1
|
Angely C, Ladant D, Planus E, Louis B, Filoche M, Chenal A, Isabey D. Functional and structural consequences of epithelial cell invasion by Bordetella pertussis adenylate cyclase toxin. PLoS One 2020; 15:e0228606. [PMID: 32392246 PMCID: PMC7213728 DOI: 10.1371/journal.pone.0228606] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/18/2020] [Indexed: 01/13/2023] Open
Abstract
Bordetella pertussis, the causative agent of whopping cough, produces an adenylate cyclase toxin (CyaA) that plays a key role in the host colonization by targeting innate immune cells which express CD11b/CD18, the cellular receptor of CyaA. CyaA is also able to invade non-phagocytic cells, via a unique entry pathway consisting in a direct translocation of its catalytic domain across the cytoplasmic membrane of the cells. Within the cells, CyaA is activated by calmodulin to produce high levels of cyclic adenosine monophosphate (cAMP) and alter cellular physiology. In this study, we explored the effects of CyaA toxin on the cellular and molecular structure remodeling of A549 alveolar epithelial cells. Using classical imaging techniques, biochemical and functional tests, as well as advanced cell mechanics method, we quantify the structural and functional consequences of the massive increase of intracellular cyclic AMP induced by the toxin: cell shape rounding associated to adhesion weakening process, actin structure remodeling for the cortical and dense components, increase in cytoskeleton stiffness, and inhibition of migration and repair. We also show that, at low concentrations (0.5 nM), CyaA could significantly impair the migration and wound healing capacities of the intoxicated alveolar epithelial cells. As such concentrations might be reached locally during B. pertussis infection, our results suggest that the CyaA, beyond its major role in disabling innate immune cells, might also contribute to the local alteration of the epithelial barrier of the respiratory tract, a hallmark of pertussis.
Collapse
Affiliation(s)
- Christelle Angely
- Equipe 13, Biomécanique & Appareil Respiratoire, Inserm U955, Créteil, France
- UMR 955, UPEC, Université Paris-Est, Créteil, France
- ERL 7000, CNRS, Créteil, France
| | - Daniel Ladant
- Unité de Biochimie des Interactions Macromoléculaires (CNRS UMR 3528), Département de Biologie Structurale et Chimie, Institut Pasteur, Paris, France
| | - Emmanuelle Planus
- Institut pour l’Avancée des Biosciences (IAB), Centre de Recherche UGA/ Inserm U1209 / CNRS UMR 5309, La Tronche, France
| | - Bruno Louis
- Equipe 13, Biomécanique & Appareil Respiratoire, Inserm U955, Créteil, France
- UMR 955, UPEC, Université Paris-Est, Créteil, France
- ERL 7000, CNRS, Créteil, France
| | - Marcel Filoche
- Equipe 13, Biomécanique & Appareil Respiratoire, Inserm U955, Créteil, France
- UMR 955, UPEC, Université Paris-Est, Créteil, France
- ERL 7000, CNRS, Créteil, France
- Laboratoire de Physique de la Matière Condensée, Ecole Polytechnique, CNRS, IP Paris, Palaiseau, France
| | - Alexandre Chenal
- Unité de Biochimie des Interactions Macromoléculaires (CNRS UMR 3528), Département de Biologie Structurale et Chimie, Institut Pasteur, Paris, France
| | - Daniel Isabey
- Equipe 13, Biomécanique & Appareil Respiratoire, Inserm U955, Créteil, France
- UMR 955, UPEC, Université Paris-Est, Créteil, France
- ERL 7000, CNRS, Créteil, France
- * E-mail:
| |
Collapse
|
2
|
Smith SA, Newby AC, Bond M. Ending Restenosis: Inhibition of Vascular Smooth Muscle Cell Proliferation by cAMP. Cells 2019; 8:cells8111447. [PMID: 31744111 PMCID: PMC6912325 DOI: 10.3390/cells8111447] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/14/2019] [Accepted: 11/14/2019] [Indexed: 12/17/2022] Open
Abstract
Increased vascular smooth muscle cell (VSMC) proliferation contributes towards restenosis after angioplasty, vein graft intimal thickening and atherogenesis. The second messenger 3′ 5′ cyclic adenosine monophosphate (cAMP) plays an important role in maintaining VSMC quiescence in healthy vessels and repressing VSMC proliferation during resolution of vascular injury. Although the anti-mitogenic properties of cAMP in VSMC have been recognised for many years, it is only recently that we gained a detailed understanding of the underlying signalling mechanisms. Stimuli that elevate cAMP in VSMC inhibit G1-S phase cell cycle progression by inhibiting expression of cyclins and preventing S-Phase Kinase Associated Protein-2 (Skp2-mediated degradation of cyclin-dependent kinase inhibitors. Early studies implicated inhibition of MAPK signalling, although this does not fully explain the anti-mitogenic effects of cAMP. The cAMP effectors, Protein Kinase A (PKA) and Exchange Protein Activated by cAMP (EPAC) act together to inhibit VSMC proliferation by inducing Cyclic-AMP Response Element Binding protein (CREB) activity and inhibiting members of the RhoGTPases, which results in remodelling of the actin cytoskeleton. Cyclic-AMP induced actin remodelling controls proliferation by modulating the activity of Serum Response Factor (SRF) and TEA Domain Transcription Factors (TEAD), which regulate expression of genes required for proliferation. Here we review recent research characterising these mechanisms, highlighting novel drug targets that may allow the anti-mitogenic properties of cAMP to be harnessed therapeutically to limit restenosis.
Collapse
Affiliation(s)
| | | | - Mark Bond
- Correspondence: ; Tel.: +44-117-3423586
| |
Collapse
|
3
|
Gao X, Zhang X, Cui L, Chen R, Zhang C, Xue J, Zhang L, He W, Li J, Wei S, Wei M, Cui H. Ginsenoside Rb1 Promotes Motor Functional Recovery and Axonal Regeneration in Post-stroke Mice through cAMP/PKA/CREB Signaling Pathway. Brain Res Bull 2019; 154:51-60. [PMID: 31715311 DOI: 10.1016/j.brainresbull.2019.10.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 09/29/2019] [Accepted: 10/19/2019] [Indexed: 11/29/2022]
Abstract
The central nervous system (CNS) has a poor self-repairing capability after injury because of the inhibition of axonal regeneration by many myelin-associated inhibitory factors. Therefore, ischemic stroke usually leads to disability. Previous studies reported that Ginsenoside Rb1 (GRb1) plays a role in neuronal protection in acute phase after ischemic stroke, but its efficacy in post-stroke and the underlying mechanism are not clear. Recent evidences demonstrated GRb1 promotes neurotransmitter release through the cAMP-depend protein kinase A (PKA) pathway, which is related to axonal regeneration. The present study aimed to determine whether GRb1 improves long-term motor functional recovery and promotes cortical axon regeneration in post-stroke. Adult male C57BL/6 mice were subjected to distal middle cerebral artery occlusion (dMCAO). GRb1 solution (5 mg/ml) or equal volume of normal saline was injected intraperitoneally for the first time at 24 h after surgery, and then daily injected until day 14. Day 3, 7, 14 and 28 after dMCAO were used as observation time points. Motor functional recovery was assessed with Rota-rod test and grid walking task. The expression of growth-associated protein 43 (GAP43) and biotinylated dextran amine (BDA) was measured to evaluate axonal regeneration. The levels of cyclic AMP (cAMP) and PKA were measured by Elisa, PKAc and phosphorylated cAMP response element protein (pCREB) were determined by western blot. Our results shown that GRb1 treatment improved motor function and increased the expression of GAP43 and BDA in ipsilesional and contralateral cortex. GRb1 significantly elevated cAMP and PKA, increased the protein expression of PKAc and pCREB. However, the effects of GRb1 were eliminated by H89 intervention (a PKA inhibitor). These results suggested that GRb1 improved functional recovery in post-stroke by stimulating axonal regeneration and brain repair. The underlying mechanism might be up-regulating the expression of cAMP/PKA/CREB pathway.
Collapse
Affiliation(s)
- Xuan Gao
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, PR China
| | - Xiangjian Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, PR China; Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, PR China.
| | - Lili Cui
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, PR China; Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, PR China
| | - Rong Chen
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, PR China; Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, PR China
| | - Cong Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, PR China
| | - Jing Xue
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, PR China
| | - Lan Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, PR China
| | - Weiliang He
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, PR China
| | - Jiamin Li
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, PR China
| | - Shanshan Wei
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, PR China
| | - Mengmeng Wei
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, PR China
| | - Hemei Cui
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, PR China
| |
Collapse
|
4
|
Divergent Regulation of Actin Dynamics and Megakaryoblastic Leukemia-1 and -2 (Mkl1/2) by cAMP in Endothelial and Smooth Muscle Cells. Sci Rep 2017. [PMID: 28623279 PMCID: PMC5473867 DOI: 10.1038/s41598-017-03337-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Proliferation and migration of vascular smooth muscle cells (VSMCs) or endothelial cell (ECs) promote or inhibit, respectively, restenosis after angioplasty, vein graft intimal thickening and atherogenesis. Here we investigated the effects of cAMP-induced cytoskeletal remodelling on the serum response factor (SRF) co-factors Megakaryoblastic Leukemia-1 and -2 (MKL1 and MKL2) and their role in controlling VSMC and EC proliferation and migration. Elevation of cAMP using forskolin, dibutyryl-cAMP (db-cAMP), BAY60-6583 or Cicaprost induced rapid cytoskeleton remodelling and inhibited proliferation and migration in VSMCs but not EC. Furthermore, elevated cAMP inhibited mitogen-induced nuclear-translocation of MKL1 and MKL2 in VSMCs but not ECs. Forskolin also significantly inhibited serum response factor (SRF)-dependent reporter gene (SRE-LUC) activity and mRNA expression of pro-proliferative and pro-migratory MKL1/2 target genes in VSMCs but not in ECs. In ECs, MKL1 was constitutively nuclear and MKL2 cytoplasmic, irrespective of mitogens or cAMP. Pharmacological or siRNA inhibition of MKL1 significantly inhibited the proliferation and migration of VSMC and EC. Our new data identifies and important contribution of MKL1/2 to explaining the strikingly different response of VSMCs and ECs to cAMP elevation. Elucidation of these pathways promises to identify targets for specific inhibition of VSMC migration and proliferation.
