101
|
Abstract
We all know about classical fibrinolysis, how plasminogen activation by either tissue-type plasminogen activator (t-PA) or urokinase-type plasminogen activator (u-PA) promotes fibrin breakdown, and how this process was harnessed for the therapeutic removal of blood clots. While this is still perfectly true and still applicable to thromboembolic conditions today, another dimension to this system came to light over two decades ago that implicated the plasminogen activating system in a context far removed from the dissolution of blood clots. This unsuspected area related to brain biology where t-PA was linked to a plethora of activities in the CNS, some of which do not necessarily require plasmin generation. Indeed, t-PA either directly or via plasmin, has been shown to not only have key roles in modulating astrocytes, neurons, microglia, and pericytes, but also to have profound effects in a number of CNS conditions, including ischaemic stroke, severe traumatic brain injury and also in neurodegenerative disorders. While compelling insights have been obtained from various animal models, the clinical relevance of aberrant expression of these components in the CNS, although strongly implied, are only just emerging. This review will cover these areas and will also discuss how the use of thrombolytic agents and anti-fibrinolytic drugs may potentially have impacts outside of their clinical intention, particularly in the CNS.
Collapse
Affiliation(s)
- R L Medcalf
- Australian Centre for Blood Diseases, Monash University, Melbourne, Vic, Australia
| |
Collapse
|
102
|
Abstract
INTRODUCTION Stroke is a major cause of disability and the fifth leading cause of death. Currently, the only approved acute medical treatment of ischemic stroke is tissue plasminogen activator (tPA), but its effectiveness is greatly predicated upon early administration of the drug. There is, therefore, an urgent need to find new therapeutic options for acute stroke. Areas covered: In this review, we summarize the role of Rho-associated coiled-coil containing kinase (ROCK) and its potential as a therapeutic target in stroke pathophysiology. ROCK is a major regulator of cell contractility, motility, and proliferation. Many of these ROCK-mediated processes in endothelial cells, vascular smooth muscle cells, pericytes, astrocytes, glia, neurons, leukocytes, and platelets are important in stroke pathophysiology, and the inhibition of such processes could improve stroke outcome. Expert commentary: ROCK is a potential therapeutic target for cardiovascular disease and ROCK inhibitors have already been approved for human use in Japan and China for the treatment of acute stroke. Further studies are needed to determine the role of ROCK isoforms in the pathophysiology of cerebral ischemia and whether there are further therapeutic benefits with selective ROCK inhibitors.
Collapse
Affiliation(s)
- Nikola Sladojevic
- a Department of Medicine, Section of Cardiology , University of Chicago , Chicago , IL , USA
| | - Brian Yu
- a Department of Medicine, Section of Cardiology , University of Chicago , Chicago , IL , USA
| | - James K Liao
- a Department of Medicine, Section of Cardiology , University of Chicago , Chicago , IL , USA
| |
Collapse
|
103
|
Mulherkar S, Firozi K, Huang W, Uddin MD, Grill RJ, Costa-Mattioli M, Robertson C, Tolias KF. RhoA-ROCK Inhibition Reverses Synaptic Remodeling and Motor and Cognitive Deficits Caused by Traumatic Brain Injury. Sci Rep 2017; 7:10689. [PMID: 28878396 PMCID: PMC5587534 DOI: 10.1038/s41598-017-11113-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 08/18/2017] [Indexed: 01/07/2023] Open
Abstract
Traumatic brain injury (TBI) causes extensive neural damage, often resulting in long-term cognitive impairments. Unfortunately, effective treatments for TBI remain elusive. The RhoA-ROCK signaling pathway is a potential therapeutic target since it is activated by TBI and can promote the retraction of dendritic spines/synapses, which are critical for information processing and memory storage. To test this hypothesis, RhoA-ROCK signaling was blocked by RhoA deletion from postnatal neurons or treatment with the ROCK inhibitor fasudil. We found that TBI impairs both motor and cognitive performance and inhibiting RhoA-ROCK signaling alleviates these deficits. Moreover, RhoA-ROCK inhibition prevents TBI-induced spine remodeling and mature spine loss. These data argue that TBI elicits pathological spine remodeling that contributes to behavioral deficits by altering synaptic connections, and RhoA-ROCK inhibition enhances functional recovery by blocking this detrimental effect. As fasudil has been safely used in humans, our results suggest that it could be repurposed to treat TBI.
Collapse
Affiliation(s)
- Shalaka Mulherkar
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Karen Firozi
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Wei Huang
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA.,The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, 733N. Broadway, Baltimore, MD, 21205, USA
| | | | - Raymond J Grill
- Department of Integrative Biology and Pharmacology, University of Texas Medical School at Houston, Houston, TX, 77030, USA.,Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Mauro Costa-Mattioli
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA.,Memory and Brain Research Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Claudia Robertson
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kimberley F Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA. .,Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
104
|
Rothschild PR, Salah S, Berdugo M, Gélizé E, Delaunay K, Naud MC, Klein C, Moulin A, Savoldelli M, Bergin C, Jeanny JC, Jonet L, Arsenijevic Y, Behar-Cohen F, Crisanti P. ROCK-1 mediates diabetes-induced retinal pigment epithelial and endothelial cell blebbing: Contribution to diabetic retinopathy. Sci Rep 2017; 7:8834. [PMID: 28821742 PMCID: PMC5562711 DOI: 10.1038/s41598-017-07329-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 06/23/2017] [Indexed: 12/18/2022] Open
Abstract
In diabetic retinopathy, the exact mechanisms leading to retinal capillary closure and to retinal barriers breakdown remain imperfectly understood. Rho-associated kinase (ROCK), an effector of the small GTPase Rho, involved in cytoskeleton dynamic regulation and cell polarity is activated by hyperglycemia. In one year-old Goto Kakizaki (GK) type 2 diabetic rats retina, ROCK-1 activation was assessed by its cellular distribution and by phosphorylation of its substrates, MYPT1 and MLC. In both GK rat and in human type 2 diabetic retinas, ROCK-1 is activated and associated with non-apoptotic membrane blebbing in retinal vessels and in retinal pigment epithelium (RPE) that respectively form the inner and the outer barriers. Activation of ROCK-1 induces focal vascular constrictions, endoluminal blebbing and subsequent retinal hypoxia. In RPE cells, actin cytoskeleton remodeling and membrane blebs in RPE cells contributes to outer barrier breakdown. Intraocular injection of fasudil, significantly reduces both retinal hypoxia and RPE barrier breakdown. Diabetes-induced cell blebbing may contribute to ischemic maculopathy and represent an intervention target.
Collapse
Affiliation(s)
- Pierre-Raphaël Rothschild
- Inserm UMR_S 1138, Team 17: From physiopathology of retinal diseases to clinical advances, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne University, University of Pierre et Marie Curie, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Paris Descartes University, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Sawsen Salah
- Inserm UMR_S 1138, Team 17: From physiopathology of retinal diseases to clinical advances, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne University, University of Pierre et Marie Curie, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Paris Descartes University, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Marianne Berdugo
- Inserm UMR_S 1138, Team 17: From physiopathology of retinal diseases to clinical advances, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne University, University of Pierre et Marie Curie, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Paris Descartes University, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Emmanuelle Gélizé
- Inserm UMR_S 1138, Team 17: From physiopathology of retinal diseases to clinical advances, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne University, University of Pierre et Marie Curie, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Paris Descartes University, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Kimberley Delaunay
- Inserm UMR_S 1138, Team 17: From physiopathology of retinal diseases to clinical advances, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne University, University of Pierre et Marie Curie, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Paris Descartes University, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Marie-Christine Naud
- Inserm UMR_S 1138, Team 17: From physiopathology of retinal diseases to clinical advances, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne University, University of Pierre et Marie Curie, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Paris Descartes University, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Christophe Klein
- Inserm UMR_S 1138, Team 17: From physiopathology of retinal diseases to clinical advances, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne University, University of Pierre et Marie Curie, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Paris Descartes University, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Alexandre Moulin
- Department of Ophthalmology of University of Lausanne 1000 Lausanne, Jules Gonin Hospital, Lausanne, Switzerland
| | - Michèle Savoldelli
- Inserm UMR_S 1138, Team 17: From physiopathology of retinal diseases to clinical advances, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne University, University of Pierre et Marie Curie, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Paris Descartes University, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Ciara Bergin
- Department of Ophthalmology of University of Lausanne 1000 Lausanne, Jules Gonin Hospital, Lausanne, Switzerland
| | - Jean-Claude Jeanny
- Inserm UMR_S 1138, Team 17: From physiopathology of retinal diseases to clinical advances, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne University, University of Pierre et Marie Curie, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Paris Descartes University, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Laurent Jonet
- Inserm UMR_S 1138, Team 17: From physiopathology of retinal diseases to clinical advances, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne University, University of Pierre et Marie Curie, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Paris Descartes University, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Yvan Arsenijevic
- Department of Ophthalmology of University of Lausanne 1000 Lausanne, Jules Gonin Hospital, Lausanne, Switzerland
| | - Francine Behar-Cohen
- Inserm UMR_S 1138, Team 17: From physiopathology of retinal diseases to clinical advances, Centre de Recherche des Cordeliers, Paris, France. .,Sorbonne University, University of Pierre et Marie Curie, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France. .,Paris Descartes University, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France. .,Department of Ophthalmology, Assistance Publique-Hopitaux de Paris, Hôtel-Dieu de Paris Hospital, 75004, Paris, France. .,INSERM U1138 Team 17, Le Centre de Recherches des Cordeliers (CRC), 75006, Paris, France. .,University of Lausanne, Lausanne, Switzerland.
| | - Patricia Crisanti
- Inserm UMR_S 1138, Team 17: From physiopathology of retinal diseases to clinical advances, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne University, University of Pierre et Marie Curie, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Paris Descartes University, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| |
Collapse
|
105
|
Zhang Y, Wu S. Effects of fasudil on pulmonary hypertension in clinical practice. Pulm Pharmacol Ther 2017; 46:54-63. [PMID: 28782712 DOI: 10.1016/j.pupt.2017.08.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 06/02/2017] [Accepted: 08/03/2017] [Indexed: 01/09/2023]
Abstract
Pulmonary hypertension (PH) is a pathophysiologic disorder that may involve multiple clinical conditions and can complicate the majority of cardiovascular and respiratory diseases. The presence of PH is associated with worse outcomes, but the efficacy of current therapy is still unsatisfactory. Because Rho-kinase (ROCK) plays an important role in the pathogenesis of PH, the ROCK inhibitor fasudil is expected to contribute to PH treatment. In animal models of PH, fasudil reduced pulmonary artery pressure (PAP) and improved survival. Furthermore, the short-term efficacy and safety of fasudil in the treatment of PH are demonstrated in clinical trials. Both PAP and pulmonary vascular resistance in patients with PH are significantly decreased by intravenous or inhaled fasudil without apparent side effect. However, no clinical trial has assessed the long-term efficacy of fasudil in the treatment of PH. Limited data suggest that the mid-term use of fasudil could improve exercise capacity and reduce in-hospital mortality. We also discuss the combined use of fasudil and other drugs for PH treatment. However, these combinations have not yet been evaluated in a clinical trial. According to animal studies, the combination of fasudil with beraprost or sildenafil shows synergistic effects, whereas the combination of fasudil with bosentan has no additional ameliorating effects on PH development.
Collapse
Affiliation(s)
- Yiqing Zhang
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China.
| | - Shangjie Wu
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
106
|
Townes-Anderson E, Wang J, Halász É, Sugino I, Pitler A, Whitehead I, Zarbin M. Fasudil, a Clinically Used ROCK Inhibitor, Stabilizes Rod Photoreceptor Synapses after Retinal Detachment. Transl Vis Sci Technol 2017; 6:22. [PMID: 28660097 PMCID: PMC5482187 DOI: 10.1167/tvst.6.3.22] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 05/08/2017] [Indexed: 12/20/2022] Open
Abstract
Purpose Retinal detachment disrupts the rod-bipolar synapse in the outer plexiform layer by retraction of rod axons. We showed that breakage is due to RhoA activation whereas inhibition of Rho kinase (ROCK), using Y27632, reduces synaptic damage. We test whether the ROCK inhibitor fasudil, used for other clinical applications, can prevent synaptic injury after detachment. Methods Detachments were made in pigs by subretinal injection of balanced salt solution (BSS) or fasudil (1, 10 mM). In some animals, fasudil was injected intravitreally after BSS-induced detachment. After 2 to 4 hours, retinae were fixed for immunocytochemistry and confocal microscopy. Axon retraction was quantified by imaging synaptic vesicle label in the outer nuclear layer. Apoptosis was analyzed using propidium iodide staining. For biochemical analysis by Western blotting, retinal explants, detached from retinal pigmented epithelium, were cultured for 2 hours. Results Subretinal injection of fasudil (10 mM) reduced retraction of rod spherules by 51.3% compared to control detachments (n = 3 pigs, P = 0.002). Intravitreal injection of 10 mM fasudil, a more clinically feasible route of administration, also reduced retraction (28.7%, n = 5, P < 0.05). Controls had no photoreceptor degeneration at 2 hours, but by 4 hours apoptosis was evident. Fasudil 10 mM reduced pyknotic nuclei by 55.7% (n = 4, P < 0.001). Phosphorylation of cofilin and myosin light chain, downstream effectors of ROCK, was decreased with 30 μM fasudil (n = 8–10 explants, P < 0.05). Conclusions Inhibition of ROCK signaling with fasudil reduced photoreceptor degeneration and preserved the rod-bipolar synapse after retinal detachment. Translational Relevance These results support the possibility, previously tested with Y27632, that ROCK inhibition may attenuate synaptic damage in iatrogenic detachments.
