1
|
Qin P, He C, Ye P, Li Q, Cai C, Li Y. PKCδ regulates the vascular biology in diabetic atherosclerosis. Cell Commun Signal 2023; 21:330. [PMID: 37974282 PMCID: PMC10652453 DOI: 10.1186/s12964-023-01361-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 10/20/2023] [Indexed: 11/19/2023] Open
Abstract
Diabetes mellitus, known for its complications, especially vascular complications, is becoming a globally serious social problem. Atherosclerosis has been recognized as a common vascular complication mechanism in diabetes. The diacylglycerol (DAG)-protein kinase C (PKC) pathway plays an important role in atherosclerosis. PKCs can be divided into three subgroups: conventional PKCs (cPKCs), novel PKCs (nPKCs), and atypical PKCs (aPKCs). The aim of this review is to provide a comprehensive overview of the role of the PKCδ pathway, an isoform of nPKC, in regulating the function of endothelial cells, vascular smooth muscle cells, and macrophages in diabetic atherosclerosis. In addition, potential therapeutic targets regarding the PKCδ pathway are summarized. Video Abstract.
Collapse
Affiliation(s)
- Peiliang Qin
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Changhuai He
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Pin Ye
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qin Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chuanqi Cai
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Yiqing Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
2
|
Ohm AM, Affandi T, Reisz JA, Caino MC, D'Alessandro A, Reyland ME. Metabolic reprogramming contributes to radioprotection by protein kinase Cδ. J Biol Chem 2023; 299:105186. [PMID: 37611829 PMCID: PMC10519828 DOI: 10.1016/j.jbc.2023.105186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 08/25/2023] Open
Abstract
Loss of protein kinase Cδ (PKCδ) activity renders cells resistant to DNA damaging agents, including irradiation; however, the mechanism(s) underlying resistance is poorly understood. Here, we have asked if metabolic reprogramming by PKCδ contributes to radioprotection. Analysis of global metabolomics showed that depletion of PKCδ affects metabolic pathways that control energy production and antioxidant, nucleotide, and amino acid biosynthesis. Increased NADPH and nucleotide production in PKCδ-depleted cells is associated with upregulation of the pentose phosphate pathway (PPP) as evidenced by increased activation of G6PD and an increase in the nucleotide precursor, 5-phosphoribosyl-1-pyrophosphate. Stable isotope tracing with U-[13C6] glucose showed reduced utilization of glucose for glycolysis in PKCδ-depleted cells and no increase in U-[13C6] glucose incorporation into purines or pyrimidines. In contrast, isotope tracing with [13C5, 15N2] glutamine showed increased utilization of glutamine for synthesis of nucleotides, glutathione, and tricarboxylic acid intermediates and increased incorporation of labeled glutamine into pyruvate and lactate. Using a glycolytic rate assay, we confirmed that anaerobic glycolysis is increased in PKCδ-depleted cells; this was accompanied by a reduction in oxidative phosphorylation, as assayed using a mitochondrial stress assay. Importantly, pretreatment of cells with specific inhibitors of the PPP or glutaminase prior to irradiation reversed radioprotection in PKCδ-depleted cells, indicating that these cells have acquired codependency on the PPP and glutamine for survival. Our studies demonstrate that metabolic reprogramming to increase utilization of glutamine and nucleotide synthesis contributes to radioprotection in the context of PKCδ inhibition.
Collapse
Affiliation(s)
- Angela M Ohm
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Trisiani Affandi
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - M Cecilia Caino
- Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Mary E Reyland
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.
| |
Collapse
|
3
|
Loots DT, Adeniji AA, Van Reenen M, Ozturk M, Brombacher F, Parihar SP. The metabolomics of a protein kinase C delta (PKCδ) knock-out mouse model. Metabolomics 2022; 18:92. [PMID: 36371785 PMCID: PMC9660189 DOI: 10.1007/s11306-022-01949-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 10/29/2022] [Indexed: 11/15/2022]
Abstract
INTRODUCTION PKCδ is ubiquitously expressed in mammalian cells and its dysregulation plays a key role in the onset of several incurable diseases and metabolic disorders. However, much remains unknown about the metabolic pathways and disturbances induced by PKC deficiency, as well as the metabolic mechanisms involved. OBJECTIVES This study aims to use metabolomics to further characterize the function of PKC from a metabolomics standpoint, by comparing the full serum metabolic profiles of PKC deficient mice to those of wild-type mice. METHODS The serum metabolomes of PKCδ knock-out mice were compared to that of a wild-type strain using a GCxGC-TOFMS metabolomics research approach and various univariate and multivariate statistical analyses. RESULTS Thirty-seven serum metabolite markers best describing the difference between PKCδ knock-out and wild-type mice were identified based on a PCA power value > 0.9, a t-test p-value < 0.05, or an effect size > 1. XERp prediction was also done to accurately select the metabolite markers within the 2 sample groups. Of the metabolite markers identified, 78.4% (29/37) were elevated and 48.65% of these markers were fatty acids (18/37). It is clear that a total loss of PKCδ functionality results in an inhibition of glycolysis, the TCA cycle, and steroid synthesis, accompanied by upregulation of the pentose phosphate pathway, fatty acids oxidation, cholesterol transport/storage, single carbon and sulphur-containing amino acid synthesis, branched-chain amino acids (BCAA), ketogenesis, and an increased cell signalling via N-acetylglucosamine. CONCLUSION The charaterization of the dysregulated serum metabolites in this study, may represent an additional tool for the early detection and screening of PKCδ-deficiencies or abnormalities.
Collapse
Affiliation(s)
- Du Toit Loots
- Human Metabolomics, North-West University, Hoffman Street, 2531, Potchefstroom, South Africa.
| | | | - Mari Van Reenen
- Human Metabolomics, North-West University, Hoffman Street, 2531, Potchefstroom, South Africa
| | - Mumin Ozturk
- Human Metabolomics, North-West University, Hoffman Street, 2531, Potchefstroom, South Africa
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town-Component, Cape Town, South Africa
| | - Frank Brombacher
- Human Metabolomics, North-West University, Hoffman Street, 2531, Potchefstroom, South Africa
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town-Component, Cape Town, South Africa
- Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Wellcome Center for Infectious Disease Research in Africa (CIDRI-Africa), Institute of Infectious Diseases and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Suraj P Parihar
- Human Metabolomics, North-West University, Hoffman Street, 2531, Potchefstroom, South Africa.
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town-Component, Cape Town, South Africa.
- Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
- Wellcome Center for Infectious Disease Research in Africa (CIDRI-Africa), Institute of Infectious Diseases and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
4
|
Gonzalez-Covarrubias V, Martínez-Martínez E, del Bosque-Plata L. The Potential of Metabolomics in Biomedical Applications. Metabolites 2022; 12:metabo12020194. [PMID: 35208267 PMCID: PMC8880031 DOI: 10.3390/metabo12020194] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/28/2022] [Accepted: 01/30/2022] [Indexed: 12/12/2022] Open
Abstract
The metabolome offers a dynamic, comprehensive, and precise picture of the phenotype. Current high-throughput technologies have allowed the discovery of relevant metabolites that characterize a wide variety of human phenotypes with respect to health, disease, drug monitoring, and even aging. Metabolomics, parallel to genomics, has led to the discovery of biomarkers and has aided in the understanding of a diversity of molecular mechanisms, highlighting its application in precision medicine. This review focuses on the metabolomics that can be applied to improve human health, as well as its trends and impacts in metabolic and neurodegenerative diseases, cancer, longevity, the exposome, liquid biopsy development, and pharmacometabolomics. The identification of distinct metabolomic profiles will help in the discovery and improvement of clinical strategies to treat human disease. In the years to come, metabolomics will become a tool routinely applied to diagnose and monitor health and disease, aging, or drug development. Biomedical applications of metabolomics can already be foreseen to monitor the progression of metabolic diseases, such as obesity and diabetes, using branched-chain amino acids, acylcarnitines, certain phospholipids, and genomics; these can assess disease severity and predict a potential treatment. Future endeavors should focus on determining the applicability and clinical utility of metabolomic-derived markers and their appropriate implementation in large-scale clinical settings.
Collapse
Affiliation(s)
| | - Eduardo Martínez-Martínez
- Laboratory of Cell Communication and Extracellular Vesicles, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico;
| | - Laura del Bosque-Plata
- Laboratory of Nutrigenetics and Nutrigenomics, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico
- Correspondence: ; Tel.: +52-55-53-50-1974
| |
Collapse
|
5
|
Protein kinase C-mediated calcium signaling as the basis for cardiomyocyte plasticity. Arch Biochem Biophys 2021; 701:108817. [PMID: 33626379 DOI: 10.1016/j.abb.2021.108817] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 02/03/2021] [Accepted: 02/14/2021] [Indexed: 01/08/2023]
Abstract
Protein kinase C is the superfamily of intracellular effector molecules which control crucial cellular functions. Here, we for the first time did the percentage estimation of all known PKC and PKC-related isozymes at the individual cadiomyocyte level. Broad spectrum of PKC transcripts is expressed in the left ventricular myocytes. In addition to the well-known 'heart-specific' PKCα, cardiomyocytes have the high expression levels of PKCN1, PKCδ, PKCD2, PKCε. In general, we detected all PKC isoforms excluding PKCη. In cardiomyocytes PKC activity tonically regulates voltage-gated Ca2+-currents, intracellular Ca2+ level and nitric oxide (NO) production. Imidazoline receptor of the first type (I1R)-mediated induction of the PKC activity positively modulates Ca2+ release through ryanodine receptor (RyR), increasing the Ca2+ leakage in the cytosol. In cardiomyocytes with the Ca2+-overloaded regions of > 9-10 μm size, the local PKC-induced Ca2+ signaling is transformed to global accompanied by spontaneous Ca2+ waves propagation across the entire cell perimeter. Such switching of Ca2+ signaling in cardiac cells can be important for the development of several cardiovascular pathologies and/or myocardial plasticity at the cardiomyocyte level.
Collapse
|
6
|
Speidel JT, Affandi T, Jones DNM, Ferrara SE, Reyland ME. Functional proteomic analysis reveals roles for PKCδ in regulation of cell survival and cell death: Implications for cancer pathogenesis and therapy. Adv Biol Regul 2020; 78:100757. [PMID: 33045516 PMCID: PMC8294469 DOI: 10.1016/j.jbior.2020.100757] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 12/18/2022]
Abstract
Protein Kinase C-δ (PKCδ), regulates a broad group of biological functions and disease processes, including well-defined roles in immune function, cell survival and apoptosis. PKCδ primarily regulates apoptosis in normal tissues and non-transformed cells, and genetic disruption of the PRKCD gene in mice is protective in many diseases and tissue damage models. However pro-survival/pro-proliferative functions have also been described in some transformed cells and in mouse models of cancer. Recent evidence suggests that the contribution of PKCδ to specific cancers may depend in part on the oncogenic context of the tumor, consistent with its paradoxical role in cell survival and cell death. Here we will discuss what is currently known about biological functions of PKCδ and potential paradigms for PKCδ function in cancer. To further understand mechanisms of regulation by PKCδ, and to gain insight into the plasticity of PKCδ signaling, we have used functional proteomics to identify pathways that are dependent on PKCδ. Understanding how these distinct functions of PKCδ are regulated will be critical for the logical design of therapeutics to target this pathway.
