1
|
Fu J, Yu MG, Li Q, Park K, King GL. Insulin's actions on vascular tissues: Physiological effects and pathophysiological contributions to vascular complications of diabetes. Mol Metab 2021; 52:101236. [PMID: 33878400 PMCID: PMC8513152 DOI: 10.1016/j.molmet.2021.101236] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/07/2021] [Accepted: 04/12/2021] [Indexed: 12/12/2022] Open
Abstract
Background Insulin has been demonstrated to exert direct and indirect effects on vascular tissues. Its actions in vascular cells are mediated by two major pathways: the insulin receptor substrate 1/2-phosphoinositide-3 kinase/Akt (IRS1/2/PI3K/Akt) pathway and the Src/mitogen-activated protein kinase (MAPK) pathway, both of which contribute to the expression and distribution of metabolites, hormones, and cytokines. Scope of review In this review, we summarize the current understanding of insulin's physiological and pathophysiological actions and associated signaling pathways in vascular cells, mainly in endothelial cells (EC) and vascular smooth muscle cells (VSMC), and how these processes lead to selective insulin resistance. We also describe insulin's potential new signaling and biological effects derived from animal studies and cultured capillary and arterial EC, VSMC, and pericytes. We will not provide a detailed discussion of insulin's effects on the myocardium, insulin's structure, or its signaling pathways' various steps, since other articles in this issue discuss these areas in depth. Major conclusions Insulin mediates many important functions on vascular cells via its receptors and signaling cascades. Its direct actions on EC and VSMC are important for transporting and communicating nutrients, cytokines, hormones, and other signaling molecules. These vascular actions are also important for regulating systemic fuel metabolism and energetics. Inhibiting or enhancing these pathways leads to selective insulin resistance, exacerbating the development of endothelial dysfunction, atherosclerosis, restenosis, poor wound healing, and even myocardial dysfunction. Targeted therapies to improve selective insulin resistance in EC and VSMC are thus needed to specifically mitigate these pathological processes. Insulin's actions in vascular cells have a significant influence on systemic metabolism. Insulin exerts its vascular effects through its receptors and signaling cascades. Inhibition or enhancement of different insulin signaling leads to selective insulin resistance. Loss of insulin's actions causes endothelial dysfunction and vascular complications in diabetes.
Collapse
Affiliation(s)
- Jialin Fu
- Dianne Nunnally Hoppes Laboratory for Diabetes Complications, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Marc Gregory Yu
- Dianne Nunnally Hoppes Laboratory for Diabetes Complications, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Qian Li
- Dianne Nunnally Hoppes Laboratory for Diabetes Complications, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Kyoungmin Park
- Dianne Nunnally Hoppes Laboratory for Diabetes Complications, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA
| | - George L King
- Dianne Nunnally Hoppes Laboratory for Diabetes Complications, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
2
|
Bogunovic N, Meekel JP, Micha D, Blankensteijn JD, Hordijk PL, Yeung KK. Impaired smooth muscle cell contractility as a novel concept of abdominal aortic aneurysm pathophysiology. Sci Rep 2019; 9:6837. [PMID: 31048749 PMCID: PMC6497672 DOI: 10.1038/s41598-019-43322-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 04/16/2019] [Indexed: 12/20/2022] Open
Abstract
Ruptured abdominal aortic aneurysms (AAA) are associated with overall mortality rates up to 90%. Despite extensive research, mechanisms leading to AAA formation and advancement are still poorly understood. Smooth muscle cells (SMC) are predominant in the aortic medial layer and maintain the wall structure. Apoptosis of SMC is a well-known phenomenon in the pathophysiology of AAA. However, remaining SMC function is less extensively studied. The aim of this study is to assess the in vitro contractility of human AAA and non-pathologic aortic SMC. Biopsies were perioperatively harvested from AAA patients (n = 21) and controls (n = 6) and clinical data were collected. Contractility was measured using Electric Cell-substrate Impedance Sensing (ECIS) upon ionomycin stimulation. Additionally, SMC of 23% (5 out of 21) of AAA patients showed impaired maximum contraction compared to controls. Also, SMC from patients who underwent open repair after earlier endovascular repair and SMC from current smokers showed decreased maximum contraction vs. controls (p = 0.050 and p = 0.030, respectively). Our application of ECIS can be used to study contractility in other vascular diseases. Finally, our study provides with first proof that impaired SMC contractility might play a role in AAA pathophysiology.
Collapse
MESH Headings
- Actins/genetics
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/physiopathology
- Apoptosis/genetics
- Apoptosis/physiology
- Calcium-Binding Proteins/genetics
- Cells, Cultured
- Cytoskeletal Proteins/genetics
- Humans
- In Vitro Techniques
- Microfilament Proteins/genetics
- Muscle Contraction/genetics
- Muscle Contraction/physiology
- Muscle Proteins/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiology
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/physiology
- Polymerase Chain Reaction
- Vimentin/genetics
- Calponins
Collapse
Affiliation(s)
- Natalija Bogunovic
- Departments of Vascular Surgery, Amsterdam University Medical Centers, location VU University Medical center, Amsterdam, The Netherlands
- Departments of Physiology, Amsterdam University Medical Centers, location VU University Medical center, Amsterdam, The Netherlands
- Departments of Clinical Genetics, Amsterdam University Medical Centers, location VU University Medical center, Amsterdam, The Netherlands
| | - Jorn P Meekel
- Departments of Vascular Surgery, Amsterdam University Medical Centers, location VU University Medical center, Amsterdam, The Netherlands
- Departments of Physiology, Amsterdam University Medical Centers, location VU University Medical center, Amsterdam, The Netherlands
| | - Dimitra Micha
- Departments of Clinical Genetics, Amsterdam University Medical Centers, location VU University Medical center, Amsterdam, The Netherlands
| | - Jan D Blankensteijn
- Departments of Vascular Surgery, Amsterdam University Medical Centers, location VU University Medical center, Amsterdam, The Netherlands
| | - Peter L Hordijk
- Departments of Physiology, Amsterdam University Medical Centers, location VU University Medical center, Amsterdam, The Netherlands
| | - Kak K Yeung
- Departments of Vascular Surgery, Amsterdam University Medical Centers, location VU University Medical center, Amsterdam, The Netherlands.