Collapse
Affiliation(s)
- Ellen Townes-Anderson
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Jianfeng Wang
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Éva Halász
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Ilene Sugino
- Institute of Ophthalmology and Visual Science, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Amy Pitler
- Department of Microbiology, Biochemistry, and Medical Genetics, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Ian Whitehead
- Department of Microbiology, Biochemistry, and Medical Genetics, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Marco Zarbin
- Institute of Ophthalmology and Visual Science, Rutgers New Jersey Medical School, Newark, NJ, USA
| |
Collapse
|
107
|
Niego B, Lee N, Larsson P, De Silva TM, Au AEL, McCutcheon F, Medcalf RL. Selective inhibition of brain endothelial Rho-kinase-2 provides optimal protection of an in vitro blood-brain barrier from tissue-type plasminogen activator and plasmin. PLoS One 2017; 12:e0177332. [PMID: 28510599 PMCID: PMC5433693 DOI: 10.1371/journal.pone.0177332] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 04/26/2017] [Indexed: 12/13/2022] Open
Abstract
Rho-kinase (ROCK) inhibition, broadly utilised in cardiovascular disease, may protect the blood-brain barrier (BBB) during thrombolysis from rt-PA-induced damage. While the use of nonselective ROCK inhibitors like fasudil together with rt-PA may be hindered by possible hypotensive side-effects and inadequate capacity to block detrimental rt-PA activity in brain endothelial cells (BECs), selective ROCK-2 inhibition may overcome these limitations. Here, we examined ROCK-2 expression in major brain cells and compared the ability of fasudil and KD025, a selective ROCK-2 inhibitor, to attenuate rt-PA-induced BBB impairment in an in vitro human model. ROCK-2 was highly expressed relative to ROCK-1 in all human and mouse brain cell types and particularly enriched in rodent brain endothelial cells and astrocytes compared to neurons. KD025 was more potent than fasudil in attenuation of rt-PA- and plasminogen-induced BBB permeation under normoxia, but especially under stroke-like conditions. Importantly, only KD025, but not fasudil, was able to block rt-PA-dependent permeability increases, morphology changes and tight junction degradation in isolated BECs. Selective ROCK-2 inhibition further diminished rt-PA-triggered myosin phosphorylation, shape alterations and matrix metalloprotease activation in astrocytes. These findings highlight ROCK-2 as the key isoform driving BBB impairment and brain endothelial damage by rt-PA and the potential of KD025 to optimally protect the BBB during thrombolysis.
Collapse
Affiliation(s)
- Be’eri Niego
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
- * E-mail:
| | - Natasha Lee
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Pia Larsson
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - T. Michael De Silva
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Melbourne, Victoria, Australia
| | - Amanda E-Ling Au
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Fiona McCutcheon
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Robert L. Medcalf
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
108
|
Abstract
Disruption of neuronal morphology contributes to the pathology of neurodegenerative disorders such as Alzheimer's disease (AD). However, the underlying molecular mechanisms are unknown. Here, we show that postnatal deletion of Cdh1, a cofactor of the anaphase-promoting complex/cyclosome (APC/C) ubiquitin ligase in neurons [Cdh1 conditional knockout (cKO)], disrupts dendrite arborization and causes dendritic spine and synapse loss in the cortex and hippocampus, concomitant with memory impairment and neurodegeneration, in adult mice. We found that the dendrite destabilizer Rho protein kinase 2 (Rock2), which accumulates in the brain of AD patients, is an APC/CCdh1 substrate in vivo and that Rock2 protein and activity increased in the cortex and hippocampus of Cdh1 cKO mice. In these animals, inhibition of Rock activity, using the clinically approved drug fasudil, prevented dendritic network disorganization, memory loss, and neurodegeneration. Thus, APC/CCdh1-mediated degradation of Rock2 maintains the dendritic network, memory formation, and neuronal survival, suggesting that pharmacological inhibition of aberrantly accumulated Rock2 may be a suitable therapeutic strategy against neurodegeneration.
Collapse
|
109
|
Karsy M, Brock A, Guan J, Taussky P, Kalani MYS, Park MS. Neuroprotective strategies and the underlying molecular basis of cerebrovascular stroke. Neurosurg Focus 2017; 42:E3. [DOI: 10.3171/2017.1.focus16522] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Stroke is a leading cause of disability in the US. Although there has been significant progress in the area of medical and surgical thrombolytic technologies, neuroprotective agents to prevent secondary cerebral injury and to minimize disability remain limited. Only limited success has been reported in preclinical and clinical trials evaluating a variety of compounds. In this review, the authors discuss the most up-to-date information regarding the underlying molecular biology of stroke as well as strategies that aim to mitigate this complex signaling cascade. Results of historical research trials involving N-methyl-d-aspartate and α-amino-3-hydroxy-5-methyl-4-isoxazole propionate receptor antagonists, clomethiazole, antioxidants, citicoline, nitric oxide, and immune regulators have laid the groundwork for current progress. In addition, more recent studies involving therapeutic hypothermia, magnesium, albumin, glyburide, uric acid, and a variety of other treatments have provided more options. The use of neuroprotective agents in combination or with existing thrombolytic treatments may be one of many exciting areas of further development. Although past trials of neuroprotective agents in ischemic stroke have been limited, significant insights into mechanisms of stroke, animal models, and trial design have incrementally improved approaches for future therapies.
Collapse
|
110
|
Rho-kinase inhibitor prevents acute injury against transient focal cerebral ischemia by enhancing the expression and function of GABA receptors in rats. Eur J Pharmacol 2017; 797:134-142. [DOI: 10.1016/j.ejphar.2017.01.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 01/11/2017] [Accepted: 01/17/2017] [Indexed: 02/01/2023]
|
111
|
Fukuta T, Asai T, Yanagida Y, Namba M, Koide H, Shimizu K, Oku N. Combination therapy with liposomal neuroprotectants and tissue plasminogen activator for treatment of ischemic stroke. FASEB J 2017; 31:1879-1890. [PMID: 28082354 DOI: 10.1096/fj.201601209r] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 01/03/2017] [Indexed: 12/24/2022]
Abstract
For ischemic stroke treatment, extension of the therapeutic time window (TTW) of thrombolytic therapy with tissue plasminogen activator (tPA) and amelioration of secondary ischemia/reperfusion (I/R) injury are most desirable. Our previous studies have indicated that liposomal delivery of neuroprotectants into an ischemic region is effective for stroke treatment. In the present study, for solving the above problems in the clinical setting, the usefulness of combination therapy with tPA and liposomal fasudil (fasudil-Lip) was investigated in ischemic stroke model rats with photochemically induced thrombosis, with clots that were dissolved by tPA. Treatment with tPA 3 h after occlusion markedly increased blood-brain barrier permeability and activated matrix metalloproteinase (MMP)-2 and -9, which are involved in cerebral hemorrhage. However, an intravenous administration of fasudil-Lip before tPA markedly suppressed the increase in permeability and the MMP activation stemming from tPA. The combination treatment showed significantly larger neuroprotective effects, even in the case of delayed tPA administration compared with each treatment alone or the tPA/fasudil-treated group. These findings suggest that treatment with fasudil-Lip before tPA could decrease the risk of tPA-derived cerebral hemorrhage and extend the TTW of tPA and that the combination therapy could be a useful therapeutic option for ischemic stroke.-Fukuta, T., Asai, T., Yanagida, Y., Namba, M., Koide, H., Shimizu, K., Oku, N. Combination therapy with liposomal neuroprotectants and tissue plasminogen activator for treatment of ischemic stroke.
Collapse
Affiliation(s)
- Tatsuya Fukuta
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Japan; and.,Japan Society for the Promotion of Science, Tokyo, Japan
| | - Tomohiro Asai
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Japan; and
| | - Yosuke Yanagida
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Japan; and
| | - Mio Namba
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Japan; and
| | - Hiroyuki Koide
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Japan; and
| | - Kosuke Shimizu
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Japan; and
| | - Naoto Oku
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Japan; and
| |
Collapse
|
112
|
Sun Z, Wu X, Li W, Peng H, Shen X, Ma L, Liu H, Li H. RhoA/rock signaling mediates peroxynitrite-induced functional impairment of Rat coronary vessels. BMC Cardiovasc Disord 2016; 16:193. [PMID: 27724862 PMCID: PMC5057502 DOI: 10.1186/s12872-016-0372-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 09/28/2016] [Indexed: 01/03/2023] Open
Abstract
Background Diabetes-induced vascular dysfunction may arise from reduced nitric oxide (NO) availability, following interaction with superoxide to form peroxynitrite. Peroxynitrite can induce formation of 3-nitrotyrosine-modified proteins. RhoA/ROCK signaling is also involved in diabetes-induced vascular dysfunction. The study aimed to investigate possible links between Rho/ROCK signaling, hyperglycemia, and peroxynitrite in small coronary arteries. Methods Rat small coronary arteries were exposed to normal (NG; 5.5 mM) or high (HG; 23 mM) D-glucose. Vascular ring constriction to 3 mM 4-aminopyridine and dilation to 1 μM forskolin were measured. Protein expression (immunohistochemistry and western blot), mRNA expression (real-time PCR), and protein activity (luminescence-based G-LISA and kinase activity spectroscopy assays) of RhoA, ROCK1, and ROCK2 were determined. Results Vascular ring constriction and dilation were smaller in the HG group than in the NG group (P < 0.05); inhibition of RhoA or ROCK partially reversed the effects of HG. Peroxynitrite impaired vascular ring constriction/dilation; this was partially reversed by inhibition of RhoA or ROCK. Protein and mRNA expressions of RhoA, ROCK1, and ROCK2 were higher under HG than NG (P < 0.05). This HG-induced upregulation was attenuated by inhibition of RhoA or ROCK (P < 0.05). HG increased RhoA, ROCK1, and ROCK2 activity (P < 0.05). Peroxynitrite also enhanced RhoA, ROCK1, and ROCK2 activity; these actions were partially inhibited by 100 μM urate (peroxynitrite scavenger). Exogenous peroxynitrite had no effect on the expression of the voltage-dependent K+ channels 1.2 and 1.5. Conclusions Peroxynitrite-induced coronary vascular dysfunction may be mediated, at least in part, through increased expressions and activities of RhoA, ROCK1, and ROCK2.
Collapse
Affiliation(s)
- Zhijun Sun
- Department of Heart Center, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China
| | - Xing Wu
- Department of Heart Center, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China
| | - Weiping Li
- Department of Heart Center, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China
| | - Hui Peng
- Department of Heart Center, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China
| | - Xuhua Shen
- Department of Heart Center, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China
| | - Lu Ma
- Beijing Key Laboratory of Metabolic Disturbance Related Cardiovascular Disease, Beijing, People's Republic of China
| | - Huirong Liu
- Beijing Key Laboratory of Metabolic Disturbance Related Cardiovascular Disease, Beijing, People's Republic of China
| | - Hongwei Li
- Department of Heart Center, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China.
| |
Collapse
|
113
|
Pellegrino PR, Schiller AM, Haack KKV, Zucker IH. Central Angiotensin-II Increases Blood Pressure and Sympathetic Outflow via Rho Kinase Activation in Conscious Rabbits. Hypertension 2016; 68:1271-1280. [PMID: 27672026 DOI: 10.1161/hypertensionaha.116.07792] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 08/25/2016] [Indexed: 01/08/2023]
Abstract
Elevated sympathetic tone and activation of the renin-angiotensin system are pathophysiologic hallmarks of hypertension, and the interactions between these systems are particularly deleterious. The importance of Rho kinase as a mediator of the effects of angiotensin-II (AngII) in the periphery is clear, but the role of Rho kinase in sympathoexcitation caused by central AngII is not well established. We hypothesized that AngII mediates its effects in the brain by the activation of the RhoA/Rho kinase pathway. Chronically instrumented, conscious rabbits received the following intracerebroventricular infusion treatments for 2 weeks via osmotic minipump: AngII, Rho kinase inhibitor Fasudil, AngII plus Fasudil, or a vehicle control. AngII increased mean arterial pressure over the course of the infusion, and this effect was prevented by the coadministration of Fasudil. AngII increased cardiac and vascular sympathetic outflow as quantified by the heart rate response to metoprolol and the depressor effect of hexamethonium; coadministration of Fasudil abolished both of these effects. AngII increased baseline renal sympathetic nerve activity in conscious animals and impaired baroreflex control of sympathetic nerve activity; again Fasudil coinfusion prevented these effects. Each of these end points showed a statistically significant interaction between AngII and Fasudil. Quantitative immunofluorescence of brain slices confirmed that Rho kinase activity was increased by AngII and decreased by Fasudil. Taken together, these data indicate that hypertension, elevated sympathetic outflow, and baroreflex dysfunction caused by central AngII are mediated by Rho kinase activation and suggest that Rho kinase inhibition may be an important therapeutic target in sympathoexcitatory cardiovascular diseases.
