1
|
Smith CEL, Streets AJ, Lake AVR, Natarajan S, Best SK, Szymanska K, Karwatka M, Stevenson T, Trowbridge R, Grant G, Grellscheid SN, Foster R, Morrison CG, Mavria G, Bond J, Ong ACM, Johnson CA. Drug and siRNA screens identify ROCK2 as a therapeutic target for ciliopathies. COMMUNICATIONS MEDICINE 2025; 5:129. [PMID: 40253509 PMCID: PMC12009310 DOI: 10.1038/s43856-025-00847-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 04/04/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND Primary cilia mediate vertebrate development and growth factor signalling. Defects in primary cilia cause inherited developmental conditions termed ciliopathies. Ciliopathies often present with cystic kidney disease, a major cause of early renal failure. Currently, only one drug, Tolvaptan, is licensed to slow the decline of renal function for the ciliopathy polycystic kidney disease. Novel therapeutic interventions are needed. METHODS We screened clinical development compounds to identify those that reversed cilia loss due to siRNA knockdown. In parallel, we undertook a whole genome siRNA-based reverse genetics phenotypic screen to identify positive modulators of cilia formation. RESULTS Using a clinical development compound screen, we identify fasudil hydrochloride. Fasudil is a generic, off-patent drug that is a potent, broadly selective Rho-associated coiled-coil-containing protein kinase (ROCK) inhibitor. In parallel, the siRNA screen identifies ROCK2 and we demonstrate that ROCK2 is a key mediator of cilium formation and function through its possible effects on actin cytoskeleton remodelling. CONCLUSIONS Our results indicate that specific ROCK2 inhibitors (e.g. belumosudil) could be repurposed for cystic kidney disease treatment. We propose that ROCK2 inhibition represents a novel, disease-modifying therapeutic approach for heterogeneous ciliopathies.
Collapse
Affiliation(s)
- Claire E L Smith
- Division of Molecular Medicine, Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Andrew J Streets
- Academic Nephrology Unit, Division of Clinical Medicine, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield, UK
| | - Alice V R Lake
- Division of Molecular Medicine, Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Subaashini Natarajan
- Division of Molecular Medicine, Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Sunayna K Best
- Division of Molecular Medicine, Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Katarzyna Szymanska
- Division of Molecular Medicine, Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Magdalena Karwatka
- Division of Molecular Medicine, Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Thomas Stevenson
- Computational Biology Unit, Department of Biosciences, University of Bergen, Bergen, Norway
| | - Rachel Trowbridge
- School of Molecular & Cellular Biology & Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Gary Grant
- Division of Molecular Medicine, Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Sushma N Grellscheid
- Computational Biology Unit, Department of Biosciences, University of Bergen, Bergen, Norway
- Department of Biosciences, University of Durham, Durham, UK
| | - Richard Foster
- School of Molecular & Cellular Biology & Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
- School of Chemistry, University of Leeds, Leeds, UK
| | - Ciaran G Morrison
- Centre for Chromosome Biology, School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Georgia Mavria
- Division of Molecular Medicine, Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Jacquelyn Bond
- St James's Campus Infrastructure Flow Cytometry and Imaging Facility, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Leeds, UK
| | - Albert C M Ong
- Academic Nephrology Unit, Division of Clinical Medicine, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield, UK
| | - Colin A Johnson
- Division of Molecular Medicine, Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK.