Collapse
|
5
|
Gerarduzzi C, He Q, Zhai B, Antoniou J, Di Battista JA. Prostaglandin E2-Dependent Phosphorylation of RAS Inhibition 1 (RIN1) at Ser 291 and 292 Inhibits Transforming Growth Factor-β-Induced RAS Activation Pathway in Human Synovial Fibroblasts: Role in Cell Migration. J Cell Physiol 2016; 232:202-15. [PMID: 27137893 DOI: 10.1002/jcp.25412] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/28/2016] [Indexed: 12/27/2022]
Abstract
Prostaglandin E2 (PGE2 )-stimulated G-protein-coupled receptor (GPCR) activation inhibits pro-fibrotic TGFβ-dependent stimulation of human fibroblast to myofibroblast transition (FMT), though the precise molecular mechanisms are not fully understood. In the present study, we describe the PGE2 -dependent suppression and reversal of TGFβ-induced events such as α-sma expression, stress fiber formation, and Ras/Raf/ERK/MAPK pathway-dependent activation of myofibroblast migration. In order to elucidate post-ligand-receptor signaling pathways, we identified a predominant PKA phosphorylation motif profile in human primary fibroblasts after treatment with exogenous PGE2 (EC50 30 nM, Vmax 100 nM), mimicked by the adenyl cyclase activator forskolin (EC50 5 μM, Vmax 10 μM). We used a global phosphoproteomic approach to identify a 2.5-fold difference in PGE2 -induced phosphorylation of proteins containing the PKA motif. Deducing the signaling pathway of our migration data, we identified Ras inhibitor 1 (RIN1) as a substrate, whereby PGE2 induced its phosphorylation at Ser291 and at Ser292 by a 5.4- and 4.8-fold increase, respectively. In a series of transient and stable over expression studies in HEK293T and HeLa cells using wild-type (wt) and mutant RIN1 (Ser291/292Ala) or Ras constructs and siRNA knock-down experiments, we showed that PGE2 -dependent phosphorylation of RIN1 resulted in the abrogation of TGFβ-induced Ras/Raf signaling activation and subsequent downstream blockade of cellular migration, emphasizing the importance of such phosphosites in PGE2 suppression of wound closure. Overexpression experiments in tandem with pull-down assays indicated that specific Ser291/292 phosphorylation of RIN1 favored binding to activated Ras. In principal, understanding PGE2 -GPCR activated signaling pathways mitigating TGFβ-induced fibrosis may lead to more evidence-based treatments against the disease. J. Cell. Physiol. 232: 202-215, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Casimiro Gerarduzzi
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts. .,Departments of Experimental Medicine, McGill University, Montréal, QC, Canada.
| | - QingWen He
- Departments of Medicine and Experimental Medicine, McGill University, Montréal, QC, Canada
| | - Beibei Zhai
- Departments of Experimental Medicine, McGill University, Montréal, QC, Canada
| | - John Antoniou
- Department of Orthopaedic Surgery, Jewish General Hospital, Montréal, QC, Canada
| | - John A Di Battista
- Departments of Medicine and Experimental Medicine, McGill University, Montréal, QC, Canada
| |
Collapse
|
6
|
Kimura TE, Duggirala A, Smith MC, White S, Sala-Newby GB, Newby AC, Bond M. The Hippo pathway mediates inhibition of vascular smooth muscle cell proliferation by cAMP. J Mol Cell Cardiol 2016; 90:1-10. [PMID: 26625714 PMCID: PMC4727789 DOI: 10.1016/j.yjmcc.2015.11.024] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 11/05/2015] [Accepted: 11/20/2015] [Indexed: 12/30/2022]
Abstract
AIMS Inhibition of vascular smooth muscle cell (VSMC) proliferation by intracellular cAMP prevents excessive neointima formation and hence angioplasty restenosis and vein-graft failure. These protective effects are mediated via actin-cytoskeleton remodelling and subsequent regulation of gene expression by mechanisms that are incompletely understood. Here we investigated the role of components of the growth-regulatory Hippo pathway, specifically the transcription factor TEAD and its co-factors YAP and TAZ in VSMC. METHODS AND RESULTS Elevation of cAMP using forskolin, dibutyryl-cAMP or the physiological agonists, Cicaprost or adenosine, significantly increased phosphorylation and nuclear export YAP and TAZ and inhibited TEAD-luciferase report gene activity. Similar effects were obtained by inhibiting RhoA activity with C3-transferase, its downstream kinase, ROCK, with Y27632, or actin-polymerisation with Latrunculin-B. Conversely, expression of constitutively-active RhoA reversed the inhibitory effects of forskolin on TEAD-luciferase. Forskolin significantly inhibited the mRNA expression of the pro-mitogenic genes, CCN1, CTGF, c-MYC and TGFB2 and this was reversed by expression of constitutively-active YAP or TAZ phospho-mutants. Inhibition of YAP and TAZ function with RNAi or Verteporfin significantly reduced VSMC proliferation. Furthermore, the anti-mitogenic effects of forskolin were reversed by overexpression of constitutively-active YAP or TAZ. CONCLUSION Taken together, these data demonstrate that cAMP-induced actin-cytoskeleton remodelling inhibits YAP/TAZ-TEAD dependent expression of pro-mitogenic genes in VSMC. This mechanism contributes novel insight into the anti-mitogenic effects of cAMP in VSMC and suggests a new target for intervention.
Collapse
Affiliation(s)
- Tomomi E Kimura
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol BS2 8HW, UK
| | - Aparna Duggirala
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol BS2 8HW, UK
| | - Madeleine C Smith
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol BS2 8HW, UK
| | - Stephen White
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol BS2 8HW, UK
| | - Graciela B Sala-Newby
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol BS2 8HW, UK
| | - Andrew C Newby
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol BS2 8HW, UK
| | - Mark Bond
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol BS2 8HW, UK.
| |
Collapse
|
7
|
SCYL2 Protects CA3 Pyramidal Neurons from Excitotoxicity during Functional Maturation of the Mouse Hippocampus. J Neurosci 2015. [PMID: 26203146 DOI: 10.1523/jneurosci.2056-14.2015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Neuronal death caused by excessive excitatory signaling, excitotoxicity, plays a central role in neurodegenerative disorders. The mechanisms regulating this process, however, are still incompletely understood. Here we show that the coated vesicle-associated kinase SCYL2/CVAK104 plays a critical role for the normal functioning of the nervous system and for suppressing excitotoxicity in the developing hippocampus. Targeted disruption of Scyl2 in mice caused perinatal lethality in the vast majority of newborn mice and severe sensory-motor deficits in mice that survived to adulthood. Consistent with a neurogenic origin of these phenotypes, neuron-specific deletion of Scyl2 also caused perinatal lethality in the majority of newborn mice and severe neurological defects in adult mice. The neurological deficits in these mice were associated with the degeneration of several neuronal populations, most notably CA3 pyramidal neurons of the hippocampus, which we analyzed in more detail. The loss of CA3 neurons occurred during the functional maturation of the hippocampus and was the result of a BAX-dependent apoptotic process. Excessive excitatory signaling was present at the onset of degeneration, and inhibition of excitatory signaling prevented the degeneration of CA3 neurons. Biochemical fractionation reveals that Scyl2-deficient mice have an altered composition of excitatory receptors at synapses. Our findings demonstrate an essential role for SCYL2 in regulating neuronal function and survival and suggest a role for SCYL2 in regulating excitatory signaling in the developing brain. Significance statement: Here we examine the in vivo function of SCYL2, an evolutionarily conserved and ubiquitously expressed protein pseudokinase thought to regulate protein trafficking along the secretory pathway, and demonstrate its importance for the normal functioning of the nervous system and for suppressing excitatory signaling in the developing brain. Together with recent studies demonstrating a role of SCYL1 in preventing motor neuron degeneration, our findings clearly establish the SCY1-like family of protein pseudokinases as key regulators of neuronal function and survival.
Collapse
|
8
|
Blirando K, Blaise R, Gorodnaya N, Rouxel C, Meilhac O, Vincent P, Limon I. The stellate vascular smooth muscle cell phenotype is induced by IL-1β via the secretion of PGE2 and subsequent cAMP-dependent protein kinase A activation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:3235-47. [PMID: 26403276 DOI: 10.1016/j.bbamcr.2015.09.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 09/11/2015] [Accepted: 09/14/2015] [Indexed: 12/12/2022]
Abstract
Atherosclerosis development is associated with morphological changes to intimal cells, leading to a stellate cell phenotype. In this study, we aimed to determine whether and how key pro-atherogenic cytokines present in atherosclerotic plaques (IL-1β, TNFα and IFNγ) could induce this phenotype, as these molecules are known to trigger the transdifferentiation of vascular smooth muscle cells (VSMCs). We found that, IL-1β was the only major inflammatory mediator tested capable of inducing a stellate morphology in VSMCs. This finding was confirmed by staining for F-actin and vinculin at focal adhesions, as these two markers were disrupted only by IL-1β. We then investigated the possible association of this IL-1β-dependent change in morphology with an increase in intracellular cAMP concentration ([cAMP]), using the FRET-based biosensor for cAMP (T)Epac(VV). Experiments in the presence of IL-1β or medium conditioned by IL-1β-treated VSMCs and pharmacological tools demonstrated that the long-term increase in intracellular cAMP concentration was induced by the secretion of an autocrine/paracrine mediator, prostaglandin E₂(PGE₂), acting through the EP4 receptor. Finally, by knocking down the expression of the regulatory subunit PKAR1α, thereby reproducing the effects of IL-1β and PGE₂ on VSMCs, we demonstrated the contribution of PKA activity to the observed behavior of VSMCs.