Collapse
Affiliation(s)
- Peter R Pellegrino
- From the Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (P.R.P., A.M.S., I.H.Z.); U.S. Army Institute of Surgical Research, Fort Sam Houston, TX (A.M.S.); and Department of Pediatrics, Emory University School of Medicine, Atlanta, GA (K.K.V.H.)
| | - Alicia M Schiller
- From the Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (P.R.P., A.M.S., I.H.Z.); U.S. Army Institute of Surgical Research, Fort Sam Houston, TX (A.M.S.); and Department of Pediatrics, Emory University School of Medicine, Atlanta, GA (K.K.V.H.)
| | - Karla K V Haack
- From the Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (P.R.P., A.M.S., I.H.Z.); U.S. Army Institute of Surgical Research, Fort Sam Houston, TX (A.M.S.); and Department of Pediatrics, Emory University School of Medicine, Atlanta, GA (K.K.V.H.)
| | - Irving H Zucker
- From the Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (P.R.P., A.M.S., I.H.Z.); U.S. Army Institute of Surgical Research, Fort Sam Houston, TX (A.M.S.); and Department of Pediatrics, Emory University School of Medicine, Atlanta, GA (K.K.V.H.).
| |
Collapse
|
114
|
De Silva TM, Kinzenbaw DA, Modrick ML, Reinhardt LD, Faraci FM. Heterogeneous Impact of ROCK2 on Carotid and Cerebrovascular Function. Hypertension 2016; 68:809-17. [PMID: 27432870 PMCID: PMC4982851 DOI: 10.1161/hypertensionaha.116.07430] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 06/11/2016] [Indexed: 01/05/2023]
Abstract
Rho kinase (ROCK) has been implicated in physiological and pathophysiological processes, including regulation of vascular function. ROCK signaling is thought to be a critical contributor to cardiovascular disease, including hypertension and effects of angiotensin II (Ang II). Two isoforms of ROCK (1 and 2) have been identified and are expressed in vascular cells. In this study, we examined the importance of ROCK2 in relation to vessel function using several models and a novel inhibitor of ROCK2. First, incubation of carotid arteries with the direct RhoA activator CN-03 or Ang II impaired endothelium-dependent relaxation by ≈40% to 50% (P<0.05) without altering endothelium-independent relaxation. Both CN-03- and Ang II-induced endothelial dysfunction was prevented by Y-27632 (an inhibitor of both ROCK isoforms) or the selective ROCK2 inhibitor SLX-2119. In contrast, SLX-2119 had little effect on contraction of carotid arteries to receptor-mediated agonists (serotonin, phenylephrine, vasopressin, or U46619). Second, in basilar arteries, SLX-2119 inhibited constriction to Ang II by ≈90% without significantly affecting responses to serotonin or KCl. Third, in isolated pressurized brain parenchymal arterioles, SLX-2119 inhibited myogenic tone in a concentration-dependent manner (eg, 1 μmol/L SLX-2119 dilated by 79±4%). Finally, SLX-2119 dilated small pial arterioles in vivo, an effect that was augmented by inhibition of nitric oxide synthase. These findings suggest that ROCK2 has major, but heterogeneous, effects on function of endothelium and vascular muscle. The data support the concept that aberrant ROCK2 signaling may be a key contributor to select aspects of large and small vessel disease, including Ang II-induced endothelial dysfunction.
Collapse
Affiliation(s)
- T Michael De Silva
- From the Departments of Internal Medicine (T.M.D.S., D.A.K., M.L.M., L.D.R., F.M.F.) and Pharmacology (F.M.F.), Francois M. Abboud Cardiovascular Center, The University of Iowa Carver College of Medicine; Iowa City Veterans Affairs Healthcare System (F.M.F.); and Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Australia (T.M.D.S.)
| | - Dale A Kinzenbaw
- From the Departments of Internal Medicine (T.M.D.S., D.A.K., M.L.M., L.D.R., F.M.F.) and Pharmacology (F.M.F.), Francois M. Abboud Cardiovascular Center, The University of Iowa Carver College of Medicine; Iowa City Veterans Affairs Healthcare System (F.M.F.); and Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Australia (T.M.D.S.)
| | - Mary L Modrick
- From the Departments of Internal Medicine (T.M.D.S., D.A.K., M.L.M., L.D.R., F.M.F.) and Pharmacology (F.M.F.), Francois M. Abboud Cardiovascular Center, The University of Iowa Carver College of Medicine; Iowa City Veterans Affairs Healthcare System (F.M.F.); and Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Australia (T.M.D.S.)
| | - Lindsey D Reinhardt
- From the Departments of Internal Medicine (T.M.D.S., D.A.K., M.L.M., L.D.R., F.M.F.) and Pharmacology (F.M.F.), Francois M. Abboud Cardiovascular Center, The University of Iowa Carver College of Medicine; Iowa City Veterans Affairs Healthcare System (F.M.F.); and Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Australia (T.M.D.S.)
| | - Frank M Faraci
- From the Departments of Internal Medicine (T.M.D.S., D.A.K., M.L.M., L.D.R., F.M.F.) and Pharmacology (F.M.F.), Francois M. Abboud Cardiovascular Center, The University of Iowa Carver College of Medicine; Iowa City Veterans Affairs Healthcare System (F.M.F.); and Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Australia (T.M.D.S.).
| |
Collapse
|
115
|
Abstract
In past decades, growing evidence from basic and clinical researches reveal that small guanosine triphosphate binding protein ras homolog gene family, member A (RhoA) and its main effector Rho-associated kinase (ROCK) play central and complex roles in cardiovascular systems, and increasing RhoA and ROCK activity is associated with a broad range of cardiovascular diseases such as congestive heart failure, atherosclerosis, and hypertension. Favorable outcomes have been observed with ROCK inhibitors treatment. In this review, we briefly summarize the pathophysiological roles of RhoA/ROCK signaling pathway on cardiovascular system, displaying the potential benefits in the cardiovascular system with controlling RhoA/ROCK signaling pathway.
Collapse
|
116
|
Henderson BW, Gentry EG, Rush T, Troncoso JC, Thambisetty M, Montine TJ, Herskowitz JH. Rho-associated protein kinase 1 (ROCK1) is increased in Alzheimer's disease and ROCK1 depletion reduces amyloid-β levels in brain. J Neurochem 2016; 138:525-31. [PMID: 27246255 DOI: 10.1111/jnc.13688] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 05/16/2016] [Accepted: 05/30/2016] [Indexed: 12/01/2022]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia and mitigating amyloid-β (Aβ) levels may serve as a rational therapeutic avenue to slow AD progression. Pharmacologic inhibition of the Rho-associated protein kinases (ROCK1 and ROCK2) is proposed to curb Aβ levels, and mechanisms that underlie ROCK2's effects on Aβ production are defined. How ROCK1 affects Aβ generation remains a critical barrier. Here, we report that ROCK1 protein levels were elevated in mild cognitive impairment due to AD (MCI) and AD brains compared to controls. Aβ42 oligomers marginally increased ROCK1 and ROCK2 protein levels in neurons but strongly induced phosphorylation of Lim kinase 1 (LIMK1), suggesting that Aβ42 activates ROCKs. RNAi depletion of ROCK1 or ROCK2 suppressed endogenous Aβ40 production in neurons, and Aβ40 levels were reduced in brains of ROCK1 heterozygous knock-out mice compared to wild-type littermate controls. ROCK1 knockdown decreased amyloid precursor protein (APP), and treatment with bafilomycin accumulated APP levels in neurons depleted of ROCK1. These observations suggest that reduction of ROCK1 diminishes Aβ levels by enhancing APP protein degradation. Collectively, these findings support the hypothesis that both ROCK1 and ROCK2 are therapeutic targets to combat Aβ production in AD. Mitigating amyloid-β (Aβ) levels is a rational strategy for Alzheimer's disease (AD) treatment, however, therapeutic targets with clinically available drugs are lacking. We hypothesize that Aβ accumulation in mild cognitive impairment because of AD (MCI) and AD activates the RhoA/ROCK pathway which in turn fuels production of Aβ. Escalation of this cycle over the course of many years may contribute to the buildup of amyloid pathology in MCI and/or AD.
Collapse
Affiliation(s)
- Benjamin W Henderson
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Erik G Gentry
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Travis Rush
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Juan C Troncoso
- Departments of Pathology and Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Madhav Thambisetty
- Unit of Clinical and Translational Neuroscience, Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland, USA
| | - Thomas J Montine
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Jeremy H Herskowitz
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Department of Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
117
|
Zychowski KE, Lucas SN, Sanchez B, Herbert G, Campen MJ. Hypoxia-induced pulmonary arterial hypertension augments lung injury and airway reactivity caused by ozone exposure. Toxicol Appl Pharmacol 2016; 305:40-45. [PMID: 27286659 DOI: 10.1016/j.taap.2016.06.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/10/2016] [Accepted: 06/03/2016] [Indexed: 11/29/2022]
Abstract
Ozone (O3)-related cardiorespiratory effects are a growing public health concern. Ground level O3 can exacerbate pre-existing respiratory conditions; however, research regarding therapeutic interventions to reduce O3-induced lung injury is limited. In patients with chronic obstructive pulmonary disease, hypoxia-associated pulmonary hypertension (HPH) is a frequent comorbidity that is difficult to treat clinically, yet associated with increased mortality and frequency of exacerbations. In this study, we hypothesized that established HPH would confer vulnerability to acute O3 pulmonary toxicity. Additionally, we tested whether improvement of pulmonary endothelial barrier integrity via rho-kinase inhibition could mitigate pulmonary inflammation and injury. To determine if O3 exacerbated HPH, male C57BL/6 mice were subject to either 3 weeks continuous normoxia (20.9% O2) or hypoxia (10.0% O2), followed by a 4-h exposure to either 1ppm O3 or filtered air (FA). As an additional experimental intervention fasudil (20mg/kg) was administered intraperitoneally prior to and after O3 exposures. As expected, hypoxia significantly increased right ventricular pressure and hypertrophy. O3 exposure in normoxic mice caused lung inflammation but not injury, as indicated by increased cellularity and edema in the lung. However, in hypoxic mice, O3 exposure led to increased inflammation and edema, along with a profound increase in airway hyperresponsiveness to methacholine. Fasudil administration resulted in reduced O3-induced lung injury via the enhancement of pulmonary endothelial barrier integrity. These results indicate that increased pulmonary vascular pressure may enhance lung injury, inflammation and edema when exposed to pollutants, and that enhancement of pulmonary endothelial barrier integrity may alleviate such vulnerability.
Collapse
Affiliation(s)
- Katherine E Zychowski
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, NM 87131, United States
| | - Selita N Lucas
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, NM 87131, United States
| | - Bethany Sanchez
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, NM 87131, United States
| | - Guy Herbert
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, NM 87131, United States
| | - Matthew J Campen
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, NM 87131, United States.
| |
Collapse
|
118
|
Abstract
Hypertensive cardiac remodeling is characterized by left ventricular hypertrophy and interstitial fibrosis, which can lead to heart failure with preserved ejection fraction. The Rho-associated coiled-coil containing kinases (ROCKs) are members of the serine/threonine protein kinase family, which mediates the downstream effects of the small GTP-binding protein RhoA. There are 2 isoforms: ROCK1 and ROCK2. They have different functions in different types of cells and tissues. There is growing evidence that ROCKs contribute to the development of cardiovascular diseases, including cardiac fibrosis, hypertrophy, and subsequent heart failure. Recent experimental studies using ROCK inhibitors, such as fasudil, have shown the benefits of ROCK inhibition in cardiac remodeling. Mice lacking each ROCK isoform also exhibit reduced myocardial fibrosis in a variety of pathological models of cardiac remodeling. Indeed, clinical studies with fasudil have suggested that ROCKs could be potential novel therapeutic targets for cardiovascular diseases. In this review, we summarize the current understanding of the roles of ROCKs in the development of cardiac fibrosis and hypertrophy and discuss their therapeutic potential for deleterious cardiac remodeling. (Circ J 2016; 80: 1491-1498).
Collapse
Affiliation(s)
- Toru Shimizu
- Section of Cardiology, Department of Medicine, University of Chicago
| | | |
Collapse
|
119
|
Rho Kinase Inhibition as a Therapeutic for Progressive Supranuclear Palsy and Corticobasal Degeneration. J Neurosci 2016; 36:1316-23. [PMID: 26818518 DOI: 10.1523/jneurosci.2336-15.2016] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
UNLABELLED Progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD) are neurodegenerative four-repeat tauopathies with no cure. Mitigating pathogenic tau levels is a rational strategy for tauopathy treatment, but therapeutic targets with clinically available drugs are lacking. Here, we report that protein levels of the Rho-associated protein kinases (ROCK1 and ROCK2), p70 S6 kinase (S6K), and mammalian target of rapamycin (mTOR) were increased in PSP and CBD brains. RNAi depletion of ROCK1 or ROCK2 reduced tau mRNA and protein level in human neuroblastoma cells. However, additional phenotypes were observed under ROCK2 knockdown, including decreased S6K and phosphorylated mTOR levels. Pharmacologic inhibition of Rho kinases in neurons diminished detergent-soluble and -insoluble tau through a combination of autophagy enhancement and tau mRNA reduction. Fasudil, a clinically approved ROCK inhibitor, suppressed rough eye phenotype and mitigated pathogenic tau levels by inducing autophagic pathways in a Drosophila model of tauopathy. Collectively, these findings highlight the Rho kinases as rational therapeutic targets to combat tau accumulation in PSP and CBD. SIGNIFICANCE STATEMENT Studies of progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD) suggest that mitigating pathogenic tau levels is a rational strategy for tauopathy treatment. In this report, the Rho-associated protein kinases (ROCK1 and ROCK2) are identified as novel drug targets for PSP and CBD. We show that elevated insoluble tau levels are associated with increased ROCK1 and ROCK2 in PSP and CBD brains, whereas experiments in cellular and animal models identify pharmacologic inhibition of ROCKs as a mechanism-based approach to reduce tau levels. Our study correlates bona fide changes in PSP and CBD brains with cellular models, identifies drug targets, and tests the therapeutic in vivo.