Collapse
Affiliation(s)
- Jordan T Speidel
- Department of Craniofacial Biology, School of Dental Medicine, USA
| | - Trisiani Affandi
- Department of Craniofacial Biology, School of Dental Medicine, USA
| | | | - Sarah E Ferrara
- University of Colorado Comprehensive Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Mary E Reyland
- Department of Craniofacial Biology, School of Dental Medicine, USA.
| |
Collapse
|
7
|
Abstract
Despite the increasing use and success of interventional coronary reperfusion strategies, morbidity and mortality from acute myocardial infarction are still substantial. Myocardial infarct size is a major determinant of prognosis in these patients. Therefore, cardioprotective strategies aim to reduce infarct size. However, a perplexing gap exists between the many preclinical studies reporting infarct size reduction with mechanical and pharmacological interventions and the poor translation into better clinical outcomes in patients. This Review revisits the pathophysiology of myocardial ischaemia-reperfusion injury, including the role of autophagy and forms of cell death such as necrosis, apoptosis, necroptosis and pyroptosis. Other cellular compartments in addition to cardiomyocytes are addressed, notably the coronary microcirculation. Preclinical and clinical research developments in mechanical and pharmacological approaches to induce cardioprotection, and their signal transduction pathways, are discussed. Additive cardioprotective interventions are advocated. For clinical translation into treatments for patients with acute myocardial infarction, who typically are of advanced age, have comorbidities and are receiving several medications, not only infarct size reduction but also attenuation of coronary microvascular obstruction, as well as longer-term targets including infarct repair and reverse remodelling, must be considered to improve patient outcomes. Future clinical trials must focus on patients who really need adjunct cardioprotection, that is, those with severe haemodynamic alterations.
Collapse
|
8
|
Marrocco V, Bogomolovas J, Ehler E, Dos Remedios CG, Yu J, Gao C, Lange S. PKC and PKN in heart disease. J Mol Cell Cardiol 2019; 128:212-226. [PMID: 30742812 PMCID: PMC6408329 DOI: 10.1016/j.yjmcc.2019.01.029] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/30/2019] [Accepted: 01/31/2019] [Indexed: 12/22/2022]
Abstract
The protein kinase C (PKC) and closely related protein kinase N (PKN) families of serine/threonine protein kinases play crucial cellular roles. Both kinases belong to the AGC subfamily of protein kinases that also include the cAMP dependent protein kinase (PKA), protein kinase B (PKB/AKT), protein kinase G (PKG) and the ribosomal protein S6 kinase (S6K). Involvement of PKC family members in heart disease has been well documented over the years, as their activity and levels are mis-regulated in several pathological heart conditions, such as ischemia, diabetic cardiomyopathy, as well as hypertrophic or dilated cardiomyopathy. This review focuses on the regulation of PKCs and PKNs in different pathological heart conditions and on the influences that PKC/PKN activation has on several physiological processes. In addition, we discuss mechanisms by which PKCs and the closely related PKNs are activated and turned-off in hearts, how they regulate cardiac specific downstream targets and pathways, and how their inhibition by small molecules is explored as new therapeutic target to treat cardiomyopathies and heart failure.
Collapse
Affiliation(s)
- Valeria Marrocco
- Division of Cardiology, School of Medicine, University of California-San Diego, La Jolla, USA
| | - Julius Bogomolovas
- Division of Cardiology, School of Medicine, University of California-San Diego, La Jolla, USA; Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Elisabeth Ehler
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, School of Cardiovascular Medicine and Sciences, British Heart Foundation Research Excellence Centre, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | | | - Jiayu Yu
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Gao
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at UCLA, University of California-Los Angeles, Los Angeles, USA.
| | - Stephan Lange
- Division of Cardiology, School of Medicine, University of California-San Diego, La Jolla, USA; University of Gothenburg, Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg, Sweden.
| |
Collapse
|
9
|
Park YS, Lee JE, Park JI, Myung CH, Lim YH, Park CK, Hwang JS. Inhibitory mechanism of ginsenoside Rh3 on granulocyte-macrophage colony-stimulating factor expression in UV-B-irradiated murine SP-1 keratinocytes. J Ginseng Res 2018; 44:274-281. [PMID: 32148409 PMCID: PMC7031754 DOI: 10.1016/j.jgr.2018.12.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 11/20/2018] [Accepted: 12/18/2018] [Indexed: 12/16/2022] Open
Abstract
Background Ultraviolet (UV) goes through the epidermis and promotes release of inflammatory cytokines in keratinocytes. Granulocyte–macrophage colony-stimulating factor (GM-CSF), one of the keratinocyte-derived cytokines, regulates proliferation and differentiation of melanocytes. Extracellular signal–regulated kinase (ERK1/2) and protein kinase C (PKC) signaling pathways regulate expression of GM-CSF. Based on these results, we found that ginsenoside Rh3 prevented GM-CSF production and release in UV-B–exposed SP-1 keratinocytes and that this inhibitory effect resulted from the reduction of PKCδ and ERK phosphorylation. Methods We investigated the mechanism by which ginsenoside Rh3 from Panax ginseng inhibited GM-CSF release from UV-B–irradiated keratinocytes. Results Treatment with 12-O-tetradecanoylphorbol-13-acetate (TPA) or UV-B induced release of GM-CSF in the SP-1 keratinocytes. To elucidate whether the change in GM-CSF expression could be related to PKC signaling, the cells were pretreated with H7, an inhibitor of PKC, and irradiated with UV-B. GM-CSF was decreased by H7 in a dose-dependent manner. When we analyzed which ginsenosides repressed GM-CSF expression among 15 ginsenosides, ginsenoside Rh3 showed the largest decline to 40% of GM-CSF expression in enzyme-linked immunosorbent assay. Western blot analysis showed that TPA enhanced the phosphorylation of PKCδ and ERK in the keratinocytes. When we examined the effect of ginsenoside Rh3, we identified that ginsenoside Rh3 inhibited the TPA-induced phosphorylation levels of PKCδ and ERK. Conclusion In summary, we found that ginsenoside Rh3 impeded UV-B–induced GM-CSF production through repression of PKCδ and ERK phosphorylation in SP-1 keratinocytes.
Collapse
Affiliation(s)
- Young Sun Park
- Department of Genetic Engineering & Graduate School of Biotechnology, Kyung Hee University, Yongin, Republic of Korea
| | - Ji Eun Lee
- Department of Genetic Engineering & Graduate School of Biotechnology, Kyung Hee University, Yongin, Republic of Korea
| | - Jong Il Park
- Department of Genetic Engineering & Graduate School of Biotechnology, Kyung Hee University, Yongin, Republic of Korea
| | - Cheol Hwan Myung
- Department of Genetic Engineering & Graduate School of Biotechnology, Kyung Hee University, Yongin, Republic of Korea
| | - Young-Ho Lim
- KGC R&D Headquarters, Daejeon, Republic of Korea
| | | | - Jae Sung Hwang
- Department of Genetic Engineering & Graduate School of Biotechnology, Kyung Hee University, Yongin, Republic of Korea
| |
Collapse
|
10
|
Abstract
Disturbances in cardiac metabolism underlie most cardiovascular diseases. Metabolomics, one of the newer omics technologies, has emerged as a powerful tool for defining changes in both global and cardiac-specific metabolism that occur across a spectrum of cardiovascular disease states. Findings from metabolomics studies have contributed to better understanding of the metabolic changes that occur in heart failure and ischemic heart disease and have identified new cardiovascular disease biomarkers. As technologies advance, the metabolomics field continues to evolve rapidly. In this review, we will discuss the current state of metabolomics technologies, including consideration of various metabolomics platforms and elements of study design; the emerging utility of stable isotopes for metabolic flux studies; and the use of metabolomics to better understand specific cardiovascular diseases, with an emphasis on recent advances in the field.
Collapse
Affiliation(s)
- Robert W McGarrah
- From the Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute (R.W.M., S.B.C., G.F.Z., S.H.S., C.B.N.)
- Division of Cardiology (R.W.M., S.H.S.)
- Department of Medicine (R.W.M., G.F.Z., S.H.S., C.B.N.)
| | - Scott B Crown
- From the Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute (R.W.M., S.B.C., G.F.Z., S.H.S., C.B.N.)
| | - Guo-Fang Zhang
- From the Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute (R.W.M., S.B.C., G.F.Z., S.H.S., C.B.N.)
- Division of Endocrinology (G.F.Z., C.B.N.)
- Department of Medicine (R.W.M., G.F.Z., S.H.S., C.B.N.)
| | - Svati H Shah
- From the Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute (R.W.M., S.B.C., G.F.Z., S.H.S., C.B.N.)
- Division of Cardiology (R.W.M., S.H.S.)
- Department of Medicine (R.W.M., G.F.Z., S.H.S., C.B.N.)
| | - Christopher B Newgard
- From the Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute (R.W.M., S.B.C., G.F.Z., S.H.S., C.B.N.)
- Division of Endocrinology (G.F.Z., C.B.N.)
- Department of Medicine (R.W.M., G.F.Z., S.H.S., C.B.N.)
- Departments of Pharmacology and Cancer Biology (C.B.N.), Duke University Medical Center, Durham, NC
| |
Collapse
|
11
|
Protein kinase C mechanisms that contribute to cardiac remodelling. Clin Sci (Lond) 2017; 130:1499-510. [PMID: 27433023 DOI: 10.1042/cs20160036] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 05/18/2016] [Indexed: 12/12/2022]
Abstract
Protein phosphorylation is a highly-regulated and reversible process that is precisely controlled by the actions of protein kinases and protein phosphatases. Factors that tip the balance of protein phosphorylation lead to changes in a wide range of cellular responses, including cell proliferation, differentiation and survival. The protein kinase C (PKC) family of serine/threonine kinases sits at nodal points in many signal transduction pathways; PKC enzymes have been the focus of considerable attention since they contribute to both normal physiological responses as well as maladaptive pathological responses that drive a wide range of clinical disorders. This review provides a background on the mechanisms that regulate individual PKC isoenzymes followed by a discussion of recent insights into their role in the pathogenesis of diseases such as cancer. We then provide an overview on the role of individual PKC isoenzymes in the regulation of cardiac contractility and pathophysiological growth responses, with a focus on the PKC-dependent mechanisms that regulate pump function and/or contribute to the pathogenesis of heart failure.
Collapse
|
12
|
Abstract
For the past century, vitamin A has been considered to serve as a precursor for retinoids that facilitate vision or as a precursor for retinoic acid (RA), a signaling molecule that modulates gene expression. However, vitamin A circulates in plasma at levels that far exceed the amount needed for vision or the synthesis of nanomolar levels of RA, and this suggests that vitamin A alcohol (i.e. retinol) may possess additional biological activity. We have pursued this question for the last 20 years, and in this chapter, we unfold the story of our quest and the data that support a novel and distinct role for vitamin A (alcohol) action. Our current model supports direct binding of vitamin A to the activation domains of serine/threonine kinases, such as protein kinase C (PKC) and Raf isoforms, where it is involved in redox activation of these proteins. Redox activation of PKCs was first described by the founders of the PKC field, but several hurdles needed to be overcome before a detailed understanding of the biochemistry could be provided. Two discoveries moved the field forward. First, was the discovery that the PKCδ isoform was activated by cytochrome c, a protein with oxidoreduction activity in mitochondria. Second, was the revelation that both PKCδ and cytochrome c are tethered to p66Shc, an adapter protein that brings the PKC zinc-finger substrate into close proximity with its oxidizing partner. Detailed characterization of the PKCδ signalosome complex was made possible by the work of many investigators. Our contribution was determining that vitamin A is a vital co-factor required to support an unprecedented redox-activation mechanism. This unique function of vitamin A is the first example of a general system that connects the one-electron redox chemistry of a heme protein (cytochrome c) with the two-electron chemistry of a classical phosphoprotein (PKCδ). Furthermore, contributions to the regulation of mitochondrial energetics attest to biological significance of vitamin A alcohol action.