Collapse
Affiliation(s)
- Karl Blirando
- Sorbonne Universités, UPMC Univ Paris 06CNRS UMR 8256 B2A, IBPS, F-75005, Paris, France
| | - Régis Blaise
- Sorbonne Universités, UPMC Univ Paris 06CNRS UMR 8256 B2A, IBPS, F-75005, Paris, France
| | - Natalia Gorodnaya
- Sorbonne Universités, UPMC Univ Paris 06CNRS UMR 8256 B2A, IBPS, F-75005, Paris, France
| | - Clotilde Rouxel
- Sorbonne Universités, UPMC Univ Paris 06CNRS UMR 8256 B2A, IBPS, F-75005, Paris, France
| | - Olivier Meilhac
- Inserm U1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI) CYROI, 2, rue Maxime Rivière, 97490 Sainte Clotilde, La Réunion, France
| | - Pierre Vincent
- Sorbonne Universités, UPMC Univ Paris 06CNRS UMR 8256 B2A, IBPS, F-75005, Paris, France
| | - Isabelle Limon
- Sorbonne Universités, UPMC Univ Paris 06CNRS UMR 8256 B2A, IBPS, F-75005, Paris, France.
| |
Collapse
|
9
|
Wang Y, Kunit T, Ciotkowska A, Rutz B, Schreiber A, Strittmatter F, Waidelich R, Liu C, Stief CG, Gratzke C, Hennenberg M. Inhibition of prostate smooth muscle contraction and prostate stromal cell growth by the inhibitors of Rac, NSC23766 and EHT1864. Br J Pharmacol 2015; 172:2905-17. [PMID: 25631101 PMCID: PMC4439884 DOI: 10.1111/bph.13099] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 01/19/2015] [Accepted: 01/20/2015] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND AND PURPOSE Medical therapy of lower urinary tract symptoms (LUTS) suggestive of benign prostatic hyperplasia (BPH) targets smooth muscle contraction in the prostate, or prostate growth. However, current therapeutic options are insufficient. Here, we investigated the role of Rac in the control of smooth muscle tone in human prostates and growth of prostate stromal cells. EXPERIMENTAL APPROACH Experiments were performed using human prostate tissues from radical prostatectomy and cultured stromal cells (WPMY-1). Expression of Rac was examined by Western blot and fluorescence staining. Effects of Rac inhibitors (NSC23766 and EHT1864) on contractility were assessed in the organ bath. The effects of Rac inhibitors were assessed by pull-down, cytotoxicity using a cell counting kit, cytoskeletal organization by phalloidin staining and cell growth using an 5-ethynyl-2'-deoxyuridine assay. KEY RESULTS Expression of Rac1-3 was observed in prostate samples from each patient. Immunoreactivity for Rac1-3 was observed in the stroma, where it colocalized with the smooth muscle marker, calponin. NSC23766 and EHT1864 significantly reduced contractions of prostate strips induced by noradrenaline, phenylephrine or electrical field stimulation. NSC23766 and EHT1864 inhibited Rac activity in WPMY-1 cells. Survival of WPMY-1 cells ranged between 64 and 81% after incubation with NSC23766 (50 or 100 μM) or EHT1864 (25 μM) for 24 h. NSC23766 and EHT1864 induced cytoskeletal disorganization in WPMY-1 cells. Both inhibitors impaired the growth of WPMY-1 cells. CONCLUSIONS AND IMPLICATIONS Rac may be a link connecting the control of prostate smooth muscle tone with proliferation of smooth muscle cells. Improvements in LUTS suggestive of BPH by Rac inhibitors appears possible.
Collapse
Affiliation(s)
- Y Wang
- Department of Urology, Ludwig Maximilian UniversityMunich, Germany
- Department of Urology, Zhujiang Hospital, Southern Medical UniversityGuangzhou, China
| | - T Kunit
- Department of Urology, Ludwig Maximilian UniversityMunich, Germany
- University Hospital for Urology and AndrologySalzburg, Austria
| | - A Ciotkowska
- Department of Urology, Ludwig Maximilian UniversityMunich, Germany
| | - B Rutz
- Department of Urology, Ludwig Maximilian UniversityMunich, Germany
| | - A Schreiber
- Department of Urology, Ludwig Maximilian UniversityMunich, Germany
| | - F Strittmatter
- Department of Urology, Ludwig Maximilian UniversityMunich, Germany
| | - R Waidelich
- Department of Urology, Ludwig Maximilian UniversityMunich, Germany
| | - C Liu
- Department of Urology, Zhujiang Hospital, Southern Medical UniversityGuangzhou, China
| | - C G Stief
- Department of Urology, Ludwig Maximilian UniversityMunich, Germany
| | - C Gratzke
- Department of Urology, Ludwig Maximilian UniversityMunich, Germany
| | - M Hennenberg
- Department of Urology, Ludwig Maximilian UniversityMunich, Germany
| |
Collapse
|
10
|
Duggirala A, Kimura TE, Sala-Newby GB, Johnson JL, Wu YJ, Newby AC, Bond M. cAMP-induced actin cytoskeleton remodelling inhibits MKL1-dependent expression of the chemotactic and pro-proliferative factor, CCN1. J Mol Cell Cardiol 2015; 79:157-68. [PMID: 25446180 PMCID: PMC4312355 DOI: 10.1016/j.yjmcc.2014.11.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 10/24/2014] [Accepted: 11/12/2014] [Indexed: 12/17/2022]
Abstract
Elevation of intracellular cAMP concentration has numerous vascular protective effects that are in part mediated via actin cytoskeleton-remodelling and subsequent regulation of gene expression. However, the mechanisms are incompletely understood. Here we investigated whether cAMP-induced actin-cytoskeleton remodelling modulates VSMC behaviour by inhibiting expression of CCN1. In cultured rat VSMC, CCN1-silencing significantly inhibited BrdU incorporation and migration in a wound healing assay. Recombinant CCN1 enhanced chemotaxis in a Boyden chamber. Adding db-cAMP, or elevating cAMP using forskolin, significantly inhibited CCN1 mRNA and protein expression in vitro; transcriptional regulation was demonstrated by measuring pre-spliced CCN1 mRNA and CCN1-promoter activity. Forskolin also inhibited CCN1 expression in balloon injured rat carotid arteries in vivo. Inhibiting RhoA activity, which regulates actin-polymerisation, by cAMP-elevation or pharmacologically with C3-transferase, or inhibiting its downstream kinase, ROCK, with Y27632, significantly inhibited CCN1 expression. Conversely, expression of constitutively active RhoA reversed the inhibitory effects of forskolin on CCN1 mRNA. Furthermore, CCN1 mRNA levels were significantly decreased by inhibiting actin-polymerisation with latrunculin B or increased by stimulating actin-polymerisation with Jasplakinolide. We next tested the role of the actin-dependent SRF co-factor, MKL1, in CCN1 expression. Forskolin inhibited nuclear translocation of MKL1 and binding of MKL1 to the CCN1 promoter. Constitutively-active MKL1 enhanced basal promoter activity of wild-type but not SRE-mutated CCN1; and prevented forskolin inhibition. Furthermore, pharmacological MKL-inhibition with CCG-1423 significantly inhibited CCN1 promoter activity as well as mRNA and protein expression. Our data demonstrates that cAMP-induced actin-cytoskeleton remodelling regulates expression of CCN1 through MKL1: it highlights a novel cAMP-dependent mechanism controlling VSMC behaviour.
Collapse
Affiliation(s)
- Aparna Duggirala
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol, BS2 8HW, UK
| | - Tomomi E Kimura
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol, BS2 8HW, UK
| | - Graciela B Sala-Newby
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol, BS2 8HW, UK
| | - Jason L Johnson
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol, BS2 8HW, UK
| | - Yih-Jer Wu
- Department of Medicine, MacKay Medical College, New Taipei, Taiwan; Cardiovascular Division, Department of Internal Medicine, MacKay Memorial Hospital, New Taipei, Taiwan
| | - Andrew C Newby
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol, BS2 8HW, UK
| | - Mark Bond
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol, BS2 8HW, UK.
| |
Collapse
|
11
|
Gerarduzzi C, He Q, Antoniou J, Di Battista JA. Quantitative phosphoproteomic analysis of signaling downstream of the prostaglandin e2/g-protein coupled receptor in human synovial fibroblasts: potential antifibrotic networks. J Proteome Res 2014; 13:5262-80. [PMID: 25223752 DOI: 10.1021/pr500495s] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The Prostaglandin E2 (PGE2) signaling mechanism within fibroblasts is of growing interest as it has been shown to prevent numerous fibrotic features of fibroblast activation with limited evidence of downstream pathways. To understand the mechanisms of fibroblasts producing tremendous amounts of PGE2 with autocrine effects, we apply a strategy of combining a wide-screening of PGE2-induced kinases with quantitative phosphoproteomics. Our large-scale proteomic approach identified a PKA signal transmitted through phosphorylation of its substrates harboring the R(R/X)X(S*/T*) motif. We documented 115 substrates, of which 72 had 89 sites with a 2.5-fold phosphorylation difference in PGE2-treated cells than in untreated cells, where approximately half of such sites were defined as being novel. They were compiled by networking software to focus on highlighted activities and to associate them with a functional readout of fibroblasts. The substrates were associated with a variety of cellular functions including cytoskeletal structures (migration/motility), regulators of G-protein coupled receptor function, protein kinases, and transcriptional/translational regulators. For the first time, we extended the PGE2 pathway into an elaborate network of interconnecting phosphoproteins, providing vital information to a once restricted signalosome. These data provide new insights into eicosanoid-initiated cell signaling with regards to the regulation of fibroblast activation and the identification of new targets for evidenced-based pharmacotherapy against fibrosis.
Collapse
Affiliation(s)
- Casimiro Gerarduzzi
- Department of Experimental Medicine, McGill University , 687 Pine Avenue West, Montreal, Quebec H3A 1A1, Canada
| | | | | | | |
Collapse
|
12
|
Kimura TE, Duggirala A, Hindmarch CCT, Hewer RC, Cui MZ, Newby AC, Bond M. Inhibition of Egr1 expression underlies the anti-mitogenic effects of cAMP in vascular smooth muscle cells. J Mol Cell Cardiol 2014; 72:9-19. [PMID: 24534707 PMCID: PMC4051994 DOI: 10.1016/j.yjmcc.2014.02.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 01/30/2014] [Accepted: 02/01/2014] [Indexed: 01/23/2023]
Abstract
AIMS Cyclic AMP inhibits vascular smooth muscle cell (VSMC) proliferation which is important in the aetiology of numerous vascular diseases. The anti-mitogenic properties of cAMP in VSMC are dependent on activation of protein kinase A (PKA) and exchange protein activated by cAMP (EPAC), but the mechanisms are unclear. METHODS AND RESULTS Selective agonists of PKA and EPAC synergistically inhibited Egr1 expression, which was essential for VSMC proliferation. Forskolin, adenosine, A2B receptor agonist BAY60-6583 and Cicaprost also inhibited Egr1 expression in VSMC but not in endothelial cells. Inhibition of Egr1 by cAMP was independent of cAMP response element binding protein (CREB) activity but dependent on inhibition of serum response element (SRE) activity. SRF binding to the Egr1 promoter was not modulated by cAMP stimulation. However, Egr1 expression was dependent on the SRF co-factors Elk1 and 4 but independent of MAL. Inhibition of SRE-dependent Egr1 expression was due to synergistic inhibition of Rac1 activity by PKA and EPAC, resulting in rapid cytoskeleton remodelling and nuclear export of ERK1/2. This was associated with de-phosphorylation of the SRF co-factor Elk1. CONCLUSION cAMP inhibits VSMC proliferation by rapidly inhibiting Egr1 expression. This occurs, at least in part, via inhibition of Rac1 activity leading to rapid actin-cytoskeleton remodelling, nuclear export of ERK1/2, impaired Elk1-phosphorylation and inhibition of SRE activity. This identifies one of the earliest mechanisms underlying the anti-mitogenic effects of cAMP in VSMC but not in endothelial cells, making it an attractive target for selective inhibition of VSMC proliferation.