Collapse
|
120
|
Bye N, Christie KJ, Turbic A, Basrai HS, Turnley AM. Rho kinase inhibition following traumatic brain injury in mice promotes functional improvement and acute neuron survival but has little effect on neurogenesis, glial responses or neuroinflammation. Exp Neurol 2016; 279:86-95. [DOI: 10.1016/j.expneurol.2016.02.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/08/2016] [Accepted: 02/15/2016] [Indexed: 12/27/2022]
|
121
|
Neuroprotection against cerebral ischemia/reperfusion injury by intravenous administration of liposomal fasudil. Int J Pharm 2016; 506:129-37. [PMID: 27107903 DOI: 10.1016/j.ijpharm.2016.04.046] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 03/25/2016] [Accepted: 04/15/2016] [Indexed: 12/31/2022]
Abstract
Fasudil, a Rho-kinase inhibitor, is a promising neuroprotectant against ischemic stroke; however, its low bioavailability is an obstacle to be overcome. Our previous study revealed that the liposomal drug delivery system is a hopeful strategy to increase the therapeutic efficacy of neuroprotectants. In the present study, the usefulness of intravenously administered liposomal fasudil for cerebral ischemia/reperfusion (I/R) injury treatment was examined in transient middle cerebral artery occlusion (t-MCAO) rats. The results showed that PEGylated liposomes of approximately 100nm in diameter accumulated more extensively in the I/R region compared with those of over 200nm. Confocal images showed that fluorescence-labeled liposomal fasudil was widely distributed in the I/R region, and was not noticeably taken up by microglia, which are well-known resident macrophages in the brain, and neuronal cells. These data indicated that liposomal fasudil mainly exerted its pharmacological activity by releasing fasudil from the liposomes in the I/R region. Moreover, liposomal fasudil effectively suppressed neutrophil invasion and brain cell damage in the t-MCAO rats, resulting in amelioration of their motor function deficits. These findings demonstrated both the importance of particle size for neuroprotectant delivery and the effectiveness of liposomal fasudil for the treatment of cerebral I/R injury.
Collapse
|
122
|
Moon JS, Lee SH, Han SH, Kim EJ, Cho H, Lee W, Kim MK, Kim TE, Park HJ, Rhee JK, Kim SJ, Cho SW, Han SH, Oh JW. Inhibition of hepatitis C virus in mouse models by lipidoid nanoparticle-mediated systemic delivery of siRNA against PRK2. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:1489-98. [PMID: 27013134 DOI: 10.1016/j.nano.2016.02.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 02/10/2016] [Accepted: 02/15/2016] [Indexed: 12/12/2022]
Abstract
Host-targeting antivirals have an advantage over direct-acting antivirals in that they have a high genetic barrier to resistance. Here, we describe in vivo anti-hepatitis C virus (HCV) efficacy of a potent siRNA targeting the protein kinase C-related kinase 2 (PRK2), which phosphorylates HCV NS5B RNA-dependent RNA polymerase and promotes HCV replication. PRK2-silencing reduced the phosphorylated NS5B level and resulted in inhibition of NS5B RdRp activity to decrease HCV genome abundance. Systemic administration of lipidoid nanoparticle-formulated PRK2 siRNA (once every three days for a total of three injections at a dose of 3mgkg(-1)) resulted in a 3.72 and 1.96 log10 reduction in serum HCV RNA titer, in mouse subcutaneous and orthotopic xenograft models for HCV replication, respectively. Our results verify the essential role of PRK2 in HCV replication and offer a host-targeting anti-HCV siRNA therapy that might be beneficial for non-responders to current treatment regimens.
Collapse
Affiliation(s)
- Jae-Su Moon
- Department of Biotechnology, Yonsei University, Seoul, Korea
| | - Seung-Hoon Lee
- Department of Biotechnology, Yonsei University, Seoul, Korea
| | - Song-Hee Han
- Department of Biotechnology, Yonsei University, Seoul, Korea
| | - Eun-Jung Kim
- Department of Biotechnology, Yonsei University, Seoul, Korea
| | - Hee Cho
- Department of Biotechnology, Yonsei University, Seoul, Korea
| | - Wooseong Lee
- Department of Biotechnology, Yonsei University, Seoul, Korea
| | - Mi-Kyung Kim
- Department of Biotechnology, Yonsei University, Seoul, Korea
| | - Tae-Eun Kim
- Department of Biotechnology, Yonsei University, Seoul, Korea
| | - Hyun-Ji Park
- Department of Biotechnology, Yonsei University, Seoul, Korea
| | - Jin-Kyu Rhee
- Western Seoul Center of Korea Basic Science Institute, Seoul, Korea
| | - Seong-Jun Kim
- Department of Biotechnology, Yonsei University, Seoul, Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul, Korea
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Korea
| | - Jong-Won Oh
- Department of Biotechnology, Yonsei University, Seoul, Korea.
| |
Collapse
|
123
|
Abeysinghe HCS, Phillips EL, Chin-Cheng H, Beart PM, Roulston CL. Modulating Astrocyte Transition after Stroke to Promote Brain Rescue and Functional Recovery: Emerging Targets Include Rho Kinase. Int J Mol Sci 2016; 17:288. [PMID: 26927079 PMCID: PMC4813152 DOI: 10.3390/ijms17030288] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 01/26/2016] [Accepted: 02/05/2016] [Indexed: 01/13/2023] Open
Abstract
Stroke is a common and serious condition, with few therapies. Whilst previous focus has been directed towards biochemical events within neurons, none have successfully prevented the progression of injury that occurs in the acute phase. New targeted treatments that promote recovery after stroke might be a better strategy and are desperately needed for the majority of stroke survivors. Cells comprising the neurovascular unit, including blood vessels and astrocytes, present an alternative target for supporting brain rescue and recovery in the late phase of stroke, since alteration in the unit also occurs in regions outside of the lesion. One of the major changes in the unit involves extensive morphological transition of astrocytes resulting in altered energy metabolism, decreased glutamate reuptake and recycling, and retraction of astrocyte end feed from both blood vessels and neurons. Whilst globally inhibiting transitional change in astrocytes after stroke is reported to result in further damage and functional loss, we discuss the available evidence to suggest that the transitional activation of astrocytes after stroke can be modulated for improved outcomes. In particular, we review the role of Rho-kinase (ROCK) in reactive gliosis and show that inhibiting ROCK after stroke results in reduced scar formation and improved functional recovery.
Collapse
Affiliation(s)
- Hima Charika S Abeysinghe
- Neurotrauma Research, Department of Medicine, St Vincent's Campus, University of Melbourne, Parkville, VIC 3065, Australia.
- Department of Surgery, St Vincent's Campus, University of Melbourne, Parkville, VIC 3065, Australia.
| | - Ellie L Phillips
- Department of Biochemistry and Molecular Biology, Bio21 Insitute, University of Melbourne, Parkville, VIC 3010, Australia.
| | - Heung Chin-Cheng
- Department of Biochemistry and Molecular Biology, Bio21 Insitute, University of Melbourne, Parkville, VIC 3010, Australia.
| | - Philip M Beart
- The Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, Parkville, VIC 3010, Australia.
| | - Carli L Roulston
- Neurotrauma Research, Department of Medicine, St Vincent's Campus, University of Melbourne, Parkville, VIC 3065, Australia.
| |
Collapse
|
124
|
Effect of a Rho Kinase Inhibitor on Cognitive Impairment Induced by Chronic Cerebral Hypoperfusion in Rats. NEUROPHYSIOLOGY+ 2016. [DOI: 10.1007/s11062-016-9548-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
125
|
Donegan RK, Lieberman RL. Discovery of Molecular Therapeutics for Glaucoma: Challenges, Successes, and Promising Directions. J Med Chem 2016; 59:788-809. [PMID: 26356532 PMCID: PMC5547565 DOI: 10.1021/acs.jmedchem.5b00828] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Glaucoma, a heterogeneous ocular disorder affecting ∼60 million people worldwide, is characterized by painless neurodegeneration of retinal ganglion cells (RGCs), resulting in irreversible vision loss. Available therapies, which decrease the common causal risk factor of elevated intraocular pressure, delay, but cannot prevent, RGC death and blindness. Notably, it is changes in the anterior segment of the eye, particularly in the drainage of aqueous humor fluid, which are believed to bring about changes in pressure. Thus, it is primarily this region whose properties are manipulated in current and emerging therapies for glaucoma. Here, we focus on the challenges associated with developing treatments, review the available experimental methods to evaluate the therapeutic potential of new drugs, describe the development and evaluation of emerging Rho-kinase inhibitors and adenosine receptor ligands that offer the potential to improve aqueous humor outflow and protect RGCs simultaneously, and present new targets and approaches on the horizon.
Collapse
Affiliation(s)
- Rebecca K Donegan
- School of Chemistry and Biochemistry, Georgia Institute of Technology , 901 Atlantic Drive NW, Atlanta, Georgia 30332-0400, United States
| | - Raquel L Lieberman
- School of Chemistry and Biochemistry, Georgia Institute of Technology , 901 Atlantic Drive NW, Atlanta, Georgia 30332-0400, United States
| |
Collapse
|
126
|
Liu FT, Yang YJ, Wu JJ, Li S, Tang YL, Zhao J, Liu ZY, Xiao BG, Zuo J, Liu W, Wang J. Fasudil, a Rho kinase inhibitor, promotes the autophagic degradation of A53T α-synuclein by activating the JNK 1/Bcl-2/beclin 1 pathway. Brain Res 2016; 1632:9-18. [DOI: 10.1016/j.brainres.2015.12.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 11/22/2015] [Accepted: 12/03/2015] [Indexed: 12/21/2022]
|
127
|
He Q, Li YH, Guo SS, Wang Y, Lin W, Zhang Q, Wang J, Ma CG, Xiao BG. Inhibition of Rho-kinase by Fasudil protects dopamine neurons and attenuates inflammatory response in an intranasal lipopolysaccharide-mediated Parkinson's model. Eur J Neurosci 2016; 43:41-52. [PMID: 26565388 DOI: 10.1111/ejn.13132] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Revised: 07/13/2015] [Accepted: 11/05/2015] [Indexed: 12/12/2022]
Abstract
Microglia activation and inflammatory factors in brain microenvironment are associated with degeneration of neurons in the substantia nigra (SN) of Parkinson's disease (PD) patients and various PD models. There is increasing evidence that the Rho/ROCK (Rho kinase) signalling pathway may play a critical role in the inflammatory response, and ROCK inhibitor has been reported to have neuroprotective effects. In this study, we examined the neuroprotective potential and possible mechanism of ROCK inhibitor Fasudil in an intranasal lipopolysaccharide (LPS)-induced PD model. ROCK was activated with LPS stimulation and inhibited by Fasudil treatment in this PD model. Behavioural tests demonstrated a clear improvement in motor performance after Fasudil treatment. Furthermore, Fasudil resulted in a significant attenuation of dopamine cell loss, α-synuclein accumulation and inflammatory response with the reversion of inflammatory M1 to anti-inflammatory M2 microglia, decreased NF-кB activation, and IL-12 and TNF-α generation in the SN and olfactory bulb in this model. This study establishes a role for Fasudil in protecting against LPS-mediated dopamine degeneration and provides a therapeutic strategy for the treatment of PD.
Collapse
Affiliation(s)
- Qing He
- Department of Neurology, Shanghai Ninth People's Hospital Affiliated Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Yan-hua Li
- Department of Neurology, Institute of Brain Science, Medical School, Shanxi Datong University, Datong, China
| | - Si-si Guo
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Ying Wang
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Wei Lin
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Qiong Zhang
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Jian Wang
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Cun-gen Ma
- Department of Neurology, Institute of Brain Science, Medical School, Shanxi Datong University, Datong, China
- '2011'Collaborative Innovation Center/Research Center of Neurobiology, Shanxi University of Traditional Chinese Medicine, Taiyuan, China
| | - Bao-Guo Xiao
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| |
Collapse
|
128
|
Hartmann S, Ridley AJ, Lutz S. The Function of Rho-Associated Kinases ROCK1 and ROCK2 in the Pathogenesis of Cardiovascular Disease. Front Pharmacol 2015; 6:276. [PMID: 26635606 PMCID: PMC4653301 DOI: 10.3389/fphar.2015.00276] [Citation(s) in RCA: 228] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 11/03/2015] [Indexed: 01/26/2023] Open
Abstract
Rho-associated kinases ROCK1 and ROCK2 are serine/threonine kinases that are downstream targets of the small GTPases RhoA, RhoB, and RhoC. ROCKs are involved in diverse cellular activities including actin cytoskeleton organization, cell adhesion and motility, proliferation and apoptosis, remodeling of the extracellular matrix and smooth muscle cell contraction. The role of ROCK1 and ROCK2 has long been considered to be similar; however, it is now clear that they do not always have the same functions. Moreover, depending on their subcellular localization, activation, and other environmental factors, ROCK signaling can have different effects on cellular function. With respect to the heart, findings in isoform-specific knockout mice argue for a role of ROCK1 and ROCK2 in the pathogenesis of cardiac fibrosis and cardiac hypertrophy, respectively. Increased ROCK activity could play a pivotal role in processes leading to cardiovascular diseases such as hypertension, pulmonary hypertension, angina pectoris, vasospastic angina, heart failure, and stroke, and thus ROCK activity is a potential new biomarker for heart disease. Pharmacological ROCK inhibition reduces the enhanced ROCK activity in patients, accompanied with a measurable improvement in medical condition. In this review, we focus on recent findings regarding ROCK signaling in the pathogenesis of cardiovascular disease, with a special focus on differences between ROCK1 and ROCK2 function.