| |
Collapse
|
2
|
Kuwano A, Ishiguro T, Nomura S, Omura Y, Hodotsuka K, Tanaka Y, Murakami M, Kawamata T, Kawashima A. Predictive factors for improvement of symptomatic cerebral vasospasm following subarachnoid hemorrhage by selective intra-arterial administration of fasudil hydrochloride. Interv Neuroradiol 2025; 31:175-180. [PMID: 36740915 PMCID: PMC12035402 DOI: 10.1177/15910199231155037] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/17/2023] [Indexed: 02/07/2023] Open
Abstract
BackgroundSymptomatic cerebral vasospasm after subarachnoid hemorrhage (SAH) is a significant cause of delayed cerebral ischemia that leads to poor outcomes. Selective intra-arterial administration of fasudil hydrochloride (IAF) has been adopted for its vasodilatory effect on spasm arteries to prevent delayed cerebral ischemia. However, its effect on clinical outcomes and predictive factors for good recovery are not fully understood. This study aimed to investigate the outcomes of selective IAF and identify predictive factors for good outcomes in patients with cerebral vasospasm after SAH.MethodsA retrospective study of 36 patients with cerebral vasospasm following SAH who underwent selective IAF at our institution between January 2014 and May 2022 was conducted. We evaluated the improvements in neurological findings before and after selective IAF. Statistical analyses were performed to determine factors associated with good outcomes.ResultsSelective IAF improved the neurological findings in 26 patients (72.2%). Pre-therapeutic absence of cerebral infarction in more than 1/3 of the spasm artery perfusion area was significantly associated with an improvement in neurological findings (p < 0.0001). Furthermore, there was a tendency for a good outcome when the age was younger (p = 0.093), and the spasm was limited to peripheral vessels (p = 0.065).ConclusionOur study indicates that selective IAF has a promising effect in improving symptomatic vasospasm, except when a large cerebral infarction exists in the spasm artery perfusion area. Early consideration of selective IAF could be recommended once patients experience symptomatic cerebral vasospasm after SAH.
Collapse
Affiliation(s)
- Atsushi Kuwano
- Department of Neurosurgery, Tokyo Women's Medical University Yachiyo Medical Center, Yachiyo-city, Japan
| | - Taichi Ishiguro
- Department of Neurosurgery, Tokyo Women's Medical University Yachiyo Medical Center, Yachiyo-city, Japan
| | - Shunsuke Nomura
- Department of Neurosurgery, Tokyo Women's Medical University Yachiyo Medical Center, Yachiyo-city, Japan
| | - Yoshihiro Omura
- Department of Neurosurgery, Tokyo Women's Medical University Yachiyo Medical Center, Yachiyo-city, Japan
| | - Kenichi Hodotsuka
- Department of Neurosurgery, Tokyo Women's Medical University Yachiyo Medical Center, Yachiyo-city, Japan
| | - Yukiko Tanaka
- Department of Neurosurgery, Tokyo Women's Medical University Yachiyo Medical Center, Yachiyo-city, Japan
| | - Masato Murakami
- Department of Neurosurgery, Tokyo Women's Medical University Yachiyo Medical Center, Yachiyo-city, Japan
| | - Takakazu Kawamata
- Department of Neurosurgery, Tokyo Women's Medical University, Shinjuku, Japan
| | - Akitsugu Kawashima
- Department of Neurosurgery, Tokyo Women's Medical University Yachiyo Medical Center, Yachiyo-city, Japan
| |
Collapse
|
3
|
Liu X, Qian Z, Li Y, Wang Y, Zhang Y, Zhang Y, Enoch IVMV. Unveiling synergies: Integrating TCM herbal medicine and acupuncture with conventional approaches in stroke management. Neuroscience 2025; 567:109-122. [PMID: 39730019 DOI: 10.1016/j.neuroscience.2024.12.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/09/2024] [Accepted: 12/21/2024] [Indexed: 12/29/2024]
Abstract
This review explores the mechanisms and treatment strategies of ischemic stroke, a leading cause of morbidity and mortality worldwide. Ischemic stroke results from the obstruction of blood flow to the brain, leading to significant neurological impairment. The paper categorizes ischemic stroke into subtypes based on etiology, including cardioembolism and large artery atherosclerosis, and discusses the challenges of current therapeutic approaches. Conventional treatments like tissue plasminogen activator (tPA) and surgical interventions are limited by narrow windows and potential complications. The review highlights the promise of acupuncture, which offers neuroprotective benefits by promoting cerebral ischemic tolerance and neural regeneration. Integrating acupuncture with conventional treatments may enhance patient outcomes. Emphasis is placed on understanding the pathophysiology to develop targeted therapies that mitigate neuronal damage and enhance recovery.
