1
|
Renton MC, McGee SL, Howlett KF. The role of protein kinase D (PKD) in obesity: Lessons from the heart and other tissues. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119814. [PMID: 39128598 DOI: 10.1016/j.bbamcr.2024.119814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/15/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024]
Abstract
Obesity causes a range of tissue dysfunctions that increases the risk for morbidity and mortality. Protein kinase D (PKD) represents a family of stress-activated intracellular signalling proteins that regulate essential processes such as cell proliferation and differentiation, cell survival, and exocytosis. Evidence suggests that PKD regulates the cellular adaptations to the obese environment in metabolically important tissues and drives the development of a variety of diseases. This review explores the role that PKD plays in tissue dysfunction in obesity, with special consideration of the development of obesity-mediated cardiomyopathy, a distinct cardiovascular disease that occurs in the absence of common comorbidities and leads to eventual heart failure and death. The downstream mechanisms mediated by PKD that could contribute to dysfunctions observed in the heart and other metabolically important tissues in obesity, and the predicted cell types involved are discussed to suggest potential targets for the development of therapeutics against obesity-related disease.
Collapse
Affiliation(s)
- Mark C Renton
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Australia; The Fralin Biomedical Research Institute at Virginia Tech Carilion, Centre for Vascular and Heart Research, Roanoke, VA, USA.
| | - Sean L McGee
- Institute for Mental and Physical Health and Clinical Translation, Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Australia.
| | - Kirsten F Howlett
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Australia.
| |
Collapse
|
2
|
Waheed‐Ullah Q, Wilsdon A, Abbad A, Rochette S, Bu'Lock F, Hitz M, Dombrowsky G, Cuello F, Brook JD, Loughna S. Effect of deletion of the protein kinase PRKD1 on development of the mouse embryonic heart. J Anat 2024; 245:70-83. [PMID: 38419169 PMCID: PMC11161829 DOI: 10.1111/joa.14033] [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: 06/29/2023] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 03/02/2024] Open
Abstract
Congenital heart disease (CHD) is the most common congenital anomaly, with an overall incidence of approximately 1% in the United Kingdom. Exome sequencing in large CHD cohorts has been performed to provide insights into the genetic aetiology of CHD. This includes a study of 1891 probands by our group in collaboration with others, which identified three novel genes-CDK13, PRKD1, and CHD4, in patients with syndromic CHD. PRKD1 encodes a serine/threonine protein kinase, which is important in a variety of fundamental cellular functions. Individuals with a heterozygous mutation in PRKD1 may have facial dysmorphism, ectodermal dysplasia and may have CHDs such as pulmonary stenosis, atrioventricular septal defects, coarctation of the aorta and bicuspid aortic valve. To obtain a greater appreciation for the role that this essential protein kinase plays in cardiogenesis and CHD, we have analysed a Prkd1 transgenic mouse model (Prkd1em1) carrying deletion of exon 2, causing loss of function. High-resolution episcopic microscopy affords detailed morphological 3D analysis of the developing heart and provides evidence for an essential role of Prkd1 in both normal cardiac development and CHD. We show that homozygous deletion of Prkd1 is associated with complex forms of CHD such as atrioventricular septal defects, and bicuspid aortic and pulmonary valves, and is lethal. Even in heterozygotes, cardiac differences occur. However, given that 97% of Prkd1 heterozygous mice display normal heart development, it is likely that one normal allele is sufficient, with the defects seen most likely to represent sporadic events. Moreover, mRNA and protein expression levels were investigated by RT-qPCR and western immunoblotting, respectively. A significant reduction in Prkd1 mRNA levels was seen in homozygotes, but not heterozygotes, compared to WT littermates. While a trend towards lower PRKD1 protein expression was seen in the heterozygotes, the difference was only significant in the homozygotes. There was no compensation by the related Prkd2 and Prkd3 at transcript level, as evidenced by RT-qPCR. Overall, we demonstrate a vital role of Prkd1 in heart development and the aetiology of CHD.
Collapse
Affiliation(s)
- Qazi Waheed‐Ullah
- School of Life Sciences, Faculty of Medicine and Health SciencesUniversity of NottinghamNottinghamUK
| | - Anna Wilsdon
- School of Life Sciences, Faculty of Medicine and Health SciencesUniversity of NottinghamNottinghamUK
| | - Aseel Abbad
- School of Life Sciences, Faculty of Medicine and Health SciencesUniversity of NottinghamNottinghamUK
| | - Sophie Rochette
- School of Life Sciences, Faculty of Medicine and Health SciencesUniversity of NottinghamNottinghamUK
| | - Frances Bu'Lock
- East Midlands Congenital Heart CentreUniversity Hospitals of Leicester NHS TrustLeicesterUK
| | - Marc‐Phillip Hitz
- Institute of Medical GeneticsCarl von Ossietzky University OldenburgOldenburgGermany
| | - Gregor Dombrowsky
- Institute of Medical GeneticsCarl von Ossietzky University OldenburgOldenburgGermany
| | - Friederike Cuello
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research CenterUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/LübeckUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - J. David Brook
- School of Life Sciences, Faculty of Medicine and Health SciencesUniversity of NottinghamNottinghamUK
| | - Siobhan Loughna
- School of Life Sciences, Faculty of Medicine and Health SciencesUniversity of NottinghamNottinghamUK
| |
Collapse
|
3
|
Sugawara Y, Mizuno Y, Oku S, Sawada Y, Goto T. Role of protein kinase D1 in vasoconstriction and haemodynamics in rats. Microvasc Res 2024; 152:104627. [PMID: 37963515 DOI: 10.1016/j.mvr.2023.104627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 10/31/2023] [Accepted: 11/06/2023] [Indexed: 11/16/2023]
Abstract
AIMS Protein kinase D (PKD), once considered an effector of protein kinase C (PKC), now plays many pathophysiological roles in various tissues. However, little is known about role of PKD in vascular function. We investigated the role of PKD in contraction of rat aorta and human aortic smooth muscle cells (HASMCs) and in haemodynamics in rats. METHODS AND RESULTS Isometric tension of rat aortic was measured to examine norepinephrine-induced contraction in the presence of PKD, PKC and Rho-kinase inhibitors. Phosphorylation of PKD1, myosin targeting subunit-1 (MYPT1), myosin light chain (MLC), CPI-17 and heat-shock protein 27 (HSP27), and actin polymerization were measured in the aorta. Phosphorylation of MYPT1 and MLC was also measured in HASMCs knocked down with specific siRNAs of PKD 1, 2 and 3. Intracellular calcium concentrations and cell shortening were measured in HASMCs. Norepinephrine-induced aortic contraction was accompanied by increased phosphorylation of PKD1, MYPT1 and MLC and actin polymerization, all of which were attenuated with PKD inhibitor CRT0066101. PKD1 phosphorylation was not inhibited by PKC inhibitor, chelerythrine or Rho kinase inhibitor, fasudil. In HASMCs, the phosphorylation of MYPT1 and MLC was attenuated by PKD1, but not PKD2, 3 knockdown. In HASMCs, CRT0066101 inhibited norepinephrine-induced cell shortening without affecting calcium concentration. Administration of CRT0066101 decreased systemic vascular resistance and blood pressure without affecting cardiac output in rats. CONCLUSIONS PKD1 may play roles in aorta contraction and haemodynamics via phosphorylation of MYPT1 and actin polymerization in a calcium-independent manner.