Collapse
Affiliation(s)
- Ulrich Hammerling
- Member Emeritus, Immunology Program, Sloan-Kettering Institute for Cancer Research, 10065, New York, NY, USA.
| |
Collapse
|
13
|
Shabrova E, Hoyos B, Vinogradov V, Kim YK, Wassef L, Leitges M, Quadro L, Hammerling U. Retinol as a cofactor for PKCδ-mediated impairment of insulin sensitivity in a mouse model of diet-induced obesity. FASEB J 2015; 30:1339-55. [PMID: 26671999 DOI: 10.1096/fj.15-281543] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 11/23/2015] [Indexed: 12/15/2022]
Abstract
We previously defined that the mitochondria-localized PKCδ signaling complex stimulates the conversion of pyruvate to acetyl-coenzyme A by the pyruvate dehydrogenase complex. We demonstrated in vitro and ex vivo that retinol supplementation enhances ATP synthesis in the presence of the PKCδ signalosome. Here, we tested in vivo if a persistent oversupply of retinol would further impair glucose metabolism in a mouse model of diet-induced insulin resistance. We crossed mice overexpressing human retinol-binding protein (hRBP) under the muscle creatine kinase (MCK) promoter (MCKhRBP) with the PKCδ(-/-) strain to generate mice with a different status of the PKCδ signalosome and retinoid levels. Mice with a functional PKCδ signalosome and elevated retinoid levels (PKCδ(+/+)hRBP) developed the most advanced stage of insulin resistance. In contrast, elevation of retinoid levels in mice with inactive PKCδ did not affect remarkably their metabolism, resulting in phenotypic similarity between PKCδ(-/-)hRBP and PKCδ(-/-) mice. Therefore, in addition to the well-defined role of PKCδ in the etiology of metabolic syndrome, we present a novel PKCδ signaling pathway that requires retinol as a metabolic cofactor and is involved in the regulation of fuel utilization in mitochondria. The distinct role in whole-body energy homeostasis establishes the PKCδ signalosome as a promising target for therapeutic intervention in metabolic disorders.
Collapse
Affiliation(s)
- Elena Shabrova
- *Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, New York, USA; Department of Food Science, Rutgers Center for Lipid Research, Rutgers University, New Brunswick, New Jersey, USA; and Biotechnology Center of Oslo, University of Oslo, Oslo, Norway
| | - Beatrice Hoyos
- *Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, New York, USA; Department of Food Science, Rutgers Center for Lipid Research, Rutgers University, New Brunswick, New Jersey, USA; and Biotechnology Center of Oslo, University of Oslo, Oslo, Norway
| | - Valerie Vinogradov
- *Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, New York, USA; Department of Food Science, Rutgers Center for Lipid Research, Rutgers University, New Brunswick, New Jersey, USA; and Biotechnology Center of Oslo, University of Oslo, Oslo, Norway
| | - Youn-Kyung Kim
- *Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, New York, USA; Department of Food Science, Rutgers Center for Lipid Research, Rutgers University, New Brunswick, New Jersey, USA; and Biotechnology Center of Oslo, University of Oslo, Oslo, Norway
| | - Lesley Wassef
- *Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, New York, USA; Department of Food Science, Rutgers Center for Lipid Research, Rutgers University, New Brunswick, New Jersey, USA; and Biotechnology Center of Oslo, University of Oslo, Oslo, Norway
| | - Michael Leitges
- *Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, New York, USA; Department of Food Science, Rutgers Center for Lipid Research, Rutgers University, New Brunswick, New Jersey, USA; and Biotechnology Center of Oslo, University of Oslo, Oslo, Norway
| | - Loredana Quadro
- *Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, New York, USA; Department of Food Science, Rutgers Center for Lipid Research, Rutgers University, New Brunswick, New Jersey, USA; and Biotechnology Center of Oslo, University of Oslo, Oslo, Norway
| | - Ulrich Hammerling
- *Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, New York, USA; Department of Food Science, Rutgers Center for Lipid Research, Rutgers University, New Brunswick, New Jersey, USA; and Biotechnology Center of Oslo, University of Oslo, Oslo, Norway
| |
Collapse
|
14
|
Li M, Vienberg SG, Bezy O, O'Neill BT, Kahn CR. Role of PKCδ in Insulin Sensitivity and Skeletal Muscle Metabolism. Diabetes 2015; 64:4023-32. [PMID: 26307588 PMCID: PMC4657586 DOI: 10.2337/db14-1891] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 08/17/2015] [Indexed: 01/06/2023]
Abstract
Protein kinase C (PKC)δ has been shown to be increased in liver in obesity and plays an important role in the development of hepatic insulin resistance in both mice and humans. In the current study, we explored the role of PKCδ in skeletal muscle in the control of insulin sensitivity and glucose metabolism by generating mice in which PKCδ was deleted specifically in muscle using Cre-lox recombination. Deletion of PKCδ in muscle improved insulin signaling in young mice, especially at low insulin doses; however, this did not change glucose tolerance or insulin tolerance tests done with pharmacological levels of insulin. Likewise, in young mice, muscle-specific deletion of PKCδ did not rescue high-fat diet-induced insulin resistance or glucose intolerance. However, with an increase in age, PKCδ levels in muscle increased, and by 6 to 7 months of age, muscle-specific deletion of PKCδ improved whole-body insulin sensitivity and muscle insulin resistance and by 15 months of age improved the age-related decline in whole-body glucose tolerance. At 15 months of age, M-PKCδKO mice also exhibited decreased metabolic rate and lower levels of some proteins of the OXPHOS complex suggesting a role for PKCδ in the regulation of mitochondrial mass at older age. These data indicate an important role of PKCδ in the regulation of insulin sensitivity and mitochondrial homeostasis in skeletal muscle with aging.
Collapse
Affiliation(s)
- Mengyao Li
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Sara G Vienberg
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA Novo Nordisk Foundation Center for Basic Metabolic Research, Copenhagen University, Copenhagen, Denmark
| | - Olivier Bezy
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Brian T O'Neill
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - C Ronald Kahn
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| |
Collapse
|
15
|
Shende P, Xu L, Morandi C, Pentassuglia L, Heim P, Lebboukh S, Berthonneche C, Pedrazzini T, Kaufmann BA, Hall MN, Rüegg MA, Brink M. Cardiac mTOR complex 2 preserves ventricular function in pressure-overload hypertrophy. Cardiovasc Res 2015; 109:103-14. [DOI: 10.1093/cvr/cvv252] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 11/06/2015] [Indexed: 11/12/2022] Open
|
16
|
Shi T, Papay RS, Perez DM. α1A-Adrenergic receptor prevents cardiac ischemic damage through PKCδ/GLUT1/4-mediated glucose uptake. J Recept Signal Transduct Res 2015; 36:261-70. [PMID: 26832303 DOI: 10.3109/10799893.2015.1091475] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
While α(1)-adrenergic receptors (ARs) have been previously shown to limit ischemic cardiac damage, the mechanisms remain unclear. Most previous studies utilized low oxygen conditions in addition to ischemic buffers with glucose deficiencies, but we discovered profound differences if the two conditions are separated. We assessed both mouse neonatal and adult myocytes and HL-1 cells in a series of assays assessing ischemic damage under hypoxic or low glucose conditions. We found that α(1)-AR stimulation protected against increased lactate dehydrogenase release or Annexin V(+) apoptosis under conditions that were due to low glucose concentration not to hypoxia. The α(1)-AR antagonist prazosin or nonselective protein kinase C (PKC) inhibitors blocked the protective effect. α(1)-AR stimulation increased (3)H-deoxyglucose uptake that was blocked with either an inhibitor to glucose transporter 1 or 4 (GLUT1 or GLUT4) or small interfering RNA (siRNA) against PKCδ. GLUT1/4 inhibition also blocked α(1)-AR-mediated protection from apoptosis. The PKC inhibitor rottlerin or siRNA against PKCδ blocked α(1)-AR stimulated GLUT1 or GLUT4 plasma membrane translocation. α(1)-AR stimulation increased plasma membrane concentration of either GLUT1 or GLUT4 in a time-dependent fashion. Transgenic mice overexpressing the α(1A)-AR but not α(1B)-AR mice displayed increased glucose uptake and increased GLUT1 and GLUT4 plasma membrane translocation in the adult heart while α(1A)-AR but not α(1B)-AR knockout mice displayed lowered glucose uptake and GLUT translocation. Our results suggest that α(1)-AR activation is anti-apoptotic and protective during cardiac ischemia due to glucose deprivation and not hypoxia by enhancing glucose uptake into the heart via PKCδ-mediated GLUT translocation that may be specific to the α(1A)-AR subtype.
Collapse
Affiliation(s)
- Ting Shi
- a Department of Molecular Cardiology , Lerner Research Institute, Cleveland Clinic Foundation , Cleveland , OH , USA
| | - Robert S Papay
- a Department of Molecular Cardiology , Lerner Research Institute, Cleveland Clinic Foundation , Cleveland , OH , USA
| | - Dianne M Perez
- a Department of Molecular Cardiology , Lerner Research Institute, Cleveland Clinic Foundation , Cleveland , OH , USA
| |
Collapse
|
17
|
Abstract
A large number of protein substrates are phosphorylated by each protein kinase under physiological and pathological conditions. However, it remains a challenge to determine which of these phosphorylated substrates of a given kinase is critical for each cellular response. Genetics enabled the generation of separation-of-function mutations that selectively cause a loss of one molecular event without affecting others, thus providing some tools to assess the importance of that one event for the measured physiological response. However, the genetic approach is laborious and not adaptable to all systems. Furthermore, pharmacological tools of the catalytic site are not optimal due to their non-selective nature. In the present brief review, we discuss some of the challenges in drug development that will regulate the multifunctional protein kinase Cδ (PKCδ).
Collapse
|
18
|
Sun X, Kumar S, Sharma S, Aggarwal S, Lu Q, Gross C, Rafikova O, Lee SG, Dasarathy S, Hou Y, Meadows ML, Han W, Su Y, Fineman JR, Black SM. Endothelin-1 induces a glycolytic switch in pulmonary arterial endothelial cells via the mitochondrial translocation of endothelial nitric oxide synthase. Am J Respir Cell Mol Biol 2014; 50:1084-95. [PMID: 24392990 DOI: 10.1165/rcmb.2013-0187oc] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Recent studies have indicated that, during the development of pulmonary hypertension (PH), there is a switch from oxidative phosphorylation to glycolysis in the pulmonary endothelium. However, the mechanisms underlying this phenomenon have not been elucidated. Endothelin (ET)-1, an endothelial-derived vasoconstrictor peptide, is increased in PH, and has been shown to play an important role in the oxidative stress associated with PH. Thus, in this study, we investigated whether there was a potential link between increases in ET-1 and mitochondrial remodeling. Our data indicate that ET-1 induces the redistribution of endothelial nitric oxide synthase (eNOS) from the plasma membrane to the mitochondria in pulmonary arterial endothelial cells, and that this was dependent on eNOS uncoupling. We also found that ET-1 disturbed carnitine metabolism, resulting in the attenuation of mitochondrial bioenergetics. However, ATP levels were unchanged due to a compensatory increase in glycolysis. Further mechanistic investigations demonstrated that ET-1 mediated the redistribution of eNOS via the phosphorylation of eNOS at Thr495 by protein kinase C δ. In addition, the glycolytic switch appeared to be dependent on mitochondrial-derived reactive oxygen species that led to the activation of hypoxia-inducible factor signaling. Finally, the cell culture data were confirmed in vivo using the monocrotaline rat model of PH. Thus, we conclude that ET-1 induces a glycolytic switch in pulmonary arterial endothelial cells via the redistribution of uncoupled eNOS to the mitochondria, and that preventing this event may be an approach for the treatment of PH.