Collapse
Affiliation(s)
- Xiliang Liu
- Department of Rehabilitation Medicine, Dezhou Traditional Chinese Medicine Hospital, Dezhou 253000, China
| | - Zhendong Qian
- Department of Rehabilitation Medicine, Dezhou Traditional Chinese Medicine Hospital, Dezhou 253000, China
| | - Yuxuan Li
- Department of Rehabilitation Medicine, Dezhou Traditional Chinese Medicine Hospital, Dezhou 253000, China
| | - Yanwei Wang
- Department of Rehabilitation Medicine, Dezhou Traditional Chinese Medicine Hospital, Dezhou 253000, China
| | - Yan Zhang
- Department of Rehabilitation Medicine, Dezhou Traditional Chinese Medicine Hospital, Dezhou 253000, China
| | - Yu Zhang
- Department of Rehabilitation Medicine, Dezhou Traditional Chinese Medicine Hospital, Dezhou 253000, China.
| | - Israel V M V Enoch
- Centre for Nanoscience and Genomics, Karunya Institute of Technology and Sciences (Deemed University), Coimbatore 641114, Tamil Nadu, India
| |
Collapse
|
4
|
Imai T, Qin T, Morais A, Sasaki Y, Erdogan T, McKerracher L, Ayata C. Isoform-selective and non-selective rho-kinase inhibitors do not affect collagenase-induced intracerebral hemorrhage outcomes in mice: Influence of sex and circadian cycle. J Cereb Blood Flow Metab 2025:271678X241312010. [PMID: 39763388 PMCID: PMC11705295 DOI: 10.1177/0271678x241312010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 12/04/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025]
Abstract
Rho-associated protein kinase (ROCK) inhibitors are therapeutic candidates in ischemic stroke and subarachnoid hemorrhage. However, their efficacy in intracerebral hemorrhage (ICH) is unknown. Here, we tested the efficacy of fasudil (10 mg/kg), an isoform-nonselective ROCK inhibitor, and NRL-1049 (10 mg/kg), a novel inhibitor with 43-fold higher selectivity for ROCK2 isoform compared with ROCK1, in a collagenase-induced ICH model in mice. Both short (1-3 days) and prolonged (14 days) therapeutic paradigms were tested using robust sample sizes in both males and females and in active and inactive circadian stages. Outcome readouts included weight loss, mortality, hematoma volume, hemispheric swelling, brain water content, BBB permeability to large molecules, and sensorimotor and cognitive function. We found the treatments safe but not efficacious in improving the hematoma volume, BBB disruption, or neurological deficits in this collagenase-induced ICH model. Intriguingly, however, induction of ICH during the active circadian stage was associated with worse tissue and behavioral outcomes compared with the inactive stage.
Collapse
Affiliation(s)
- Takahiko Imai
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Tao Qin
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Andreia Morais
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Yuichi Sasaki
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Taylan Erdogan
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | | | - Cenk Ayata
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| |
Collapse
|
5
|
Peng B, Mohammed FS, Tang X, Liu J, Sheth KN, Zhou J. Nanotechnology approaches to drug delivery for the treatment of ischemic stroke. Bioact Mater 2025; 43:145-161. [PMID: 39386225 PMCID: PMC11462157 DOI: 10.1016/j.bioactmat.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024] Open
Abstract
Ischemic stroke is a major global public health concern that lacks effective treatment options. A significant challenge lies in delivering therapeutic agents to the brain due to the restrictive nature of the blood-brain barrier (BBB). The BBB's selectivity hampers the delivery of therapeutically relevant quantities of agents to the brain, resulting in a lack of FDA-approved pharmacotherapies for stroke. In this article, we review therapeutic agents that have been evaluated in clinical trials or are currently undergoing clinical trials. Subsequently, we survey strategies for synthesizing and engineering nanoparticles (NPs) for drug delivery to the ischemic brain. We then provide insights into the potential clinical translation of nanomedicine, offering a perspective on its transformative role in advancing stroke treatment strategies. In summary, existing literature suggests that drug delivery represents a major barrier for clinical translation of stroke pharmacotherapies. While nanotechnology has shown significant promise in addressing this challenge, further advancements aimed at improving delivery efficiency and simplifying formulations are necessary for successful clinical translation.