Collapse
Affiliation(s)
- Yoh Sugawara
- Department of Anaesthesiology and Critical Care Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yusuke Mizuno
- Department of Anaesthesiology and Critical Care Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Shinya Oku
- Department of Anaesthesiology and Critical Care Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yuri Sawada
- Department of Anaesthesiology and Critical Care Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takahisa Goto
- Department of Anaesthesiology and Critical Care Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
4
|
Connelly JA, Zhang X, Chen Y, Chao Y, Shi Y, Jacob TC, Wang QJ. Protein kinase D2 confers neuroprotection by promoting AKT and CREB activation in ischemic stroke. Neurobiol Dis 2023; 187:106305. [PMID: 37730136 PMCID: PMC10836334 DOI: 10.1016/j.nbd.2023.106305] [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: 06/18/2023] [Revised: 09/12/2023] [Accepted: 09/16/2023] [Indexed: 09/22/2023] Open
Abstract
Ischemic stroke, constituting 80-90% of all strokes, is a leading cause of death and long-term disability in adults. There is an urgent need to discover new targets and therapies for this devastating condition. Protein kinase D (PKD), as a key target of diacylglycerol involved in ischemic responses, has not been well studied in ischemic stroke, particularly PKD2. In this study, we found that PKD2 expression and activity were significantly upregulated in the ipsilateral side of the brain after transient focal cerebral ischemia, which coincides with the upregulation of PKD2 in primary neurons in response to in vitro ischemia, implying a potential role of PKD2 in neuronal survival in ischemic stroke. Using kinase-dead PKD2 knock-in (PKD2-KI) mice, we examined whether loss of PKD2 activity affected stroke outcomes in mice subjected to 1 h of transient middle cerebral artery occlusion (tMCAO) and 24 h of reperfusion. Our data demonstrated that PKD2-KI mice exhibited larger infarction volumes and worsened neurological scores, indicative of increased brain injury, as compared to the wild-type (WT) mice, confirming a neuroprotective role of PKD2 in ischemia/reperfusion (I/R) injury. Mouse primary neurons obtained from PKD2-KI mice also exhibited increased cell death as compared to the WT neurons when subjected to in vitro ischemia. We have further identified AKT and CREB as two main signaling nodes through which PKD2 regulates neuronal survival during I/R injury. In summary, PKD2 confers neuroprotection in ischemic stroke by promoting AKT and CREB activation and targeted activation of PKD2 may benefit neuronal survival in ischemic stroke.
Collapse
Affiliation(s)
- Jaclyn A Connelly
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, USA
| | - Xuejing Zhang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, USA
| | - Yuzhou Chen
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, USA
| | - Yapeng Chao
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, USA
| | - Yejie Shi
- Department of Neurology, University of Pittsburgh, Pittsburgh, USA
| | - Tija C Jacob
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, USA
| | - Q Jane Wang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, USA.
| |
Collapse
|
5
|
Miao LN, Pan D, Shi J, Du JP, Chen PF, Gao J, Yu Y, Shi DZ, Guo M. Role and Mechanism of PKC-δ for Cardiovascular Disease: Current Status and Perspective. Front Cardiovasc Med 2022; 9:816369. [PMID: 35242825 PMCID: PMC8885814 DOI: 10.3389/fcvm.2022.816369] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/11/2022] [Indexed: 12/18/2022] Open
Abstract
Protein kinase C (PKC) is a protein kinase with important cellular functions. PKC-δ, a member of the novel PKC subfamily, has been well-documented over the years. Activation of PKC-δ plays an important regulatory role in myocardial ischemia/reperfusion (IRI) injury and myocardial fibrosis, and its activity and expression levels can regulate pathological cardiovascular diseases such as atherosclerosis, hypertension, cardiac hypertrophy, and heart failure. This article aims to review the structure and function of PKC-δ, summarize the current research regarding its activation mechanism and its role in cardiovascular disease, and provide novel insight into further research on the role of PKC-δ in cardiovascular diseases.
Collapse
Affiliation(s)
- Li-na Miao
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Deng Pan
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Junhe Shi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jian-peng Du
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Peng-fei Chen
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Gao
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanqiao Yu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Da-Zhuo Shi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Da-Zhuo Shi
| | - Ming Guo
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- Ming Guo
| |
Collapse
|
6
|
Steinberg SF. Decoding the Cardiac Actions of Protein Kinase D Isoforms. Mol Pharmacol 2021; 100:558-567. [PMID: 34531296 DOI: 10.1124/molpharm.121.000341] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/07/2021] [Indexed: 11/22/2022] Open
Abstract
Protein kinase D (PKD) consists of a family of three structurally related enzymes that play key roles in a wide range of biological functions that contribute to the evolution of cardiac hypertrophy and heart failure. PKD1 (the founding member of this enzyme family) has been implicated in the phosphorylation of substrates that regulate cardiac hypertrophy, contraction, and susceptibility to ischemia/reperfusion injury, and de novo PRKD1 (protein kinase D1 gene) mutations have been identified in patients with syndromic congenital heart disease. However, cardiomyocytes coexpress all three PKDs. Although stimulus-specific activation patterns for PKD1, PKD2, and PKD3 have been identified in cardiomyocytes, progress toward identifying PKD isoform-specific functions in the heart have been hampered by significant gaps in our understanding of the molecular mechanisms that regulate PKD activity. This review incorporates recent conceptual breakthroughs in our understanding of various alternative mechanisms for PKD activation, with an emphasis on recent evidence that PKDs activate certain effector responses as dimers, to consider the role of PKD isoforms in signaling pathways that drive cardiac hypertrophy and ischemia/reperfusion injury. The focus is on whether the recently identified activation mechanisms that enhance the signaling repertoire of PKD family enzymes provide novel therapeutic strategies to target PKD enzymes and prevent or slow the evolution of cardiac injury and pathological cardiac remodeling. SIGNIFICANCE STATEMENT: PKD isoforms regulate a large number of fundamental biological processes, but the understanding of the biological actions of individual PKDs (based upon studies using adenoviral overexpression or gene-silencing methods) remains incomplete. This review focuses on dimerization, a recently identified mechanism for PKD activation, and the notion that this mechanism provides a strategy to develop novel PKD-targeted pharmaceuticals that restrict proliferation, invasion, or angiogenesis in cancer and prevent or slow the evolution of cardiac injury and pathological cardiac remodeling.
Collapse
|
7
|
Giardoglou P, Bournele D, Park M, Kanoni S, Dedoussis GV, Steinberg SF, Deloukas P, Beis D. A zebrafish forward genetic screen identifies an indispensable threonine residue in the kinase domain of PRKD2. Biol Open 2021; 10:bio.058542. [PMID: 33597201 PMCID: PMC7969590 DOI: 10.1242/bio.058542] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Protein kinase D2 belongs to a family of evolutionarily conserved enzymes regulating several biological processes. In a forward genetic screen for zebrafish cardiovascular mutants, we identified a mutation in the prkd2 gene. Homozygous mutant embryos develop as wild type up to 36 h post-fertilization and initiate blood flow, but fail to maintain it, resulting in a complete outflow tract stenosis. We identified a mutation in the prkd2 gene that results in a T757A substitution at a conserved residue in the kinase domain activation loop (T714A in human PRKD2) that disrupts catalytic activity and drives this phenotype. Homozygous mutants survive without circulation for several days, allowing us to study the extreme phenotype of no intracardiac flow, in the background of a functional heart. We show dysregulation of atrioventricular and outflow tract markers in the mutants and higher sensitivity to the Calcineurin inhibitor, Cyclosporin A. Finally we identify TBX5 as a potential regulator of PRKD2. Our results implicate PRKD2 catalytic activity in outflow tract development in zebrafish. This article has an associated First Person interview with the first author of the paper. Summary: We identified, through a zebrafish forward screen, an evolutionarily conserved residue in the catalytic domain of protein kinase D2 and its homologues.