Collapse
Affiliation(s)
- Xutong Sun
- 1 Pulmonary Disease Program, Vascular Biology Center, and
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Greene MW, Burrington CM, Lynch DT, Davenport SK, Johnson AK, Horsman MJ, Chowdhry S, Zhang J, Sparks JD, Tirrell PC. Lipid metabolism, oxidative stress and cell death are regulated by PKC delta in a dietary model of nonalcoholic steatohepatitis. PLoS One 2014; 9:e85848. [PMID: 24454937 PMCID: PMC3893275 DOI: 10.1371/journal.pone.0085848] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 12/03/2013] [Indexed: 12/31/2022] Open
Abstract
Steatosis, oxidative stress, and apoptosis underlie the development of nonalcoholic steatohepatitis (NASH). Protein kinase C delta (PKCδ) has been implicated in fatty liver disease and is activated in the methionine and choline-deficient (MCD) diet model of NASH, yet its pathophysiological importance towards steatohepatitis progression is uncertain. We therefore addressed the role of PKCδ in the development of steatosis, inflammation, oxidative stress, apoptosis, and fibrosis in an animal model of NASH. We fed PKCδ−/− mice and wildtype littermates a control or MCD diet. PKCδ−/− primary hepatocytes were used to evaluate the direct effects of fatty acids on hepatocyte lipid metabolism gene expression. A reduction in hepatic steatosis and triglyceride levels were observed between wildtype and PKCδ−/− mice fed the MCD diet. The hepatic expression of key regulators of β-oxidation and plasma triglyceride metabolism was significantly reduced in PKCδ−/− mice and changes in serum triglyceride were blocked in PKCδ−/− mice. MCD diet-induced hepatic oxidative stress and hepatocyte apoptosis were reduced in PKCδ−/− mice. MCD diet-induced NADPH oxidase activity and p47phox membrane translocation were blunted and blocked, respectively, in PKCδ−/− mice. Expression of pro-apoptotic genes and caspase 3 and 9 cleavage in the liver of MCD diet fed PKCδ−/− mice were blunted and blocked, respectively. Surprisingly, no differences in MCD diet-induced fibrosis or pro-fibrotic gene expression were observed in 8 week MCD diet fed PKCδ−/− mice. Our results suggest that PKCδ plays a role in key pathological features of fatty liver disease but not ultimately in fibrosis in the MCD diet model of NASH.
Collapse
Affiliation(s)
- Michael W. Greene
- Boshell Diabetes and Metabolic Disease Research Program, Auburn University, Auburn, Alabama, United States of America
- College of Human Sciences, Auburn University, Auburn, Alabama, United States of America
- Bassett Research Institute, Bassett Medical Center, Bassett Healthcare Network, Cooperstown, New York, United States of America
- * E-mail:
| | - Christine M. Burrington
- Bassett Research Institute, Bassett Medical Center, Bassett Healthcare Network, Cooperstown, New York, United States of America
| | - Darin T. Lynch
- Bassett Research Institute, Bassett Medical Center, Bassett Healthcare Network, Cooperstown, New York, United States of America
| | - Samantha K. Davenport
- Department of Pathology, Bassett Medical Center, Bassett Healthcare Network, Cooperstown, New York, United States of America
| | - Andrew K. Johnson
- Bassett Research Institute, Bassett Medical Center, Bassett Healthcare Network, Cooperstown, New York, United States of America
| | - Melissa J. Horsman
- Bassett Research Institute, Bassett Medical Center, Bassett Healthcare Network, Cooperstown, New York, United States of America
| | - Saleem Chowdhry
- Department of Internal Medicine, Bassett Medical Center, Bassett Healthcare Network, Cooperstown, New York, United States of America
| | - Jian Zhang
- College of Human Sciences, Auburn University, Auburn, Alabama, United States of America
| | - Janet D. Sparks
- University of Rochester Medical Center, School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Paul C. Tirrell
- Department of Internal Medicine, Bassett Medical Center, Bassett Healthcare Network, Cooperstown, New York, United States of America
| |
Collapse
|
20
|
Xia L, Wang TD, Shen SM, Zhao M, Sun H, He Y, Xie L, Wu ZX, Han SF, Wang LS, Chen GQ. Phosphoproteomics study on the activated PKCδ-induced cell death. J Proteome Res 2013; 12:4280-301. [PMID: 23879269 DOI: 10.1021/pr400089v] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The proteolytic activation of protein kinase Cδ (PKCδ) generates a catalytic fragment called PKCδ-CF, which induces cell death. However, the mechanisms underlying PKCδ-CF-mediated cell death are largely unknown. On the basis of an engineering leukemic cell line with inducible expression of PKCδ-CF, here we employ SILAC-based quantitative phosphoproteomics to systematically and dynamically investigate the overall phosphorylation events during cell death triggered by PKCδ-CF expression. Totally, 3000 phosphorylation sites were analyzed. Considering the fact that early responses to PKCδ-CF expression initiate cell death, we sought to identify pathways possibly related directly with PKCδ by further analyzing the data set of phosphorylation events that occur in the initiation stage of cell death. Interacting analysis of this data set indicates that PKCδ-CF triggers complicated networks to initiate cell death, and motif analysis and biochemistry verification reveal that several kinases in the downstream of PKCδ conduct these networks. By analysis of the specific sequence motif of kinase-substrate, we also find 59 candidate substrates of PKCδ from the up-regulated phosphopeptides, of which 12 were randomly selected for in vitro kinase assay and 9 were consequently verified as substrates of PKCδ. To our greatest understanding, this study provides the most systematic analysis of phosphorylation events initiated by the cleaved activated PKCδ, which would vastly extend the profound understanding of PKCδ-directed signal pathways in cell death. The MS data have been deposited to the ProteomeXchange with identifier PXD000225.
Collapse
Affiliation(s)
- Li Xia
- The Department of Pathophysiology and Shanghai Universities E-Institute for Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine (SJTU-SM) , Shanghai, P.R. China , 200025
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Figueira TR, Barros MH, Camargo AA, Castilho RF, Ferreira JCB, Kowaltowski AJ, Sluse FE, Souza-Pinto NC, Vercesi AE. Mitochondria as a source of reactive oxygen and nitrogen species: from molecular mechanisms to human health. Antioxid Redox Signal 2013; 18:2029-74. [PMID: 23244576 DOI: 10.1089/ars.2012.4729] [Citation(s) in RCA: 315] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mitochondrially generated reactive oxygen species are involved in a myriad of signaling and damaging pathways in different tissues. In addition, mitochondria are an important target of reactive oxygen and nitrogen species. Here, we discuss basic mechanisms of mitochondrial oxidant generation and removal and the main factors affecting mitochondrial redox balance. We also discuss the interaction between mitochondrial reactive oxygen and nitrogen species, and the involvement of these oxidants in mitochondrial diseases, cancer, neurological, and cardiovascular disorders.
Collapse
Affiliation(s)
- Tiago R Figueira
- Department of Clinical Pathology, Faculty of Medical Sciences, State University of Campinas, Campinas, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Davis J, Maillet M, Miano JM, Molkentin JD. Lost in transgenesis: a user's guide for genetically manipulating the mouse in cardiac research. Circ Res 2012; 111:761-77. [PMID: 22935533 DOI: 10.1161/circresaha.111.262717] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The advent of modern mouse genetics has benefited many fields of diseased-based research over the past 20 years, none perhaps more profoundly than cardiac biology. Indeed, the heart is now arguably one of the easiest tissues to genetically manipulate, given the availability of an ever-growing tool chest of molecular reagents/promoters and "facilitator" mouse lines. It is now possible to modify the expression of essentially any gene or partial gene product in the mouse heart at any time, either gain or loss of function. This review is designed as a handbook for the nonmouse geneticist and/or junior investigator to permit the successful manipulation of any gene or RNA product in the heart, while avoiding artifacts. In the present review, guidelines, pitfalls, and limitations are presented so that rigorous and appropriate examination of cardiac genotype-phenotype relationships can be performed. This review uses examples from the field to illustrate the vast spectrum of experimental and design details that must be considered when using genetically modified mouse models to study cardiac biology.
Collapse
Affiliation(s)
- Jennifer Davis
- Department of Pediatrics, University of Cincinnati, Howard Hughes Medical Institute, Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way, S4.409, Cincinnati, OH 45229, USA
| | | | | | | |
Collapse
|
23
|
Abstract
The conventional reductionist approach to cardiovascular research investigates individual candidate factors or linear signalling pathways but ignores more complex interactions in biological systems. The advent of molecular profiling technologies that focus on a global characterization of whole complements allows an exploration of the interconnectivity of pathways during pathophysiologically relevant processes, but has brought about the issue of statistical analysis and data integration. Proteins identified by differential expression as well as those in protein–protein interaction networks identified through experiments and through computational modelling techniques can be used as an initial starting point for functional analyses. In combination with other ‘-omics’ technologies, such as transcriptomics and metabolomics, proteomics explores different aspects of disease, and the different pillars of observations facilitate the data integration in disease-specific networks. Ultimately, a systems biology approach may advance our understanding of cardiovascular disease processes at a ‘biological pathway’ instead of a ‘single molecule’ level and accelerate progress towards disease-modifying interventions.
Collapse
Affiliation(s)
- Sarah R Langley
- King's British Heart Foundation Centre, King's College London, 125 Coldharbour Lane, London SE5 9NU, UK
| | | | | | | | | |
Collapse
|
24
|
Pula G, Perera S, Prokopi M, Sidibe A, Boulanger CM, Mayr M. Proteomic analysis of secretory proteins and vesicles in vascular research. Proteomics Clin Appl 2012; 2:882-91. [PMID: 21136886 DOI: 10.1002/prca.200800040] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The release of proteins and membrane vesicles in the bloodstream regulates diverse vascular processes, both physiological, such as angiogenesis and haemostasis, and pathological, such as atherosclerosis and atherothrombosis. Proteomics, beside its canonical application for the expression profiling in cells and organs, can be applied to the study of secreted proteins and microvesicles, which play a significant role in the homeostasis of the vasculature, and the development of the atherosclerotic disease.