- Departments of Physiology, Amsterdam University Medical Centers, location VU University Medical center, Amsterdam, The Netherlands.
| |
Collapse
|
3
|
Jin HF, Liu XW, Tang YM, Tang LJ, Wang YL, Du CQ. Effects of total flavones from Dendranthema morifolium on vasocontraction and proliferation of vascular smooth muscle cells. Mol Med Rep 2015; 13:989-93. [PMID: 26648162 DOI: 10.3892/mmr.2015.4576] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 10/14/2015] [Indexed: 11/06/2022] Open
Abstract
Pharmacological studies have shown that the active components in Dendranthema morifolium exhibit protective effects against ischemia/reperfusion injury; however, its pharmacological action on blood vessels has not yet been investigated. The purpose of the present study was to assess the effects of the total flavones extracted from D. morifolium (Ramat.) Tzvel. cv. Hangju (FDM) on the vasocontraction and proliferation of vascular smooth muscle cells (VSMCs). The tension of rat thoracic aortic rings was measured using a mechanical force transducer attached to a recording system. FDM induced a dose‑dependent relaxation of rings with endothelium pre‑contracted by either phenylephrine (PE; 10(‑6) mol/l) or a high concentration of potassium chloride (KCl; 60 mmol/l). FDM did not significantly affect the vasorelaxant effects on mechanically removed endothelium. In endothelium‑denuded aortic rings depolarized by 60 mmol/l KCl, FDM inhibited the contraction induced by Ca2+. FDM reduced the transient contraction caused by PE in a Ca2+‑free solution, but did not affect the contraction induced by phorbol ester. Furthermore, FDM inhibited the proliferation of VSMCs with or without growth stimulation by insulin. In conclusion, that the vasorelaxation induced by FDM in rat aortic rings is not dependent on the endothelium but is mediated via a reduction of the influx of extracellular Ca2+ through the voltage‑dependent and receptor‑operated channels and via the inhibition of the release of intracellular Ca2+ in VSMCs. The anti‑proliferative activity of FDM suggests that it may be beneficial in inhibiting atherosclerosis.
Collapse
Affiliation(s)
- Hong-Feng Jin
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, P.R. China
| | - Xiao-Wei Liu
- Department of Cardiology, The First Clinical Medical Institute of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yi-Ming Tang
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, P.R. China
| | - Li-Jiang Tang
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, P.R. China
| | - Ya-Li Wang
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, P.R. China
| | - Chang-Qing Du
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, P.R. China
| |
Collapse
|
4
|
Chiang S, Breen DM, Guo J, Mori Y, Giacca A. Local insulin application on the carotid artery inhibits neointima formation. Can J Physiol Pharmacol 2013; 91:1086-94. [DOI: 10.1139/cjpp-2013-0038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Anti-mitogenic agents currently used to prevent restenosis in drug-eluting stents delay re-endothelialization. Delayed re-endothelialization is now considered as the main cause of late stent thrombosis with drug-eluting stents, which emphasizes the need for new treatments. We have shown that systemic insulin treatment decreases neointimal growth and accelerates re-endothelialization after arterial injury in a rat model of restenosis. However, systemic insulin treatment cannot be given to non-diabetic individuals because of the risk of hypoglycemia. Thus, we investigated whether local insulin treatment is also effective in reducing neointimal growth after arterial injury. Rats were given local vehicle or local insulin delivered via Pluronic gel applied around the carotid artery immediately following balloon injury. Plasma glucose and systemic insulin levels were not affected by local insulin treatment. Insulin decreased intimal area at 28 days (P < 0.05) and also inhibited vascular smooth muscle cell migration by 60% at 4 days (P < 0.05). NPH (a longer-lasting insulin) also decreased neointimal area. These results indicate that local insulin treatment can lead to decreased restenosis, suggesting a protective vascular effect of insulin in vivo and that local insulin treatment, possibly via insulin-eluting stents, may be clinically relevant.
Collapse
Affiliation(s)
- Simon Chiang
- Department of Physiology, Medical Science Building, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| | - Danna M. Breen
- Department of Physiology, Medical Science Building, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| | - June Guo
- Department of Physiology, Medical Science Building, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| | - Yusaku Mori
- Department of Physiology, Medical Science Building, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
- Division of Diabetes, Metabolism and Endocrinology, Showa University, Shinagawa, Tokyo 142-0064, Japan
| | - Adria Giacca
- Department of Physiology, Medical Science Building, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
5
|
Schwappacher R, Rangaswami H, Su-Yuo J, Hassad A, Spitler R, Casteel DE. cGMP-dependent protein kinase Iβ regulates breast cancer cell migration and invasion via interaction with the actin/myosin-associated protein caldesmon. J Cell Sci 2013; 126:1626-36. [PMID: 23418348 DOI: 10.1242/jcs.118190] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The two isoforms of type I cGMP-dependent protein kinase (PKGIα and PKGIβ) differ in their first ∼100 amino acids, giving each isoform unique dimerization and autoinhibitory domains. The dimerization domains form coiled-coil structures and serve as platforms for isoform-specific protein-protein interactions. Using the PKGIβ dimerization domain as an affinity probe in a proteomic screen, we identified the actin/myosin-associated protein caldesmon (CaD) as a PKGIβ-specific binding protein. PKGIβ phosphorylated human CaD on serine 12 in vitro and in intact cells. Phosphorylation on serine 12 or mutation of serine 12 to glutamic acid (S12E) reduced the interaction between CaD and myosin IIA. Because CaD inhibits myosin ATPase activity and regulates cell motility, we examined the effects of PKGIβ and CaD on cell migration and invasion. Inhibition of the NO/cGMP/PKG pathway reduced migration and invasion of human breast cancer cells, whereas PKG activation enhanced their motility and invasion. siRNA-mediated knockdown of endogenous CaD had pro-migratory and pro-invasive effects in human breast cancer cells. Reconstituting cells with wild-type CaD slowed migration and invasion; however, CaD containing a phospho-mimetic S12E mutation failed to reverse the pro-migratory and pro-invasive activity of CaD depletion. Our data suggest that PKGIβ enhances breast cancer cell motility and invasive capacity, at least in part, by phosphorylating CaD. These findings identify a pro-migratory and pro-invasive function for PKGIβ in human breast cancer cells, suggesting that PKGIβ is a potential target for breast cancer treatment.
Collapse
Affiliation(s)
- Raphaela Schwappacher
- Department of Medicine and Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | |
Collapse
|
6
|
Shoukry A, El-Sherbieny I, Swelam E. Association of insulin resistance, insulin and leptin levels with coronary in-stent restenosis. Egypt Heart J 2012. [DOI: 10.1016/j.ehj.2011.08.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
7
|
Singer HA. Ca2+/calmodulin-dependent protein kinase II function in vascular remodelling. J Physiol 2011; 590:1349-56. [PMID: 22124148 DOI: 10.1113/jphysiol.2011.222232] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Vascular smooth muscle (VSM) undergoes a phenotypic switch in response to injury, a process that contributes to pathophysiological vascular wall remodelling. VSM phenotype switching is a consequence of changes in gene expression, including an array of ion channels and pumps affecting spatiotemporal features of intracellular Ca(2+) signals. Ca(2+) signalling promotes vascular wall remodelling by regulating cell proliferation, motility, and/or VSM gene transcription, although the mechanisms are not clear. In this review, the functions of multifunctional Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) in VSM phenotype switching and synthetic phenotype function are considered. CaMKII isozymes have complex structural and autoregulatory properties. Vascular injury in vivo results in rapid changes in CaMKII isoform expression with reduced expression of CaMKIIγ and upregulation of CaMKIIδ in medial wall VSM. SiRNA-mediated suppression of CaMKIIδ or gene deletion attenuates VSM proliferation and consequent neointimal formation. In vitro studies support functions for CaMKII in the regulation of cell proliferation, motility and gene expression via phosphorylation of CREB1 and HDACIIa/MEF2 complexes. These studies support the concept, and provide potential mechanisms, whereby Ca(2+) signalling through CaMKIIδ promotes VSM phenotype transitions and vascular remodelling.