Collapse
Affiliation(s)
- Panagiota Giardoglou
- Zebrafish Disease Model lab, Biomedical Research Foundation Academy of Athens, Athens 115 27, Greece.,Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, Athens 176 71, Greece
| | - Despina Bournele
- Zebrafish Disease Model lab, Biomedical Research Foundation Academy of Athens, Athens 115 27, Greece
| | - Misun Park
- Department of Pharmacology, Columbia University, New York 100 27, USA
| | - Stavroula Kanoni
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Clinical Pharmacology Centre, Queen Mary University of London, London, EC1M 6BQ, UK
| | - George V Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, Athens 176 71, Greece
| | - Susan F Steinberg
- Department of Pharmacology, Columbia University, New York 100 27, USA
| | - Panos Deloukas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Clinical Pharmacology Centre, Queen Mary University of London, London, EC1M 6BQ, UK.,Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah 222 52, Saudi Arabia
| | - Dimitris Beis
- Zebrafish Disease Model lab, Biomedical Research Foundation Academy of Athens, Athens 115 27, Greece
| |
Collapse
|
8
|
Renton MC, McGee SL, Howlett KF. The role of protein kinase D (PKD) in intracellular nutrient sensing and regulation of adaptive responses to the obese environment. Obes Rev 2021; 22:e13145. [PMID: 32929844 DOI: 10.1111/obr.13145] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/19/2020] [Accepted: 09/01/2020] [Indexed: 12/16/2022]
Abstract
Obesity is associated with ectopic accumulation of lipids, which is implicated in the development of insulin resistance, type 2 diabetes mellitus and cardiovascular disease. As the global prevalence of obesity continues to rise, it is becoming increasingly important to understand the underlying cellular mechanisms of this disease. Protein kinase D (PKD) is an intracellular signalling kinase with well characterized roles in intracellular vesicle transport and secretion, cancer cell proliferation and cardiac hypertrophy. However, emerging evidence also highlights PKD as a novel nutrient sensor. PKD activation is mediated by the accumulation of the lipid intermediate diacylglycerol, and PKD activity in the liver, heart and adipose tissue increases upon feeding. In obesity, PKD signalling is linked to reduced insulin signalling and dysfunction in adipose tissue, liver and heart, whilst in the pancreas, PKD is essential for the compensatory increase in glucose-stimulated insulin secretion from β-cells during obesity. Collectively, these studies reveal aspects of PKD signalling that are involved in the tissue-specific responses to obesity. This review summarizes the emerging evidence suggesting that PKD plays an important role in regulating the adaptive response to the obese environment.
Collapse
Affiliation(s)
- Mark C Renton
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Australia
| | - Sean L McGee
- Institute for Mental and Physical Health and Clinical Translation, Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Australia
| | - Kirsten F Howlett
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Australia
| |
Collapse
|
9
|
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
|
10
|
De Jong KA, Hall LG, Renton MC, Connor T, Martin SD, Kowalski GM, Shaw CS, Bruce CR, Howlett KF, McGee SL. Loss of protein kinase D activity demonstrates redundancy in cardiac glucose metabolism and preserves cardiac function in obesity. Mol Metab 2020; 42:101105. [PMID: 33099046 PMCID: PMC7680779 DOI: 10.1016/j.molmet.2020.101105] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/18/2020] [Accepted: 10/20/2020] [Indexed: 12/17/2022] Open
Abstract
Objective Protein kinase D (PKD) signaling has been implicated in stress-induced cardiac remodeling and function as well as metabolic processes including contraction-mediated cardiac glucose uptake. PKD has recently emerged as a nutrient-sensing kinase that is activated in high-lipid environments, such as in obesity. However, the role of PKD signaling in cardiac glucose metabolism and cardiac function in both normal and obese conditions remains unknown. Methods A cardiac-specific and inducible dominant negative (DN) PKD mouse model was developed. Echocardiography was used to assess cardiac function, while metabolic phenotyping was performed, including stable isotope metabolomics on cardiac tissue in mice fed either regular chow or a high-fat diet (43% calories from fat). Results Cardiac PKD activity declined by ∼90% following DN PKD induction in adult mice. The mice had diminished basal cardiac glucose clearance, suggesting impaired contraction-mediated glucose uptake, but normal cardiac function. In obesity studies, systolic function indices were reduced in control mice, but not in cardiac DN PKD mice. Using targeted stable isotope metabolomic analyses, no differences in glucose flux through glycolysis or the TCA cycle were observed between groups. Conclusions The data show that PKD contributes to cardiac dysfunction in obesity and highlight the redundancy in cardiac glucose metabolism that maintains cardiac glucose flux in vivo. The data suggest that impairments in contraction-mediated glucose uptake are unlikely to drive cardiac dysfunction in both normal and metabolic disease states. Cardiac protein kinase D (PKD) is required for contraction-mediated glucose uptake. PKD is not essential for normal cardiac function. Loss of PKD activity does not alter cardiac glucose flux in normal or obese mice. Loss of cardiac PKD activity preserves cardiac function in obesity.
Collapse
Affiliation(s)
- Kirstie A De Jong
- Institute for Mental and Physical Health and Clinical Translation, Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Australia; Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany
| | - Liam G Hall
- Institute for Mental and Physical Health and Clinical Translation, Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Australia
| | - Mark C Renton
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Australia
| | - Timothy Connor
- Institute for Mental and Physical Health and Clinical Translation, Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Australia
| | - Sheree D Martin
- Institute for Mental and Physical Health and Clinical Translation, Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Australia
| | - Greg M Kowalski
- Institute for Mental and Physical Health and Clinical Translation, Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Australia; Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Australia
| | - Christopher S Shaw
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Australia
| | - Clinton R Bruce
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Australia
| | - Kirsten F Howlett
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Australia
| | - Sean L McGee
- Institute for Mental and Physical Health and Clinical Translation, Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Australia.
| |
Collapse
|
11
|
Kolczynska K, Loza-Valdes A, Hawro I, Sumara G. Diacylglycerol-evoked activation of PKC and PKD isoforms in regulation of glucose and lipid metabolism: a review. Lipids Health Dis 2020; 19:113. [PMID: 32466765 PMCID: PMC7257441 DOI: 10.1186/s12944-020-01286-8] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/14/2020] [Indexed: 12/15/2022] Open
Abstract
Protein kinase C (PKC) and Protein kinase D (PKD) isoforms can sense diacylglycerol (DAG) generated in the different cellular compartments in various physiological processes. DAG accumulates in multiple organs of the obese subjects, which leads to the disruption of metabolic homeostasis and the development of diabetes as well as associated diseases. Multiple studies proved that aberrant activation of PKCs and PKDs contributes to the development of metabolic diseases. DAG-sensing PKC and PKD isoforms play a crucial role in the regulation of metabolic homeostasis and therefore might serve as targets for the treatment of metabolic disorders such as obesity and diabetes.
Collapse
Affiliation(s)
- Katarzyna Kolczynska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warszawa, Poland
| | - Angel Loza-Valdes
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warszawa, Poland
| | - Izabela Hawro
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warszawa, Poland
| | - Grzegorz Sumara
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warszawa, Poland.
| |
Collapse
|
12
|
Tóth AD, Schell R, Lévay M, Vettel C, Theis P, Haslinger C, Alban F, Werhahn S, Frischbier L, Krebs-Haupenthal J, Thomas D, Gröne HJ, Avkiran M, Katus HA, Wieland T, Backs J. Inflammation leads through PGE/EP 3 signaling to HDAC5/MEF2-dependent transcription in cardiac myocytes. EMBO Mol Med 2019; 10:emmm.201708536. [PMID: 29907596 PMCID: PMC6034133 DOI: 10.15252/emmm.201708536] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The myocyte enhancer factor 2 (MEF2) regulates transcription in cardiac myocytes and adverse remodeling of adult hearts. Activators of G protein-coupled receptors (GPCRs) have been reported to activate MEF2, but a comprehensive analysis of GPCR activators that regulate MEF2 has to our knowledge not been performed. Here, we tested several GPCR agonists regarding their ability to activate a MEF2 reporter in neonatal rat ventricular myocytes. The inflammatory mediator prostaglandin E2 (PGE2) strongly activated MEF2. Using pharmacological and protein-based inhibitors, we demonstrated that PGE2 regulates MEF2 via the EP3 receptor, the βγ subunit of Gi/o protein and two concomitantly activated downstream pathways. The first consists of Tiam1, Rac1, and its effector p21-activated kinase 2, the second of protein kinase D. Both pathways converge on and inactivate histone deacetylase 5 (HDAC5) and thereby de-repress MEF2. In vivo, endotoxemia in MEF2-reporter mice induced upregulation of PGE2 and MEF2 activation. Our findings provide an unexpected new link between inflammation and cardiac remodeling by de-repression of MEF2 through HDAC5 inactivation, which has potential implications for new strategies to treat inflammatory cardiomyopathies.