Collapse
Affiliation(s)
- Giordano Pula
- Cardiovascular Division, King's College London, London, UK
| | | | | | | | | | | |
Collapse
|
25
|
Wu-Zhang AX, Murphy AN, Bachman M, Newton AC. Isozyme-specific interaction of protein kinase Cδ with mitochondria dissected using live cell fluorescence imaging. J Biol Chem 2012; 287:37891-906. [PMID: 22988234 DOI: 10.1074/jbc.m112.412635] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
PKCδ signaling to mitochondria has been implicated in both mitochondrial apoptosis and metabolism. However, the mechanism by which PKCδ interacts with mitochondria is not well understood. Using FRET-based imaging, we show that PKCδ interacts with mitochondria by a novel and isozyme-specific mechanism distinct from its canonical recruitment to other membranes such as the plasma membrane or Golgi. Specifically, we show that PKCδ interacts with mitochondria following stimulation with phorbol esters or, in L6 myocytes, with insulin via a mechanism that requires two steps. In the first step, PKCδ translocates acutely to mitochondria by a mechanism that requires its C1A and C1B domains and a Leu-Asn sequence in its turn motif. In the second step, PKCδ is retained at mitochondria by a mechanism that depends on its C2 domain, a unique Glu residue in its activation loop, intrinsic catalytic activity, and the mitochondrial membrane potential. In contrast, of these determinants, only the C1B domain is required for the phorbol ester-stimulated translocation of PKCδ to other membranes. PKCδ also basally localizes to mitochondria and increases mitochondrial respiration via many of the same determinants that promote its agonist-evoked interaction. PKCδ localized to mitochondria has robust activity, as revealed by a FRET reporter of PKCδ-specific activity (δCKAR). These data support a model in which multiple determinants unique to PKCδ drive a specific interaction with mitochondria that promotes mitochondrial respiration.
Collapse
Affiliation(s)
- Alyssa X Wu-Zhang
- Department of Pharmacology, University of California San Diego, La Jolla, California 92093, USA
| | | | | | | |
Collapse
|
26
|
Chichger H, Grinnell KL, Casserly B, Chung CS, Braza J, Lomas-Neira J, Ayala A, Rounds S, Klinger JR, Harrington EO. Genetic disruption of protein kinase Cδ reduces endotoxin-induced lung injury. Am J Physiol Lung Cell Mol Physiol 2012; 303:L880-8. [PMID: 22983354 DOI: 10.1152/ajplung.00169.2012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The pathogenesis of acute lung injury and acute respiratory distress syndrome is characterized by sequestration of leukocytes in lung tissue, disruption of capillary integrity, and pulmonary edema. PKCδ plays a critical role in RhoA-mediated endothelial barrier function and inflammatory responses. We used mice with genetic deletion of PKCδ (PKCδ(-/-)) to assess the role of PKCδ in susceptibility to LPS-induced lung injury and pulmonary edema. Under baseline conditions or in settings of increased capillary hydrostatic pressures, no differences were noted in the filtration coefficients (k(f)) or wet-to-dry weight ratios between PKCδ(+/+) and PKCδ(-/-) mice. However, at 24 h after exposure to LPS, the k(f) values were significantly higher in lungs isolated from PKCδ(+/+) than PKCδ(-/-) mice. In addition, bronchoalveolar lavage fluid obtained from LPS-exposed PKCδ(+/+) mice displayed increased protein and cell content compared with LPS-exposed PKCδ(-/-) mice, but similar changes in inflammatory cytokines were measured. Histology indicated elevated LPS-induced cellularity and inflammation within PKCδ(+/+) mouse lung parenchyma relative to PKCδ(-/-) mouse lungs. Transient overexpression of catalytically inactive PKCδ cDNA in the endothelium significantly attenuated LPS-induced endothelial barrier dysfunction in vitro and increased k(f) lung values in PKCδ(+/+) mice. However, transient overexpression of wild-type PKCδ cDNA in PKCδ(-/-) mouse lung vasculature did not alter the protective effects of PKCδ deficiency against LPS-induced acute lung injury. We conclude that PKCδ plays a role in the pathological progression of endotoxin-induced lung injury, likely mediated through modulation of inflammatory signaling and pulmonary vascular barrier function.
Collapse
Affiliation(s)
- Havovi Chichger
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island 02908, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Protein kinase cδ in apoptosis: a brief overview. Arch Immunol Ther Exp (Warsz) 2012; 60:361-72. [PMID: 22918451 DOI: 10.1007/s00005-012-0188-8] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2011] [Accepted: 08/06/2012] [Indexed: 12/21/2022]
Abstract
Protein kinase C-delta (PKCδ), a member of the lipid-regulated serine/threonine PKC family, has been implicated in a wide range of important cellular processes. In the past decade, the critical role of PKCδ in the regulation of both intrinsic and extrinsic apoptosis pathways has been widely explored. In most cases, over-expression or activation of PKCδ results in the induction of apoptosis. The phosphorylations and multiple cell organelle translocations of PKCδ initiate apoptosis by targeting multiple downstream effectors. During apoptosis, PKCδ is proteolytically cleaved by caspase-3 to generate a constitutively activated catalytic fragment, which amplifies apoptosis cascades in nucleus and mitochondria. However, PKCδ also exerts its anti-apoptotic and pro-survival roles in some cases. Therefore, the complicated role of PKCδ in apoptosis appears to be stimulus and cell type dependent. This review is mainly focused on how PKCδ gets activated in diverse ways in response to apoptotic signals and how PKCδ targets different downstream regulators to sponsor or restrain apoptosis induction.
Collapse
|
28
|
Burniston JG, Kenyani J, Wastling JM, Burant CF, Qi NR, Koch LG, Britton SL. Proteomic analysis reveals perturbed energy metabolism and elevated oxidative stress in hearts of rats with inborn low aerobic capacity. Proteomics 2011; 11:3369-79. [PMID: 21751351 DOI: 10.1002/pmic.201000593] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Selection on running capacity has created rat phenotypes of high-capacity runners (HCRs) that have enhanced cardiac function and low-capacity runners (LCRs) that exhibit risk factors of metabolic syndrome. We analysed hearts of HCRs and LCRs from generation 22 of selection using DIGE and identified proteins from MS database searches. The running capacity of HCRs was six-fold greater than LCRs. DIGE resolved 957 spots and proteins were unambiguously identified in 369 spots. Protein expression profiling detected 67 statistically significant (p<0.05; false discovery rate <10%, calculated using q-values) differences between HCRs and LCRs. Hearts of HCR rats exhibited robust increases in the abundance of each enzyme of the β-oxidation pathway. In contrast, LCR hearts were characterised by the modulation of enzymes associated with ketone body or amino acid metabolism. LCRs also exhibited enhanced expression of antioxidant enzymes such as catalase and greater phosphorylation of α B-crystallin at serine 59, which is a common point of convergence in cardiac stress signalling. Thus, proteomic analysis revealed selection on low running capacity is associated with perturbations in cardiac energy metabolism and provided the first evidence that the LCR cardiac proteome is exposed to greater oxidative stress.
Collapse
Affiliation(s)
- Jatin G Burniston
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK.
| | | | | | | | | | | | | |
Collapse
|
29
|
Pyruvate kinase type M2: A key regulator of the metabolic budget system in tumor cells. Int J Biochem Cell Biol 2011; 43:969-80. [DOI: 10.1016/j.biocel.2010.02.005] [Citation(s) in RCA: 480] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Revised: 01/24/2010] [Accepted: 02/08/2010] [Indexed: 12/17/2022]
|
30
|
Hiding in plain sight: uncovering a new function of vitamin A in redox signaling. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1821:241-7. [PMID: 21763457 DOI: 10.1016/j.bbalip.2011.06.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Revised: 05/27/2011] [Accepted: 06/15/2011] [Indexed: 01/05/2023]
Abstract
The protein kinase Cδ signalosome modulates the generation of acetyl-Coenzyme A from glycolytic sources. This module is composed of four interlinked components: PKCδ, the signal adapter p66Shc, cytochrome c, and vitamin A. It resides in the intermembrane space of mitochondria, and is at the center of a feedback loop that senses upstream the redox balance between oxidized and reduced cytochrome c as a measure of the workload of the respiratory chain, and transmits a forward signal to the pyruvate dehydrogenase complex to adjust the flux of fuel entering the tricarboxylic acid cycle. The novel role of vitamin A as co-activator and potential electron carrier, required for redox activation of PKCδ, is discussed. This article is part of a Special Issue entitled Retinoid and Lipid Metabolism.
Collapse
|
31
|
Mayr M, May D, Gordon O, Madhu B, Gilon D, Yin X, Xing Q, Drozdov I, Ainali C, Tsoka S, Xu Q, Griffiths J, Horrevoets A, Keshet E. Metabolic homeostasis is maintained in myocardial hibernation by adaptive changes in the transcriptome and proteome. J Mol Cell Cardiol 2011; 50:982-90. [PMID: 21354174 PMCID: PMC3107937 DOI: 10.1016/j.yjmcc.2011.02.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Revised: 01/31/2011] [Accepted: 02/14/2011] [Indexed: 11/18/2022]
Abstract
A transgenic mouse model for conditional induction of long-term hibernation via myocardium-specific expression of a VEGF-sequestering soluble receptor allowed the dissection of the hibernation process into an initiation and a maintenance phase. The hypoxic initiation phase was characterized by peak levels of K(ATP) channel and glucose transporter 1 (GLUT1) expression. Glibenclamide, an inhibitor of K(ATP) channels, blocked GLUT1 induction. In the maintenance phase, tissue hypoxia and GLUT1 expression were reduced. Thus, we employed a combined "-omics" approach to resolve this cardioprotective adaptation process. Unguided bioinformatics analysis on the transcriptomic, proteomic and metabolomic datasets confirmed that anaerobic glycolysis was affected and that the observed enzymatic changes in cardiac metabolism were directly linked to hypoxia-inducible factor (HIF)-1 activation. Although metabolite concentrations were kept relatively constant, the combination of the proteomic and transcriptomic dataset improved the statistical confidence of the pathway analysis by 2 orders of magnitude. Importantly, proteomics revealed a reduced phosphorylation state of myosin light chain 2 and cardiac troponin I within the contractile apparatus of hibernating hearts in the absence of changes in protein abundance. Our study demonstrates how combining different "-omics" datasets aids in the identification of key biological pathways: chronic hypoxia resulted in a pronounced adaptive response at the transcript and the protein level to keep metabolite levels steady. This preservation of metabolic homeostasis is likely to contribute to the long-term survival of the hibernating myocardium.