Collapse
Affiliation(s)
- Harold A Singer
- Center for Cardiovascular Sciences, Albany Medical College (MC-8), 47 New Scotland Avenue, Albany, NY 12208, USA.
| |
Collapse
|
8
|
Kim YH, Kim YS, Park SY, Park CH, Choi WS, Cho GJ. CaMKII regulates pericyte loss in the retina of early diabetic mouse. Mol Cells 2011; 31:289-93. [PMID: 21331776 PMCID: PMC3932701 DOI: 10.1007/s10059-011-0038-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Revised: 12/30/2010] [Accepted: 02/10/2011] [Indexed: 01/01/2023] Open
Abstract
Inducible nitric oxide synthase (iNOS) is an essential mediator in diabetic vascular lesions and known to be regulated by activation of Ca(2+)/calmodulin-dependent protein kinase II (CaMKII). The aim of this study was to investigate whether CaMKII affects iNOS-mediated pericyte death in the retina of diabetic mice with early stage disease. Total- and phospho-CaMKII, iNOS, and active caspase-3 protein levels were assessed by Western blotting, and CaMKII activity was measured by kinase assay. iNOS-related pericyte death was assessed by double immunofluorescent staining for iNOS and α-smooth muscle actin, followed by the TUNEL assay. Autocamtide-2-related inhibitory peptide (AIP), a specific inhibitor of CaMKII, was injected into the right vitreous 2 days before sacrifice of mice, to examine the effect of CaMKII inactivation in diabetic retinas. The levels of total- and phospho-CaMKII, iNOS, and active caspase-3 protein, and CaMKII activity were significantly increased in the diabetic retinas compared with those of control retinas. Furthermore, TUNEL-positive signals colocalized with iNOS-immunoreactive pericytes in the same retinas. However, inactivation of CaMKII by AIP treatment inhibited all these changes, which was accompanied by less pericyte loss. Our results demonstrate that CaMKII contributes to iNOS-related death of pericytes in the diabetic retina and that inactivation of this enzyme may be a potential treatment for retinal vascular lesion.
Collapse
Affiliation(s)
| | | | | | | | - Wan Sung Choi
- Department of Anatomy and Neurobiology, School of Medicine, Brain Korea 21 Biomedical Center, Gyeongsang National University, Jinju 660-751, Korea
| | - Gyeong Jae Cho
- Department of Anatomy and Neurobiology, School of Medicine, Brain Korea 21 Biomedical Center, Gyeongsang National University, Jinju 660-751, Korea
| |
Collapse
|
9
|
Guo D, Zhang JJ, Huang XY. A new Rac/PAK/GC/cGMP signaling pathway. Mol Cell Biochem 2009; 334:99-103. [PMID: 19937092 DOI: 10.1007/s11010-009-0327-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Accepted: 11/04/2009] [Indexed: 10/20/2022]
Abstract
Guanosine 3',5'-cyclic monophosphate (cGMP) and small GTPase Rac are critical regulators of cell functions. Recently, Rac has been shown to use its downstream effector p21-activated kinase (PAK) to directly activate transmembrane guanylyl cyclases (GCs). This novel Rac/PAK/GC/cGMP signaling pathway bridges Rac and cGMP, and provides a general molecular mechanism for diverse receptors to regulate physiological functions such as cell migration through elevating the cellular cGMP level.
Collapse
Affiliation(s)
- Dagang Guo
- Department of Physiology, Cornell University Weill Medical College, New York, NY 10065, USA
| | | | | |
Collapse
|
10
|
Bouallegue A, Pandey NR, Srivastava AK. CaMKII knockdown attenuates H2O2-induced phosphorylation of ERK1/2, PKB/Akt, and IGF-1R in vascular smooth muscle cells. Free Radic Biol Med 2009; 47:858-66. [PMID: 19545622 DOI: 10.1016/j.freeradbiomed.2009.06.022] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2008] [Revised: 05/19/2009] [Accepted: 06/18/2009] [Indexed: 12/23/2022]
Abstract
We have shown earlier a requirement for Ca(2+) and calmodulin (CaM) in the H(2)O(2)-induced activation of extracellular signal-regulated kinases 1 and 2 (ERK1/2) and protein kinase B (PKB), key mediators of growth-promoting, proliferative, and hypertrophic responses in vascular smooth muscle cells (VSMC). Because the effect of CaM is mediated through CaM-dependent protein kinase II (CaMKII), we have investigated here the potential role of CaMKII in H(2)O(2)-induced ERK1/2 and PKB phosphorylation by using pharmacological inhibitors of CaM and CaMKII, a CaMKII inhibitor peptide, and siRNA knockdown strategies for CaMKII alpha. Calmidazolium and W-7, antagonists of CaM, as well as KN-93, a specific inhibitor of CaMKII, attenuated H(2)O(2)-induced responses of ERK1/2 and PKB phosphorylation in a dose-dependent fashion. Similar to H(2)O(2), calmidazolium and KN-93 also exhibited an inhibitory effect on glucose/glucose oxidase-induced phosphorylation of ERK1/2 and PKB in these cells. Transfection of VSMC with CaMKII autoinhibitory peptide corresponding to the autoinhibitory domain (aa 281-309) of CaMKII and with siRNA of CaMKII alpha attenuated the H(2)O(2)-induced phosphorylation of ERK1/2 and PKB. In addition, calmidazolium and KN-93 blocked H(2)O(2)-induced Pyk2 and insulin-like growth factor-1 receptor (IGF-1R) phosphorylation. Moreover, treatment of VSMC with CaMKII alpha siRNA abolished the H(2)O(2)-induced IGF-1R phosphorylation. H(2)O(2) treatment also induced Thr(286) phosphorylation of CaMKII, which was inhibited by both calmidazolium and KN-93. These results demonstrate that CaMKII plays a critical upstream role in mediating the effects of H(2)O(2) on ERK1/2, PKB, and IGF-1R phosphorylation.