Collapse
Affiliation(s)
- Svenja Hartmann
- Institute of Pharmacology, University Medical Center Göttingen, Georg-August-University Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research, Göttingen, Germany
- Randall Division of Cell and Molecular Biophysics, King’s College London, London, UK
| | - Anne J. Ridley
- Randall Division of Cell and Molecular Biophysics, King’s College London, London, UK
| | - Susanne Lutz
- Institute of Pharmacology, University Medical Center Göttingen, Georg-August-University Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research, Göttingen, Germany
| |
Collapse
|
129
|
Loirand G. Rho Kinases in Health and Disease: From Basic Science to Translational Research. Pharmacol Rev 2015; 67:1074-95. [PMID: 26419448 DOI: 10.1124/pr.115.010595] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Rho-associated kinases ROCK1 and ROCK2 are key regulators of actin cytoskeleton dynamics downstream of Rho GTPases that participate in the control of important physiologic functions, S including cell contraction, migration, proliferation, adhesion, and inflammation. Several excellent review articles dealing with ROCK function and regulation have been published over the past few years. Although a brief overview of general molecular, biochemical, and functional properties of ROCKs is included, an effort has been made to produce an original work by collecting and synthesizing recent studies aimed at translating basic discoveries from cell and experimental models into knowledge of human physiology, pathophysiological mechanisms, and medical therapeutics. This review points out the specificity and distinct roles of ROCK1 and ROCK2 isoforms highlighted in the last few years. Results obtained from genetically modified mice and genetic analysis in humans are discussed. This review also addresses the involvement of ROCKs in human diseases and the potential use of ROCK activity as a biomarker or a pharmacological target for specific inhibitors.
Collapse
Affiliation(s)
- Gervaise Loirand
- Institut National de la Santé et de la Recherche Médicale UMR1087, Université de Nantes, CHU Nantes, l'institut du thorax, Nantes, France
| |
Collapse
|
130
|
Kajikawa M, Noma K, Nakashima A, Maruhashi T, Iwamoto Y, Matsumoto T, Iwamoto A, Oda N, Hidaka T, Kihara Y, Aibara Y, Chayama K, Sasaki S, Kato M, Dote K, Goto C, Liao JK, Higashi Y. Rho-associated kinase activity is an independent predictor of cardiovascular events in acute coronary syndrome. Hypertension 2015; 66:892-9. [PMID: 26283039 PMCID: PMC4989242 DOI: 10.1161/hypertensionaha.115.05587] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 07/25/2015] [Indexed: 11/16/2022]
Abstract
Rho-associated kinases play an important role in a variety of cellular functions. Although Rho-associated kinase activity has been shown to be an independent predictor for future cardiovascular events in a general population, there is no information on Rho-associated kinase activity in patients with acute coronary syndrome. We evaluated leukocyte Rho-associated kinase activity by Western blot analysis in 73 patients with acute coronary syndrome and 73 age- and gender-matched control subjects. Rho-associated kinase activity within 2 hours of acute coronary syndrome onset was higher in patients with acute coronary syndrome than in the control subjects (0.95±0.55 versus 0.69±0.31; P<0.001). Rho-associated kinase activity promptly increased from 0.95±0.55 to 1.11±0.81 after 3 hours and reached a peak of 1.21±0.76 after 1 day (P=0.03 and P=0.03, respectively) and then gradually decreased to 0.83±0.52 after 7 days, 0.78±0.42 after 14 days, and 0.72±0.30 after 6 months (P=0.22, P=0.29, and P=0.12, respectively). During a median follow-up period of 50.8 months, 31 first major cardiovascular events (death from cardiovascular causes, myocardial infarction, ischemic stroke, and coronary revascularization) occurred. After adjustment for age, sex, cardiovascular risk factors, and concomitant treatment with statins, increased Rho-associated kinase activity was associated with increasing risk of first major cardiovascular events (hazard ratio, 4.56; 95% confidence interval, 1.98–11.34; P<0.001). These findings suggest that Rho-associated kinase activity is dramatically changed after acute coronary syndrome and that Rho-associated kinase activity could be a useful biomarker to predict cardiovascular events in Japanese patients with acute coronary syndrome.
Collapse
Affiliation(s)
- Masato Kajikawa
- From the Department of Cardiovascular Medicine, Graduate School of Biomedical Sciences (M.K., T. Maruhashi, Y.I., T. Matsumoto, A. I., N.O., T.H., Y.K.), Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine (K.N., Y.H.), and Department of Gastroenterology and Metabolism, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences (K.C.), Hiroshima University, Hiroshima, Japan; Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (K.N., A.N., Y.H.); Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan (S.S., M.K., K.D.); Department of Physical Therapy, Hirohsima International University, Hiroshima, Japan (C.G.); and Section of Cardiology, University of Chicago Medical Center, IL (J.K.L.)
| | - Kensuke Noma
- From the Department of Cardiovascular Medicine, Graduate School of Biomedical Sciences (M.K., T. Maruhashi, Y.I., T. Matsumoto, A. I., N.O., T.H., Y.K.), Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine (K.N., Y.H.), and Department of Gastroenterology and Metabolism, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences (K.C.), Hiroshima University, Hiroshima, Japan; Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (K.N., A.N., Y.H.); Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan (S.S., M.K., K.D.); Department of Physical Therapy, Hirohsima International University, Hiroshima, Japan (C.G.); and Section of Cardiology, University of Chicago Medical Center, IL (J.K.L.)
| | - Ayumu Nakashima
- From the Department of Cardiovascular Medicine, Graduate School of Biomedical Sciences (M.K., T. Maruhashi, Y.I., T. Matsumoto, A. I., N.O., T.H., Y.K.), Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine (K.N., Y.H.), and Department of Gastroenterology and Metabolism, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences (K.C.), Hiroshima University, Hiroshima, Japan; Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (K.N., A.N., Y.H.); Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan (S.S., M.K., K.D.); Department of Physical Therapy, Hirohsima International University, Hiroshima, Japan (C.G.); and Section of Cardiology, University of Chicago Medical Center, IL (J.K.L.)
| | - Tatsuya Maruhashi
- From the Department of Cardiovascular Medicine, Graduate School of Biomedical Sciences (M.K., T. Maruhashi, Y.I., T. Matsumoto, A. I., N.O., T.H., Y.K.), Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine (K.N., Y.H.), and Department of Gastroenterology and Metabolism, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences (K.C.), Hiroshima University, Hiroshima, Japan; Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (K.N., A.N., Y.H.); Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan (S.S., M.K., K.D.); Department of Physical Therapy, Hirohsima International University, Hiroshima, Japan (C.G.); and Section of Cardiology, University of Chicago Medical Center, IL (J.K.L.)
| | - Yumiko Iwamoto
- From the Department of Cardiovascular Medicine, Graduate School of Biomedical Sciences (M.K., T. Maruhashi, Y.I., T. Matsumoto, A. I., N.O., T.H., Y.K.), Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine (K.N., Y.H.), and Department of Gastroenterology and Metabolism, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences (K.C.), Hiroshima University, Hiroshima, Japan; Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (K.N., A.N., Y.H.); Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan (S.S., M.K., K.D.); Department of Physical Therapy, Hirohsima International University, Hiroshima, Japan (C.G.); and Section of Cardiology, University of Chicago Medical Center, IL (J.K.L.)
| | - Takeshi Matsumoto
- From the Department of Cardiovascular Medicine, Graduate School of Biomedical Sciences (M.K., T. Maruhashi, Y.I., T. Matsumoto, A. I., N.O., T.H., Y.K.), Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine (K.N., Y.H.), and Department of Gastroenterology and Metabolism, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences (K.C.), Hiroshima University, Hiroshima, Japan; Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (K.N., A.N., Y.H.); Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan (S.S., M.K., K.D.); Department of Physical Therapy, Hirohsima International University, Hiroshima, Japan (C.G.); and Section of Cardiology, University of Chicago Medical Center, IL (J.K.L.)
| | - Akimichi Iwamoto
- From the Department of Cardiovascular Medicine, Graduate School of Biomedical Sciences (M.K., T. Maruhashi, Y.I., T. Matsumoto, A. I., N.O., T.H., Y.K.), Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine (K.N., Y.H.), and Department of Gastroenterology and Metabolism, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences (K.C.), Hiroshima University, Hiroshima, Japan; Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (K.N., A.N., Y.H.); Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan (S.S., M.K., K.D.); Department of Physical Therapy, Hirohsima International University, Hiroshima, Japan (C.G.); and Section of Cardiology, University of Chicago Medical Center, IL (J.K.L.)
| | - Nozomu Oda
- From the Department of Cardiovascular Medicine, Graduate School of Biomedical Sciences (M.K., T. Maruhashi, Y.I., T. Matsumoto, A. I., N.O., T.H., Y.K.), Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine (K.N., Y.H.), and Department of Gastroenterology and Metabolism, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences (K.C.), Hiroshima University, Hiroshima, Japan; Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (K.N., A.N., Y.H.); Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan (S.S., M.K., K.D.); Department of Physical Therapy, Hirohsima International University, Hiroshima, Japan (C.G.); and Section of Cardiology, University of Chicago Medical Center, IL (J.K.L.)
| | - Takayuki Hidaka
- From the Department of Cardiovascular Medicine, Graduate School of Biomedical Sciences (M.K., T. Maruhashi, Y.I., T. Matsumoto, A. I., N.O., T.H., Y.K.), Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine (K.N., Y.H.), and Department of Gastroenterology and Metabolism, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences (K.C.), Hiroshima University, Hiroshima, Japan; Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (K.N., A.N., Y.H.); Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan (S.S., M.K., K.D.); Department of Physical Therapy, Hirohsima International University, Hiroshima, Japan (C.G.); and Section of Cardiology, University of Chicago Medical Center, IL (J.K.L.)
| | - Yasuki Kihara
- From the Department of Cardiovascular Medicine, Graduate School of Biomedical Sciences (M.K., T. Maruhashi, Y.I., T. Matsumoto, A. I., N.O., T.H., Y.K.), Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine (K.N., Y.H.), and Department of Gastroenterology and Metabolism, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences (K.C.), Hiroshima University, Hiroshima, Japan; Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (K.N., A.N., Y.H.); Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan (S.S., M.K., K.D.); Department of Physical Therapy, Hirohsima International University, Hiroshima, Japan (C.G.); and Section of Cardiology, University of Chicago Medical Center, IL (J.K.L.)
| | - Yoshiki Aibara
- From the Department of Cardiovascular Medicine, Graduate School of Biomedical Sciences (M.K., T. Maruhashi, Y.I., T. Matsumoto, A. I., N.O., T.H., Y.K.), Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine (K.N., Y.H.), and Department of Gastroenterology and Metabolism, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences (K.C.), Hiroshima University, Hiroshima, Japan; Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (K.N., A.N., Y.H.); Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan (S.S., M.K., K.D.); Department of Physical Therapy, Hirohsima International University, Hiroshima, Japan (C.G.); and Section of Cardiology, University of Chicago Medical Center, IL (J.K.L.)
| | - Kazuaki Chayama
- From the Department of Cardiovascular Medicine, Graduate School of Biomedical Sciences (M.K., T. Maruhashi, Y.I., T. Matsumoto, A. I., N.O., T.H., Y.K.), Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine (K.N., Y.H.), and Department of Gastroenterology and Metabolism, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences (K.C.), Hiroshima University, Hiroshima, Japan; Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (K.N., A.N., Y.H.); Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan (S.S., M.K., K.D.); Department of Physical Therapy, Hirohsima International University, Hiroshima, Japan (C.G.); and Section of Cardiology, University of Chicago Medical Center, IL (J.K.L.)
| | - Shota Sasaki
- From the Department of Cardiovascular Medicine, Graduate School of Biomedical Sciences (M.K., T. Maruhashi, Y.I., T. Matsumoto, A. I., N.O., T.H., Y.K.), Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine (K.N., Y.H.), and Department of Gastroenterology and Metabolism, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences (K.C.), Hiroshima University, Hiroshima, Japan; Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (K.N., A.N., Y.H.); Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan (S.S., M.K., K.D.); Department of Physical Therapy, Hirohsima International University, Hiroshima, Japan (C.G.); and Section of Cardiology, University of Chicago Medical Center, IL (J.K.L.)
| | - Masaya Kato
- From the Department of Cardiovascular Medicine, Graduate School of Biomedical Sciences (M.K., T. Maruhashi, Y.I., T. Matsumoto, A. I., N.O., T.H., Y.K.), Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine (K.N., Y.H.), and Department of Gastroenterology and Metabolism, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences (K.C.), Hiroshima University, Hiroshima, Japan; Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (K.N., A.N., Y.H.); Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan (S.S., M.K., K.D.); Department of Physical Therapy, Hirohsima International University, Hiroshima, Japan (C.G.); and Section of Cardiology, University of Chicago Medical Center, IL (J.K.L.)
