this syndrome. Although PH primarily affects the arteries, venous disease is increasingly recognized as an important entity. Moreover, prognosis in PH is determined largely by the status of the right ventricle, rather than the levels of pulmonary artery pressures. It is increasingly clear that although vasospasm plays a role, PH is an obstructive lung panvasculopathy. Disordered metabolism and mitochondrial structure, inflammation, and dysregulation of growth factors lead to a proliferative, apoptosis-resistant state. These abnormalities may be acquired, genetically mediated as a result of mutations in bone morphogenetic protein receptor-2 or activin-like kinase-1, or epigenetically inherited (as a result of epigenetic silencing of genes such as superoxide dismutase-2). There is a pressing need to better understand how the pathobiology leads to severe disease in some patients versus mild PH in others. Recent recognition of a potential role of acquired abnormalities of mitochondrial metabolism in the right ventricular myocytes and pulmonary vascular cells suggests new therapeutic approaches, diagnostic modalities, and biomarkers. Finally, dissection of the role of pulmonary inflammation in the initiation and promotion of PH has revealed a complex yet fascinating interplay with pulmonary vascular remodeling, promising to lead to novel therapeutics and diagnostics. Emerging concepts are also relevant to the pathobiology of PH, including a role for bone marrow and circulating progenitor cells and microribonucleic acids. Continued interest in the interface of the genetic basis of PH and cellular and molecular pathogenetic links should further expand our understanding of the disease.
Collapse
Affiliation(s)
- Rubin M Tuder
- Program in Translational Lung Research, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.
| | - Stephen L Archer
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Peter Dorfmüller
- Department of Pathology, Marie Lannelongue Hospital, University Paris-Sud, Le Plessis-Robinson, France
| | - Serpil C Erzurum
- Lerner Research Institute and Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - Christophe Guignabert
- INSERM UMR 999, LabEx LERMIT, Marie Lannelongue Hospital and University Paris-Sud, School of Medicine, Kremlin-Bicêtre, France
| | | | - Marlene Rabinovitch
- Cardiovascular Institute and Department of Pediatrics and The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, California
| | - Ralph Schermuly
- Excellence Cluster Cardio-Pulmonary System, German Lung Center, Universities of Giessen and Marburg Lung Center, Justus-Liebig-University, Giessen, Germany
| | - Kurt R Stenmark
- Cardiovascular Pulmonary Laboratory, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom.
| |
Collapse
|
346
|
Aravamudan B, Kiel A, Freeman M, Delmotte P, Thompson M, Vassallo R, Sieck GC, Pabelick CM, Prakash YS. Cigarette smoke-induced mitochondrial fragmentation and dysfunction in human airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 2014; 306:L840-54. [PMID: 24610934 DOI: 10.1152/ajplung.00155.2013] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The balance between mitochondrial fission and fusion is crucial for mitochondria to perform its normal cellular functions. We hypothesized that cigarette smoke (CS) disrupts this balance and enhances mitochondrial dysfunction in the airway. In nonasthmatic human airway smooth muscle (ASM) cells, CS extract (CSE) induced mitochondrial fragmentation and damages their networked morphology in a concentration-dependent fashion, via increased expression of mitochondrial fission protein dynamin-related protein 1 (Drp1) and decreased fusion protein mitofusin (Mfn) 2. CSE effects on Drp1 vs. Mfn2 and mitochondrial network morphology involved reactive oxygen species (ROS), activation of extracellular signal-regulated kinase (ERK), phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt), protein kinase C (PKC) and proteasome pathways, as well as transcriptional regulation via factors such as NF-κB and nuclear erythroid 2-related factor 2. Inhibiting Drp1 prevented CSE effects on mitochondrial networks and ROS generation, whereas blocking Mfn2 had the opposite, detrimental effect. In ASM from asmatic patients, mitochondria exhibited substantial morphological defects at baseline and showed increased Drp1 but decreased Mfn2 expression, with exacerbating effects of CSE. Overall, these results highlight the importance of mitochondrial networks and their regulation in the context of cellular changes induced by insults such as inflammation (as in asthma) or CS. Altered mitochondrial fission/fusion proteins have a further potential to influence parameters such as ROS and cell proliferation and apoptosis relevant to airway diseases.
Collapse
Affiliation(s)
- Bharathi Aravamudan
- Division of Anesthesia Research, Dept. of Anesthesiology, 4-184 W. Joseph SMH, Mayo Clinic, Rochester, MN 55905.
| | | | | | | | | | | | | | | | | |
Collapse
|
347
|
Endothelial progenitor cells and pulmonary arterial hypertension. Heart Lung Circ 2014; 23:595-601. [PMID: 24680485 DOI: 10.1016/j.hlc.2014.02.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Accepted: 02/17/2014] [Indexed: 01/23/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease characterised by lung endothelial cell dysfunction and vascular remodelling. A number of studies now suggest that endothelial progenitor cells (EPCs) may induce neovascularisation and could be a promising approach for cell based therapy for PAH. On the contrary EPCs may contribute to pulmonary vascular remodelling, particularly in end-stage pulmonary disease. This review article will provide a brief summary of the relationship between PAH and EPCs, the application of the EPCs to PAH and highlight the potential clinical application of the EPCs cell therapy to PAH.