Collapse
Affiliation(s)
- Ali Bouallegue
- Laboratory of Cell Signaling, Montreal Diabetes Research Centre, Centre de Recherche, Centre Hospitalier de l'Université de Montréal, Technopole Angus Campus, Montreal, Canada
| | | | | |
Collapse
|
11
|
Breen DM, Chan KK, Dhaliwall JK, Ward MR, Al Koudsi N, Lam L, De Souza M, Ghanim H, Dandona P, Stewart DJ, Bendeck MP, Giacca A. Insulin increases reendothelialization and inhibits cell migration and neointimal growth after arterial injury. Arterioscler Thromb Vasc Biol 2009; 29:1060-6. [PMID: 19359661 DOI: 10.1161/atvbaha.109.185447] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Insulin has both growth-promoting and protective vascular effects in vitro, however the predominant effect in vivo is unclear. We investigated the effects of insulin in vivo on neointimal growth after arterial injury. METHODS AND RESULTS Rats were given subcutaneous control (C) or insulin implants (3U/d;I) 3 days before arterial (carotid or aortic) balloon catheter injury. Normoglycemia was maintained by oral glucose and, after surgery, by intraperitoneal glucose infusion (saline in C). Insulin decreased intimal area (P<0.01) but did not change intimal cell proliferation or apoptosis. However, insulin inhibited cell migration into the intima (P<0.01) and increased expression of smooth muscle cell (SMC) differentiation markers (P<0.05). Insulin also increased reendothelialization (P<0.01) and the number of circulating progenitor cells (P<0.05). CONCLUSIONS These results are the first demonstration that insulin has a protective effect on both SMC and endothelium in vivo, resulting in inhibition of neointimal growth after vessel injury.
Collapse
Affiliation(s)
- Danna M Breen
- Department of Physiology, University of Toronto. Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Hultman KA, Scott AW, Johnson SL. Small molecule modifier screen for kit-dependent functions in zebrafish embryonic melanocytes. Zebrafish 2009; 5:279-87. [PMID: 19133826 DOI: 10.1089/zeb.2008.0542] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Zebrafish is gaining popularity as a vertebrate model for screening small molecules that affect specific phenotypes or genetic pathways. In this study, we present a targeted drug screen to identify drug modifiers of the melanocyte migration defect of a temperature-sensitive allele of the Kit receptor tyrosine kinase, kit(ts). We first test two candidate drugs, the phosphatidylinositol-3-kinase kinase inhibitor (LY294002) and the Erk/MAP kinase inhibitor (PD98059), for their effect on melanocyte migration and survival. We find that LY294002 enhances the migration defect of kit(ts), implicating the phosphatidylinositol-3-kinase kinase pathway in promoting kit-dependent melanocyte migration, but not survival. We then used the kit(ts)-sensitized genetic background to screen a panel of 1280 pharmacologically active drugs to identify drug enhancers and suppressors of the kit(ts) melanocyte migration defect. We identified three drug enhancers of migration, two of which, Papaverine and Isoliquiritigenin, specifically enhance the kit(ts) migration defect, while 8-DPAT affected both melanocyte migration and survival. These drugs now provide additional experimental tools for investigating the mechanisms of kit-promoted melanocyte migration and survival in the zebrafish embryo.
Collapse
Affiliation(s)
- Keith A Hultman
- Department of Genetics, Washington University School of Medicine, 4566 Scott Ave., St. Louis, MO 63110, USA
| | | | | |
Collapse
|
13
|
Suzuki H, Kimura K, Shirai H, Eguchi K, Higuchi S, Hinoki A, Ishimaru K, Brailoiu E, Dhanasekaran DN, Stemmle LN, Fields TA, Frank GD, Autieri MV, Eguchi S. Endothelial nitric oxide synthase inhibits G12/13 and rho-kinase activated by the angiotensin II type-1 receptor: implication in vascular migration. Arterioscler Thromb Vasc Biol 2008; 29:217-24. [PMID: 19095998 DOI: 10.1161/atvbaha.108.181024] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Although, endothelial nitric oxide (NO) synthase (eNOS) is believed to antagonize vascular remodeling induced by the angiotensin II (AngII) type-1 receptor, the exact signaling mechanism remains unclear. METHODS AND RESULTS By expressing eNOS to vascular smooth muscle cells (VSMCs) via adenovirus, we investigated a signal transduction mechanism of the eNOS gene transfer in preventing vascular remodeling induced by AngII. We found marked inhibition of AngII-induced Rho/Rho-kinase activation and subsequent VSMC migration by eNOS gene transfer whereas G(q)-dependent transactivation of the epidermal growth factor receptor by AngII remains intact. This could be explained by the specific inhibition of G(12/13) activation by eNOS-mediated G(12/13) phosphorylation. CONCLUSIONS The eNOS/NO cascade specifically targets the Rho/Rho-kinase system via inhibition of G(12/13) to prevent vascular migration induced by AngII, representing a novel signal cross-talk in cardiovascular protection by NO.
Collapse
Affiliation(s)
- Hiroyuki Suzuki
- Cardiovascular Research Center, Department of Physiology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Roztocil E, Nicholl SM, Davies MG. Insulin-induced epidermal growth factor activation in vascular smooth muscle cells is ADAM-dependent. Surgery 2008; 144:245-51. [PMID: 18656632 DOI: 10.1016/j.surg.2008.03.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2008] [Accepted: 03/14/2008] [Indexed: 10/22/2022]
Abstract
BACKGROUND With the rise in metabolic syndrome, understanding the role of insulin signaling within the cells of vasculature has become more important but yet remains poorly defined. This study examines the role of insulin actions on a pivotal cross-talk receptor, epidermal growth factor receptor (EGFR). EGFR is transactivated by both G-protein-coupled receptors and receptor-linked tyrosine kinases and is key to many of their responses. OBJECTIVE To determine the pathway of EGFR transactivation by insulin in human vascular smooth muscle cells (VSMC). METHODS VSMC were cultured in vitro. Assays of EGFR phosphorylation were examined in response to insulin in the presence and absence of the plasmin inhibitors (e-aminocaproic acid and aprotinin) matrix metalloprotease (MMP) inhibitor GM6001, the A disintegrin and metalloproteinase domain (ADAM) inhibitors tumor necrosis factor-alpha protease inhibitor (TAPI)-0 and TAPI-1, heparin-binding epidermal growth factor (HB-EGF) inhibitor, CRM197, HB-EGF inhibitory antibodies, EGF inhibitory antibodies, and the EGFR inhibitor AG1478. RESULTS Insulin induced time-dependent EGFR phosphorylation, which was inhibited by AG1478 in a concentration-dependent manner. Application of the plasmin inhibitors did not block the response. EGFR phosphorylation by insulin was blocked by inhibition of MMP activity and the ligand HB-EGF. The presence of the ADAM inhibitors, TAPI-0 and TAPI-1 significantly decreased EGFR activation. EGFR phosphorylation by EGF was not interrupted by inhibition of plasmin, MMPs TAPIs, or HB-EGF. Direct blockade of the EGFR prevented activation by both insulin and EGF. CONCLUSION Insulin can induce transactivation of EGFR by an ADAM-mediated, HB-EGF-dependent process. This is the first description of cross-talk via ADAM between insulin and EGFR in VSMC. Targeting a pivotal cross-talk receptor such as EGFR, which can be transactivated by both G-protein-coupled receptors and receptor tyrosine kinases is an attractive molecular target.