Collapse
Affiliation(s)
- András D Tóth
- Department of Molecular Cardiology and Epigenetics, Heidelberg University, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany.,Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Richard Schell
- Department of Molecular Cardiology and Epigenetics, Heidelberg University, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany.,Department of Cardiology, Heidelberg University, Heidelberg, Germany
| | - Magdolna Lévay
- DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany.,Experimental Pharmacology, European Center of Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christiane Vettel
- DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany.,Experimental Pharmacology, European Center of Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Philipp Theis
- Department of Molecular Cardiology and Epigenetics, Heidelberg University, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany
| | - Clemens Haslinger
- Department of Molecular Cardiology and Epigenetics, Heidelberg University, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany
| | - Felix Alban
- Department of Molecular Cardiology and Epigenetics, Heidelberg University, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany
| | - Stefanie Werhahn
- Department of Molecular Cardiology and Epigenetics, Heidelberg University, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany
| | - Lina Frischbier
- Department of Molecular Cardiology and Epigenetics, Heidelberg University, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany
| | - Jutta Krebs-Haupenthal
- Department of Molecular Cardiology and Epigenetics, Heidelberg University, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany
| | - Dominique Thomas
- Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt, Germany
| | - Hermann-Josef Gröne
- Department of Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany
| | - Metin Avkiran
- Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, The Rayne Institute, St Thomas' Hospital, London, UK
| | - Hugo A Katus
- Department of Cardiology, Heidelberg University, Heidelberg, Germany
| | - Thomas Wieland
- DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany.,Experimental Pharmacology, European Center of Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Johannes Backs
- Department of Molecular Cardiology and Epigenetics, Heidelberg University, Heidelberg, Germany .,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany
| |
Collapse
|
13
|
Roy A, Veroli MV, Prasad S, Wang QJ. Protein Kinase D2 Modulates Cell Cycle By Stabilizing Aurora A Kinase at Centrosomes. Mol Cancer Res 2018; 16:1785-1797. [PMID: 30018032 PMCID: PMC9923726 DOI: 10.1158/1541-7786.mcr-18-0641] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 06/18/2018] [Accepted: 07/10/2018] [Indexed: 11/16/2022]
Abstract
Aurora A kinase (AURKA) is a master cell-cycle regulator that is often dysregulated in human cancers. Its overexpression has been associated with genome instability and oncogenic transformation. The protein kinase D (PKD) family is an emerging therapeutic target of cancer. Aberrant PKD activation has been implicated in tumor growth and survival, yet the underlying mechanisms remain to be elucidated. This study identified, for the first time, a functional crosstalk between PKD2 and Aurora A kinase in cancer cells. The data demonstrate that PKD2 is catalytically active during the G2-M phases of the cell cycle, and inactivation or depletion of PKD2 causes delay in mitotic entry due to downregulation of Aurora A, an effect that can be rescued by overexpression of Aurora A. Moreover, PKD2 localizes in the centrosome with Aurora A by binding to γ-tubulin. Knockdown of PKD2 caused defects in centrosome separation, elongated G2 phase, mitotic catastrophe, and eventually cell death via apoptosis. Mechanistically, PKD2 interferes with Fbxw7 function to protect Aurora A from ubiquitin- and proteasome-dependent degradation. Taken together, these results identify PKD as a cell-cycle checkpoint kinase that positively modulates G2-M transition through Aurora A kinase in mammalian cells.Implications: PKD2 is a novel cell-cycle regulator that promotes G2-M transition by modulating Aurora A kinase stability in cancer cells and suggests the PKD2/Aurora A kinase regulatory axis as new therapeutic targets for cancer treatment. Mol Cancer Res; 16(11); 1785-97. ©2018 AACR.
Collapse
Affiliation(s)
- Adhiraj Roy
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA
| | - Maria Victoria Veroli
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA
| | - Sahdeo Prasad
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA.,Department of Biotechnology and Immunotherapeutics, Texas Tech University, Amarillo, TX 79106
| | - Qiming Jane Wang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
| |
Collapse
|
14
|
Simsek Papur O, Sun A, Glatz JFC, Luiken JJFP, Nabben M. Acute and Chronic Effects of Protein Kinase-D Signaling on Cardiac Energy Metabolism. Front Cardiovasc Med 2018; 5:65. [PMID: 29930945 PMCID: PMC5999788 DOI: 10.3389/fcvm.2018.00065] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 05/17/2018] [Indexed: 02/05/2023] Open
Abstract
Protein kinase-D (PKD) is increasingly recognized as a key regulatory signaling hub in cardiac glucose uptake and also a major player in the development of hypertrophy. Glucose is one of the predominant energy substrates for the heart to support contraction. However, a cardiac substrate switch toward glucose over-usage is associated with the development of cardiac hypertrophy. Hence, regulation of PKD activity must be strictly coordinated. This review provides mechanistic insights into the acute and chronic regulatory functions of PKD signaling in the healthy and hypertrophied heart. First an overview of the activation pathways of PKD1, the most abundant isoform in the heart, is provided. Then the various regulatory roles of the PKD isoforms in the heart in relation to cardiac glucose and fatty acid metabolism, contraction, morphology, function, and the development of cardiac hypertrophy are described. Finally, these findings are integrated and the possibility of targeting this kinase as a novel strategy to combat cardiac diseases is discussed.
Collapse
Affiliation(s)
- Ozlenen Simsek Papur
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands.,Department of Molecular Medicine, Institute of Health Science, Dokuz Eylul University, Izmir, Turkey
| | - Aomin Sun
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Jan F C Glatz
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Joost J F P Luiken
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Miranda Nabben
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
15
|
Jhun BS, O‐Uchi J, Adaniya SM, Mancini TJ, Cao JL, King ME, Landi AK, Ma H, Shin M, Yang D, Xu X, Yoon Y, Choudhary G, Clements RT, Mende U, Sheu S. Protein kinase D activation induces mitochondrial fragmentation and dysfunction in cardiomyocytes. J Physiol 2018; 596:827-855. [PMID: 29313986 PMCID: PMC5830422 DOI: 10.1113/jp275418] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 01/02/2018] [Indexed: 01/06/2023] Open
Abstract
KEY POINTS Abnormal mitochondrial morphology and function in cardiomyocytes are frequently observed under persistent Gq protein-coupled receptor (Gq PCR) stimulation. Cardiac signalling mechanisms for regulating mitochondrial morphology and function under pathophysiological conditions in the heart are still poorly understood. We demonstrate that a downstream kinase of Gq PCR, protein kinase D (PKD) induces mitochondrial fragmentation via phosphorylation of dynamin-like protein 1 (DLP1), a mitochondrial fission protein. The fragmented mitochondria enhance reactive oxygen species generation and permeability transition pore opening in mitochondria, which initiate apoptotic signalling activation. This study identifies a novel PKD-specific substrate in cardiac mitochondria and uncovers the role of PKD on cardiac mitochondria, with special emphasis on the molecular mechanism(s) underlying mitochondrial injury with abnormal mitochondrial morphology under persistent Gq PCR stimulation. These findings provide new insights into the molecular basis of cardiac mitochondrial physiology and pathophysiology, linking Gq PCR signalling with the regulation of mitochondrial morphology and function. ABSTRACT Regulation of mitochondrial morphology is crucial for the maintenance of physiological functions in many cell types including cardiomyocytes. Small and fragmented mitochondria are frequently observed in pathological conditions, but it is still unclear which cardiac signalling pathway is responsible for regulating the abnormal mitochondrial morphology in cardiomyocytes. Here we demonstrate that a downstream kinase of Gq protein-coupled receptor (Gq PCR) signalling, protein kinase D (PKD), mediates pathophysiological modifications in mitochondrial morphology and function, which consequently contribute to the activation of apoptotic signalling. We show that Gq PCR stimulation induced by α1 -adrenergic stimulation mediates mitochondrial fragmentation in a fission- and PKD-dependent manner in H9c2 cardiac myoblasts and rat neonatal cardiomyocytes. Upon Gq PCR stimulation, PKD translocates from the cytoplasm to the outer mitochondrial membrane (OMM) and phosphorylates a mitochondrial fission protein, dynamin-like protein 1 (DLP1), at S637. PKD-dependent phosphorylation of DLP1 initiates DLP1 association with the OMM, which then enhances mitochondrial fragmentation, mitochondrial superoxide generation, mitochondrial permeability transition pore opening and apoptotic signalling. Finally, we demonstrate that DLP1 phosphorylation at S637 by PKD occurs in vivo using ventricular tissues from transgenic mice with cardiac-specific overexpression of constitutively active Gαq protein. In conclusion, Gq PCR-PKD signalling induces mitochondrial fragmentation and dysfunction via PKD-dependent DLP1 phosphorylation in cardiomyocytes. This study is the first to identify a novel PKD-specific substrate, DLP1 in mitochondria, as well as the functional role of PKD in cardiac mitochondria. Elucidation of these molecular mechanisms by which PKD-dependent enhanced fission mediates cardiac mitochondrial injury will provide novel insight into the relationship among mitochondrial form, function and Gq PCR signalling.