| | - Keigo Dote
- From the Department of Cardiovascular Medicine, Graduate School of Biomedical Sciences (M.K., T. Maruhashi, Y.I., T. Matsumoto, A. I., N.O., T.H., Y.K.), Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine (K.N., Y.H.), and Department of Gastroenterology and Metabolism, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences (K.C.), Hiroshima University, Hiroshima, Japan; Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (K.N., A.N., Y.H.); Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan (S.S., M.K., K.D.); Department of Physical Therapy, Hirohsima International University, Hiroshima, Japan (C.G.); and Section of Cardiology, University of Chicago Medical Center, IL (J.K.L.)
| | - Chikara Goto
- From the Department of Cardiovascular Medicine, Graduate School of Biomedical Sciences (M.K., T. Maruhashi, Y.I., T. Matsumoto, A. I., N.O., T.H., Y.K.), Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine (K.N., Y.H.), and Department of Gastroenterology and Metabolism, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences (K.C.), Hiroshima University, Hiroshima, Japan; Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (K.N., A.N., Y.H.); Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan (S.S., M.K., K.D.); Department of Physical Therapy, Hirohsima International University, Hiroshima, Japan (C.G.); and Section of Cardiology, University of Chicago Medical Center, IL (J.K.L.)
| | - James K Liao
- From the Department of Cardiovascular Medicine, Graduate School of Biomedical Sciences (M.K., T. Maruhashi, Y.I., T. Matsumoto, A. I., N.O., T.H., Y.K.), Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine (K.N., Y.H.), and Department of Gastroenterology and Metabolism, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences (K.C.), Hiroshima University, Hiroshima, Japan; Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (K.N., A.N., Y.H.); Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan (S.S., M.K., K.D.); Department of Physical Therapy, Hirohsima International University, Hiroshima, Japan (C.G.); and Section of Cardiology, University of Chicago Medical Center, IL (J.K.L.)
| | - Yukihito Higashi
- From the Department of Cardiovascular Medicine, Graduate School of Biomedical Sciences (M.K., T. Maruhashi, Y.I., T. Matsumoto, A. I., N.O., T.H., Y.K.), Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine (K.N., Y.H.), and Department of Gastroenterology and Metabolism, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences (K.C.), Hiroshima University, Hiroshima, Japan; Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (K.N., A.N., Y.H.); Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan (S.S., M.K., K.D.); Department of Physical Therapy, Hirohsima International University, Hiroshima, Japan (C.G.); and Section of Cardiology, University of Chicago Medical Center, IL (J.K.L.).
| |
Collapse
|
131
|
RNAi profiling of primary human AML cells identifies ROCK1 as a therapeutic target and nominates fasudil as an antileukemic drug. Blood 2015; 125:3760-8. [PMID: 25931586 DOI: 10.1182/blood-2014-07-590646] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 04/22/2015] [Indexed: 01/07/2023] Open
Abstract
Acute myeloid leukemia (AML) is characterized by a marked genetic heterogeneity, which complicates the development of novel therapeutics. The delineation of pathways essential within an individual patient's mutational background might overcome this limitation and facilitate personalized treatment. We report the results of a large-scale lentiviral loss-of-function RNA interference (RNAi) screen in primary leukemic cells. Stringent validation identified 6 genes (BNIPL1, ROCK1, RPS13, STK3, SNX27, WDHD1) whose knockdown impaired growth and viability of the cells. Dependence on these genes was not caused by mutation or overexpression, and although some of the candidates seemed to be rather patient specific, others were essential in cells isolated from other AML patients. In addition to the phenotype observed after ROCK1 knockdown, treatment with the approved ROCK inhibitor fasudil resulted in increased apoptosis and decreased viability of primary AML cells. In contrast to observations in some other malignancies, ROCK1 inhibition did not foster growth of immature malignant progenitors but was toxic to this cell fraction in feeder coculture and xenotransplant experiments, indicating a distinct effect of ROCK1 inhibition on leukemic progenitors. We conclude that large-scale RNAi screens in primary patient-derived cells are feasible and can complement other methods for personalized cancer therapies, such as expression and mutation profiling.
Collapse
|
132
|
Role of L-type Ca(2+) channels, sarcoplasmic reticulum and Rho kinase in rat basilar artery contractile properties in a new model of subarachnoid hemorrhage. Vascul Pharmacol 2015; 72:64-72. [PMID: 25937251 DOI: 10.1016/j.vph.2015.04.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 03/20/2015] [Accepted: 04/22/2015] [Indexed: 11/22/2022]
Abstract
We have previously described that L-type Ca(2+) channels' (LTCCs) activation and metabotropic Ca(2+) release from the sarcoplasmic reticulum (SR) regulate RhoA/Rho kinase (ROCK) activity and sustained arterial contraction. We have investigated whether this signaling pathway can be altered in a new experimental model of subarachnoid hemorrhage (SAH). For this purpose, arterial reactivity was evaluated on days 1 to 5 after surgery. A significant increase of basal tone, measured 4 and 60min after normalization, was observed on day 5 after SAH and at 60min on days 2 and 3 after SAH. This phenomenon was suppressed with LTCCs and ROCK inhibitors. We have also studied arterial rings vasoreactivity in response to high K(+) solutions. Interestingly, there were no significant differences in the phasic component of the high K(+)-induced contraction between sham and SAH groups, whereas a significant increase in the sustained contraction was observed on day 5 after SAH. This latter component was sensitive to fasudil, and selectively reduced by low nifedipine concentration, and phospholipase C and SR-ATPase inhibitors. Therefore, our data suggest that the metabotropic function of LTCCs is potentiated in SAH. Our results could provide a new strategy to optimize the pharmacological treatment of this pathological process.
Collapse
|
133
|
Mikelis CM, Simaan M, Ando K, Fukuhara S, Sakurai A, Amornphimoltham P, Masedunskas A, Weigert R, Chavakis T, Adams RH, Offermanns S, Mochizuki N, Zheng Y, Gutkind JS. RhoA and ROCK mediate histamine-induced vascular leakage and anaphylactic shock. Nat Commun 2015; 6:6725. [PMID: 25857352 PMCID: PMC4394241 DOI: 10.1038/ncomms7725] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 02/22/2015] [Indexed: 01/08/2023] Open
Abstract
Histamine-induced vascular leakage is an integral component of many highly prevalent human diseases, including allergies, asthma, and anaphylaxis. Yet, how histamine induces the disruption of the endothelial barrier is not well defined. By using genetically modified animal models, pharmacologic inhibitors, and a synthetic biology approach, here we show that the small GTPase RhoA mediates histamine-induced vascular leakage. Histamine causes the rapid formation of focal adherens junctions, disrupting the endothelial barrier by acting on H1R Gαq-coupled receptors, which is blunted in endothelial Gαq/11 KO mice. Interfering with RhoA and ROCK function abolishes endothelial permeability, while phospholipase Cβ plays a limited role. Moreover, endothelial-specific RhoA gene deletion prevents vascular leakage and passive cutaneous anaphylaxis in vivo, and ROCK inhibitors protect from lethal systemic anaphylaxis. This study supports a key role for the RhoA signaling circuitry in vascular permeability, thereby identifying novel pharmacological targets for many human diseases characterized by aberrant vascular leakage.
Collapse
Affiliation(s)
- Constantinos M Mikelis
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - May Simaan
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Koji Ando
- Department of Cell Biology, CREST-JST, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565-8565, Japan
| | - Shigetomo Fukuhara
- Department of Cell Biology, CREST-JST, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565-8565, Japan
| | - Atsuko Sakurai
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Panomwat Amornphimoltham
- Intracellular Membrane Trafficking Unit, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Andrius Masedunskas
- Intracellular Membrane Trafficking Unit, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Roberto Weigert
- Intracellular Membrane Trafficking Unit, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry, Faculty of Medicine, Technische Universität Dresden, Dresden 01307, Germany
| | - Ralf H Adams
- 1] Department of Tissue Morphogenesis, Max-Planck Institute for Molecular Biomedicine, Münster D-48149, Germany [2] Faculty of Medicine, University of Münster, Münster 48149, Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max-Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Naoki Mochizuki
- Department of Cell Biology, CREST-JST, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565-8565, Japan
| | - Yi Zheng
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital, University of Cincinnati College of Medicine, Cincinnati, Ohio 45229, USA
| | - J Silvio Gutkind
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
134
|
Saito A, Inoue M, Kon H, Imaruoka S, Basaki K, Midorikawa H, Sasaki T, Nishijima M. Effectiveness of intraarterial administration of fasudil hydrochloride for preventing symptomatic vasospasm after subarachnoid hemorrhage. ACTA NEUROCHIRURGICA. SUPPLEMENT 2015; 120:297-301. [PMID: 25366640 DOI: 10.1007/978-3-319-04981-6_50] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
OBJECTIVE We examined the effect of intraarterial administration of fasudil hydrochloride (IAFC), a Rho kinase inhibitor, for the prevention of symptomatic vasospasm after SAH by evaluating cerebral circulation. METHODS We evaluated IAFC cases of 57 sides of 38 patients (12 men and 26 women, average age 60.2 years old) diagnosed with aneurysmal subarachnoid hemorrhage (SAH) from February 2012 to November 2012. All cases were treated by clipping or coil embolization within 48 h after onset. Indication for IAFC was the existence of a spastic change on follow-up digital subtraction angiography (DSA) compared with that of onset. RESULTS Clipping was performed in 30 cases and coil embolization in 8 cases. IAFC was performed an average of 6.6 days after onset. Color gradient mapping demonstrated reduction of the circulation time after IAFC compared with before IAFC on 39 sides, no change on 15 sides, and extension on 3 sides. Average arterial circulation time before IAFC was 2.25 ± 0.57 s and after IAFC was 1.95 ± 0.55 s. IAFC significantly shortened average arterial circulation (P = 0.005). No case developed symptomatic vasospasm after IAFC. CONCLUSION IAFC significantly reduced the cerebral circulation time after aneurysmal SAH and might be effective for the prevention of symptomatic vasospasm.
Collapse
Affiliation(s)
- Atsushi Saito
- Departments of Neurosurgery and Radiology, Aomori Prefectural Central Hospital, 2-1-1 Higashitsukurimichi, Aomori, 0308553, Japan,
| | | | | | | | | | | | | | | |
Collapse
|
135
|
Bath PM, Wardlaw JM. Pharmacological treatment and prevention of cerebral small vessel disease: a review of potential interventions. Int J Stroke 2015; 10:469-78. [PMID: 25727737 PMCID: PMC4832291 DOI: 10.1111/ijs.12466] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 01/06/2015] [Indexed: 12/21/2022]
Abstract
Small vessel disease encompasses lacunar stroke, white matter hyperintensities, lacunes and microbleeds. It causes a quarter of all ischemic strokes, is the commonest cause of vascular dementia, and the cause is incompletely understood. Vascular prophylaxis, as appropriate for large artery disease and cardioembolism, includes antithrombotics, and blood pressure and lipid lowering; however, these strategies may not be effective for small vessel disease, or are already used routinely so precluding further detailed study. Further, intensive antiplatelet therapy is known to be hazardous in small vessel disease through enhanced bleeding. Whether acetylcholinesterase inhibitors, which delay the progression of Alzheimer's dementia, are relevant in small vessel disease remains unclear. Potential prophylactic and treatment strategies might be those that target brain microvascular endothelium and the blood brain barrier, microvascular function and neuroinflammation. Potential interventions include endothelin antagonists, neurotrophins, nitric oxide donors and phosphodiesterase 5 inhibitors, peroxisome proliferator‐activated receptor‐gamma agonists, and prostacyclin mimics and phosphodiesterase 3 inhibitors. Several drugs that have relevant properties are licensed for other disorders, offering the possibility of drug repurposing. Others are in development. Since influencing multiple targets may be most effective, using multiple agents and/or those that have multiple effects may be preferable. We focus on potential small vessel disease mechanistic targets, summarize drugs that have relevant actions, and review data available from randomized trials on their actions and on the available evidence for their use in lacunar stroke.
Collapse
Affiliation(s)
- Philip M Bath
- Stroke Trials Unit, Division of Clinical Neuroscience, University of Nottingham, Nottingham, UK
| | - Joanna M Wardlaw
- Division of Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
136
|
Bond LM, Sellers JR, McKerracher L. Rho kinase as a target for cerebral vascular disorders. Future Med Chem 2015; 7:1039-53. [PMID: 26062400 PMCID: PMC4656981 DOI: 10.4155/fmc.15.45] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The development of novel pharmaceutical treatments for disorders of the cerebral vasculature is a serious unmet medical need. These vascular disorders are typified by a disruption in the delicate Rho signaling equilibrium within the blood vessel wall. In particular, Rho kinase overactivation in the smooth muscle and endothelial layers of the vessel wall results in cytoskeletal modifications that lead to reduced vascular integrity and abnormal vascular growth. Rho kinase is thus a promising target for the treatment of cerebral vascular disorders. Indeed, preclinical studies indicate that Rho kinase inhibition may reduce the formation/growth/rupture of both intracranial aneurysms and cerebral cavernous malformations.