Collapse
Affiliation(s)
- Elisa Roztocil
- Vascular Biology and Therapeutics Program, Methodist DeBakey Heart and Vascular Center, Department of Cardiovascular Surgery, The Methodist Hospital, Houston, TX 77030, USA
| | | | | |
Collapse
|
15
|
Mercure MZ, Ginnan R, Singer HA. CaM kinase II delta2-dependent regulation of vascular smooth muscle cell polarization and migration. Am J Physiol Cell Physiol 2008; 294:C1465-75. [PMID: 18385282 DOI: 10.1152/ajpcell.90638.2007] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Previous studies indicate involvement of the multifunctional Ca2+/calmodulin-dependent protein kinase II (CaMKII) in vascular smooth muscle (VSM) cell migration. In the present study, molecular loss-of-function studies were used specifically to assess the role of the predominant CaMKII delta2 isoform on VSM cell migration using a scratch wound healing assay. Targeted CaMKII delta2 knockdown using siRNA or inhibition of activity by overexpressing a kinase-negative mutant resulted in attenuation of VSM cell migration. Temporal and spatial assessments of kinase autophosphorylation indicated rapid and transient activation in response to wounding, in addition to a sustained activation in the leading edge of migrating and spreading cells. Furthermore, siRNA-mediated suppression of CaMKII delta2 resulted in the inhibition of wound-induced Rac activation and Golgi reorganization, and disruption of leading edge morphology, indicating an important function for CaMKII delta2 in regulating VSM cell polarization. Numerous previous reports link activation of CaMKII to ERK1/2 signaling in VSM. Wound-induced ERK1/2 activation was also found to be dependent on CaMKII; however, ERK activity did not account for effects of CaMKII in regulating Golgi polarization, indicating alternative mechanisms by which CaMKII affects the complex events involved in cell migration. Wounding a VSM cell monolayer results in CaMKII delta2 activation, which positively regulates VSM cell polarization and downstream signaling, including Rac and ERK1/2 activation, leading to cell migration.
Collapse
Affiliation(s)
- Melissa Z Mercure
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208, USA
| | | | | |
Collapse
|
16
|
Yang M, Kahn AM. Insulin-inhibited and stimulated cultured vascular smooth muscle cell migration are related to divergent effects on protein phosphatase-2A and autonomous calcium/calmodulin-dependent protein kinase II. Atherosclerosis 2007; 196:227-233. [PMID: 17553505 DOI: 10.1016/j.atherosclerosis.2007.04.050] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2006] [Revised: 03/30/2007] [Accepted: 04/30/2007] [Indexed: 11/26/2022]
Abstract
Insulin, in the permissive presence of nitric oxide (NO), stimulates cGMP production which inhibits autonomous calcium/calmodulin-dependent protein kinase II (CaM kinase II) thereby inhibiting cultured vascular smooth muscle cell (VSMC) migration. In the presence of angiotensin II (Ang II), insulin stimulates NAD(P)H oxidase activity leading to increased VSMC migration. We wished to see whether insulin-stimulated cGMP stimulates protein phosphatase-2A (PP-2A) thereby inhibiting autonomous CaM kinase II and migration, and whether insulin, in the presence of Ang II, inhibits PP-2A and stimulates autonomous CaM kinase II in a NAD(P)H oxidase-dependent manner. One nanomole per litre of insulin in the presence of NO, or 50 micromol/L 8-Br-cGMP stimulated PP-2A activity by 46+/-6 and 247+/-23%, respectively (both P<0.05), and 8-Br-cGMP inhibited autonomous CaM kinase II activity by 67+/-9% (P<0.05) by a 10 nmol/L okadaic acid-sensitive pathway. Insulin plus Ang II inhibited PP-2A activity by 57+/-7% (P<0.05) and stimulated autonomous CaM kinase II activity by 120+/-14% (P<0.05), both by an apocynin-sensitive pathway. 8-Br-cGMP-inhibited VSMC migration was blocked by okadaic acid. It is concluded that insulin in the presence of NO stimulates cGMP which stimulates PP-2A activity causing inhibition of autonomous CaM kinase II activity and thus VSMC migration, and that insulin in the presence of Ang II inhibits PP-2A and stimulates autonomous CaM kinase II activities by a NAD(P)H oxidase-dependent mechanism which are associated with insulin-stimulated NAD(P)H oxidase-dependent migration.
Collapse
Affiliation(s)
- Ming Yang
- Division of Nephrology and Hypertension, Department of Medicine, Medical School, University of Texas Health Science Center, Houston, United States
| | - Andrew M Kahn
- Division of Nephrology and Hypertension, Department of Medicine, Medical School, University of Texas Health Science Center, Houston, United States.
| |
Collapse
|
17
|
Jones RJ, Jourd'heuil D, Salerno JC, Smith SME, Singer HA. iNOS regulation by calcium/calmodulin-dependent protein kinase II in vascular smooth muscle. Am J Physiol Heart Circ Physiol 2007; 292:H2634-42. [PMID: 17293490 DOI: 10.1152/ajpheart.01247.2006] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nitric oxide synthase (NOS) expression is regulated transcriptionally in response to cytokine induction and posttranslationally by palmitoylation and trafficking into perinuclear aggresome-like structures. We investigated the effects of multifunctional calcium/calmodulin-dependent protein kinase II protein kinase (CaMKII) on inducible NOS (iNOS) trafficking in cultured rat aortic vascular smooth muscle cells (VSMCs). Immunofluorescence and confocal microscopy demonstrated colocalization of iNOS and CaMKIIdelta(2) with a perinuclear distribution and concentration in aggresome-like structures identified by colocalization with gamma-tubulin. Furthermore, CaMKIIdelta(2) coimmunoprecipitated with iNOS in a CaMKII activity-dependent manner. Addition of Ca(2+)-mobilizing stimuli expected to activate CaMKII; a purinergic agonist (UTP) or calcium ionophore (ionomycin) caused a general redistribution of iNOS from cytosolic to membrane and nuclear fractions. Similarly, adenoviral expression of a constitutively active CaMKIIdelta(2) mutant altered iNOS localization, shifting iNOS from the cytosolic fraction. Suppression of CaMKIIdelta(2) using an adenovirus expressing a short hairpin, small interfering RNA increased nuclear iNOS localization in resting cells but inhibited ionomycin-induced translocation of iNOS to the nucleus. Following addition of these chronic and acute CaMKII modulators, there were fewer aggresome-like structures containing iNOS. All of the treatments that chronically affected CaMKII activity or expression significantly inhibited iNOS-specific activity following cytokine induction. The results suggest that CaMKIIdelta(2) may be an important regulator of iNOS trafficking and activity in VSMCs.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/cytology
- Aorta, Thoracic/metabolism
- Benzylamines/pharmacology
- Calcium/metabolism
- Calcium-Calmodulin-Dependent Protein Kinase Type 2
- Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors
- Calcium-Calmodulin-Dependent Protein Kinases/genetics
- Calcium-Calmodulin-Dependent Protein Kinases/metabolism
- Cell Membrane/enzymology
- Cell Nucleus/enzymology
- Cells, Cultured
- Cytokines/metabolism
- Cytoplasm/enzymology
- Enzyme Induction
- Interferon-gamma/metabolism
- Interleukin-1beta/metabolism
- Ionomycin/pharmacology