Collapse
MESH Headings
- Adenosine/pharmacology
- Aminopyridines/pharmacology
- Animals
- Cell Proliferation/drug effects
- Colforsin/pharmacology
- Cyclic AMP/pharmacology
- Cyclic AMP Response Element-Binding Protein/genetics
- Cyclic AMP Response Element-Binding Protein/metabolism
- Cyclic AMP-Dependent Protein Kinases/genetics
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Early Growth Response Protein 1/antagonists & inhibitors
- Early Growth Response Protein 1/genetics
- Early Growth Response Protein 1/metabolism
- Epoprostenol/analogs & derivatives
- Epoprostenol/pharmacology
- Gene Expression Regulation
- Guanine Nucleotide Exchange Factors/genetics
- Guanine Nucleotide Exchange Factors/metabolism
- Human Umbilical Vein Endothelial Cells/cytology
- Human Umbilical Vein Endothelial Cells/drug effects
- Human Umbilical Vein Endothelial Cells/metabolism
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Organ Specificity
- Primary Cell Culture
- Protein Binding
- Rats
- Rats, Sprague-Dawley
- Serum Response Factor/genetics
- Serum Response Factor/metabolism
- Signal Transduction
- ets-Domain Protein Elk-1/genetics
- ets-Domain Protein Elk-1/metabolism
- rac1 GTP-Binding Protein/genetics
- rac1 GTP-Binding Protein/metabolism
Collapse
Affiliation(s)
- Tomomi E Kimura
- Bristol Heart Institute, University of Bristol, Bristol BS2 8HW, UK
| | - Aparna Duggirala
- Bristol Heart Institute, University of Bristol, Bristol BS2 8HW, UK
| | - Charles C T Hindmarch
- Laboratory for Integrative Neuroscience & Endocrinology, University of Bristol, Bristol BS2 8HW, UK; University of Malaya, Department of Physiology, Faculty of Medicine, Kuala Lumpur, Malaysia
| | - Richard C Hewer
- Bristol Heart Institute, University of Bristol, Bristol BS2 8HW, UK
| | - Mei-Zhen Cui
- Department of Pathobiology, College of Veterinary Medicine, The University of Tennessee, USA
| | - Andrew C Newby
- Bristol Heart Institute, University of Bristol, Bristol BS2 8HW, UK
| | - Mark Bond
- Bristol Heart Institute, University of Bristol, Bristol BS2 8HW, UK.
| |
Collapse
|
13
|
Sun Z, Huang S, Li Z, Meininger GA. Zyxin is involved in regulation of mechanotransduction in arteriole smooth muscle cells. Front Physiol 2012; 3:472. [PMID: 23267329 PMCID: PMC3526782 DOI: 10.3389/fphys.2012.00472] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 12/03/2012] [Indexed: 01/16/2023] Open
Abstract
Zyxin is a focal adhesion protein that has been implicated in the modulation of cell adhesion and motility, and is hypothesized to be a mechano-sensor in integrin-mediated responses to mechanical force. To test the functional role of zyxin in the mechanotransduction of microvascular smooth muscle cells (VSMC), we utilized atomic force microscopy (AFM) to apply localized pulling forces to VSMC through a fibronectin (FN) focal adhesion induced by a FN-coated bead on cell surface. Application of force with the AFM induced an increase of zyxin accumulation at the site of the FN-bead focal adhesion that accompanied the VSMC contractile response. Whereas, reduction of zyxin expression by using a zyxin-shRNA construct abolished the VSMC contractile response to AFM pulling forces, even though the zyxin-silenced VSMCs displayed increased adhesion to FN in both AFM adhesion assays and cell adhesion assays. The reduced zyxin expression significantly impaired cell spreading and reorganization of the actin cytoskeleton that could indicate a possible underlying reason for the loss of a contractile response to mechanical force. Consistent with these observations, in zyxin-silenced VSMC, we also observed a reduced expression of Rac1, which plays an important role in the actin reorganization in VSMC, but increased thyroid receptor-interacting proteins (TRIP6) and FAK expression, the latter being a major protein that promote cell adhesion. In conclusion, these data support an important enabling role for zyxin in VSMCs ability to mechanically respond to applied force.
Collapse
Affiliation(s)
- Zhe Sun
- Dalton Cardiovascular Research Center and Department of Medical Pharmacology and Physiology, University of Missouri Columbia, MO, USA
| | | | | | | |
Collapse
|
14
|
Palamà IE, Coluccia AML, Gigli G, Riehle M. Modulation of alignment and differentiation of skeletal myoblasts by biomimetic materials. Integr Biol (Camb) 2012; 4:1299-309. [DOI: 10.1039/c2ib20133j] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
15
|
Fuseler JW, Valarmathi MT. Modulation of the migration and differentiation potential of adult bone marrow stromal stem cells by nitric oxide. Biomaterials 2011; 33:1032-43. [PMID: 22071099 DOI: 10.1016/j.biomaterials.2011.10.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Accepted: 10/11/2011] [Indexed: 11/29/2022]
Abstract
Nitric oxide (NO) is a diffusible free radical, which serves as a pluripotent intracellular messenger in numerous cell systems. NO has been demonstrated to regulate actin dependent cellular functions and functions as a putative inductive agent in directing stem cells differentiation. In this study, we investigated the effect of exogenous NO on the kinetics of movement and morphological changes in adult bone marrow stromal cells (BMSCs) in a wound healing model of cellular migration. Cellular migration and morphological changes were determined by measurement of changes in the area and fractal dimension of BMSCs monolayer as a function of time in the presence of an NO donor (S-Nitroso-N-Acetyl-D,L-Penicillamine, SNAP) compared to untreated BMSCs. Response of the BMSCs' actin cytoskeleton and desmin to NO was assessed by determining changes in their integrated optical density (IOD) and fractal dimension at 24 h and 7 days. NO suppressed BMSCs' migration accompanied by a reduction in cell size, with maintenance of their stellate to polygonal morphology. In response to NO, the actin cytoskeleton expressed an increase in randomness but maintained a constant amount of F-actin relative to the cell size. The presence of NO also induced an increase in randomly organized cytoplasmic desmin. These data suggest that NO has an apparent inductive effect on adult BMSCs and is capable of initiating phenotypic change at the gross cellular, cytoskeletal and molecular levels. It is apparent, however, that additional factors or conditions are required to further drive the differentiation of adult BMSCs into specific phenotypes, such as cardiomyocytes.
Collapse
Affiliation(s)
- John W Fuseler
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | | |
Collapse
|
16
|
β-Adrenergic inhibition of contractility in L6 skeletal muscle cells. PLoS One 2011; 6:e22304. [PMID: 21829455 PMCID: PMC3145637 DOI: 10.1371/journal.pone.0022304] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Accepted: 06/23/2011] [Indexed: 12/24/2022] Open
Abstract
The β-adrenoceptors (β-ARs) control many cellular processes. Here, we show that β-ARs inhibit calcium depletion-induced cell contractility and subsequent cell detachment of L6 skeletal muscle cells. The mechanism underlying the cell detachment inhibition was studied by using a quantitative cell detachment assay. We demonstrate that cell detachment induced by depletion of extracellular calcium is due to myosin- and ROCK-dependent contractility. The β-AR inhibition of L6 skeletal muscle cell detachment was shown to be mediated by the β2-AR and increased cAMP but was surprisingly not dependent on the classical downstream effectors PKA or Epac, nor was it dependent on PKG, PI3K or PKC. However, inhibition of potassium channels blocks the β2-AR mediated effects. Furthermore, activation of potassium channels fully mimicked the results of β2-AR activation. In conclusion, we present a novel finding that β2-AR signaling inhibits contractility and thus cell detachment in L6 skeletal muscle cells by a cAMP and potassium channel dependent mechanism.
Collapse
|
17
|
Yao M, Roberts DD, Isenberg JS. Thrombospondin-1 inhibition of vascular smooth muscle cell responses occurs via modulation of both cAMP and cGMP. Pharmacol Res 2011; 63:13-22. [PMID: 20971192 PMCID: PMC3026097 DOI: 10.1016/j.phrs.2010.10.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 10/14/2010] [Accepted: 10/14/2010] [Indexed: 12/21/2022]
Abstract
Nitric oxide (NO) drives pro-survival responses in vascular cells and limits platelet adhesion, enhancing blood flow and minimizing thrombosis. The matricellular protein thrombospondin-1 (TSP1), through interaction with its receptor CD47, inhibits soluble guanylyl cyclase (sGC) activation by NO in vascular cells. In vascular smooth muscle cells (VSMCs) both intracellular cGMP and cAMP regulate adhesion, contractility, proliferation, and migration. cGMP can regulate cAMP through feedback control of hydrolysis. Inhibition of the cAMP phosphodiesterase-4 selectively interfered with the ability of exogenous TSP1 to block NO-driven VSMC adhesion but not cGMP accumulation, suggesting that cAMP also contributes to VSMC regulation by TSP1. Inhibition of phosphodiesterase-4 was sufficient to elevate cAMP levels, and inhibiting guanylyl cyclase or phosphodiesterase-3, or adding exogenous TSP1 reversed this increase in cAMP. Thus, TSP1 regulates VSMC cAMP levels in part via cGMP-dependent inhibition of phosphodiesterase-3. Additionally basal cAMP levels were consistently elevated in both VSMCs and skeletal muscle from TSP1 null mice, and treating null cells with exogenous TSP1 suppressed cAMP levels to those of wild type cells. TSP1 inhibited both forskolin and isoproterenol stimulated increases in cAMP in VSMCs. TSP1 also abrogated forskolin and isoproterenol stimulated vasodilation. Consistent with its ability to directly limit adenylyl cyclase-activated vasodilation, TSP1 also limited cAMP-induced dephosphorylation of myosin light chain-2. These findings demonstrate that TSP1 limits both cGMP and cAMP signaling pathways and functional responses in VSMCs and arteries, by both phosphodiesterase-dependent cross talk between these second messengers and by inhibition of adenylyl cyclase activation.