Collapse
Affiliation(s)
- Manuel Mayr
- King's British Heart Foundation Centre, King's College London, London, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Up-regulation and redistribution of protein kinase C-δ in chronically hypoxic heart. Mol Cell Biochem 2010; 345:271-82. [DOI: 10.1007/s11010-010-0581-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Accepted: 08/27/2010] [Indexed: 12/29/2022]
|
33
|
Nguyen TT, Ogbi M, Yu Q, Fishman JB, Thomas W, Harvey BJ, Fulton D, Johnson JA. Modulation of the protein kinase Cdelta interaction with the "d" subunit of F1F0-ATP synthase in neonatal cardiac myocytes: development of cell-permeable, mitochondrially targeted inhibitor and facilitator peptides. J Biol Chem 2010; 285:22164-73. [PMID: 20460381 PMCID: PMC2903377 DOI: 10.1074/jbc.m109.077578] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2009] [Revised: 05/10/2010] [Indexed: 11/06/2022] Open
Abstract
The F(1)F(0)-ATP synthase provides approximately 90% of cardiac ATP, yet little is known regarding its regulation under normal or pathological conditions. Previously, we demonstrated that protein kinase Cdelta (PKCdelta) inhibits F(1)F(0) activity via an interaction with the "d" subunit of F(1)F(0)-ATP synthase (dF(1)F(0)) in neonatal cardiac myocytes (NCMs) (Nguyen, T., Ogbi, M., and Johnson, J. A. (2008) J. Biol. Chem. 283, 29831-29840). We have now identified a dF(1)F(0)-derived peptide (NH(2)-(2)AGRKLALKTIDWVSF(16)-COOH) that inhibits PKCdelta binding to dF(1)F(0) in overlay assays. We have also identified a second dF(1)F(0)-derived peptide (NH(2)-(111)RVREYEKQLEKIKNMI(126)-COOH) that facilitates PKCdelta binding to dF(1)F(0). Incubation of NCMs with versions of these peptides containing HIV-Tat protein transduction and mammalian mitochondrial targeting sequences resulted in their delivery into mitochondria. Preincubation of NCMs, with 10 nm extracellular concentrations of the mitochondrially targeted PKCdelta-dF(1)F(0) interaction inhibitor, decreased 100 nm 4beta-phorbol 12-myristate 13-acetate (4beta-PMA)-induced co-immunoprecipitation of PKCdelta with dF(1)F(0) by 50 +/- 15% and abolished the 30 nm 4beta-PMA-induced inhibition of F(1)F(0)-ATPase activity. A scrambled sequence (inactive) peptide, which contained HIV-Tat and mitochondrial targeting sequences, was without effect. In contrast, the cell-permeable, mitochondrially targeted PKCdelta-dF(1)F(0) facilitator peptide by itself induced the PKCdelta-dF(1)F(0) co-immunoprecipitation and inhibited F(1)F(0)-ATPase activity. In in vitro PKC add-back experiments, the PKCdelta-F(1)F(0) inhibitor blocked PKCdelta-mediated inhibition of F(1)F(0)-ATPase activity, whereas the facilitator induced inhibition. We have developed the first cell-permeable, mitochondrially targeted modulators of the PKCdelta-dF(1)F(0) interaction in NCMs. These novel peptides will improve our understanding of cardiac F(1)F(0) regulation and may have potential as therapeutics to attenuate cardiac injury.
Collapse
Affiliation(s)
- Tiffany T. Nguyen
- From the Department of Pharmacology and Toxicology, School of Medicine, and Program in Regenerative Medicine, Institute of Molecular Medicine and Genetics, and
| | - Mourad Ogbi
- From the Department of Pharmacology and Toxicology, School of Medicine, and Program in Regenerative Medicine, Institute of Molecular Medicine and Genetics, and
| | - Qilin Yu
- From the Department of Pharmacology and Toxicology, School of Medicine, and Program in Regenerative Medicine, Institute of Molecular Medicine and Genetics, and
| | | | - Warren Thomas
- the Department of Molecular Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Brian J. Harvey
- the Department of Molecular Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - David Fulton
- From the Department of Pharmacology and Toxicology, School of Medicine, and Program in Regenerative Medicine, Institute of Molecular Medicine and Genetics, and
- The Vascular Biology Center, Medical College of Georgia, Augusta, Georgia 30912-2300
| | - John A. Johnson
- From the Department of Pharmacology and Toxicology, School of Medicine, and Program in Regenerative Medicine, Institute of Molecular Medicine and Genetics, and
| |
Collapse
|
34
|
Folmes CD, Sawicki G, Cadete VJ, Masson G, Barr AJ, Lopaschuk GD. Novel O-palmitolylated beta-E1 subunit of pyruvate dehydrogenase is phosphorylated during ischemia/reperfusion injury. Proteome Sci 2010; 8:38. [PMID: 20618950 PMCID: PMC2909933 DOI: 10.1186/1477-5956-8-38] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Accepted: 07/09/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND During and following myocardial ischemia, glucose oxidation rates are low and fatty acids dominate as a source of oxidative metabolism. This metabolic phenotype is associated with contractile dysfunction during reperfusion. To determine the mechanism of this reliance on fatty acid oxidation as a source of ATP generation, a functional proteomics approach was utilized. RESULTS 2-D gel electrophoresis of mitochondria from working rat hearts subjected to 25 minutes of global no flow ischemia followed by 40 minutes of aerobic reperfusion identified 32 changes in protein abundance compared to aerobic controls. Of the five proteins with the greatest change in abundance, two were increased (long chain acyl-coenzyme A dehydrogenase (48 +/- 1 versus 39 +/- 3 arbitrary units, n = 3, P < 0.05) and alpha subunit of ATP synthase (189 +/- 15 versus 113 +/- 23 arbitrary units, n = 3, P < 0.05)), while two were decreased (24 kDa subunit of NADH-ubiquinone oxidoreductase (94 +/- 7 versus 127 +/- 9 arbitrary units, n = 3, P < 0.05) and D subunit of ATP synthase (230 +/- 11 versus 368 +/- 47 arbitrary units, n = 3, P < 05)). Two forms of pyruvate dehydrogenase betaE1 subunit, the rate-limiting enzyme for glucose oxidation, were also identified. The protein level of the more acidic form of pyruvate dehydrogenase was reduced during reperfusion (37 +/- 4 versus 56 +/- 7 arbitrary units, n = 3, P < 05), while the more basic form remained unchanged. The more acidic isoform was found to be O-palmitoylated, while both isoforms exhibited ischemia/reperfusion-induced phosphorylation. In silico analysis identified the putative kinases as the insulin receptor kinase for the more basic form and protein kinase Czeta or protein kinase A for the more acidic form. These modifications of pyruvate dehydrogenase are associated with a 35% decrease in glucose oxidation during reperfusion. CONCLUSIONS Cardiac ischemia/reperfusion induces significant changes to a number of metabolic proteins of the mitochondrial proteome. In particular, ischemia/reperfusion induced the post-translational modification of pyruvate dehydrogenase, the rate-limiting step of glucose oxidation, which is associated with a 35% decrease in glucose oxidation during reperfusion. Therefore these post-translational modifications may have important implications in the regulation of myocardial energy metabolism.
Collapse
Affiliation(s)
- Clifford Dl Folmes
- Cardiovascular Research Group and the Departments of Pharmacology and Pediatrics, The University of Alberta, Edmonton, Alberta, Canada
| | - Grzegorz Sawicki
- Department of Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.,Department of Clinical Chemistry, Medical University of Wroclaw, Wroclaw, Poland
| | - Virgilio Jj Cadete
- Department of Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Grant Masson
- Cardiovascular Research Group and the Departments of Pharmacology and Pediatrics, The University of Alberta, Edmonton, Alberta, Canada
| | - Amy J Barr
- Cardiovascular Research Group and the Departments of Pharmacology and Pediatrics, The University of Alberta, Edmonton, Alberta, Canada
| | - Gary D Lopaschuk
- Cardiovascular Research Group and the Departments of Pharmacology and Pediatrics, The University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
35
|
Puntmann VO, Mayr M. Phenotyping transgenic animals—An integrated readout of pathophysiology by combining proteomics and metabolomics with cardiovascular imaging. J Mol Cell Cardiol 2010; 48:571-3. [DOI: 10.1016/j.yjmcc.2009.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2009] [Revised: 12/03/2009] [Accepted: 12/03/2009] [Indexed: 10/20/2022]
|
36
|
Arrell DK, Zlatkovic J, Kane GC, Yamada S, Terzic A. ATP-sensitive K+ channel knockout induces cardiac proteome remodeling predictive of heart disease susceptibility. J Proteome Res 2010; 8:4823-34. [PMID: 19673485 DOI: 10.1021/pr900561g] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Forecasting disease susceptibility requires detection of maladaptive signatures prior to onset of overt symptoms. A case-in-point are cardiac ATP-sensitive K+ (K(ATP)) channelopathies, for which the substrate underlying disease vulnerability remains to be identified. Resolving molecular pathobiology, even for single genetic defects, mandates a systems platform to reliably diagnose disease predisposition. High-throughput proteomic analysis was here integrated with network biology to decode consequences of Kir6.2 K(ATP) channel pore deletion. Differential two-dimensional gel electrophoresis reproducibly resolved >800 protein species from hearts of asymptomatic wild-type and Kir6.2-knockout counterparts. K(ATP) channel ablation remodeled the cardiac proteome, significantly altering 71 protein spots, from which 102 unique identities were assigned following hybrid linear ion trap quadrupole-Orbitrap tandem mass spectrometry. Ontological annotation stratified the K(ATP) channel-dependent protein cohort into a predominant bioenergetic module (63 resolved identities), with additional focused sets representing signaling molecules (6), oxidoreductases (8), chaperones (6), and proteins involved in catabolism (6), cytostructure (8), and transcription and translation (5). Protein interaction mapping, in conjunction with expression level changes, localized a K(ATP) channel-associated subproteome within a nonstochastic scale-free network. Global assessment of the K(ATP) channel deficient environment verified the primary impact on metabolic pathways and revealed overrepresentation of markers associated with cardiovascular disease. Experimental imposition of graded stress precipitated exaggerated structural and functional myocardial defects in the Kir6.2-knockout, decreasing survivorship and validating the forecast of disease susceptibility. Proteomic cartography thus provides an integral view of molecular remodeling in the heart induced by K(ATP) channel deletion, establishing a systems approach that predicts outcome at a presymptomatic stage.
Collapse
Affiliation(s)
- D Kent Arrell
- Marriott Heart Disease Research Program, Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | |
Collapse
|
37
|
Iff J, Wang W, Sajic T, Oudry N, Gueneau E, Hopfgartner G, Varesio E, Szanto I. Differential proteomic analysis of STAT6 knockout mice reveals new regulatory function in liver lipid homeostasis. J Proteome Res 2010; 8:4511-24. [PMID: 19663508 DOI: 10.1021/pr9003272] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Increased inflammatory signaling is a key feature of metabolic disorders. In this context, the role of increased pro-inflammatory signals has been extensively studied. By contrast, no efforts have been dedicated to study the contrasting scenario: the attenuation of anti-inflammatory signals and their role in metabolic homeostasis. IL-4 and IL-13 are anti-inflammatory cytokines signaling through the Signal Transducer and Activator of Transcription 6 (STAT6). Our study was aimed at evaluating the lack of STAT6 signaling on liver homeostasis. To this end we analyzed the liver proteome of wild type and STAT6 knock-out mice using 2D nanoscale LC-MS/MS with iTRAQ labeling technique. The coordinated changes in proteins identified by this quantitative proteome analysis indicated disturbed lipid homeostasis and a state of hepatocellular stress. Most significantly, the expression of the liver fatty acid binding protein (FABP1) was increased in the knock-out mice. In line with the elevated FABP1 expression we found latent liver lipid accumulation in the STAT6-deficient mice which was further aggravated when mice were challenged by a high fat diet. In conclusion, our study revealed a so far uncharacterized role for STAT6 in regulating liver lipid homeostasis and demonstrates the importance of anti-inflammatory signaling in the defense against the development of liver steatosis.