- Ionophores/pharmacology
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Nitric Oxide Synthase Type II/biosynthesis
- Protein Kinase Inhibitors/pharmacology
- Protein Transport
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Rats
- Rats, Sprague-Dawley
- Signal Transduction/drug effects
- Sulfonamides/pharmacology
- Tumor Necrosis Factor-alpha/metabolism
- Uridine Triphosphate/metabolism
Collapse
Affiliation(s)
- Rachel J Jones
- Center for Cardiovascular Sciences, Albany Medical College, 43 New Scotland Ave., Albany, NY 12208, USA
| | | | | | | | | |
Collapse
|
18
|
Piggott LA, Hassell KA, Berkova Z, Morris AP, Silberbach M, Rich TC. Natriuretic peptides and nitric oxide stimulate cGMP synthesis in different cellular compartments. J Gen Physiol 2006; 128:3-14. [PMID: 16769793 PMCID: PMC2151547 DOI: 10.1085/jgp.200509403] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2005] [Accepted: 05/23/2006] [Indexed: 11/20/2022] Open
Abstract
Cyclic nucleotide-gated (CNG) channels are a family of ion channels activated by the binding of cyclic nucleotides. Endogenous channels have been used to measure cyclic nucleotide signals in photoreceptor outer segments and olfactory cilia for decades. Here we have investigated the subcellular localization of cGMP signals by monitoring CNG channel activity in response to agonists that activate either particulate or soluble guanylyl cyclase. CNG channels were heterologously expressed in either human embryonic kidney (HEK)-293 cells that stably overexpress a particulate guanylyl cyclase (HEK-NPRA cells), or cultured vascular smooth muscle cells (VSMCs). Atrial natriuretic peptide (ANP) was used to activate the particulate guanylyl cyclase and the nitric oxide donor S-nitroso-n-acetylpenicillamine (SNAP) was used to activate the soluble guanylyl cyclase. CNG channel activity was monitored by measuring Ca2+ or Mn2+ influx through the channels using the fluorescent dye, fura-2. We found that in HEK-NPRA cells, ANP-induced increases in cGMP levels activated CNG channels in a dose-dependent manner (0.05-10 nM), whereas SNAP (0.01-100 microM) induced increases in cGMP levels triggered little or no activation of CNG channels (P < 0.01). After pretreatment with 100 microM 3-isobutyl-1-methylxanthine (IBMX), a nonspecific phosphodiesterase inhibitor, ANP-induced Mn2+ influx through CNG channels was significantly enhanced, while SNAP-induced Mn2+ influx remained small. In contrast, we found that in the presence of IBMX, both 1 nM ANP and 100 microM SNAP triggered similar increases in total cGMP levels. We next sought to determine if cGMP signals are compartmentalized in VSMCs, which endogenously express particulate and soluble guanylyl cyclase. We found that 10 nM ANP induced activation of CNG channels more readily than 100 muM SNAP; whereas 100 microM SNAP triggered higher levels of total cellular cGMP accumulation. These results suggest that cGMP signals are spatially segregated within cells, and that the functional compartmentalization of cGMP signals may underlie the unique actions of ANP and nitric oxide.
Collapse
Affiliation(s)
- Leslie A Piggott
- Program in Cell and Regulatory Biology, The University of Texas Graduate School of Biomedical Sciences at Houston, 77225, USA
| | | | | | | | | | | |
Collapse
|
19
|
Monti LD, Setola E, Fragasso G, Camisasca RP, Lucotti P, Galluccio E, Origgi A, Margonato A, Piatti P. Metabolic and endothelial effects of trimetazidine on forearm skeletal muscle in patients with type 2 diabetes and ischemic cardiomyopathy. Am J Physiol Endocrinol Metab 2006; 290:E54-E59. [PMID: 16174656 DOI: 10.1152/ajpendo.00083.2005] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The aim of the present study was to evaluate the effect of prolonged inhibition of beta-oxidation on glucose and lipid muscle forearm metabolism and cGMP and endothelin-1 forearm release in patients with type 2 diabetes mellitus and ischemic cardiomyopathy. Fifteen patients were randomly allocated in a double-blind cross-over parallel study with trimetazidine (20 mg tid) or placebo lasting 15 days. At the end of each period, all patients underwent euglycemic hyperinsulinemic clamps with forearm indirect calorimetry and endothelial balance of vasodilator and vasoconstricor factors. Compared with placebo, trimetazidine induced 1) an increase in insulin-induced forearm glucose uptake and glucose oxidation accompanied by a reduction in forearm lipid oxidation and citrate release and 2) a decrease of endothelin-1 release paralleled by a significant increase in forearm cGMP release. Forearm glucose oxidation significantly correlated with cGMP release (r=0.37, P<0.04), whereas forearm lipid oxidation positively correlated with endothelin-1 release (r=0.40, P<0.03). In conclusion, for the first time, we demonstrated that insulin-induced forearm glucose oxidation and forearm cGMP release were increased whereas forearm endothelin-1 release was decreased during trimetazidine treatment. Muscle's metabolic and vascular effects of trimetazidine add new interest in the use of trimetazidine in type 2 diabetic patients with cardiovascular disease.
Collapse
Affiliation(s)
- Lucilla D Monti
- Laboratory L20, Core Lab., Diabetology, Endocrinology and Metabolic Disease Unit, IRCCS H San Raffaele, Via Olgettina 60, 20132 Milan, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Kang DH, Han L, Ouyang X, Kahn AM, Kanellis J, Li P, Feng L, Nakagawa T, Watanabe S, Hosoyamada M, Endou H, Lipkowitz M, Abramson R, Mu W, Johnson RJ. Uric acid causes vascular smooth muscle cell proliferation by entering cells via a functional urate transporter. Am J Nephrol 2005; 25:425-33. [PMID: 16113518 DOI: 10.1159/000087713] [Citation(s) in RCA: 169] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2005] [Accepted: 07/05/2005] [Indexed: 11/19/2022]
Abstract
BACKGROUND Soluble uric acid stimulates vascular smooth muscle cell (VSMC) proliferation by activating mitogen-activated protein kinases, and stimulating COX-2 and PDGF synthesis. The mechanism by which uric acid enters the VSMC is not known. We hypothesized that uric acid enters via transporters similar to that observed in the kidney. METHODS We studied the uptake of uric acid into rat VSMC under polarized and depolarized conditions and in the presence of organic anion transport (OAT) inhibitors (probenecid and benzbromarone) or p-aminohippurate (PAH). We also examined the ability of probenecid to inhibit uric acid-induced VSMC proliferation and monocyte chemoattractant protein-1 (MCP-1) synthesis. RESULTS (14)C-Urate uptake was shown in VSMC and was enhanced under depolarized conditions. (14)C-Uric acid uptake was inhibited by probenecid and benzbromarone, as well as by unlabelled urate and PAH. Probenecid blocked VSMC proliferation and MCP-1 expression in response to uric acid. VSMC did not express rOAT1-3, rOAT-5 or URAT-1 mRNA by PCR, but did express the voltage-sensitive transporter (UAT) by both PCR and RNase protection assay. CONCLUSIONS Urate enters VSMC by both voltage-sensitive and OAT pathways, and the uptake, cell proliferation and MCP-1 expression can be blocked by OAT inhibitors. The specific transporter(s) responsible for the urate uptake remains to be determined.