Collapse
MESH Headings
- Actins/metabolism
- Animals
- CD47 Antigen/metabolism
- Cells, Cultured
- Cyclic AMP/metabolism
- Cyclic GMP/metabolism
- Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism
- Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism
- Endothelial Cells/metabolism
- Guanylate Cyclase
- Humans
- Hydrolysis
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Skeletal/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Nitric Oxide/metabolism
- Phosphodiesterase Inhibitors/pharmacology
- Rats
- Receptors, Cytoplasmic and Nuclear
- Signal Transduction/drug effects
- Soluble Guanylyl Cyclase
- Thrombospondin 1/deficiency
- Thrombospondin 1/genetics
- Thrombospondin 1/metabolism
- Vasodilation/drug effects
- Vasodilator Agents/pharmacology
Collapse
Affiliation(s)
- Mingyi Yao
- Vascular Medicine Institute of the University of Pittsburgh, Pittsburgh, PA
| | - David D. Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jeff S. Isenberg
- Vascular Medicine Institute of the University of Pittsburgh, Pittsburgh, PA
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
18
|
Hewer RC, Sala-Newby GB, Wu YJ, Newby AC, Bond M. PKA and Epac synergistically inhibit smooth muscle cell proliferation. J Mol Cell Cardiol 2010; 50:87-98. [PMID: 20971121 PMCID: PMC3093616 DOI: 10.1016/j.yjmcc.2010.10.010] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 10/08/2010] [Accepted: 10/11/2010] [Indexed: 12/14/2022]
Abstract
Cyclic AMP signalling promotes VSMC quiescence in healthy vessels and during vascular healing following injury. Cyclic AMP inhibits VSMC proliferation via mechanisms that are not fully understood. We investigated the role of PKA and Epac signalling on cAMP-induced inhibition of VSMC proliferation. cAMP-mediated growth arrest was PKA-dependent. However, selective PKA activation with 6-Benzoyl-cAMP did not inhibit VSMC proliferation, indicating a requirement for additional pathways. Epac activation using the selective cAMP analogue 8-CPT-2′-O-Me-cAMP, did not affect levels of hyperphosphorylated Retinoblastoma (Rb) protein, a marker of G1-S phase transition, or BrdU incorporation, despite activation of the Epac-effector Rap1. However, 6-Benzoyl-cAMP and 8-CPT-2′-O-Me-cAMP acted synergistically to inhibit Rb-hyperphosphorylation and BrdU incorporation, indicating that both pathways are required for growth inhibition. Consistent with this, constitutively active Epac increased Rap1 activity and synergised with 6-Benzoyl-cAMP to inhibit VSMC proliferation. PKA and Epac synergised to inhibit phosphorylation of ERK and JNK. Induction of stellate morphology, previously associated with cAMP-mediated growth arrest, was also dependent on activation of both PKA and Epac. Rap1 inhibition with Rap1GAP or siRNA silencing did not negate forskolin-induced inhibition of Rb-hyperphosphorylation, BrdU incorporation or stellate morphology. This data demonstrates for the first time that Epac synergises with PKA via a Rap1-independent mechanism to mediate cAMP-induced growth arrest in VSMC. This work highlights the role of Epac as a major player in cAMP-dependent growth arrest in VSMC.
Collapse
|
19
|
Miller IS, Lynch I, Dowling D, Dawson KA, Gallagher WM. Surface-induced cell signaling events control actin rearrangements and motility. J Biomed Mater Res A 2010; 93:493-504. [PMID: 19585567 DOI: 10.1002/jbm.a.32530] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Understanding the interrelationship between material surface properties and the biological response to such materials remains a fundamental scientific challenge, as well as being of considerable practical importance in medicine. Through the use of a homologous series of copolymers of increasing hydrophobicity, we aimed to illuminate the interplay between material surface hydrophobicity and signalling events within cells in contact with this model system. Extending previous work, we hereby unravel key pathways controlling cell motility and the formation of a stellate phenotype, following interaction with polymer-coated surfaces. We reveal a comparative increase in cellular motility with increasing surface hydrophilicity, conjoint with an arrest in cell cycle progression. We also show an anomalous turnover of actin within the cell as a function of changing surface hydrophobicity. Finally, we show that cyclic adenosine monophosphate may be an effector of the cellular phenotype, as its production is increased in response to changes in the surface properties. These results highlight important signaling events which control actin rearrangements and the subsequent motility and its effectors.
Collapse
Affiliation(s)
- Ian S Miller
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | | | | | |
Collapse
|
20
|
The T3SS effector EspT defines a new category of invasive enteropathogenic E. coli (EPEC) which form intracellular actin pedestals. PLoS Pathog 2009; 5:e1000683. [PMID: 20011125 PMCID: PMC2782363 DOI: 10.1371/journal.ppat.1000683] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Accepted: 11/05/2009] [Indexed: 02/07/2023] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) strains are defined as extracellular pathogens which nucleate actin rich pedestal-like membrane extensions on intestinal enterocytes to which they intimately adhere. EPEC infection is mediated by type III secretion system effectors, which modulate host cell signaling. Recently we have shown that the WxxxE effector EspT activates Rac1 and Cdc42 leading to formation of membrane ruffles and lamellipodia. Here we report that EspT-induced membrane ruffles facilitate EPEC invasion into non-phagocytic cells in a process involving Rac1 and Wave2. Internalized EPEC resides within a vacuole and Tir is localized to the vacuolar membrane, resulting in actin polymerization and formation of intracellular pedestals. To the best of our knowledge this is the first time a pathogen has been shown to induce formation of actin comets across a vacuole membrane. Moreover, our data breaks the dogma of EPEC as an extracellular pathogen and defines a new category of invasive EPEC. Enteropathogenic E. coli (EPEC) is an important diarrheal pathogen responsible for significant infant mortality in the developing world and is increasingly associated with sporadic outbreaks in the developed world. The virulence strategy of EPEC revolves around a conserved Type 3 secretion system (T3SS) which translocates bacterial effector proteins directly into host cells. EPEC is considered to be a non-invasive pathogen which intimately adheres to host cells and polymerizes actin rich pedestals on which extracellular bacteria rest. Recently we have identified the T3SS effector EspT which activates the mammalian Rho GTPases Rac1 and Cdc42, resulting in the formation of membrane ruffles and lamellipodia. In this study we dissect the signaling pathway utilized by EspT to nucleate membrane ruffles and demonstrate that these ruffles can promote EPEC invasion of host cells. Furthermore, we show that internalized EPEC are bound within a vacuole. We also report for the first time the ability of a bacterial pathogen to form actin comet tails across a vacuole membrane. In addition to providing novel insights into the subversion of cellular signaling by invasive pathogens, our study also breaks the long held dogma of EPEC as an extracellular pathogen and will have implications on how future EPEC infections are diagnosed and treated.
Collapse
|
21
|
Defawe OD, Kim S, Chen L, Huang D, Kenagy RD, Renné T, Walter U, Daum G, Clowes AW. VASP phosphorylation at serine239 regulates the effects of NO on smooth muscle cell invasion and contraction of collagen. J Cell Physiol 2009; 222:230-7. [PMID: 19798690 DOI: 10.1002/jcp.21942] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Nitric oxide triggers cGMP-dependent kinase-mediated phosphorylation of the actin regulator vasodilator-stimulated phosphoprotein (VASP) at residue serine239. The function of this phosphorylation for smooth muscle cell (SMC) adhesion, spreading, matrix contraction, and invasion is not well understood. We reconstituted VASP deficient SMC with wild-type VASP (wt-VASP) or VASP mutants that mimic "locked" serine239 phosphorylation (S239D-VASP) or "blocked" serine239 phosphorylation (S239A-VASP). Collagen gel contraction was reduced in S239D-VASP compared to S239A-VASP and wt-VASP expressing cells and nitric oxide (NO) stimulation decreased gel contraction of wt-VASP reconstituted SMC. Invasion of collagen was enhanced in S239D-VASP and NO-stimulated wild-type SMCs compared to S239A-VASP expressing cells. Expression of S239D-VASP impaired SMC attachment to collagen, reduced the number of membrane protrusions, and caused cell rounding compared to expression of S239A-VASP. Treatment of wt-VASP reconstituted SMCs with NO exerted similar effects as expression of S239D-VASP. As unstimulated cells were spreading on collagen S239A-VASP and wt-VASP localized to actin fibers whereas S239D-VASP was enriched in the cytosol. NO interferes with SMC invasion and contraction of collagen matrices. This requires phosphorylation of VASP on serine239, which reduces VASP binding to actin fibers. These findings support the conclusion that VASP phosphorylation at serine239 regulates cytoskeleton remodeling.
Collapse
Affiliation(s)
- Olivier D Defawe
- Department of Surgery, University of Washington, Seattle, Washington 98109, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Peters MF, Scott CW. Evaluating Cellular Impedance Assays for Detection of GPCR Pleiotropic Signaling and Functional Selectivity. ACTA ACUST UNITED AC 2009; 14:246-55. [DOI: 10.1177/1087057108330115] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
G-protein—coupled receptors can couple to different signal transduction pathways in different cell types (termed cell-specific signaling) and can activate different signaling pathways depending on the receptor conformation(s) stabilized by the activating ligand (functional selectivity). These concepts offer potential for developing pathway-specific drugs that increase efficacy and reduce side effects. Despite significant interest, functional selectivity has been difficult to exploit in drug discovery, in part due to the burden of multiple assays. Cellular impedance assays use an emerging technology that can qualitatively distinguish Gs, Gi/o, and Gq signaling in a single assay and is thereby suited for studying these pharmacological concepts. Cellular impedance confirmed cell-specific Gs and Gq coupling for the melanocortin-4 receptor and dual Gi and Gs signaling with the cannabinoid-1 (CB1) receptor. The balance of Gi versus Gs signaling depended on the cell line. In CB1-HEKs, Giand Gs-like responses combined to yield a novel impedance profile demonstrating the dynamic nature of these traces. Cellspecific signaling was observed with endogenous D1 receptor in U-2 cells and SK-N-MC cells, yet the pharmacological profile of partial and full agonists was similar in both cell lines. We conclude that the dynamic impedance profile encodes valuable relative signaling information and is sufficiently robust to help evaluate cell-specific signaling and functional selectivity. ( Journal of Biomolecular Screening 2009:246-255)
Collapse
Affiliation(s)
- Matthew F. Peters
- Lead Generation Department, AstraZeneca Pharmaceuticals LP, Wilmington, Delaware,
| | - Clay W. Scott
- Lead Generation Department, AstraZeneca Pharmaceuticals LP, Wilmington, Delaware
| |
Collapse
|
23
|
Wu YJ, Yeh HI, Hou CJY, Tsai CH, Newby AC, Bond M. Beyond Oncogenesis: The Role of S-Phase Kinase-Associated Protein-2 (SKP2) In Vascular Restenosis. INT J GERONTOL 2008. [DOI: 10.1016/s1873-9598(09)70004-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
24
|
Bulgin RR, Arbeloa A, Chung JCS, Frankel G. EspT triggers formation of lamellipodia and membrane ruffles through activation of Rac-1 and Cdc42. Cell Microbiol 2008; 11:217-29. [PMID: 19016787 PMCID: PMC2688677 DOI: 10.1111/j.1462-5822.2008.01248.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Subversion of the eukaryotic cell cytoskeleton is a virulence strategy employed by many bacterial pathogens. Due to the pivotal role of Rho GTPases in actin dynamics they are common targets of bacterial effector proteins and toxins. IpgB1, IpgB2 (Shigella), SifA, SifB (Salmonella) and Map and EspM (attaching and effacing pathogens) constitute a family of type III secretion system effectors that subverts small GTPase signalling pathways. In this study we identified and characterized EspT from Citrobacter rodentium that triggers formation of lamellipodia on Swiss 3T3 and membrane ruffles on HeLa cells, which are reminiscent of the membrane ruffles induced by IpgB1. Ectopic expression of EspT and IpgB1, but not EspM, resulted in a mitochondrial localization. Using dominant negative constructs we found that EspT-induced actin remodelling is dependent on GTP-bound Rac-1 and Cdc42 but not ELMO or Dock180, which are hijacked by IpgB1 in order to form a Rac-1 specific guanine nucleotide exchange factor. Using pull-down assays with the Rac-1 and Cdc42 binding domains of Pak and WASP we demonstrate that EspT is capable of activating both Rac-1 and Cdc42. These results suggest that EspT modulates the host cell cytoskeleton through coactivation of Rac-1 and Cdc42 by a distinct mechanism.