Collapse
|
348
|
Affiliation(s)
- Bradford G Hill
- From the Institute of Molecular Cardiology, Department of Medicine, Diabetes and Obesity Center, Department of Biochemistry and Molecular Biology, and Department of Physiology and Biophysics, University of Louisville, KY
| |
Collapse
|
349
|
Nakahira K, Cloonan SM, Mizumura K, Choi AMK, Ryter SW. Autophagy: a crucial moderator of redox balance, inflammation, and apoptosis in lung disease. Antioxid Redox Signal 2014; 20:474-94. [PMID: 23879400 PMCID: PMC3894710 DOI: 10.1089/ars.2013.5373] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
SIGNIFICANCE Autophagy is a fundamental cellular process that functions in the turnover of subcellular organelles and protein. Activation of autophagy may represent a cellular defense against oxidative stress, or related conditions that cause accumulation of damaged proteins or organelles. Selective forms of autophagy can maintain organelle populations or remove aggregated proteins. Autophagy can increase survival during nutrient deficiency and play a multifunctional role in host defense, by promoting pathogen clearance and modulating innate and adaptive immune responses. RECENT ADVANCES Autophagy has been described as an inducible response to oxidative stress. Once believed to represent a random process, recent studies have defined selective mechanisms for cargo assimilation into autophagosomes. Such mechanisms may provide for protein aggregate detoxification and mitochondrial homeostasis during oxidative stress. Although long studied as a cellular phenomenon, recent advances implicate autophagy as a component of human diseases. Altered autophagy phenotypes have been observed in various human diseases, including lung diseases such as chronic obstructive lung disease, cystic fibrosis, pulmonary hypertension, and idiopathic pulmonary fibrosis. CRITICAL ISSUES Although autophagy can represent a pro-survival process, in particular, during nutrient starvation, its role in disease pathogenesis may be multifunctional and complex. The relationship of autophagy to programmed cell death pathways is incompletely defined and varies with model system. FUTURE DIRECTIONS Activation or inhibition of autophagy may be used to alter the progression of human diseases. Further resolution of the mechanisms by which autophagy impacts the initiation and progression of diseases may lead to the development of therapeutics specifically targeting this pathway.
Collapse
Affiliation(s)
- Kiichi Nakahira
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital , Harvard Medical School, Boston, Massachusetts
| | | | | | | | | |
Collapse
|
350
|
Fessel JP, Flynn CR, Robinson LJ, Penner NL, Gladson S, Kang CJ, Wasserman DH, Hemnes AR, West JD. Hyperoxia synergizes with mutant bone morphogenic protein receptor 2 to cause metabolic stress, oxidant injury, and pulmonary hypertension. Am J Respir Cell Mol Biol 2013; 49:778-87. [PMID: 23742019 DOI: 10.1165/rcmb.2012-0463oc] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) has been associated with a number of different but interrelated pathogenic mechanisms. Metabolic and oxidative stresses have been shown to play important pathogenic roles in a variety of model systems. However, many of these relationships remain at the level of association. We sought to establish a direct role for metabolic stress and oxidant injury in the pathogenesis of PAH. Mice that universally express a disease-causing mutation in bone morphogenic protein receptor 2 (Bmpr2) were exposed to room air or to brief daily hyperoxia (95% oxygen for 3 h) for 6 weeks, and were compared with wild-type animals undergoing identical exposures. In both murine tissues and cultured endothelial cells, the expression of mutant Bmpr2 was sufficient to cause oxidant injury that was particularly pronounced in mitochondrial membranes. With the enhancement of mitochondrial generation of reactive oxygen species by hyperoxia, oxidant injury was substantially enhanced in mitochondrial membranes, even in tissues distant from the lung. Hyperoxia, despite its vasodilatory actions in the pulmonary circulation, significantly worsened the PAH phenotype (elevated right ventricular systolic pressure, decreased cardiac output, and increased pulmonary vascular occlusion) in Bmpr2 mutant animals. These experiments demonstrate that oxidant injury and metabolic stress contribute directly to disease development, and provide further evidence for PAH as a systemic disease with life-limiting cardiopulmonary manifestations.
Collapse
Affiliation(s)
- Joshua P Fessel
- 1 Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine
| | | | | | | | | | | | | | | | | |
Collapse
|