Collapse
Affiliation(s)
- Duk-Hee Kang
- Division of Nephrology, Ewha Women's University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Frank GD, Eguchi S, Motley ED. The role of reactive oxygen species in insulin signaling in the vasculature. Antioxid Redox Signal 2005; 7:1053-61. [PMID: 15998260 DOI: 10.1089/ars.2005.7.1053] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Although there is an abundance of evidence suggesting that insulin resistance plays a significant role in the vasculature, the precise mechanistic role involved still remains unclear. In this review, we discuss the current background of insulin resistance in the context of insulin signaling and action in the vasculature. Also, studies suggest that insulin resistance, diabetes, and cardiovascular disease all share a common involvement with oxidative stress. Recently, we reported that lysophosphatidylcholine, a major bioactive product of oxidized low-density lipoprotein, and angiotensin II, a vasoactive hormone and a potent inducer of reactive oxygen species (ROS), negatively regulate insulin signaling in vascular smooth muscle cells (VSMCs). In endothelial cells, insulin stimulates the release of nitric oxide, which results in VSMC relaxation and inhibition of atherosclerosis. Other data suggest that angiotensin II inhibits the vasodilator effects of insulin through insulin receptor substrate-1 phosphorylation at Ser312 and Ser616. Moreover, ROS impair insulin-induced vasorelaxation by neutralizing nitric oxide to form peroxynitrite. Thus, evidence is growing to enable us to better understand mechanistically the relationship between insulin/insulin resistance and ROS in the vasculature, and the impact they have on cardiovascular disease.
Collapse
Affiliation(s)
- Gerald D Frank
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | |
Collapse
|
22
|
Netherton SJ, Maurice DH. Vascular endothelial cell cyclic nucleotide phosphodiesterases and regulated cell migration: implications in angiogenesis. Mol Pharmacol 2005; 67:263-72. [PMID: 15475573 DOI: 10.1124/mol.104.004853] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Angiogenesis is necessary during embryonic development and wound healing but can be detrimental in pathologies, including cancer. Because initiation of angiogenesis involves migration and proliferation of vascular endothelial cells (VECs) and cAMP-elevating agents inhibit these events, such agents may represent a novel therapeutic avenue to controlling angiogenesis. Intracellular cAMP levels are regulated by their synthesis by adenylyl cyclases and hydrolysis by cyclic nucleotide phosphodiesterases (PDEs). In this report, we show that human VECs express variants of PDE2, PDE3, PDE4, and PDE5 families and demonstrate that the levels of these enzymes differ in VECs derived from aorta, umbilical vein, and microvascular structures. Selective inhibition of PDE2 did not increase cAMP in any VECs, whether in the absence or presence of forskolin, but it did inhibit migration of all VECs studied. Inhibition of PDE4 activity decreased migration, and in conjunction with forskolin, increased cAMP in all VECs studied. PDE3 inhibition potentiated forskolin-induced increases in cAMP and inhibited migration in VECs derived from aorta and umbilical vein but not in microvascular VECs. In experiments with combinations of PDE2, PDE3, and PDE4 inhibitors, a complex interaction between the abilities of these agents to limit human VEC migration was observed. Overall, our data are consistent with the hypothesis that PDE subtype inhibition allows different effects in distinct VEC populations and indicate that these agents may represent novel therapeutic agents to limit angiogenesis in complex human diseases.
Collapse
MESH Headings
- 1-Methyl-3-isobutylxanthine/pharmacology
- 2',3'-Cyclic-Nucleotide Phosphodiesterases/genetics
- 2',3'-Cyclic-Nucleotide Phosphodiesterases/metabolism
- 3',5'-Cyclic-AMP Phosphodiesterases/genetics
- 3',5'-Cyclic-AMP Phosphodiesterases/metabolism
- 3',5'-Cyclic-GMP Phosphodiesterases/genetics
- 3',5'-Cyclic-GMP Phosphodiesterases/metabolism
- Aorta
- Cell Movement/physiology
- Cyclic AMP/metabolism
- Cyclic AMP/physiology
- Cyclic Nucleotide Phosphodiesterases, Type 2
- Cyclic Nucleotide Phosphodiesterases, Type 3
- Cyclic Nucleotide Phosphodiesterases, Type 4
- Cyclic Nucleotide Phosphodiesterases, Type 5
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/physiology
- Humans
- Kinetics
- Microcirculation/physiology
- Neovascularization, Physiologic/physiology
- Phosphoric Diester Hydrolases/genetics
- Phosphoric Diester Hydrolases/metabolism
- Quinolones/pharmacology
- RNA, Messenger/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Umbilical Veins
Collapse
Affiliation(s)
- Stuart J Netherton
- Department of Pharmacology and Toxicology, Botterell Hall, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | | |
Collapse
|
23
|
Harvey BP, Banga SS, Ozer HL. Regulation of the Multifunctional Ca2+/Calmodulin-dependent Protein Kinase II by the PP2C Phosphatase PPM1F in Fibroblasts. J Biol Chem 2004; 279:24889-98. [PMID: 15140879 DOI: 10.1074/jbc.m400656200] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The regulation of the multifunctional calcium/calmodulin dependent protein kinase II (CaMKII) by serine/threonine protein phosphatases has been extensively studied in neuronal cells; however, this regulation has not been investigated previously in fibroblasts. We cloned a cDNA from SV40-transformed human fibroblasts that shares 80% homology to a rat calcium/calmodulin-dependent protein kinase phosphatase that encodes a PPM1F protein. By using extracts from transfected cells, PPM1F, but not a mutant (R326A) in the conserved catalytic domain, was found to dephosphorylate in vitro a peptide corresponding to the auto-inhibitory region of CaMKII. Further analyses demonstrated that PPM1F specifically dephosphorylates the phospho-Thr-286 in autophosphorylated CaMKII substrate and thus deactivates the CaMKII in vitro. Coimmunoprecipitation of CaMKII with PPM1F indicates that the two proteins can interact intracellularly. Binding of PPM1F to CaMKII involves multiple regions and is not dependent on intact phosphatase activity. Furthermore, overexpression of PPM1F in fibroblasts caused a reduction in the CaMKII-specific phosphorylation of the known substrate vimentin(Ser-82) following induction of the endogenous CaM kinase. These results identify PPM1F as a CaM kinase phosphatase within fibroblasts, although it may have additional functions intracellularly since it has been presented elsewhere as POPX2 and hFEM-2. We conclude that PPM1F, possibly together with the other previously described protein phosphatases PP1 and PP2A, can regulate the activity of CaMKII. Moreover, because PPM1F dephosphorylates the critical autophosphorylation site of CaMKII, we propose that this phosphatase plays a key role in the regulation of the kinase intracellularly.