Collapse
Affiliation(s)
- Lisa M Bond
- BioAxone BioSciences, Inc., 10 Rogers Street, Suite 101, Kendall Square, Cambridge, MA 02142, USA
- Laboratory of Molecular Physiology, National Heart, Lung & Blood Institute, Bethesda, MD 20892, USA
| | - James R Sellers
- Laboratory of Molecular Physiology, National Heart, Lung & Blood Institute, Bethesda, MD 20892, USA
| | - Lisa McKerracher
- BioAxone BioSciences, Inc., 10 Rogers Street, Suite 101, Kendall Square, Cambridge, MA 02142, USA
| |
Collapse
|
137
|
Xiao JW, Zhu XY, Wang QG, Zhang DZ, Cui CS, Zhang P, Chen HY, Meng LL. Acute Effects of Rho-Kinase Inhibitor Fasudil on Pulmonary Arterial Hypertension in Patients With Congenital Heart Defects. Circ J 2015; 79:1342-8. [DOI: 10.1253/circj.cj-14-1015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Jia-wang Xiao
- Department of Congenital Heart Disease, General Hospital of Shenyang Military Area Command
| | - Xian-yang Zhu
- Department of Congenital Heart Disease, General Hospital of Shenyang Military Area Command
| | - Qi-guang Wang
- Department of Congenital Heart Disease, General Hospital of Shenyang Military Area Command
| | - Duan-zhen Zhang
- Department of Congenital Heart Disease, General Hospital of Shenyang Military Area Command
| | - Chun-Sheng Cui
- Department of Congenital Heart Disease, General Hospital of Shenyang Military Area Command
| | - Po Zhang
- Department of Congenital Heart Disease, General Hospital of Shenyang Military Area Command
| | - Huo-yuan Chen
- Department of Congenital Heart Disease, General Hospital of Shenyang Military Area Command
| | - Li-li Meng
- Department of Congenital Heart Disease, General Hospital of Shenyang Military Area Command
| |
Collapse
|
138
|
The fibrinolytic system-more than fibrinolysis? Transfus Med Rev 2014; 29:102-9. [PMID: 25576010 DOI: 10.1016/j.tmrv.2014.09.006] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 09/08/2014] [Accepted: 09/12/2014] [Indexed: 01/05/2023]
Abstract
The fibrinolytic system, known for its ability to regulate the activation of the zymogen plasminogen into active plasmin, has been primarily associated with the removal of fibrin and blood clots. Tissue-type plasminogen activator, the most well-recognized plasminogen activator, was harnessed for therapeutic benefit against thromboembolic disorders more than 30 years ago, whereas inhibition of this system has been proven effective for certain bleeding disorders. However, in recent years, new and unexpected functional roles for this system have been identified mostly in relation to the central nervous system that are both unrelated and independent of fibrin degradation and clot removal. Hence, it seems reasonable to ask whether agents used to modify components or activities of the fibrinolytic system have any clinical consequences unrelated to their intended use in hemostasis. This review will provide an overview of these new features of the fibrinolytic system and will also focus on prospective considerations in the use of fibrinolytic and antifibrinolytic agents.
Collapse
|
139
|
Lee HS, Kim KS, Lim HS, Choi M, Kim HK, Ahn HY, Shin JC, Joe YA. Priming Wharton's Jelly-Derived Mesenchymal Stromal/Stem Cells With ROCK Inhibitor Improves Recovery in an Intracerebral Hemorrhage Model. J Cell Biochem 2014; 116:310-9. [DOI: 10.1002/jcb.24969] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 08/29/2014] [Indexed: 12/21/2022]
Affiliation(s)
- Hyun-Sun Lee
- Cancer Research Institute and Department of Medical Life Sciences; College of Medicine; The Catholic University of Korea; Seoul 137-701 Korea
| | - Kwang S. Kim
- Cancer Research Institute and Department of Medical Life Sciences; College of Medicine; The Catholic University of Korea; Seoul 137-701 Korea
| | - Hee-Suk Lim
- Cancer Research Institute and Department of Medical Life Sciences; College of Medicine; The Catholic University of Korea; Seoul 137-701 Korea
| | - Moran Choi
- Cancer Research Institute and Department of Medical Life Sciences; College of Medicine; The Catholic University of Korea; Seoul 137-701 Korea
| | - Hyun-Kyung Kim
- Cancer Research Institute and Department of Medical Life Sciences; College of Medicine; The Catholic University of Korea; Seoul 137-701 Korea
| | - Hyun-Young Ahn
- Department of Obstetrics and Gynecology; College of Medicine; The Catholic University of Korea; Seoul 137-701 Korea
| | - Jong-Chul Shin
- Department of Obstetrics and Gynecology; College of Medicine; The Catholic University of Korea; Seoul 137-701 Korea
| | - Young Ae Joe
- Cancer Research Institute and Department of Medical Life Sciences; College of Medicine; The Catholic University of Korea; Seoul 137-701 Korea
| |
Collapse
|
140
|
Fasudil regulates T cell responses through polarization of BV-2 cells in mice experimental autoimmune encephalomyelitis. Acta Pharmacol Sin 2014; 35:1428-38. [PMID: 25263338 DOI: 10.1038/aps.2014.68] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 04/22/2014] [Indexed: 01/22/2023]
Abstract
AIM Fasudil, a selective Rho kinase (ROCK) inhibitor, has been shown to alleviate the severity of experimental autoimmune encephalomyelitis (EAE) via attenuating demyelination and neuroinflammation. The aim of this study was to investigate the effects of fasudil on interactions between macrophages/microglia and T cells in a mice EAE model. METHODS Mouse BV-2 microglia were treated with IFN-γ and fasudil. Cell viability was detected with MTT assay. BV-2 microglia polarization was analyzed using flow cytometry. Cytokines and other proteins were detected with ELISA and Western blotting, respectively. Mice were immunized with MOG35-55 to induce EAE, and then treated with fasudil (40 mg/kg, ip) every other day from d 3 to d 27 pi. Encephalomyelitic T cells were prepared from the spleen of mice immunized with MOG35-55 on d 9 pi. RESULTS Treatment of mouse BV-2 microglia with fasudil (15 μg/mL) induced significant phenotype polarization and functional plasticity, shifting M1 to M2 polarization. When co-cultured with the encephalomyelitic T cells, fasudil-treated BV-2 microglia significantly inhibited the proliferation of antigen-reactive T cells, and down-regulated IL-17-expressing CD4(+) T cells and IL-17 production. Furthermore, fasudil-treated BV-2 microglia significantly up-regulated CD4(+)CD25(high) and CD4(+)IL-10(+) regulatory T cells (Tregs) and IL-10 production, suggesting that the encephalomyelitic T cells had converted to Tregs. In EAE mice, fasudil administration significantly decreased both CD11b(+)iNOS(+) and CD11b(+)TNF-α(+) M1 microglia, and increased CD11b(+)IL-10(+) M2 microglia. CONCLUSION Fasudil polarizes BV-2 microglia into M2 cells, which convert the encephalomyelitic T cells into Tregs in the mice EAE model.
Collapse
|
141
|
Abstract
BACKGROUND It is unclear whether blood pressure should be altered actively during the acute phase of stroke. This is an update of a Cochrane review first published in 1997, and previously updated in 2001 and 2008. OBJECTIVES To assess the clinical effectiveness of altering blood pressure in people with acute stroke, and the effect of different vasoactive drugs on blood pressure in acute stroke. SEARCH METHODS We searched the Cochrane Stroke Group Trials Register (last searched in February 2014), the Cochrane Database of Systematic reviews (CDSR) and the Cochrane Central Register of Controlled Trials (CENTRAL) (The Cochrane Library 2014, Issue 2), MEDLINE (Ovid) (1966 to May 2014), EMBASE (Ovid) (1974 to May 2014), Science Citation Index (ISI, Web of Science, 1981 to May 2014) and the Stroke Trials Registry (searched May 2014). SELECTION CRITERIA Randomised controlled trials of interventions that aimed to alter blood pressure compared with control in participants within one week of acute ischaemic or haemorrhagic stroke. DATA COLLECTION AND ANALYSIS Two review authors independently applied the inclusion criteria, assessed trial quality and extracted data. The review authors cross-checked data and resolved discrepancies by discussion to reach consensus. We obtained published and unpublished data where available. MAIN RESULTS We included 26 trials involving 17,011 participants (8497 participants were assigned active therapy and 8514 participants received placebo/control). Not all trials contributed to each outcome. Most data came from trials that had a wide time window for recruitment; four trials gave treatment within six hours and one trial within eight hours. The trials tested alpha-2 adrenergic agonists (A2AA), angiotensin converting enzyme inhibitors (ACEI), angiotensin receptor antagonists (ARA), calcium channel blockers (CCBs), nitric oxide (NO) donors, thiazide-like diuretics, and target-driven blood pressure lowering. One trial tested phenylephrine.At 24 hours after randomisation oral ACEIs reduced systolic blood pressure (SBP, mean difference (MD) -8 mmHg, 95% confidence interval (CI) -17 to 1) and diastolic blood pressure (DBP, MD -3 mmHg, 95% CI -9 to 2), sublingual ACEIs reduced SBP (MD -12.00 mm Hg, 95% CI -26 to 2) and DBP (MD -2, 95%CI -10 to 6), oral ARA reduced SBP (MD -1 mm Hg, 95% CI -3 to 2) and DBP (MD -1 mm Hg, 95% CI -3 to 1), oral beta blockers reduced SBP (MD -14 mm Hg; 95% CI -27 to -1) and DBP (MD -1 mm Hg, 95% CI -9 to 7), intravenous (iv) beta blockers reduced SBP (MD -5 mm Hg, 95% CI -18 to 8) and DBP (-5 mm Hg, 95% CI -13 to 3), oral CCBs reduced SBP (MD -13 mmHg, 95% CI -43 to 17) and DBP (MD -6 mmHg, 95% CI -14 to 2), iv CCBs reduced SBP (MD -32 mmHg, 95% CI -65 to 1) and DBP (MD -13, 95% CI -31 to 6), NO donors reduced SBP (MD -12 mmHg, 95% CI -19 to -5) and DBP (MD -3, 95% CI -4 to -2) while phenylephrine, non-significantly increased SBP (MD 21 mmHg, 95% CI -13 to 55) and DBP (MD 1 mmHg, 95% CI -15 to 16).Blood pressure lowering did not reduce death or dependency either by drug class (OR 0.98, 95% CI 0.92 to 1.05), stroke type (OR 0.98, 95% CI 0.92 to 1.05) or time to treatment (OR 0.98, 95% CI 0.92 to 1.05). Treatment within six hours of stroke appeared effective in reducing death or dependency (OR 0.86, 95% CI 0.76 to 0.99) but not death (OR 0.70, 95% CI 0.38 to 1.26) at the end of the trial. Although death or dependency did not differ between people who continued pre-stroke antihypertensive treatment versus those who stopped it temporarily (worse outcome with continuing treatment, OR 1.06, 95% CI 0.91 to 1.24), disability scores at the end of the trial were worse in participants randomised to continue treatment (Barthel Index, MD -3.2, 95% CI -5.8, -0.6). AUTHORS' CONCLUSIONS There is insufficient evidence that lowering blood pressure during the acute phase of stroke improves functional outcome. It is reasonable to withhold blood pressure-lowering drugs until patients are medically and neurologically stable, and have suitable oral or enteral access, after which drugs can than be reintroduced. In people with acute stroke, CCBs, ACEI, ARA, beta blockers and NO donors each lower blood pressure while phenylephrine probably increases blood pressure. Further trials are needed to identify which people are most likely to benefit from early treatment, in particular whether treatment started very early is beneficial.
Collapse
Affiliation(s)
- Philip MW Bath
- University of NottinghamStroke, Division of Clinical NeuroscienceCity Hospital CampusNottinghamUKNG5 1PB
| | - Kailash Krishnan
- University of NottinghamStroke, Division of Clinical NeuroscienceCity Hospital CampusNottinghamUKNG5 1PB
| | | |
Collapse
|
142
|
Cui Q, Zhang Y, Chen H, Li J. Rho kinase: A new target for treatment of cerebral ischemia/reperfusion injury. Neural Regen Res 2014; 8:1180-9. [PMID: 25206412 PMCID: PMC4107606 DOI: 10.3969/j.issn.1673-5374.2013.13.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Accepted: 02/20/2013] [Indexed: 01/08/2023] Open
Abstract
Rho kinase inhibitor fasudil hydrochloride has been shown to reduce cerebral vasospasm, to inhibit inflammation and apoptosis and to promote the recovery of neurological function. However, the effect of fasudil hydrochloride on claudin-5 protein expression has not been reported after cerebral ischemia/reperfusion. Therefore, this study sought to explore the effects of fasudil hydrochloride on blood-brain barrier permeability, growth-associated protein-43 and claudin-5 protein expression, and to further understand the neuroprotective effect of fasudil hydrochloride. A focal cerebral ischemia/reperfusion model was established using the intraluminal suture technique. Fasudil hydrochloride (15 mg/kg) was intraperitoneally injected once a day. Neurological deficit was evaluated using Longa's method. Changes in permeability of blood-brain barrier were measured using Evans blue. Changes in RhoA, growth-associated protein-43 and claudin-5 protein expression were detected using immunohistochemistry and western blotting. Results revealed that fasudil hydrochloride noticeably contributed to the recovery of neurological function, improved the function of blood-brain barrier, inhibited RhoA protein expression, and upregulated growth-associated protein-43 and claudin-5 protein expression following cerebral ischemia/reperfusion. Results indicated that Rho kinase exhibits a certain effect on neurovascular damage following cerebral ischemia/reperfusion. Intervention targeted Rho kinase might be a new therapeutic target in the treatment of cerebral ischemia/reperfusion.