Collapse
Affiliation(s)
- Joël Iff
- Department of Cellular Physiology and Metabolism, University of Geneva, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Yin X, Cuello F, Mayr U, Hao Z, Hornshaw M, Ehler E, Avkiran M, Mayr M. Proteomics analysis of the cardiac myofilament subproteome reveals dynamic alterations in phosphatase subunit distribution. Mol Cell Proteomics 2009; 9:497-509. [PMID: 20037178 PMCID: PMC2849712 DOI: 10.1074/mcp.m900275-mcp200] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Myofilament proteins are responsible for cardiac contraction. The myofilament subproteome, however, has not been comprehensively analyzed thus far. In the present study, cardiomyocytes were isolated from rodent hearts and stimulated with endothelin-1 and isoproterenol, potent inducers of myofilament protein phosphorylation. Subsequently, cardiomyocytes were “skinned,” and the myofilament subproteome was analyzed using a high mass accuracy ion trap tandem mass spectrometer (LTQ Orbitrap XL) equipped with electron transfer dissociation. As expected, a small number of myofilament proteins constituted the majority of the total protein mass with several known phosphorylation sites confirmed by electron transfer dissociation. More than 600 additional proteins were identified in the cardiac myofilament subproteome, including kinases and phosphatase subunits. The proteomic comparison of myofilaments from control and treated cardiomyocytes suggested that isoproterenol treatment altered the subcellular localization of protein phosphatase 2A regulatory subunit B56α. Immunoblot analysis of myocyte fractions confirmed that β-adrenergic stimulation by isoproterenol decreased the B56α content of the myofilament fraction in the absence of significant changes for the myosin phosphatase target subunit isoforms 1 and 2 (MYPT1 and MYPT2). Furthermore, immunolabeling and confocal microscopy revealed the spatial redistribution of these proteins with a loss of B56α from Z-disc and M-band regions but increased association of MYPT1/2 with A-band regions of the sarcomere following β-adrenergic stimulation. In summary, we present the first comprehensive proteomics data set of skinned cardiomyocytes and demonstrate the potential of proteomics to unravel dynamic changes in protein composition that may contribute to the neurohormonal regulation of myofilament contraction.
Collapse
Affiliation(s)
- Xiaoke Yin
- King's British Heart Foundation Centre, King's College London, London SE5 9NU, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Jacquet S, Yin X, Sicard P, Clark J, Kanaganayagam GS, Mayr M, Marber MS. Identification of cardiac myosin-binding protein C as a candidate biomarker of myocardial infarction by proteomics analysis. Mol Cell Proteomics 2009; 8:2687-99. [PMID: 19721077 DOI: 10.1074/mcp.m900176-mcp200] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Acute myocardial infarction (AMI) is a common cause of death for which effective treatments are available provided that diagnosis is rapid. The current diagnostic gold standards are circulating cardiac troponins I and T. However, their slow release delays diagnosis, and their persistence limits their utility in the identification of reinfarction. The aim was to identify candidate biomarkers of AMI. Isolated mouse hearts were perfused with oxygenated protein-free buffer, and coronary effluent was collected after ischemia or during matched normoxic perfusion. Effluents were analyzed using proteomics approaches based on one- or two-dimensional initial separation. Of the 459 proteins identified after ischemia with one-dimensional separation, 320 were not detected in the control coronary effluent. Among these were all classic existing biomarkers of AMI. We also identified the cardiac isoform of myosin-binding protein C in its full-length form and as a 40-kDa degradation product. This protein was not detected in the other murine organs examined, increased markedly with even trivial myocardial infarction, and could be detected in the plasma after myocardial infarction in vivo, a profile compatible with a biomarker of AMI. Two-dimensional fluorescence DIGE of ischemic and control coronary effluents identified more than 200 asymmetric spots verified by swapping dyes. Once again existing biomarkers of injury were confirmed as well as posttranslational modifications of antioxidant proteins such as peroxiredoxins. Perfusing hearts with protein-free buffers provides a platform of graded ischemic injury that allows detailed analysis of protein release and identification of candidate cardiac biomarkers like myosin-binding protein C.
Collapse
Affiliation(s)
- Sebastien Jacquet
- King's College London British Heart Foundation Centre, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
40
|
Nguyen T, Ogbi M, Johnson JA. Delta protein kinase C interacts with the d subunit of the F1F0 ATPase in neonatal cardiac myocytes exposed to hypoxia or phorbol ester. Implications for F1F0 ATPase regulation. J Biol Chem 2008; 283:29831-40. [PMID: 18725417 PMCID: PMC2573058 DOI: 10.1074/jbc.m801642200] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2008] [Revised: 08/06/2008] [Indexed: 11/06/2022] Open
Abstract
Mitochondrial protein kinase C isozymes have been reported to mediate both cardiac ischemic preconditioning and ischemia/reperfusion injury. In addition, cardiac preconditioning improves the recovery of ATP levels after ischemia/reperfusion injury. We have, therefore, evaluated protein kinase C modulation of the F(1)F(0) ATPase in neonatal cardiac myocytes. Exposure of cells to 3 or 100 nM 4beta-phorbol 12-myristate-13-acetate induced co-immunoprecipitation of delta protein kinase C (but not alpha, epsilon, or zeta protein kinase C) with the d subunit of the F(1)F(0) ATPase. This co-immunoprecipitation correlated with 40+/-3% and 72+/-9% inhibitions of oligomycin-sensitive F(1)F(0) ATPase activity, respectively. We observed prominent expression of delta protein kinase C in cardiac myocyte mitochondria, which was enhanced following a 4-h hypoxia exposure. In contrast, hypoxia decreased mitochondrial zetaPKC levels by 85+/-1%. Following 4 h of hypoxia, F(1)F(0) ATPase activity was inhibited by 75+/-9% and delta protein kinase C co-immunoprecipitated with the d subunit of F(1)F(0) ATPase. In vitro incubation of protein kinase C with F(1)F(0) ATPase enhanced F(1)F(0) activity in the absence of protein kinase C activators and inhibited it in the presence of activators. Recombinant delta protein kinase C also inhibited F(1)F(0) ATPase activity. Protein kinase C overlay assays revealed delta protein kinase C binding to the d subunit of F(1)F(0) ATPase, which was modulated by diacylglycerol, phosphatidylserine, and cardiolipin. Our results suggest a novel regulation of the F(1)F(0) ATPase by the delta protein kinase C isozyme.
Collapse
Affiliation(s)
- Tiffany Nguyen
- Department of Pharmacology and Toxicology, School of Medicine, Medical College of Georgia, Augusta, Georgia 30912-2300, USA
| | | | | |
Collapse
|
41
|
Mayr M, Liem D, Zhang J, Li X, Avliyakulov NK, Yang JI, Young G, Vondriska TM, Ladroue C, Madhu B, Griffiths JR, Gomes A, Xu Q, Ping P. Proteomic and metabolomic analysis of cardioprotection: Interplay between protein kinase C epsilon and delta in regulating glucose metabolism of murine hearts. J Mol Cell Cardiol 2008; 46:268-77. [PMID: 19027023 DOI: 10.1016/j.yjmcc.2008.10.008] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2008] [Revised: 09/29/2008] [Accepted: 10/02/2008] [Indexed: 02/02/2023]
Abstract
We applied a combined proteomic and metabolomic approach to obtain novel mechanistic insights in PKCvarepsilon-mediated cardioprotection. Mitochondrial and cytosolic proteins from control and transgenic hearts with constitutively active or dominant negative PKCvarepsilon were analyzed using difference in-gel electrophoresis (DIGE). Among the differentially expressed proteins were creatine kinase, pyruvate kinase, lactate dehydrogenase, and the cytosolic isoforms of aspartate amino transferase and malate dehydrogenase, the two enzymatic components of the malate aspartate shuttle, which are required for the import of reducing equivalents from glycolysis across the inner mitochondrial membrane. These enzymatic changes appeared to be dependent on PKCvarepsilon activity, as they were not observed in mice expressing inactive PKCvarepsilon. High-resolution proton nuclear magnetic resonance ((1)H-NMR) spectroscopy confirmed a pronounced effect of PKCvarepsilon activity on cardiac glucose and energy metabolism: normoxic hearts with constitutively active PKCvarepsilon had significantly lower concentrations of glucose, lactate, glutamine and creatine, but higher levels of choline, glutamate and total adenosine nucleotides. Moreover, the depletion of cardiac energy metabolites was slower during ischemia/reperfusion injury and glucose metabolism recovered faster upon reperfusion in transgenic hearts with active PKCvarepsilon. Notably, inhibition of PKCvarepsilon resulted in compensatory phosphorylation and mitochondrial translocation of PKCdelta. Taken together, our findings are the first evidence that PKCvarepsilon activity modulates cardiac glucose metabolism and provide a possible explanation for the synergistic effect of PKCdelta and PKCvarepsilon in cardioprotection.
Collapse
Affiliation(s)
- Manuel Mayr
- Cardiovascular Division, BHF Centre, King's College, London, 125 Coldharbour Lane, London SE5 9NU, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Affiliation(s)
- Manuel Mayr
- From the Cardiovascular Division, King’s College, London School of Medicine, King’s College London, UK
| |
Collapse
|
43
|
Schröder E, Brennan JP, Eaton P. Cardiac peroxiredoxins undergo complex modifications during cardiac oxidant stress. Am J Physiol Heart Circ Physiol 2008; 295:H425-33. [PMID: 18502910 PMCID: PMC2494773 DOI: 10.1152/ajpheart.00017.2008] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Peroxiredoxins (Prdxs), a family of antioxidant and redox-signaling proteins, are plentiful within the heart; however, their cardiac functions are poorly understood. These studies were designed to characterize the complex changes in Prdxs induced by oxidant stress in rat myocardium. Hydrogen peroxide, a Prdx substrate, was used as the model oxidant pertinent to redox signaling during health and to injury at higher concentrations. Rat hearts were aerobically perfused with a broad concentration range of hydrogen peroxide by the Langendorff method, homogenized, and analyzed by immunoblotting. Heart extracts were also analyzed by size-exclusion chromatography under nondenaturing conditions. Hydrogen peroxide-induced changes in disulfide bond formation, nonreversible oxidation of cysteine (hyperoxidation), and subcellular localization were determined. Hydrogen peroxide induced an array of changes in the myocardium, including formation of disulfide bonds that were intermolecular for Prdx1, Prdx2, and Prdx3 but intramolecular within Prdx5. For Prdx1, Prdx2, and Prdx5, disulfide bond formation can be approximated to an EC50 of 10–100, 1–10, and 100–1,000 μM peroxide, respectively. Hydrogen peroxide induced hyperoxidation, not just within monomeric Prdx (by SDS-PAGE), but also within Prdx disulfide dimers, and reflects a flexibility within the dimeric unit. Prdx oxidation was also associated with movement from the cytosolic to the membrane and myofilament-enriched fractions. In summary, Prdxs undergo a complex series of redox-dependent structural changes in the heart in response to oxidant challenge with its substrate hydrogen peroxide.