Collapse
Affiliation(s)
- Bohdan P Harvey
- Department of Microbiology and Molecular Genetics, University of Medicine and Dentistry of New Jersey (UMDNJ)-New Jersey Medical School and UMDNJ-Graduate School of Biomedical Sciences, Newark, New Jersey 07101, USA
| | | | | |
Collapse
|
24
|
Hum D, Besnard S, Sanchez R, Devost D, Gossard F, Hamet P, Tremblay J. Characterization of a cGMP-response element in the guanylyl cyclase/natriuretic peptide receptor A gene promoter. Hypertension 2004; 43:1270-8. [PMID: 15096467 DOI: 10.1161/01.hyp.0000126920.93207.53] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Previous studies have shown that atrial natriuretic peptide (ANP) can inhibit transcription of its receptor, guanylyl cyclase A, by a mechanism dependent on cGMP and have suggested the presence of a putative cGMP-response element (cGMP-RE) in the Npr1 gene promoter. To localize and characterize the putative cis-acting element, we have subcloned a 1520-bp fragment of the rat Npr1 promoter in an expression vector containing the luciferase reporter gene. Several fragments, generated by exonuclease III-directed deletions, were transiently transfected into cells to measure their promoter activity. Deletion from -1520 to -1396 of a 1520-bp-long Npr1 promoter led to a 5-fold increase in luciferase activity. Subsequent deletion to the position -1307 resulted in a decrease of luciferase activity by 90%. Preincubation of cells with 100 nM of ANP or 100 microM 8-bromo-cGMP inhibited luciferase activity of the 1520-bp and 1396-bp-long fragments, but not the activity of the 1307-bp fragment, suggesting that the cGMP-RE is localized between positions -1396 and -1307. The cGMP regulatory region was narrowed by gel shift assays and footprinting to position -1372 to -1354 from the transcription start site of Npr1 and indicated its interaction with transcriptional factor(s). Cross-competition experiments with mutated oligonucleotides led to the definition of a consensus sequence (-1372 AaAtRKaNTTCaAcAKTY -1354) for the novel cGMP-RE, which is conserved in the human (75% identity) and mouse (95% identity) Npr1 promoters.
Collapse
Affiliation(s)
- David Hum
- Centre de recherche, Centre hospitalier de l'Université de Montréal-Hôtel-Dieu, Montréal, Québec, Canada
| | | | | | | | | | | | | |
Collapse
|
25
|
Pfleiderer PJ, Lu KK, Crow MT, Keller RS, Singer HA. Modulation of vascular smooth muscle cell migration by calcium/ calmodulin-dependent protein kinase II-delta 2. Am J Physiol Cell Physiol 2004; 286:C1238-45. [PMID: 14761894 DOI: 10.1152/ajpcell.00536.2003] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Previous studies demonstrated a requirement for multifunctional Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) in PDGF-stimulated vascular smooth muscle (VSM) cell migration. In the present study, molecular approaches were used specifically to assess the role of the predominant CaMKII isoform (delta(2) or delta(C)) on VSM cell migration. Kinase-negative (K43A) and constitutively active (T287D) mutant forms of CaMKII delta(2) were expressed using recombinant adenoviruses. CaMKII activities were evaluated in vitro by using a peptide substrate and in intact cells by assessing the phosphorylation of overexpressed phospholamban on Thr(17), a CaMKII-selective phosphorylation site. Expression of kinase-negative CaMKII delta(2) inhibited substrate phosphorylation both in vitro and in the intact cell, indicating a dominant-negative function with respect to exogenous substrate. However, overexpression of the kinase-negative mutant failed to inhibit endogenous CaMKII delta(2) autophosphorylation on Thr(287) after activation of cells with ionomycin, and in fact, these subunits served as a substrate for the endogenous kinase. Constitutively active CaMKII delta(2) phosphorylated substrate in vitro without added Ca(2+)/calmodulin and in the intact cell without added Ca(2+)-dependent stimuli, but it inhibited autophosphorylation of endogenous CaMKII delta(2) on Thr(287). Basal and PDGF-stimulated cell migration was significantly enhanced in cells expressing kinase-negative CaMKII delta(2), an effect opposite that of KN-93, a chemical inhibitor of CaMKII activation. Expression of the constitutively active CaMKII delta(2) mutant inhibited PDGF-stimulated cell migration. These studies point to a role for the CaMKII delta(2) isoform in regulating VSM cell migration. An inclusive interpretation of results using both pharmacological and molecular approaches raises the hypothesis that CaMKII delta(2) autophosphorylation may play an important role in PDGF-stimulated VSM cell migration.
Collapse
Affiliation(s)
- Paul J Pfleiderer
- Center for Cardiovascular Sciences, Albany Medical College, NY 12208, USA
| | | | | | | | | |
Collapse
|
26
|
Yang M, Yang Y, Zhang S, Kahn AM. Insulin-stimulated hydrogen peroxide increases guanylate cyclase activity in vascular smooth muscle. Hypertension 2003; 42:569-73. [PMID: 12963680 DOI: 10.1161/01.hyp.0000092441.27668.bd] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Insulin resistance is associated with vascular disease. Physiological concentrations of insulin inhibit cultured vascular smooth muscle cell (VSMC) contraction and migration by increasing nitric oxide (NO)-stimulated cGMP accumulation. The failure to do so in insulin-resistant states may aggravate vascular disease. We sought to determine the mechanism of insulin's increase in cGMP accumulation. Isobutylmethylxanthine, an inhibitor of phosphodiesterase activity, inhibited the decline in cGMP levels measured by immunoassay in cGMP-loaded cultured rat aortic VSMCs, but 1 nmol insulin did not. Thus, insulin's increase in cGMP accumulation is due to stimulated production, not inhibited hydrolysis and/or efflux. Insulin, which increases the NADH/NAD+ ratio in these cells, stimulated superoxide anion (O2-) accumulation measured by lucigenin luminescence to 256+/-25% (P<0.05) by a process that was blocked by the NADH oxidase inhibitor diphenyliodonium (DPI) and enhanced by the superoxide dismutase inhibitor diethyldithiocarbonate (DETCA). Insulin also stimulated hydrogen peroxide (H2O2) accumulation measured by horseradish peroxidase/luminol luminescence to 221+/-22% (P<0.05) by a DETCA-sensitive mechanism. H2O2 (100 micromol/L) in the absence of insulin increased NO-stimulated cGMP accumulation to 151+/-11% (P<0.05). Insulin alone increased NO-stimulated cGMP accumulation to 183+/-17% (P<0.05), and this was blocked by either DPI or DETCA. We conclude that insulin increases NADH oxidase-derived O2- production in cultured rat VSMCs. This did not cause the expected scavenging of NO resulting in the reduction of NO-stimulated guanylate cyclase activity, but enough O2- was metabolized to H2O2 to increase overall NO-stimulated cGMP production.
Collapse
Affiliation(s)
- Ming Yang
- Division of Renal Diseases and Hypertension, Department of Medicine, The University of Texas Health Science Center, Houston, Tex 77030, USA
| | | | | | | |
Collapse
|