Collapse
Affiliation(s)
- Richard R Bulgin
- Centre for Molecular Microbiology and Infection, Division of Cell and Molecular Biology, Imperial College, London SW7 2AZ, UK
| | | | | | | |
Collapse
|
25
|
Thakar RG, Chown MG, Patel A, Peng L, Kumar S, Desai TA. Contractility-dependent modulation of cell proliferation and adhesion by microscale topographical cues. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2008; 4:1416-1424. [PMID: 18711756 DOI: 10.1002/smll.200701302] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Engineering of cellular assembly on biomaterial scaffolds by utilizing microscale topographical cues has emerged as a powerful strategy in cardiovascular tissue engineering and regenerative medicine. However, the mechanisms through which these cues are processed to yield changes in canonical cell behaviors remain unclear. Previously, we showed that when mixtures of cardiomyocytes and fibroblasts were cultured on polydimethylsiloxane surfaces studded with microscale pillars (micropegs), fibroblast proliferation was dramatically suppressed, which suggests that the micropegs could be exploited to minimize fibrosis and scar formation. Here, we demonstrate that this effect relies on altered adhesive and micromechanical interactions between individual cells and micropegs. First, we show that the proliferation of a cell physically attached to a micropeg is significantly lower than that of a cell cultured on a featureless region of the substrate. Micropeg adhesion is accompanied by a marked elongation in cell and nuclear shape. When fibroblast contractility is pharmacologically attenuated through low-dose inhibition of either Rho-associated kinase or myosin light chain kinase, the potency with which micropeg adhesion suppresses cell proliferation is significantly reduced. Together, our results support a model in which cell fate decisions may be directly manipulated within tissue engineering scaffolds by the inclusion of microtopographical structures that alter cellular mechanics.
Collapse
Affiliation(s)
- Rahul G Thakar
- Department of Physiology, University of California, San Francisco 203C Byers Hall Box 2520, 1700 4th Street San Francisco, CA 94158-2330, USA
| | | | | | | | | | | |
Collapse
|
26
|
Yokoyama U, Minamisawa S, Quan H, Akaike T, Jin M, Otsu K, Ulucan C, Wang X, Baljinnyam E, Takaoka M, Sata M, Ishikawa Y. Epac1 is upregulated during neointima formation and promotes vascular smooth muscle cell migration. Am J Physiol Heart Circ Physiol 2008; 295:H1547-55. [PMID: 18689492 DOI: 10.1152/ajpheart.01317.2007] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Vascular remodeling after mechanoinjury largely depends on the migration of smooth muscle cells, an initial key step to wound healing. However, the role of the second messenger system, in particular, the cAMP signal, in regulating such remodeling remains controversial. Exchange protein activated by cAMP (Epac) has been identified as a new target molecule of the cAMP signal, which is independent from PKA. We thus examined whether Epac plays a distinct role from PKA in vascular remodeling. To examine the role of Epac and PKA in migration, we used primary culture smooth muscle cells from both the fetal and adult rat aorta. A cAMP analog selective to PKA, 8-(4-parachlorophenylthio)-cAMP (pCPT-cAMP), decreased cell migration, whereas an Epac-selective analog, 8-pCPT-2'-O-Me-cAMP, enhanced migration. Adenovirus-mediated gene transfer of PKA decreased cell migration, whereas that of Epac1 significantly enhanced cell migration. Striking morphological differences were observed between pCPT-cAMP- and 8-pCPT-2'-O-Me-cAMP-treated aortic smooth muscle cells. Furthermore, overexpression of Epac1 enhanced the development of neointimal formation in fetal rat aortic tissues in organ culture. When the mouse femoral artery was injured mechanically in vivo, we found that the expression of Epac1 was upregulated in vascular smooth muscle cells, whereas that of PKA was downregulated with the progress of neointimal thickening. Our findings suggest that Epac1, in opposition to PKA, increases vascular smooth muscle cell migration. Epac may thus play an important role in advancing vascular remodeling and restenosis upon vascular injury.
Collapse
Affiliation(s)
- Utako Yokoyama
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Bond M, Wu YJ, Sala-Newby GB, Newby AC. Rho GTPase, Rac1, regulates Skp2 levels, vascular smooth muscle cell proliferation, and intima formation in vitro and in vivo. Cardiovasc Res 2008; 80:290-8. [PMID: 18599477 DOI: 10.1093/cvr/cvn188] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
AIMS Vascular smooth muscle cell (VSMC) proliferation contributes to intima formation after angioplasty or venous by-pass grafting, and during atherosclerosis. VSMC proliferation requires degradation of p27(Kip1) promoted by S-phase kinase-associated protein-2 (Skp2), an F-box protein component of the Skp-Cullin-F-box(Skp2) ubiquitin-ligase. We investigated the role of Rac(1) in the regulation of Skp2 in rat VSMC. METHODS AND RESULTS Rat carotid balloon injury increased Rac(1) activity. Rho GTPase inhibition with Clostridium difficile Toxin B or specific Rac(1) inhibition with adenovirus-mediated expression of dominant-negative Rac(1) reduced Skp2 levels, and VSMC proliferation in vitro and intima formation in vivo following carotid balloon injury. Inhibition of Skp2 expression and proliferation by dominant-negative Rac(1) was reversed by exogenous Skp2. Elevation of endogenous adenosine 3',5'-cyclic monophosphate (cAMP) with forskolin-inhibited Rac(1) activity, reduced Skp2, increased p27(Kip1) and inhibited VSMC proliferation, effects that were reversed by constitutively active Rac(1). These effects were independent of Rac(1) Cdc42/Rac interactive binding (CRIB)-domain effector proteins but associated with Rac(1)-dependent actin polymerization. CONCLUSION Rac(1) activity regulates VSMC proliferation by controlling Skp2 levels. Activation of Rac(1) induced by balloon injury in vivo increases Skp2 levels, which promotes VSMC proliferation and intima formation. Inhibition of this novel pathway underlies the negative effects of cAMP on VSMC proliferation.
Collapse
Affiliation(s)
- Mark Bond
- Bristol Heart Institute, University of Bristol, Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK.
| | | | | | | |
Collapse
|
28
|
Muzaffar S, Shukla N, Bond M, Newby AC, Angelini GD, Sparatore A, Del Soldato P, Jeremy JY. Exogenous hydrogen sulfide inhibits superoxide formation, NOX-1 expression and Rac1 activity in human vascular smooth muscle cells. J Vasc Res 2008; 45:521-8. [PMID: 18463417 DOI: 10.1159/000129686] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2007] [Accepted: 12/19/2007] [Indexed: 12/31/2022] Open
Abstract
The activity of NADPH oxidase (NOX) is blocked by nitric oxide (NO). Hydrogen sulfide (H(2)S) is also produced by blood vessels. It is reasonable to suggest that H(2)S may have similar actions to NO on NOX. In order to test this hypothesis, the effect of sodium hydrosulfide (NaHS) on O(2)(-) formation, the expression of NOX-1 (a catalytic subunit of NOX) and Rac(1) activity (essential for full NOX activity) in isolated vascular smooth muscle cells (hVSMCs) was investigated. hVSMCs were incubated with the thromboxane A(2) analogue U46619 +/- NaHS for 1 or 16 h, and O(2)(-) formation, NOX-1 expression and Rac(1) activity were assessed. The possible interaction between H(2)S and NO was also studied by using an NO synthase inhibitor, L-NAME, and an NO donor, DETA-NONOate. The role of K(ATP) channels was studied by using glibenclamide. NaHS inhibited O(2)(-) formation following incubation of 1 h (IC(50), 30 nM) and 16 h (IC(50), 20 nM), blocked NOX-1 expression and inhibited Rac(1) activity. These inhibitory effects of NaHS were mediated by the cAMP-protein-kinase-A axis. Exogenous H(2)S prevents NOX-driven intravascular oxidative stress through an a priori inhibition of Rac(1) and downregulation of NOX-1 protein expression, an effect mediated by activation of the adenylylcyclase-cAMP-protein-kinase-G system by H(2)S.
Collapse
Affiliation(s)
- Saima Muzaffar
- Bristol Heart Institute, University of Bristol, Bristol, UK.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
The cyclic AMP effector Epac integrates pro- and anti-fibrotic signals. Proc Natl Acad Sci U S A 2008; 105:6386-91. [PMID: 18434542 DOI: 10.1073/pnas.0801490105] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Scar formation occurs during the late stages of the inflammatory response but, when excessive, produces fibrosis that can lead to functional and structural damage of tissues. Here, we show that the profibrogenic agonist, transforming growth factor beta1, transcriptionally decreases expression of Exchange protein activated by cAMP 1 (Epac1) in fibroblasts/fibroblast-like cells from multiple tissues (i.e., cardiac, lung, and skin fibroblasts and hepatic stellate cells). Overexpression of Epac1 inhibits transforming growth factor beta1-induced collagen synthesis, indicating that a decrease of Epac1 expression appears to be necessary for the fibrogenic phenotype, an idea supported by evidence that Epac1 expression in cardiac fibroblasts is inhibited after myocardial infarction. Epac and protein kinase A, a second mediator of cAMP action, have opposite effects on migration but both inhibit synthesis of collagen and DNA by fibroblasts. Epac is preferentially activated by low concentrations of cAMP and stimulates migration via the small G protein Rap1 but inhibits collagen synthesis in a Rap1-independent manner. The regulation of Epac expression and activation thus appear to be critical for the integration of pro- and anti-fibrotic signals and for the regulation of fibroblast function.