Collapse
Affiliation(s)
- Ewald Schröder
- Dept. of Cardiology, St. Thomas' Hospital, King's College London, London SE1 7EH, UK
| | | | | |
Collapse
|
44
|
Pyruvate Kinase Type M2: A Key Regulator Within the Tumour Metabolome and a Tool for Metabolic Profiling of Tumours. ONCOGENES MEET METABOLISM 2008:99-124. [DOI: 10.1007/2789_2008_091] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
45
|
Guo D, Nguyen T, Ogbi M, Tawfik H, Ma G, Yu Q, Caldwell RW, Johnson JA. Protein kinase C-epsilon coimmunoprecipitates with cytochrome oxidase subunit IV and is associated with improved cytochrome-c oxidase activity and cardioprotection. Am J Physiol Heart Circ Physiol 2007; 293:H2219-30. [PMID: 17660387 DOI: 10.1152/ajpheart.01306.2006] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
We have utilized an in situ rat coronary ligation model to establish a PKC-epsilon cytochrome oxidase subunit IV (COIV) coimmunoprecipitation in myocardium exposed to ischemic preconditioning (PC). Ischemia-reperfusion (I/R) damage and PC protection were confirmed using tetrazolium-based staining methods and serum levels of cardiac troponin I. Homogenates prepared from the regions at risk (RAR) and not at risk (RNAR) for I/R injury were fractionated into cell-soluble (S), 600 g low-speed centrifugation (L), Percoll/Optiprep density gradient-purified mitochondrial (M), and 100,000 g particulate (P) fractions. COIV immunoreactivity and cytochrome-c oxidase activity measurements estimated the percentages of cellular mitochondria in S, L, M, and P fractions to be 0, 55, 29, and 16%, respectively. We observed 18, 3, and 3% of PKC-delta, -epsilon, and -zeta isozymes in the M fraction under basal conditions. Following PC, we observed a 61% increase in PKC-epsilon levels in the RAR M fraction compared with the RNAR M fraction. In RAR mitochondria, we also observed a 2.8-fold increase in PKC-epsilon serine 729 phosphoimmunoreactivity (autophosphorylation), indicating the presence of activated PKC-epsilon in mitochondria following PC. PC administered before prolonged I/R induced a 1.9-fold increase in the coimmunoprecipitation of COIV, with anti-PKC-epsilon antisera and a twofold enhancement of cytochrome-c oxidase activity. Our results suggest that PKC-epsilon may interact with COIV as a component of the cardioprotection in PC. Induction of this interaction may provide a novel therapeutic target for protecting the heart from I/R damage.
Collapse
Affiliation(s)
- Dehuang Guo
- Department of Pharmacology & Toxicology, School of Medicine, Medical College of Georgia, Augusta, Georgia 30912-2300, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Meier BW, Gomez JD, Kirichenko OV, Thompson JA. Mechanistic basis for inflammation and tumor promotion in lungs of 2,6-di-tert-butyl-4-methylphenol-treated mice: electrophilic metabolites alkylate and inactivate antioxidant enzymes. Chem Res Toxicol 2007; 20:199-207. [PMID: 17305404 PMCID: PMC2570584 DOI: 10.1021/tx060214f] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
An established model for mechanistic analysis of lung carcinogenesis involves administration of 3-methylcholanthrene to mice followed by several weekly injections of the tumor promoter 2,6-di-tert-butyl-4-methylphenol (BHT). BHT is metabolized to quinone methides (QMs) responsible for promoting tumor formation. QMs are strongly electrophilic and readily form adducts with proteins. The goal of the present study was to identify adducted proteins in the lungs of mice injected with BHT and to assess the potential impact of these modifications on tumorigenesis. Cytosolic proteins from treated mouse lungs were separated by two-dimensional electrophoresis, adducts detected by immunoblotting, and proteins identified by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Eight adducts were detected in the lungs of most, or all, of six experimental groups of BALB mice. Of these adducts, several were structural proteins, but others, namely, peroxiredoxin 6 (Prx6), Cu,Zn-superoxide dismutase (SOD1), carbonyl reductase, and selenium-binding protein 1, have direct or indirect antioxidant functions. When the 9000g supernatant fraction of mouse lung was treated with BHT-QM (2,6-di-tert-butyl-4-methylene-2,5-cyclohexadienone), substantial lipid peroxidation and increases in hydrogen peroxide and superoxide formation were observed. Studies with human Prx6 and bovine SOD1 demonstrated inhibition of enzyme activity concomitant with adduct formation. LC-MS/MS analysis of digests of adducted Prx6 demonstrated adduction of both Cys 91 and Cys 47; the latter residue is essential for peroxidatic activity. Analysis of QM-treated bovine SOD1 by matrix-assisted laser desorption/ionization time-of-flight MS demonstrated the predominance of a monoadduct at His 78. This study provides evidence that indicates Prx6, SOD1, and possibly other antioxidant enzymes in mouse lung are inhibited by BHT-derived QMs leading to enhanced levels of reactive oxygen species and inflammation and providing a mechanistic basis for the effects of BHT on lung tumorigenesis.
Collapse
Affiliation(s)
| | | | | | - John A. Thompson
- To whom correspondence should be addressed. Tel. 303−315−6167. Fax: 303−315−0274. E-mail:
| |
Collapse
|
47
|
Arikawa E, Ma RCW, Isshiki K, Luptak I, He Z, Yasuda Y, Maeno Y, Patti ME, Weir GC, Harris RA, Zammit VA, Tian R, King GL. Effects of insulin replacements, inhibitors of angiotensin, and PKCbeta's actions to normalize cardiac gene expression and fuel metabolism in diabetic rats. Diabetes 2007; 56:1410-20. [PMID: 17363743 DOI: 10.2337/db06-0655] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
High-density oligonucleotide arrays were used to compare gene expression of rat hearts from control, untreated diabetic, and diabetic groups treated with islet cell transplantation (ICT), protein kinase C (PKC)beta inhibitor ruboxistaurin, or ACE inhibitor captopril. Among the 376 genes that were differentially expressed between untreated diabetic and control hearts included key metabolic enzymes that account for the decreased glucose and increased free fatty acid utilization in the diabetic heart. ICT or insulin replacements reversed these gene changes with normalization of hyperglycemia, dyslipidemia, and cardiac PKC activation in diabetic rats. Surprisingly, both ruboxistaurin and ACE inhibitors improved the metabolic gene profile (confirmed by real-time RT-PCR and protein analysis) and ameliorated PKC activity in diabetic hearts without altering circulating metabolites. Functional assessments using Langendorff preparations and (13)C nuclear magnetic resonance spectroscopy showed a 36% decrease in glucose utilization and an impairment in diastolic function in diabetic rat hearts, which were normalized by all three treatments. In cardiomyocytes, PKC inhibition attenuated fatty acid-induced increases in the metabolic genes PDK4 and UCP3 and also prevented fatty acid-mediated inhibition of basal and insulin-stimulated glucose oxidation. Thus, PKCbeta or ACE inhibitors may ameliorate cardiac metabolism and function in diabetes partly by normalization of fuel metabolic gene expression directly in the myocardium.
Collapse
Affiliation(s)
- Emi Arikawa
- Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Mayr M, Madhu B, Xu Q. Proteomics and Metabolomics Combined in Cardiovascular Research. Trends Cardiovasc Med 2007; 17:43-8. [PMID: 17292045 DOI: 10.1016/j.tcm.2006.11.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2006] [Revised: 11/28/2006] [Accepted: 11/29/2006] [Indexed: 11/22/2022]
Abstract
Proteomics and metabolomics offer a nonbiased suite of tools to address pathophysiologic mechanisms from various levels by integrating signal transduction, cellular metabolism, and phenotype analysis. To link alterations of cellular proteins to metabolism and function, we have recently combined proteomic and metabolomic techniques. Examples, including genetic manipulation, ischemic preconditioning, atherosclerosis, and stem cell differentiation, are discussed to illustrate how the combination of these updated technologies may advance our understanding of cardiovascular biology.
Collapse
Affiliation(s)
- Manuel Mayr
- Cardiovascular Division, King's College, London SE5 9NU, UK.
| | | | | |
Collapse
|
49
|
Siwko S, Mochly-Rosen D. Use of a novel method to find substrates of protein kinase C delta identifies M2 pyruvate kinase. Int J Biochem Cell Biol 2007; 39:978-87. [PMID: 17337233 PMCID: PMC1931518 DOI: 10.1016/j.biocel.2007.01.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2006] [Revised: 01/12/2007] [Accepted: 01/17/2007] [Indexed: 11/24/2022]
Abstract
Protein kinase C (PKC) family members have been implicated in numerous cellular processes. However, identifying the substrates of each PKC isozyme remains a challenge. Here, we describe a method using two-dimensional (2D) isoelectric focusing gel electrophoresis to identify substrates of delta PKC (deltaPKC) in MCF-7 breast carcinoma cells. We show that M2 pyruvate kinase is a substrate of deltaPKC, and further characterize the interaction between M2 pyruvate kinase and deltaPKC in MCF-7 cells by immunoprecipitation. deltaPKC activation in vitro or in cells did not appear to alter the enzyme activity or polymerization of M2 pyruvate kinase.
Collapse
Affiliation(s)
- Stefan Siwko
- Cancer Biology Program, CCSR Room 3145, 269 Campus Dr., Stanford University, Stanford, CA 94305, USA.
| | | |
Collapse
|
50
|
Andreadou I, Iliodromitis EK, Mikros E, Constantinou M, Agalias A, Magiatis P, Skaltsounis AL, Kamber E, Tsantili-Kakoulidou A, Kremastinos DT. The olive constituent oleuropein exhibits anti-ischemic, antioxidative, and hypolipidemic effects in anesthetized rabbits. J Nutr 2006; 136:2213-9. [PMID: 16857843 DOI: 10.1093/jn/136.8.2213] [Citation(s) in RCA: 176] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Thgoal of this study was to evaluate the efficacy of the antioxidant olive constituent, oleuropein, on infarct size, oxidative damage, and the metabolic profile in rabbits subjected to ischemia. Oleuropein, 10 or 20 mg/(kg x d), was administered to 8 groups that consumed a normal or hypercholesterolemic diet for 6 wk or only the higher dose for 3 wk. Circulating levels of malondialdehyde, protein carbonyl, nitrite+nitrate, cholesterol, triglycerides, SOD activity, and the metabolic profile were measured using 1H NMR spectra. In rabbits that consumed the normal diet, the infarct size (percentage of infarct to risk areas) was reduced by the administration of 10 mg oleuropein/(kg x d) (16.1 +/- 2.9%) or 20 mg oleuropein/(kg x d) for 3 wk (21.7 +/- 2.2%) or for 6 wk (24.3 +/- 1.3%) compared with the control group (48.05 +/- 2.0%, P < 0.05). Only the higher dose of 20 mg/(kg x d) reduced the infarct size in hypercholesterolemic rabbits (34.7 +/- 4.4% for 6 wk and 34.8 +/- 6.1% for 3 wk) compared with the cholesterol-fed control group (52.8 +/- 2.4%, P < 0.05). Oleuropein decreased the plasma lipid peroxidation product and protein carbonyl concentrations compared with the control groups, in which these factors increased relative to baseline due to ischemia and reperfusion. Furthermore, in rabbits administered oleuropein, RBC superoxide dismutase activity did not change during ischemia and reperfusion. This activity was significantly higher than in both control groups in which it was reduced by ischemia and reperfusion compared with baseline. Treatment for 6 wk with both doses of oleuropein reduced total cholesterol and triglyceride concentrations. 1H NMR spectra revealed a different profile of glycolysis metabolites in the oleuropein-treated groups compared with the controls. Oleuropein, for 3 or 6 wk, reduced the infarct size, conferred strong antioxidant protection and reduced the circulating lipids. This is the first experimental study in vivo that suggests the possibility of using an olive constituent in the treatment of ischemia.
Collapse
Affiliation(s)
- Ioanna Andreadou
- Second University Department of Cardiology, Medical School, Attikon General Hospital, University of Athens, Athens, Greece.
| | | | | | | | | | | | | | | | | | | |
Collapse
|