Collapse
Affiliation(s)
- Qinghong Cui
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Yongbo Zhang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Hui Chen
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Jimei Li
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| |
Collapse
|
143
|
Takata M, Tanaka H, Kimura M, Nagahara Y, Tanaka K, Kawasaki K, Seto M, Tsuruma K, Shimazawa M, Hara H. Fasudil, a rho kinase inhibitor, limits motor neuron loss in experimental models of amyotrophic lateral sclerosis. Br J Pharmacol 2014; 170:341-51. [PMID: 23763343 DOI: 10.1111/bph.12277] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 05/28/2013] [Accepted: 06/01/2013] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder with no effective treatment. Fasudil hydrochloride (fasudil), a potent rho kinase (ROCK) inhibitor, is useful for the treatment of ischaemic diseases. In previous reports, fasudil improved pathology in mouse models of Alzheimer's disease and spinal muscular atrophy, but there is no evidence in that it can affect ALS. We therefore investigated its effects on experimental models of ALS. EXPERIMENTAL APPROACH In mice motor neuron (NSC34) cells, the neuroprotective effect of hydroxyfasudil (M3), an active metabolite of fasudil, and its mechanism were evaluated. Moreover, the effects of fasudil, 30 and 100 mg·kg(-1), administered via drinking water to mutant superoxide dismutase 1 (SOD1(G93A)) mice were tested by measuring motor performance, survival time and histological changes, and its mechanism investigated. KEY RESULTS M3 prevented motor neuron cell death induced by SOD1(G93A). Furthermore, M3 suppressed both the increase in ROCK activity and phosphorylated phosphatase and tensin homologue deleted on chromosome 10 (PTEN), and the reduction in phosphorylated Akt induced by SOD1(G93A). These effects of M3 were attenuated by treatment with a PI3K inhibitor (LY294002). Moreover, fasudil slowed disease progression, increased survival time and reduced motor neuron loss, in SOD1(G93A) mice. Fasudil also attenuated the increase in ROCK activity and PTEN, and the reduction in Akt in SOD1(G93A) mice. CONCLUSIONS AND IMPLICATIONS These findings indicate that fasudil may be effective at suppressing motor neuron degeneration and symptom progression in ALS. Hence, fasudil may have potential as a therapeutic agent for ALS treatment.
Collapse
Affiliation(s)
- M Takata
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
144
|
Abstract
Rho kinase (ROCK) is a major downstream effector of the small GTPase RhoA. ROCK family, consisting of ROCK1 and ROCK2, plays central roles in the organization of actin cytoskeleton and is involved in a wide range of fundamental cellular functions, such as contraction, adhesion, migration, proliferation, and apoptosis. Due to the discovery of effective inhibitors, such as fasudil and Y27632, the biological roles of ROCK have been extensively explored with particular attention on the cardiovascular system. In many preclinical models of cardiovascular diseases, including vasospasm, arteriosclerosis, hypertension, pulmonary hypertension, stroke, ischemia-reperfusion injury, and heart failure, ROCK inhibitors have shown a remarkable efficacy in reducing vascular smooth muscle cell hypercontraction, endothelial dysfunction, inflammatory cell recruitment, vascular remodeling, and cardiac remodeling. Moreover, fasudil has been used in the clinical trials of several cardiovascular diseases. The continuing utilization of available pharmacological inhibitors and the development of more potent or isoform-selective inhibitors in ROCK signaling research and in treating human diseases are escalating. In this review, we discuss the recent molecular, cellular, animal, and clinical studies with a focus on the current understanding of ROCK signaling in cardiovascular physiology and diseases. We particularly note that emerging evidence suggests that selective targeting ROCK isoform based on the disease pathophysiology may represent a novel therapeutic approach for the disease treatment including cardiovascular diseases.
Collapse
|
145
|
Porras-González C, González-Rodríguez P, Calderón-Sánchez E, López-Barneo J, Ureña J. Low-dose combination of Rho kinase and L-type Ca2+ channel antagonists for selective inhibition of depolarization-induced sustained arterial contraction. Eur J Pharmacol 2014; 732:130-8. [DOI: 10.1016/j.ejphar.2014.02.046] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 01/27/2014] [Accepted: 02/23/2014] [Indexed: 11/29/2022]
|
146
|
Gibson CL, Srivastava K, Sprigg N, Bath PMW, Bayraktutan U. Inhibition of Rho-kinase protects cerebral barrier from ischaemia-evoked injury through modulations of endothelial cell oxidative stress and tight junctions. J Neurochem 2014; 129:816-26. [PMID: 24528233 DOI: 10.1111/jnc.12681] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 01/20/2014] [Accepted: 02/07/2014] [Indexed: 12/22/2022]
Abstract
Ischaemic strokes evoke blood-brain barrier (BBB) disruption and oedema formation through a series of mechanisms involving Rho-kinase activation. Using an animal model of human focal cerebral ischaemia, this study assessed and confirmed the therapeutic potential of Rho-kinase inhibition during the acute phase of stroke by displaying significantly improved functional outcome and reduced cerebral lesion and oedema volumes in fasudil- versus vehicle-treated animals. Analyses of ipsilateral and contralateral brain samples obtained from mice treated with vehicle or fasudil at the onset of reperfusion plus 4 h post-ischaemia or 4 h post-ischaemia alone revealed these benefits to be independent of changes in the activity and expressions of oxidative stress- and tight junction-related parameters. However, closer scrutiny of the same parameters in brain microvascular endothelial cells subjected to oxygen-glucose deprivation ± reperfusion revealed marked increases in prooxidant NADPH oxidase enzyme activity, superoxide anion release and in expressions of antioxidant enzyme catalase and tight junction protein claudin-5. Cotreatment of cells with Y-27632 prevented all of these changes and protected in vitro barrier integrity and function. These findings suggest that inhibition of Rho-kinase after acute ischaemic attacks improves cerebral integrity and function through regulation of endothelial cell oxidative stress and reorganization of intercellular junctions. Inhibition of Rho-kinase (ROCK) activity in a mouse model of human ischaemic stroke significantly improved functional outcome while reducing cerebral lesion and oedema volumes compared to vehicle-treated counterparts. Studies conducted with brain microvascular endothelial cells exposed to OGD ± R in the presence of Y-27632 revealed restoration of intercellular junctions and suppression of prooxidant NADPH oxidase activity as important factors in ROCK inhibition-mediated BBB protection.
Collapse
Affiliation(s)
- Claire L Gibson
- School of Psychology, University of Leicester, Leicester, UK
| | | | | | | | | |
Collapse
|
147
|
Sawada N, Liao JK. Rho/Rho-associated coiled-coil forming kinase pathway as therapeutic targets for statins in atherosclerosis. Antioxid Redox Signal 2014; 20:1251-67. [PMID: 23919640 PMCID: PMC3934442 DOI: 10.1089/ars.2013.5524] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
SIGNIFICANCE The 3-hydroxy-methylglutaryl coenzyme A reductase inhibitors or statins are important therapeutic agents for lowering serum cholesterol levels. However, recent studies suggest that statins may exert atheroprotective effects beyond cholesterol lowering. These so-called "pleiotropic effects" include effects of statins on vascular and inflammatory cells. Thus, it is important to understand whether other signaling pathways that are involved in atherosclerosis could be targets of statins, and if so, whether individuals with "overactivity" of these pathways could benefit from statin therapy, regardless of serum cholesterol level. RECENT ADVANCES Statins inhibit the synthesis of isoprenoids, which are important for the function of the Rho/Rho-associated coiled-coil containing kinase (ROCK) pathway. Indeed, recent studies suggest that inhibition of the Rho/ROCK pathway by statins could lead to improved endothelial function and decreased vascular inflammation and atherosclerosis. Thus, the Rho/ROCK pathway has emerged as an important target of statin therapy for reducing atherosclerosis and possibly cardiovascular disease. CRITICAL ISSUES Because atherosclerosis is both a lipid and an inflammatory disease, it is important to understand how inhibition of Rho/ROCK pathway could contribute to statins' antiatherosclerotic effects. FUTURE DIRECTIONS The role of ROCKs (ROCK1 and ROCK2) in endothelial, smooth muscle, and inflammatory cells needs to be determined in the context of atherogenesis. This could lead to the development of specific ROCK1 or ROCK2 inhibitors, which could have greater therapeutic benefits with less toxicity. Also, clinical trials will need to be performed to determine whether inhibition of ROCKs, with and without statins, could lead to further reduction in atherosclerosis and cardiovascular disease.
Collapse
Affiliation(s)
- Naoki Sawada
- 1 GCOE Program and Department of Molecular Endocrinology and Metabolism, Tokyo Medical and Dental University , Tokyo, Japan
| | | |
Collapse
|
148
|
Pedder H, Vesterinen HM, Macleod MR, Wardlaw JM. Systematic review and meta-analysis of interventions tested in animal models of lacunar stroke. Stroke 2014; 45:563-70. [PMID: 24385271 DOI: 10.1161/strokeaha.113.003128] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND AND PURPOSE A total of 25% of strokes are lacunar, and these are pathophysiologically different from large artery strokes. Despite emerging evidence of a substantial impact on physical disability and dementia, little attention has been paid to the development of specific treatments. The optimal use of the animal models of lacunar stroke used to test candidate interventions is not known. METHODS We conducted a systematic review and meta-analysis of studies testing candidate interventions in animal models of lacunar stroke. We used random-effects meta-analysis to assess the impact of study characteristics and trim and fill to seek evidence of publication bias. RESULTS The efficacy of 43 distinct interventions was described in 57 publications. The median number of quality checklist items scored was 3 of 8 (interquartile range, 2-4). Many models reflected mechanisms of limited relevance to lacunar stroke. Meta-analysis of results from 27 studies showed that on average, infarct size and neurobehavioral outcome were improved by 34.2% (24.1-44.2) and 0.82 standardized mean difference (0.51-1.14), respectively. Four interventions improved both infarct size and neurobehavioral outcome but there were insufficient data for this finding to be considered robust. For infarct size, efficacy was lower in studies reporting blinding and higher in studies reporting randomization. For neurobehavior, efficacy was lower in randomized studies. For infarct size there was evidence of publication bias. CONCLUSIONS No intervention has yet been tested in sufficient range and depth to support translation to clinical trial. There is limited reporting of measures to reduce the risk of bias and evidence for a substantial publications bias.
Collapse
Affiliation(s)
- Hugo Pedder
- From the Department of Clinical Neurosciences, Western General Hospital, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | | | | | | |
Collapse
|
149
|
Fukuda T, Narahara Y, Kanazawa H, Matsushita Y, Kidokoro H, Itokawa N, Kondo C, Atsukawa M, Nakatsuka K, Sakamoto C. Effects of fasudil on the portal and systemic hemodynamics of patients with cirrhosis. J Gastroenterol Hepatol 2014; 29:325-9. [PMID: 24033356 DOI: 10.1111/jgh.12360] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/02/2013] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIM Fasudil, a Rho-kinase inhibitor, has been shown to reduce portal venous pressure in cirrhotic rats. However, its effects on portal and systemic hemodynamics have not been investigated in cirrhotic patients with portal hypertension. The aim of this study was to assess the effects of fasudil on the portal and systemic hemodynamics of cirrhotic patients with portal hypertension. METHODS Twenty-three patients with cirrhosis and portal hypertension were studied. Systemic and portal hemodynamics were measured prior to and 50 min after the initiation of intravenous administration of 30 mg fasudil (n = 15) or placebo (n = 8). RESULTS After fasudil, there were significant decreases in both mean arterial pressure (P < 0.05) and systemic vascular resistance (P < 0.05), whereas the heart rate increased significantly (P < 0.05). There was a significant decrease in the hepatic venous pressure gradient (P < 0.05). Portal vascular resistance also decreased significantly (P < 0.01). Placebo caused no significant effects. There were no symptomatic reactions caused by changes in the mean arterial pressure or heart rate after fasudil. CONCLUSIONS In cirrhotic patients with portal hypertension, fasudil lowers portal vascular resistance, resulting in decreased portal venous pressure with reducing arterial pressure.
Collapse
Affiliation(s)
- Takeshi Fukuda
- Division of Gastroenterology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Pharmacologic inhibition of ROCK2 suppresses amyloid-β production in an Alzheimer's disease mouse model. J Neurosci 2014; 33:19086-98. [PMID: 24305806 DOI: 10.1523/jneurosci.2508-13.2013] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia and has no cure. Genetic, cell biological, and biochemical studies suggest that reducing amyloid-β (Aβ) production may serve as a rational therapeutic avenue to delay or prevent AD progression. Inhibition of RhoA, a Rho GTPase family member, is proposed to curb Aβ production. However, a barrier to this hypothesis has been the limited understanding of how the principal downstream effectors of RhoA, Rho-associated, coiled-coil containing protein kinase (ROCK) 1 and ROCK2, modulate Aβ generation. Here, we report that ROCK1 knockdown increased endogenous human Aβ production, whereas ROCK2 knockdown decreased Aβ levels. Inhibition of ROCK2 kinase activity, using an isoform-selective small molecule (SR3677), suppressed β-site APP cleaving enzyme 1 (BACE1) enzymatic action and diminished production of Aβ in AD mouse brain. Immunofluorescence and confocal microscopy analyses revealed that SR3677 alters BACE1 endocytic distribution and promotes amyloid precursor protein (APP) traffic to lysosomes. Moreover, SR3677 blocked ROCK2 phosphorylation of APP at threonine 654 (T654); in neurons, T654 was critical for APP processing to Aβ. These observations suggest that ROCK2 inhibition reduces Aβ levels through independent mechanisms. Finally, ROCK2 protein levels were increased in asymptomatic AD, mild cognitive impairment, and AD brains, demonstrating that ROCK2 levels change in the earliest stages of AD and remain elevated throughout disease progression. Collectively, these findings highlight ROCK2 as a mechanism-based therapeutic target to combat Aβ production in AD.
Collapse
|