Collapse
|
30
|
Muzaffar S, Shukla N, Bond M, Sala-Newby G, Angelini GD, Newby AC, Jeremy JY. Acute inhibition of superoxide formation and Rac1 activation by nitric oxide and iloprost in human vascular smooth muscle cells in response to the thromboxane A2 analogue, U46619. Prostaglandins Leukot Essent Fatty Acids 2008; 78:247-55. [PMID: 18420399 PMCID: PMC2850987 DOI: 10.1016/j.plefa.2008.01.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2007] [Revised: 01/15/2008] [Accepted: 01/18/2008] [Indexed: 01/07/2023]
Abstract
BACKGROUND The over-production of superoxide (O(2)(-)) derived from NADPH oxidase (NOX) plays a central role in cardiovascular diseases. By contrast, nitric oxide (NO) and prostacyclin (PGI(2)) are vasculoprotective. The effect of the NO donor, NONOate and iloprost on O(2)(-) formation, p47(phox) and Rac(1) activation in human vascular smooth muscle cells (hVSMCs) was investigated. METHODS hVSMCs were incubated with 10nM thromboxane A(2) analogue, U46619 for 16h, and then with apocynin (a NOX inhibitor), NONOate or iloprost for 1h and O(2)(-) measured spectrophometrically. The role of cyclic AMP and cyclic GMP was examined by co-incubation of drugs with protein kinase (PK) A and G inhibitors listed above. Rac(1) was studied using pull-down assays. RESULTS NONOate and iloprost inhibited O(2)(-) formation, acutely, effects blocked by inhibition of PKG and PKA, respectively. Rac(1) and p47(phox) activation and translocation to the plasma membrane was completely inhibited by NONOate and iloprost, effects again reversed by co-incubation with PKG or PKA inhibitors. CONCLUSIONS NO and PGI(2) block the acute activity of NOX in hVSMCs via the cGMP-PKG axis (for NO) and by the cAMP-PKA axis (for iloprost) through inhibition of Rac(1) and p47(phox) translocation. These findings have implications in the pathophysiology and treatment of CVD.
Collapse
Affiliation(s)
- S Muzaffar
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Bristol, UK.
| | | | | | | | | | | | | |
Collapse
|
31
|
Arbeloa A, Bulgin RR, MacKenzie G, Shaw RK, Pallen MJ, Crepin VF, Berger CN, Frankel G. Subversion of actin dynamics by EspM effectors of attaching and effacing bacterial pathogens. Cell Microbiol 2008; 10:1429-41. [PMID: 18331467 PMCID: PMC2610399 DOI: 10.1111/j.1462-5822.2008.01136.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Rho GTPases are common targets of bacterial toxins and type III secretion system effectors. IpgB1 and IpgB2 of Shigella and Map of enteropathogenic (EPEC) and enterohemorrhagic (EHEC) Escherichia coli were recently grouped together on the basis that they share a conserved WxxxE motif. In this study, we characterized six WxxxE effectors from attaching and effacing pathogens: TrcA and EspM1 of EPEC strain B171, EspM1 and EspM2 of EHEC strain Sakai and EspM2 and EspM3 of Citrobacter rodentium. We show that EspM2 triggers formation of global parallel stress fibres, TrcA and EspM1 induce formation of localized parallel stress fibres and EspM3 triggers formation of localized radial stress fibres. Using EspM2 and EspM3 as model effectors, we report that while substituting the conserved Trp with Ala abolished activity, conservative Trp to Tyr or Glu to Asp substitutions did not affect stress-fibre formation. We show, using dominant negative constructs and chemical inhibitors, that the activity of EspM2 and EspM3 is RhoA and ROCK-dependent. Using Rhotekin pull-downs, we have shown that EspM2 and EspM3 activate RhoA; translocation of EspM2 and EspM3 triggered phosphorylation of cofilin. These results suggest that the EspM effectors modulate actin dynamics by activating the RhoA signalling pathway.
Collapse
Affiliation(s)
- Ana Arbeloa
- Division of Cell and Molecular Biology, Imperial College London, London SW7 2AZ, UK
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Nicola C, Lala PK, Chakraborty C. Prostaglandin E2-mediated migration of human trophoblast requires RAC1 and CDC42. Biol Reprod 2008; 78:976-82. [PMID: 18235104 DOI: 10.1095/biolreprod.107.065433] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The invasion of maternal decidua and uterine spiral arteries by a trophoblast subpopulation called extravillous trophoblast (EVT) is essential for the establishment of a normal placenta and an adequate blood flow toward the fetus. Derangements in these processes underlie pregnancy-related diseases like preeclampsia and intrauterine growth restriction. Many growth factors, growth factor binding proteins, and extracellular matrix components can positively or negatively regulate the proliferation, migration, and/or invasiveness of these EVT cells. RHO GTPases, including RHOA, RAC1, and CDC42, are ubiquitous proteins that control cytoskeletal changes by forming stress fibers and projecting lamellipodia and filopodia during cellular migration. We had previously shown that prostaglandin (PG) E(2) produced in abundance by the decidua promotes the migration of first-trimester human EVTs by increasing the intracellular concentration of calcium and activating calpain. Using our well-characterized immortalized EVT cell line, HTR-8/SVneo, as well as villus explants from first-trimester placentae, this study examined the role of RHO GTPases RAC1 and CDC42 in PGE(2)-mediated migratory responses of these cells. Though a RAC1 inhibitor, NSC23766 as well as RAC1 knockdown by siRNA decreased the migration of HTR-8/SVneo cells in a Transwell migration assay, this inhibition could not be restored by PGE(2) or 17-phenyl trinor PGE(2) (PGE receptor PTGER1 agonist) or PGE(1) Alcohol (PGE receptor PTGER4 agonist). Similar results were noted for EVT cell spreading in villus explants. Furthermore, CDC42 silencing using siRNA inhibited PGE(2)-induced migration of HTR-8/SVneo cells. Finally, the treatment of EVT cells with PGE(2), PTGER1 agonist, or PTGER4 agonist activated RAC1 and CDC42 at 10 min, suggesting that RAC1 and CDC42 play an essential role in PGE(2)-mediated migration of human EVTs.
Collapse
Affiliation(s)
- Catalin Nicola
- Departments of Anatomy and Cell Biology and Pathology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1
| | | | | |
Collapse
|
33
|
Ohnishi H, Miyake M, Kamitani S, Horiguchi Y. The morphological changes in cultured cells caused by Bordetella pertussis adenylate cyclase toxin. FEMS Microbiol Lett 2007; 279:174-9. [PMID: 18179583 DOI: 10.1111/j.1574-6968.2007.01028.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Bordetella pertussis is the causative agent for human whooping cough. It was found that Bordetella pertussis infection caused a change in shape from flat to round in L2 cells, which are derived from rat type 2 alveolar cells. This phenomenon was reproduced using the culture supernatant of B. pertussis, and bacterium-free adenylate cyclase toxin (CyaA) was identified as the factor responsible. A purified preparation of wild-type CyaA but not an enzyme-dead mutant caused the cell rounding. It was examined whether CyaA causes similar morphological changes in various cultured cell lines. L2, EBL, HEK293T, MC3T3-E1, NIH 3T3, and Vero cells were rounded by the toxin whereas Caco-2, Eph4, and MDCK cells were not, although all these cells showed a significant elevation of the intracellular cAMP level in response to CyaA treatment, which indicates that there is no quantitative correlation between the rounding phenotype and the intracellular cAMP level. CyaA has been believed to target various immunocompetent cells and support the establishment of the bacterial infection by subverting the host immune responses. The possibility that CyaA may also affect tissue cells such as respiratory epithelial cells and may be involved in the pathogenesis of the bacterial infection is also indicated.
Collapse
Affiliation(s)
- Hiroe Ohnishi
- Department of Molecular Bacteriology, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, Japan
| | | | | | | |
Collapse
|
34
|
Zhu J, Sun N, Aoudjit L, Li H, Kawachi H, Lemay S, Takano T. Nephrin mediates actin reorganization via phosphoinositide 3-kinase in podocytes. Kidney Int 2007; 73:556-66. [PMID: 18033240 DOI: 10.1038/sj.ki.5002691] [Citation(s) in RCA: 150] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Nephrin is a slit diaphragm protein critical for structural and functional integrity of visceral glomerular epithelial cells (podocytes) and is known to be tyrosine phosphorylated by Src family kinases. We studied the role of phosphoinositide 3-kinase (PI3K), activated via the phosphorylation of nephrin, in actin cytoskeletal reorganization of cultured rat podocytes. Phosphorylation of rat nephrin by the Fyn kinase markedly increased its interaction with a regulatory subunit of PI3K. Stable transfection of rat nephrin in the podocytes with podocin led to nephrin tyrosine phosphorylation, PI3K-dependent phosphorylation of Akt, increased Rac1 activity, and an altered actin cytoskeleton with decreased stress fibers and increased lamellipodia. These changes were reversed with an inhibitor of PI3K and not seen when the nephrin-mutant Y1152F replaced wild-type nephrin. Rac1 and Akt1 contributed to lamellipodia formation and decreased stress fibers, respectively. Finally, in the rat model of puromycin aminonucleoside nephrosis, nephrin tyrosine phosphorylation, nephrin-PI3K association, and glomerular Akt phosphorylation were all decreased. Our results suggest that PI3K is involved in nephrin-mediated actin reorganization in podocytes. Disturbed nephrin-PI3K interactions may contribute to abnormal podocyte morphology and proteinuria.