Collapse
Affiliation(s)
- Bong Sook Jhun
- Cardiovascular Research CenterRhode Island HospitalProvidenceRIUSA
- Department of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRIUSA
| | - Jin O‐Uchi
- Cardiovascular Research CenterRhode Island HospitalProvidenceRIUSA
- Department of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRIUSA
| | - Stephanie M. Adaniya
- Cardiovascular Research CenterRhode Island HospitalProvidenceRIUSA
- Department of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRIUSA
| | - Thomas J. Mancini
- Vascular Research LaboratoryProvidence VA Medical CenterProvidenceRIUSA
| | - Jessica L. Cao
- Cardiovascular Research CenterRhode Island HospitalProvidenceRIUSA
- Department of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRIUSA
| | - Michelle E. King
- Cardiovascular Research CenterRhode Island HospitalProvidenceRIUSA
- Department of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRIUSA
| | - Amy K. Landi
- Cardiovascular Research CenterRhode Island HospitalProvidenceRIUSA
| | - Hanley Ma
- Cardiovascular Research CenterRhode Island HospitalProvidenceRIUSA
- Department of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRIUSA
| | - Milla Shin
- Cardiovascular Research CenterRhode Island HospitalProvidenceRIUSA
- Department of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRIUSA
| | - Donqin Yang
- Cardiovascular Research CenterRhode Island HospitalProvidenceRIUSA
- Department of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRIUSA
| | - Xiaole Xu
- Center for Translational Medicine, Department of MedicineThomas Jefferson UniversityPhiladelphiaPAUSA
| | - Yisang Yoon
- Department of Physiology, Medical College of GeorgiaAugusta UniversityAugustaGAUSA
| | - Gaurav Choudhary
- Department of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRIUSA
- Vascular Research LaboratoryProvidence VA Medical CenterProvidenceRIUSA
| | - Richard T. Clements
- Vascular Research LaboratoryProvidence VA Medical CenterProvidenceRIUSA
- Department of SurgeryRhode Island Hospital and Warren Alpert School of Brown UniversityProvidenceRIUSA
| | - Ulrike Mende
- Cardiovascular Research CenterRhode Island HospitalProvidenceRIUSA
- Department of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRIUSA
| | - Shey‐Shing Sheu
- Center for Translational Medicine, Department of MedicineThomas Jefferson UniversityPhiladelphiaPAUSA
| |
Collapse
|
16
|
Protein kinase D-dependent CXCR4 down-regulation upon BCR triggering is linked to lymphadenopathy in chronic lymphocytic leukaemia. Oncotarget 2018; 7:41031-41046. [PMID: 27127886 PMCID: PMC5173040 DOI: 10.18632/oncotarget.9031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 04/16/2016] [Indexed: 02/07/2023] Open
Abstract
In Chronic Lymphocytic Leukemia (CLL), infiltration of lymph nodes by leukemic cells is observed in patients with progressive disease and adverse outcome. We have previously demonstrated that B-cell receptor (BCR) engagement resulted in CXCR4 down-regulation in CLL cells, correlating with a shorter progression-free survival in patients. In this study, we show a simultaneous down-regulation of CXCR4, CXCR5 and CD62L upon BCR triggering. While concomitant CXCR4 and CXCR5 down-regulation involves PKDs, CD62L release relies on PKC activation. BCR engagement induces PI3K-δ-dependent phosphorylation of PKD2 and 3, which in turn phosphorylate CXCR4 Ser324/325. Moreover, upon BCR triggering, PKD phosphorylation levels correlate with the extent of membrane CXCR4 decrease. Inhibition of PKD activity restores membrane expression of CXCR4 and migration towards CXCL12 in BCR-responsive cells in vitro. In terms of pathophysiology, BCR-dependent CXCR4 down-regulation is observed in leukemic cells from patients with enlarged lymph nodes, irrespective of their IGHV mutational status. Taken together, our results demonstrate that PKD-mediated CXCR4 internalization induced by BCR engagement in B-CLL is associated with lymph node enlargement and suggest PKD as a potential druggable target for CLL therapeutics.
Collapse
|
17
|
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
|
18
|
Roy A, Ye J, Deng F, Wang QJ. Protein kinase D signaling in cancer: A friend or foe? Biochim Biophys Acta Rev Cancer 2017; 1868:283-294. [PMID: 28577984 DOI: 10.1016/j.bbcan.2017.05.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 05/26/2017] [Accepted: 05/27/2017] [Indexed: 12/18/2022]
Abstract
Protein kinase D is a family of evolutionarily conserved serine/threonine kinases that belongs to the Ca++/Calmodulin-dependent kinase superfamily. Signal transduction pathways mediated by PKD can be triggered by a variety of stimuli including G protein-coupled receptor agonists, growth factors, hormones, and cellular stresses. The regulatory mechanisms and physiological roles of PKD have been well documented including cell proliferation, survival, migration, angiogenesis, regulation of gene expression, and protein/membrane trafficking. However, its precise roles in disease progression, especially in cancer, remain elusive. A plethora of studies documented the cell- and tissue-specific expressions and functions of PKD in various cancer-associated biological processes, while the causes of the differential effects of PKD have not been thoroughly investigated. In this review, we have discussed the structural-functional properties, activation mechanisms, signaling pathways and physiological functions of PKD in the context of human cancer. Additionally, we have provided a comprehensive review of the reported tumor promoting or tumor suppressive functions of PKD in several major cancer types and discussed the discrepancies that have been raised on PKD as a major regulator of malignant transformation.
Collapse
Affiliation(s)
- Adhiraj Roy
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA
| | - Jing Ye
- Department of Anesthesiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Fan Deng
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Qiming Jane Wang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA.
| |
Collapse
|
19
|
Cobbaut M, Derua R, Döppler H, Lou HJ, Vandoninck S, Storz P, Turk BE, Seufferlein T, Waelkens E, Janssens V, Van Lint J. Differential regulation of PKD isoforms in oxidative stress conditions through phosphorylation of a conserved Tyr in the P+1 loop. Sci Rep 2017; 7:887. [PMID: 28428613 PMCID: PMC5430542 DOI: 10.1038/s41598-017-00800-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/13/2017] [Indexed: 01/06/2023] Open
Abstract
Protein kinases are essential molecules in life and their crucial function requires tight regulation. Many kinases are regulated via phosphorylation within their activation loop. This loop is embedded in the activation segment, which additionally contains the Mg2+ binding loop and a P + 1 loop that is important in substrate binding. In this report, we identify Abl-mediated phosphorylation of a highly conserved Tyr residue in the P + 1 loop of protein kinase D2 (PKD2) during oxidative stress. Remarkably, we observed that the three human PKD isoforms display very different degrees of P + 1 loop Tyr phosphorylation and we identify one of the molecular determinants for this divergence. This is paralleled by a different activation mechanism of PKD1 and PKD2 during oxidative stress. Tyr phosphorylation in the P + 1 loop of PKD2 increases turnover for Syntide-2, while substrate specificity and the role of PKD2 in NF-κB signaling remain unaffected. Importantly, Tyr to Phe substitution renders the kinase inactive, jeopardizing its use as a non-phosphorylatable mutant. Since large-scale proteomics studies identified P + 1 loop Tyr phosphorylation in more than 70 Ser/Thr kinases in multiple conditions, our results do not only demonstrate differential regulation/function of PKD isoforms under oxidative stress, but also have implications for kinase regulation in general.