Collapse
Affiliation(s)
- Bin Peng
- Department of Neurosurgery, New Haven, CT, 06510, USA
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Farrah S. Mohammed
- Department of Neurosurgery, New Haven, CT, 06510, USA
- Department of Biomedical Engineering, New Haven, CT, 06510, USA
| | - Xiangjun Tang
- Department of Neurosurgery, New Haven, CT, 06510, USA
- Department of Neurosurgery, Taihe Hospital, Hubei, 442000, PR China
| | - Jia Liu
- Department of Neurosurgery, New Haven, CT, 06510, USA
| | - Kevin N. Sheth
- Department of Neurosurgery, New Haven, CT, 06510, USA
- Department of Neurology, Yale University, New Haven, CT, 06510, USA
| | - Jiangbing Zhou
- Department of Neurosurgery, New Haven, CT, 06510, USA
- Department of Biomedical Engineering, New Haven, CT, 06510, USA
| |
Collapse
|
6
|
Bachman NP, Ketelhut NB, Blomquist M, Terwoord JD. Rho-kinase inhibition reduces systolic blood pressure and forearm vascular resistance in healthy older adults: a double-blind, randomized, placebo-controlled pilot study. GeroScience 2024; 46:6317-6329. [PMID: 38888876 PMCID: PMC11494619 DOI: 10.1007/s11357-024-01240-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024] Open
Abstract
Rho-kinase has been implicated in the development of hypertension in preclinical studies and may contribute to age-related blood pressure elevation. This study tested the hypothesis that Rho-kinase contributes to elevated systolic blood pressure (SBP) in healthy older adults. Young (18-30 years, 6F/6M) and older (60-80 years, 7F/6M) adults were enrolled in a double-blind, placebo-controlled crossover study using intravenous fasudil infusion to inhibit Rho-kinase. Fasudil lowered SBP in older adults compared to placebo (saline) (2-h post-infusion: 125 ± 4 vs. 133 ± 4 mmHg, P < 0.05), whereas fasudil had no impact on SBP in young adults. Immediately following fasudil infusion, there was a transient reduction in mean arterial pressure (MAP) in young adults that was no longer evident 1-h post-infusion. In older adults, MAP remained lower throughout the fasudil visit compared to placebo (2-h post-infusion: 93 ± 3 vs. 100 ± 3 mmHg, P < 0.05) such that age-related differences in SBP and MAP were abolished. Aortic stiffness (carotid-femoral pulse wave velocity) was not altered by fasudil when central MAP was included as a covariate in analyses. Fasudil reduced forearm vascular resistance in older (2-h post-infusion: 3.3 ± 0.4 vs. 4.8 ± 0.6 mmHg/ml/min, P < 0.05) but not young (4.0 ± 0.6 vs. 3.8 ± 0.5 mmHg/ml/min) adults, which was accompanied by an increase in brachial artery diameter only in older adults. Brachial artery flow-mediated dilation was not affected by fasudil in either group. These findings indicate that Rho-kinase inhibition reduces SBP in healthy older but not young adults, which is associated with a concomitant reduction in forearm vascular resistance.
Collapse
Affiliation(s)
- Nate P Bachman
- Department of Kinesiology, Colorado Mesa University, Grand Junction, CO, USA
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
| | - Nathaniel B Ketelhut
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
| | - Michael Blomquist
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
- Biomedical Sciences Department, Rocky Vista University, 255 E. Center St., Ivins, UT, 84738, USA
| | - Janée D Terwoord
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA.