Collapse
Affiliation(s)
- J Zhu
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
35
|
Komalavilas P, Penn RB, Flynn CR, Thresher J, Lopes LB, Furnish EJ, Guo M, Pallero MA, Murphy-Ullrich JE, Brophy CM. The small heat shock-related protein, HSP20, is a cAMP-dependent protein kinase substrate that is involved in airway smooth muscle relaxation. Am J Physiol Lung Cell Mol Physiol 2007; 294:L69-78. [PMID: 17993590 DOI: 10.1152/ajplung.00235.2007] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Activation of the cAMP/cAMP-dependent PKA pathway leads to relaxation of airway smooth muscle (ASM). The purpose of this study was to examine the role of the small heat shock-related protein HSP20 in mediating PKA-dependent ASM relaxation. Human ASM cells were engineered to constitutively express a green fluorescent protein-PKA inhibitory fusion protein (PKI-GFP) or GFP alone. Activation of the cAMP-dependent signaling pathways by isoproterenol (ISO) or forskolin led to increases in the phosphorylation of HSP20 in GFP but not PKI-GFP cells. Forskolin treatment in GFP but not PKI-GFP cells led to a loss of central actin stress fibers and decreases in the number of focal adhesion complexes. This loss of stress fibers was associated with dephosphorylation of the actin-depolymerizing protein cofilin in GFP but not PKI-GFP cells. To confirm that phosphorylated HSP20 plays a role in PKA-induced ASM relaxation, intact strips of bovine ASM were precontracted with serotonin followed by ISO. Activation of the PKA pathway led to relaxation of bovine ASM, which was associated with phosphorylation of HSP20 and dephosphorylation of cofilin. Finally, treatment with phosphopeptide mimetics of HSP20 possessing a protein transduction domain partially relaxed precontracted bovine ASM strips. In summary, ISO-induced phosphorylation of HSP20 or synthetic phosphopeptide analogs of HSP20 decreases phosphorylation of cofilin and disrupts actin in ASM, suggesting that one possible mechanism by which HSP20 mediates ASM relaxation is via regulation of actin filament dynamics.
Collapse
Affiliation(s)
- Padmini Komalavilas
- Center for Metabolic Biology, College of Liberal Arts and Sciences, Arizona State University, PO Box 873704, Tempe, AZ 85287-3704, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Thomas PE, Peters-Golden M, White ES, Thannickal VJ, Moore BB. PGE(2) inhibition of TGF-beta1-induced myofibroblast differentiation is Smad-independent but involves cell shape and adhesion-dependent signaling. Am J Physiol Lung Cell Mol Physiol 2007; 293:L417-28. [PMID: 17557799 PMCID: PMC2846428 DOI: 10.1152/ajplung.00489.2006] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Myofibroblasts are pathogenic in pulmonary fibrotic disease due to their exuberant production of matrix rich in collagen that interferes with gas exchange and the ability of these cells to contract and distort the alveolar space. Transforming growth factor-beta1 (TGF-beta1) is a well-known inducer of myofibroblast differentiation. TGF-beta1-induced transformation of fibroblasts to apoptosis-resistant myofibroblasts is adhesion-dependent and focal adhesion kinase (FAK)-mediated. Prostaglandin E(2) (PGE(2)) inhibits this differentiation via E prostanoid receptor 2 (EP2) signaling and cAMP elevation, but whether PGE(2) does so by interfering with TGF-beta1 signaling is unknown. Thus we examined the effects of PGE(2) in the presence and absence of TGF-beta1 stimulation on candidate signaling pathways in human lung fibroblasts. We now demonstrate that PGE(2) does not interfere with TGF-beta1-induced Smad phosphorylation or its translocation to the nucleus. Rather, PGE(2) has dramatic effects on cell shape and cytoskeletal architecture and disrupts the formation of appropriate focal adhesions. PGE(2) treatment diminishes TGF-beta1-induced phosphorylation of paxillin, STAT-3, and FAK and, in turn, limits activation of the protein kinase B (PKB/Akt) pathway. These alterations do not, however, result in increased apoptosis within the first 24 h of treatment. Interestingly, the effects of PGE(2) stimulation alone do not always mirror the effects of PGE(2) in the presence of TGF-beta1, indicating that the context for EP2 signaling is different in the presence of TGF-beta1. Taken together, our results demonstrate that PGE(2) has the potential to limit TGF-beta1-induced myofibroblast differentiation via adhesion-dependent, but Smad-independent, pathways.
Collapse
Affiliation(s)
- Peedikayil E Thomas
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | | | | | | | | |
Collapse
|
37
|
Zhang H, Cybulsky AV, Aoudjit L, Zhu J, Li H, Lamarche-Vane N, Takano T. Role of Rho-GTPases in complement-mediated glomerular epithelial cell injury. Am J Physiol Renal Physiol 2007; 293:F148-56. [PMID: 17376765 DOI: 10.1152/ajprenal.00294.2006] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Visceral glomerular epithelial cells (GEC) are essential for maintenance of normal glomerular permselectivity. The actin cytoskeleton is a key determinant of GEC morphology and function. In the rat passive Heymann nephritis (PHN) model of membranous nephropathy, complement C5b-9 induces nonlytic GEC injury associated with morphological changes of GEC and proteinuria. The current study addresses the role of Rho family of small GTPases in complement-mediated GEC injury. When cultured rat GEC were stimulated with complement C5b-9 for 18 h, RhoA activity increased, whereas Rac1/Cdc42 activities decreased, compared with control cells. Similar changes in Rho-GTPase activities were observed in glomeruli from rats with PHN. The amount of active p190RhoGAP, a negative upstream regulator of RhoA, was decreased in complement-stimulated GEC, potentially contributing to increased RhoA activity. To address the functional effects of Rho-GTPases, GEC were transfected with constitutively active (CA) or dominant negative (DN) Rho-GTPase mutants. GEC transfected with CA-RhoA showed a smaller and round contour and prominent cortical F-actin. In contrast, GEC transfected with CA-Rac1 demonstrated morphological changes that resembled process formation. In addition, expression of CA-RhoA attenuated complement-mediated cytotoxicity, whereas cytotoxicity was augmented by DN-RhoA. Thus exposure of GEC to complement alters the balance of RhoA, Rac1, and Cdc42 activities. The activity of Rac1 may contribute to process formation, while activation of RhoA (e.g., in the setting of complement attack), with or without blunting of Rac1 activity, may have an opposite effect, i.e., contribute to foot process effacement. Activation of RhoA increases the resistance of GEC to complement-mediated injury.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Medicine, McGill University Health Centre, Montreal, Quebec H3A 2B4
| | | | | | | | | | | | | |
Collapse
|
38
|
You F, Osawa Y, Hayashi SI, Nakashima S. Immediate early gene IEX-1 induces astrocytic differentiation of U87-MG human glioma cells. J Cell Biochem 2007; 100:256-65. [PMID: 16960879 DOI: 10.1002/jcb.21082] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The immediate early response gene IEX-1 is involved in the regulation of apoptosis and cell growth. In order to increase the apoptotic sensitivity to chemotherapeutic drugs and gamma-ray, we attempted to establish U87-MG human glioma cell line expressing IEX-1. Unexpectedly, however, transfection of IEX-1 into U87-MG glioma cells resulted in morphological changes to astrocytic phenotype and increase in glial differentiation marker proteins, S-100 and glial fibrillary acidic protein (GFAP). Glial cell differentiation was used to examine in rat C6 glioma cell line, since this cell line express astrocytic phenotypes by increase in intracellular cAMP concentration. Stimulation of human U87-MG glioma cells by membrane-permeable dibutyryl cAMP (dbcAMP) not only elicited their morphological changes but also induced expression of IEX-1 as well as S-100 and GFAP. H89, an inhibitor of protein kinase A (PKA), blocked dbcAMP-induced morphological changes of U87-MG cells and expression of IEX-1. In contrast, morphological changes and expression of S-100 and GFAP induced by IEX-1 were not affected by H89. Morphological changes induced by dbcAMP were totally abolished by functional disruption of IEX-1 expression by anti-sense RNA. These results indicate that IEX-1 plays an important role in astrocytic differentiation of human glioma cells and that IEX-1 functions at downstream of PKA.
Collapse
Affiliation(s)
- Fukka You
- Department of Cell Signaling, Division of Cell and Molecular Biology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan
| | | | | | | |
Collapse
|
39
|
Gao SY, Li CY, Shimokawa T, Terashita T, Matsuda S, Yaoita E, Kobayashi N. Rho-family small GTPases are involved in forskolin-induced cell-cell contact formation of renal glomerular podocytes in vitro. Cell Tissue Res 2007; 328:391-400. [PMID: 17265067 DOI: 10.1007/s00441-006-0365-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2006] [Accepted: 12/05/2006] [Indexed: 01/01/2023]
Abstract
Intercellular adhesions between renal glomerular epithelial cells (also called podocytes) are necessary for the proper function of the glomerular filtration barrier. Although our knowledge of the molecular composition of podocyte cell-cell contact sites has greatly progressed, the underlying molecular mechanism regulating the formation of these cell-cell contacts remains largely unknown. We have used forskolin, an activator of adenylyl cyclase that elevates the level of intracellular cAMP, to investigate the effect of cAMP and three Rho-family small GTPases (RhoA, Cdc42, and Rac1) on the regulation of cell-cell contact formation in a murine podocyte cell line. Transmission electron microscopy and the immunostaining of cell adhesion molecules and actin-associated proteins have revealed a structural change at the site of cell-cell contact following forskolin treatment. The activity of the Rho-family small GTPases before and after forskolin treatment has been evaluated with a glutathione-S-transferase pull-down assay. Forskolin reinforces the integrity of cell-cell contacts, resulting in the closure of an intercellular adhesion zipper, accompanied by a redistribution of cell adhesion molecules and actin-associated proteins in a continuous linear pattern at cell-cell contacts. The Rho-family small GTPases Rac1 and Cdc42 are activated during closure of the adhesion zipper, whereas RhoA is suppressed. Thus, cAMP promotes the assembly of cell-cell contacts between podocytes via a mechanism that probably involves Rho-family small GTPases.
Collapse
Affiliation(s)
- Shuang-Yan Gao
- Department of Anatomy and Embryology, School of Medicine, Ehime University, To-on City, Ehime, Japan
| | | | | | | | | | | | | |
Collapse
|
40
|
Houslay MD. The long and short of vascular smooth muscle phosphodiesterase-4 as a putative therapeutic target. Mol Pharmacol 2005; 68:563-7. [PMID: 15958394 DOI: 10.1124/mol.105.015719] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In this issue, Tilley and Maurice (p. 596) show that differentiation of vascular smooth muscle cells to a proliferative phenotype is associated with a profound up-regulation of specific phosphodiesterase-4 (PDE4) isoforms because of increased histone acetylation. The increased PDE4 activity is seen as preventing cAMP from inhibiting the enhanced proliferation, migration, and production of extracellular matrix seen in activated VSMC. This Perspective examines the proposal that selective inhibition of PDE4D1/2 could find use in adjunctive pharmacotherapy after percutaneous coronary interventions and, in addition, discusses the recent genetic evidence that PDE4D7 may provide a therapeutic target in stroke.
Collapse
Affiliation(s)
- Miles D Houslay
- Molecular Pharmacology Group, Division of Biochemistry and Molecular Biology, IBLS, Wolfson Building, University of Glasgow, Glasgow G12 8QQ, Scotland, UK.
| |
Collapse
|