Collapse
Affiliation(s)
- Mathias Cobbaut
- Department of Cellular and Molecular Medicine, Faculty of Medicine, KU Leuven, Leuven, Belgium.,Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
| | - Rita Derua
- Department of Cellular and Molecular Medicine, Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Heike Döppler
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Hua Jane Lou
- Department of Pharmacology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Sandy Vandoninck
- Department of Cellular and Molecular Medicine, Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Peter Storz
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Benjamin E Turk
- Department of Pharmacology, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Etienne Waelkens
- Department of Cellular and Molecular Medicine, Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Veerle Janssens
- Department of Cellular and Molecular Medicine, Faculty of Medicine, KU Leuven, Leuven, Belgium.,Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
| | - Johan Van Lint
- Department of Cellular and Molecular Medicine, Faculty of Medicine, KU Leuven, Leuven, Belgium. .,Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium.
| |
Collapse
|
20
|
Wood BM, Bossuyt J. Emergency Spatiotemporal Shift: The Response of Protein Kinase D to Stress Signals in the Cardiovascular System. Front Pharmacol 2017; 8:9. [PMID: 28174535 PMCID: PMC5258689 DOI: 10.3389/fphar.2017.00009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 01/04/2017] [Indexed: 12/12/2022] Open
Abstract
Protein Kinase D isoforms (PKD 1-3) are key mediators of neurohormonal, oxidative, and metabolic stress signals. PKDs impact a wide variety of signaling pathways and cellular functions including actin dynamics, vesicle trafficking, cell motility, survival, contractility, energy substrate utilization, and gene transcription. PKD activity is also increasingly linked to cancer, immune regulation, pain modulation, memory, angiogenesis, and cardiovascular disease. This increasing complexity and diversity of PKD function, highlights the importance of tight spatiotemporal control of the kinase via protein–protein interactions, post-translational modifications or targeting via scaffolding proteins. In this review, we focus on the spatiotemporal regulation and effects of PKD signaling in response to neurohormonal, oxidant and metabolic signals that have implications for myocardial disease. Precise targeting of these mechanisms will be crucial in the design of PKD-based therapeutic strategies.
Collapse
Affiliation(s)
- Brent M Wood
- Department of Pharmacology, University of California, Davis, Davis CA, USA
| | - Julie Bossuyt
- Department of Pharmacology, University of California, Davis, Davis CA, USA
| |
Collapse
|
21
|
Haque ZK, Wang DZ. How cardiomyocytes sense pathophysiological stresses for cardiac remodeling. Cell Mol Life Sci 2016; 74:983-1000. [PMID: 27714411 DOI: 10.1007/s00018-016-2373-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 09/01/2016] [Accepted: 09/19/2016] [Indexed: 12/14/2022]
Abstract
In the past decades, the cardiovascular community has laid out the fundamental signaling cascades that become awry in the cardiomyocyte during the process of pathologic cardiac remodeling. These pathways are initiated at the cell membrane and work their way to the nucleus to mediate gene expression. Complexity is multiplied as the cardiomyocyte is subjected to cross talk with other cells as well as a barrage of extracellular stimuli and mechanical stresses. In this review, we summarize the signaling cascades that play key roles in cardiac function and then we proceed to describe emerging concepts of how the cardiomyocyte senses the mechanical and environmental stimuli to transition to the deleterious genetic program that defines pathologic cardiac remodeling. As a highlighting example of these processes, we illustrate the transition from a compensated hypertrophied myocardium to a decompensated failing myocardium, which is clinically manifested as decompensated heart failure.
Collapse
Affiliation(s)
- Zaffar K Haque
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 1260 John F. Enders Research Bldg, 320 Longwood Ave, Boston, MA, 02115, USA.
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 1260 John F. Enders Research Bldg, 320 Longwood Ave, Boston, MA, 02115, USA
| |
Collapse
|
22
|
Qiu W, Steinberg SF. Phos-tag SDS-PAGE resolves agonist- and isoform-specific activation patterns for PKD2 and PKD3 in cardiomyocytes and cardiac fibroblasts. J Mol Cell Cardiol 2016; 99:14-22. [PMID: 27515283 DOI: 10.1016/j.yjmcc.2016.08.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/05/2016] [Accepted: 08/06/2016] [Indexed: 01/16/2023]
Abstract
Protein kinase D (PKD) consists of a family of three structurally related enzymes that are co-expressed in the heart and have important roles in many biological responses. PKD1 is activated by pro-hypertrophic stimuli and has been implicated in adverse cardiac remodeling. Efforts to define the cardiac actions of PKD2 and PKD3 have been less successful at least in part because conventional methods provide a general screen for PKD activation but are poorly suited to resolve activation patterns for PKD2 or PKD3. This study uses Phos-tag SDS-PAGE, a method that exaggerates phosphorylation-dependent mobility shifts, to overcome this technical limitation. Phos-tag SDS-PAGE resolves PKD1 as distinct molecular species (indicative of pools of enzyme with distinct phosphorylation profiles) in unstimulated cardiac fibroblasts and cardiomyocytes; as a result, attempts to track PKD1 mobility shifts that result from agonist activation were only moderately successful. In contrast, PKD2 and PKD3 are recovered from resting cardiac fibroblasts and cardiomyocytes as single molecular species; both enzymes display robust mobility shifts in Phos-tag SDS-PAGE in response to treatment with sphingosine-1-phosphate, thrombin, PDGF, or H2O2. Studies with GF109203X implicate protein kinase C activity in the stimulus-dependent pathways that activate PKD2/PKD3 in both cardiac fibroblasts and cardiomyocytes. Studies with C3 toxin identify a novel role for Rho in the sphingosine-1-phosphate and thrombin receptor-dependent pathways that lead to the phosphorylation of PKD2/3 and the downstream substrate CREB in cardiomyocytes. In conclusion, Phos-tag SDS-PAGE provides a general screen for stimulus-specific changes in PKD2 and PKD3 phosphorylation and exposes a novel role for these enzymes in specific stress-dependent pathways that influence cardiac remodeling.
Collapse
Affiliation(s)
- Weihua Qiu
- Department of Pharmacology, Columbia University, New York, NY 10032, United States
| | - Susan F Steinberg
- Department of Pharmacology, Columbia University, New York, NY 10032, United States.
| |
Collapse
|
23
|
Scaffold protein enigma homolog activates CREB whereas a short splice variant prevents CREB activation in cardiomyocytes. Cell Signal 2015; 27:2425-33. [PMID: 26365342 DOI: 10.1016/j.cellsig.2015.09.007] [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: 09/03/2015] [Accepted: 09/09/2015] [Indexed: 01/27/2023]
Abstract
Enigma Homolog (ENH1 or Pdlim5) is a scaffold protein composed of an N-terminal PDZ domain and three LIM domains at the C-terminal end. The enh gene encodes for several splice variants with opposing functions. ENH1 promotes cardiomyocytes hypertrophy whereas ENH splice variants lacking LIM domains prevent it. ENH1 interacts with various Protein Kinase C (PKC) isozymes and Protein Kinase D1 (PKD1). In addition, the binding of ENH1's LIM domains to PKC is sufficient to activate the kinase without stimulation. The downstream events of the ENH1-PKC/PKD1 complex remain unknown. PKC and PKD1 are known to phosphorylate the transcription factor cAMP-response element binding protein (CREB). We tested whether ENH1 could play a role in the activation of CREB. We found that, in neonatal rat ventricular cardiomyocytes, ENH1 interacts with CREB, is necessary for the phosphorylation of CREB at ser133, and the activation of CREB-dependent transcription. On the contrary, the overexpression of ENH3, a LIM-less splice variant, inhibited the phosphorylation of CREB. ENH3 overexpression or shRNA knockdown of ENH1 prevented the CREB-dependent transcription. Our results thus suggest that ENH1 plays an essential role in CREB's activation and dependent transcription in cardiomyocytes. At the opposite, ENH3 prevents the CREB transcriptional activity. In conclusion, these results provide a first molecular explanation to the opposing functions of ENH splice variants.
Collapse
|
24
|
Chien PTY, Lin CC, Hsiao LD, Yang CM. c-Src/Pyk2/EGFR/PI3K/Akt/CREB-activated pathway contributes to human cardiomyocyte hypertrophy: Role of COX-2 induction. Mol Cell Endocrinol 2015; 409:59-72. [PMID: 25869400 DOI: 10.1016/j.mce.2015.04.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 03/23/2015] [Accepted: 04/07/2015] [Indexed: 12/27/2022]
Abstract
Thrombin and COX-2 regulating cardiac hypertrophy are via various signaling cascades. Several transcriptional factors including CREB involve in COX-2 expression. However, the interplay among thrombin, CREB, and COX-2 in primary human neonatal ventricular cardiomyocytes remains unclear. In this study, thrombin-induced COX-2 promoter activity, mRNA and protein expression, and PGE2 synthesis were attenuated by pretreatment with the inhibitors of c-Src (PP1), Pyk2 (PF431396), EGFR (AG1478), PI3K/Akt (LY294002/SH-5), and p300 (GR343), or transfection with siRNAs of c-Src, Pyk2, EGFR, p110, Akt, CREB, and p300. Moreover, thrombin-stimulated phosphorylation of c-Src, Pyk2, EGFR, Akt, CREB and p300 was attenuated by their respective inhibitors. These results indicate that thrombin-induced COX-2 expression is mediated through PAR-1/c-Src/Pyk2/EGFR/PI3K/Akt linking to CREB and p300 cascades. Functionally, thrombin-induced hypertrophy and ANF/BNP release were, at least in part, mediated through a PAR-1/COX-2-dependent pathway. We uncover the importance of COX-2 regarding human cardiomyocyte hypertrophy that will provide a therapeutic intervention in cardiovascular diseases.
Collapse
Affiliation(s)
- Peter Tzu-Yu Chien
- Graduate Institute of Biomedical Sciences, Health Ageing Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan; Department of Physiology and Pharmacology, Health Ageing Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Chih-Chung Lin
- Department of Anesthetics, Chang Gung Memorial Hospital at Lin-Kou and College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Li-Der Hsiao
- Department of Anesthetics, Chang Gung Memorial Hospital at Lin-Kou and College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Chuen-Mao Yang
- Graduate Institute of Biomedical Sciences, Health Ageing Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan; Department of Physiology and Pharmacology, Health Ageing Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan.
| |
Collapse
|
25
|
Venardos K, De Jong KA, Elkamie M, Connor T, McGee SL. The PKD inhibitor CID755673 enhances cardiac function in diabetic db/db mice. PLoS One 2015; 10:e0120934. [PMID: 25798941 PMCID: PMC4370864 DOI: 10.1371/journal.pone.0120934] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 02/09/2015] [Indexed: 01/06/2023] Open
Abstract
The development of diabetic cardiomyopathy is a key contributor to heart failure and mortality in obesity and type 2 diabetes (T2D). Current therapeutic interventions for T2D have limited impact on the development of diabetic cardiomyopathy. Clearly, new therapies are urgently needed. A potential therapeutic target is protein kinase D (PKD), which is activated by metabolic insults and implicated in the regulation of cardiac metabolism, contractility and hypertrophy. We therefore hypothesised that PKD inhibition would enhance cardiac function in T2D mice. We first validated the obese and T2D db/db mouse as a model of early stage diabetic cardiomyopathy, which was characterised by both diastolic and systolic dysfunction, without overt alterations in left ventricular morphology. These functional characteristics were also associated with increased PKD2 phosphorylation in the fed state and a gene expression signature characteristic of PKD activation. Acute administration of the PKD inhibitor CID755673 to normal mice reduced both PKD1 and 2 phosphorylation in a time and dose-dependent manner. Chronic CID755673 administration to T2D db/db mice for two weeks reduced expression of the gene expression signature of PKD activation, enhanced indices of both diastolic and systolic left ventricular function and was associated with reduced heart weight. These alterations in cardiac function were independent of changes in glucose homeostasis, insulin action and body composition. These findings suggest that PKD inhibition could be an effective strategy to enhance heart function in obese and diabetic patients and provide an impetus for further mechanistic investigations into the role of PKD in diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Kylie Venardos
- Metabolic Remodelling Laboratory, Metabolic Research Unit, School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Kirstie A. De Jong
- Metabolic Remodelling Laboratory, Metabolic Research Unit, School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Mansour Elkamie
- Metabolic Remodelling Laboratory, Metabolic Research Unit, School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Timothy Connor
- Metabolic Remodelling Laboratory, Metabolic Research Unit, School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Sean L. McGee
- Metabolic Remodelling Laboratory, Metabolic Research Unit, School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
- Program for Metabolism and Inflammation, Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
- * E-mail:
| |
Collapse
|
26
|
Nichols CB, Chang CW, Ferrero M, Wood BM, Stein ML, Ferguson AJ, Ha D, Rigor RR, Bossuyt S, Bossuyt J. β-adrenergic signaling inhibits Gq-dependent protein kinase D activation by preventing protein kinase D translocation. Circ Res 2014; 114:1398-409. [PMID: 24643961 PMCID: PMC4031034 DOI: 10.1161/circresaha.114.303870] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 03/18/2014] [Indexed: 12/16/2022]
Abstract
RATIONALE Both β-adrenergic receptor (β-AR) and Gq-coupled receptor (GqR) agonist-driven signaling play key roles in the events, leading up to and during cardiac dysfunction. How these stimuli interact at the level of protein kinase D (PKD), a nodal point in cardiac hypertrophic signaling, remains unclear. OBJECTIVE To assess the spatiotemporal dynamics of PKD activation in response to β-AR signaling alone and on coactivation with GqR-agonists. This will test our hypothesis that compartmentalized PKD signaling reconciles disparate findings of PKA facilitation and inhibition of PKD activation. METHODS AND RESULTS We report on the spatial and temporal profiles of PKD activation using green fluorescent protein-tagged PKD (wildtype or mutant S427E) and targeted fluorescence resonance energy transfer-based biosensors (D-kinase activity reporters) in adult cardiomyocytes. We find that β-AR/PKA signaling drives local nuclear activation of PKD, without preceding sarcolemmal translocation. We also discover pronounced interference of β-AR/cAMP/PKA signaling on GqR-induced translocation and activation of PKD throughout the cardiomyocyte. We attribute these effects to direct, PKA-dependent phosphorylation of PKD-S427. We also show that phosphomimetic substitution of S427 likewise impedes GqR-induced PKD translocation and activation. In neonatal myocytes, S427E inhibits GqR-evoked cell growth and expression of hypertrophic markers. Finally, we show altered S427 phosphorylation in transverse aortic constriction-induced hypertrophy. CONCLUSIONS β-AR signaling triggers local nuclear signaling and inhibits GqR-mediated PKD activation by preventing its intracellular translocation. PKA-dependent phosphorylation of PKD-S427 fine-tunes the PKD responsiveness to GqR-agonists, serving as a key integration point for β-adrenergic and Gq-coupled stimuli.
Collapse
MESH Headings
- Adrenergic beta-Agonists/pharmacology
- Animals
- Cardiomegaly/enzymology
- Cardiomegaly/pathology
- Cells, Cultured
- Cyclic AMP/metabolism
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Disease Models, Animal
- Enzyme Activation
- Fluorescence Resonance Energy Transfer
- GTP-Binding Protein alpha Subunits, Gq-G11/metabolism
- Genes, Reporter
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mutation
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Phosphorylation
- Protein Kinase C/genetics
- Protein Kinase C/metabolism
- Protein Transport
- Rabbits
- Rats
- Receptors, Adrenergic, beta/drug effects
- Receptors, Adrenergic, beta/metabolism
- Recombinant Fusion Proteins/metabolism
- Signal Transduction/drug effects
- Time Factors
- Transfection
Collapse
Affiliation(s)
| | - Chia-Wei Chang
- Department of Pharmacology, University of California, Davis, CA
- Department of Physiology, Loyola University Chicago, Maywood, IL
| | - Maura Ferrero
- Department of Pharmacology, University of California, Davis, CA
| | | | | | | | - Derrick Ha
- Department of Pharmacology, University of California, Davis, CA
| | - Robert R. Rigor
- Department of Pharmacology, University of California, Davis, CA
| | - Sven Bossuyt
- Aalto University School of Science and Technology, Helsinki, Finland
| | - Julie Bossuyt
- Department of Pharmacology, University of California, Davis, CA
| |
Collapse
|
27
|
Abstract
Oxidative stress accompanies a wide spectrum of clinically important cardiac disorders, including ischemia/reperfusion, diabetes mellitus, and hypertensive heart disease. Although reactive oxygen species (ROS) can activate signaling pathways that contribute to ischemic preconditioning and cardioprotection, high levels of ROS induce structural modifications of the sarcomere that impact on pump function and the pathogenesis of heart failure. However, the precise nature of the redox-dependent change in contractility is determined by the source/identity of the oxidant species, the level of oxidative stress, and the chemistry/position of oxidant-induced posttranslational modifications on individual proteins within the sarcomere. This review focuses on various ROS-induced posttranslational modifications of myofilament proteins (including direct oxidative modifications of myofilament proteins, myofilament protein phosphorylation by ROS-activated signaling enzymes, and myofilament protein cleavage by ROS-activated proteases) that have been implicated in the control of cardiac contractility.
Collapse
Affiliation(s)
- Susan F Steinberg
- Department of Pharmacology, College of Physicians and Surgeons, Columbia University, 630 W. 168 St, New York, NY 10032, USA.
| |
Collapse
|
28
|
Chiou YS, Sang S, Cheng KH, Ho CT, Wang YJ, Pan MH. Peracetylated (−)-epigallocatechin-3-gallate (AcEGCG) potently prevents skin carcinogenesis by suppressing the PKD1-dependent signaling pathway in CD34 + skin stem cells and skin tumors. Carcinogenesis 2013; 34:1315-22. [DOI: 10.1093/carcin/bgt042] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
|
29
|
Lynch CM, Leandry LA, Matheny RW. Lysophosphatidic acid-stimulated phosphorylation of PKD2 is mediated by PI3K p110β and PKCδ in myoblasts. J Recept Signal Transduct Res 2012; 33:41-8. [DOI: 10.3109/10799893.2012.752005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
30
|
Abstract
Protein kinase C (PKC) isoforms have emerged as important regulators of cardiac contraction, hypertrophy, and signaling pathways that influence ischemic/reperfusion injury. This review focuses on newer concepts regarding PKC isoform-specific activation mechanisms and actions that have implications for the development of PKC-targeted therapeutics.
Collapse
Affiliation(s)
- Susan F Steinberg
- Department of Pharmacology, Columbia University, New York, New York, USA.
| |
Collapse
|
31
|
Rybin VO, Guo J, Harleton E, Zhang F, Steinberg SF. Regulatory domain determinants that control PKD1 activity. J Biol Chem 2012; 287:22609-15. [PMID: 22582392 DOI: 10.1074/jbc.m112.379719] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The canonical pathway for protein kinase D1 (PKD1) activation by growth factor receptors involves diacylglycerol binding to the C1 domain and protein kinase C-dependent phosphorylation at the activation loop. PKD1 then autophosphorylates at Ser(916), a modification frequently used as a surrogate marker of PKD1 activity. PKD1 also is cleaved by caspase-3 at a site in the C1-PH interdomain during apoptosis; the functional consequences of this cleavage event remain uncertain. This study shows that PKD1-Δ1-321 (an N-terminal deletion mutant lacking the C1 domain and flanking sequence that models the catalytic fragment that accumulates during apoptosis) and PKD1-CD (the isolated catalytic domain) display high basal Ser(916) autocatalytic activity and robust activity toward CREBtide (a peptide substrate) but little to no activation loop autophosphorylation and no associated activity toward protein substrates, such as cAMP-response element binding protein and cardiac troponin I. In contrast, PKD1-ΔPH (a PH domain deletion mutant) is recovered as a constitutively active enzyme, with high basal autocatalytic activity and high basal activity toward peptide and protein substrates. These results indicate that individual regions in the regulatory domain act in a distinct manner to control PKD1 activity. Finally, cell-based studies show that PKD1-Δ1-321 does not substitute for WT-PKD1 as an in vivo activator of cAMP-response element binding protein and ERK phosphorylation. Proteolytic events that remove the C1 domain (but not the autoinhibitory PH domain) limit maximal PKD1 activity toward physiologically relevant protein substrates and lead to a defect in PKD1-dependent cellular responses.
Collapse
Affiliation(s)
- Vitalyi O Rybin
- Department of Pharmacology, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | | | | | | | | |
Collapse
|
32
|
Abstract
Protein kinase D1 (PKD1) is a stress-activated serine/threonine kinase that plays a vital role in various physiologically important biological processes, including cell growth, apoptosis, adhesion, motility, and angiogenesis. Dysregulated PKD1 expression also contributes to the pathogenesis of certain cancers and cardiovascular disorders. Studies to date have focused primarily on the canonical membrane-delimited pathway for PKD1 activation by G protein-coupled receptors or peptide growth factors. Here, agonist-dependent increases in diacylglycerol accumulation lead to the activation of protein kinase C (PKC) and PKC-dependent phosphorylation of PKD1 at two highly conserved serine residues in the activation loop; this modification increases PKD1 catalytic activity, as assessed by PKD1 autophosphorylation at a consensus phosphorylation motif at the extreme C terminus. However, recent studies expose additional controls and consequences for PKD1 activation loop and C-terminal phosphorylation as well as additional autophosphorylation reactions and trans-phosphorylations (by PKC and other cellular enzymes) that contribute to the spatiotemporal control of PKD1 signaling in cells. This review focuses on the multisite phosphorylations that are known or predicted to influence PKD1 catalytic activity and may also influence docking interactions with cellular scaffolds and trafficking to signaling microdomains in various subcellular compartments. These modifications represent novel targets for the development of PKD1-directed pharmaceuticals for the treatment of cancers and cardiovascular disorders.
Collapse
Affiliation(s)
- Susan F Steinberg
- Department of Pharmacology, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
33
|
Fu Y, Rubin CS. Protein kinase D: coupling extracellular stimuli to the regulation of cell physiology. EMBO Rep 2011; 12:785-96. [PMID: 21738220 DOI: 10.1038/embor.2011.139] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 06/17/2011] [Indexed: 01/07/2023] Open
Abstract
Protein kinase D (PKD) mediates the actions of stimuli that promote diacylglycerol (DAG) biogenesis. By phosphorylating effectors that regulate transcription, fission and polarized transport of Golgi vesicles, as well as cell migration and survival after oxidative stress, PKDs substantially expand the range of physiological processes controlled by DAG. Dysregulated PKDs have been linked to pathologies including heart hypertrophy and cancer invasiveness. Our understanding of PKD regulation by trans- and autophosphorylation, as well as the subcellular dynamics of PKD substrate phosphorylation, have increased markedly. Selective PKD inhibitors provide new, powerful tools for elucidating the physiological roles of PKDs and potentially treating cardiac disease and cancer.
Collapse
Affiliation(s)
- Ya Fu
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, USA
| | | |
Collapse
|
34
|
Phan D, Stratton MS, Khai Huynh Q, McKinsey TA. A novel protein kinase C target site in protein kinase D is phosphorylated in response to signals for cardiac hypertrophy. Biochem Biophys Res Commun 2011; 411:335-41. [DOI: 10.1016/j.bbrc.2011.06.143] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Accepted: 06/20/2011] [Indexed: 01/19/2023]
|