- Biomedical Sciences Department, Rocky Vista University, 255 E. Center St., Ivins, UT, 84738, USA.
| |
Collapse
|
7
|
Koch JC, Leha A, Bidner H, Cordts I, Dorst J, Günther R, Zeller D, Braun N, Metelmann M, Corcia P, De La Cruz E, Weydt P, Meyer T, Großkreutz J, Soriani MH, Attarian S, Weishaupt JH, Weyen U, Kuttler J, Zurek G, Rogers ML, Feneberg E, Deschauer M, Neuwirth C, Wuu J, Ludolph AC, Schmidt J, Remane Y, Camu W, Friede T, Benatar M, Weber M, Lingor P. Safety, tolerability, and efficacy of fasudil in amyotrophic lateral sclerosis (ROCK-ALS): a phase 2, randomised, double-blind, placebo-controlled trial. Lancet Neurol 2024; 23:1133-1146. [PMID: 39424560 DOI: 10.1016/s1474-4422(24)00373-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/18/2024] [Accepted: 08/29/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Fasudil is a small molecule inhibitor of Rho-associated kinase (ROCK) and is approved for the treatment of subarachnoid haemorrhage. In preclinical studies, fasudil has been shown to attenuate neurodegeneration, modulate neuroinflammation, and foster axonal regeneration. We aimed to investigate the safety, tolerability, and efficacy of fasudil in patients with amyotrophic lateral sclerosis. METHODS ROCK-ALS was a phase 2, randomised, double-blind, placebo-controlled trial conducted at 19 amyotrophic lateral sclerosis centres in Germany, France, and Switzerland. Individuals (aged 18-80 years) with at least probable amyotrophic lateral sclerosis (as per the revised El Escorial criteria), a disease duration of 6-24 months, and a slow vital capacity greater than 65% of predicted normal were eligible for inclusion. Patients were randomly assigned (1:1:1) to receive 30 mg (15 mg twice daily) or 60 mg (30 mg twice daily) fasudil or matched placebo intravenously for 20 days over a 4-week period. Follow-up assessments were performed at 45, 90, and 180 days after treatment initiation. The co-primary endpoints were safety until day 180 (defined as the proportion without drug-related serious adverse events) and tolerability during the treatment period (defined as the proportion who did not discontinue treatment due to suspected drug-related adverse events). The primary analyses were carried out in the intention-to-treat population, which included all participants who entered the treatment phase. This trial is registered at ClinicalTrials.gov (NCT03792490) and Eudra-CT (2017-003676-31) and is now completed. FINDINGS Between Feb 20, 2019, and April 20, 2022, 120 participants were enrolled and randomised; two individuals assigned fasudil 30 mg withdrew consent before the baseline visit. Thus, the intention-to-treat population comprised 35 in the fasudil 30 mg group, 39 in the fasudil 60 mg group, and 44 in the placebo group. The estimated proportion without a drug-related serious adverse event was 1·00 (95% CI 0·91 to 1·00) with placebo, 1·00 (0·89 to 1·00) with fasudil 30 mg, and 1·00 (0·90 to 1·00) with fasudil 60 mg; the difference in proportions was 0·00 (95% CI -0·11 to 0·10; p>0·99) for fasudil 30 mg versus placebo and 0·00 (-0·10 to 0·10; p>0·99) for fasudil 60 mg versus placebo. Treatment tolerability (the estimated proportion who did not discontinue) was 0·93 (95% CI 0·81 to 0·99) with placebo, 1·00 (0·90 to 1·00) with fasudil 30 mg, and 0·90 (0·76 to 0·97) with fasudil 60 mg; the difference in proportions was 0·07 (95% CI -0·05 to 0·20; p=0·25) for fasudil 30 mg versus placebo, and -0·03 (-0·18 to 0·10; p=0·70) for fasudil 60 mg versus placebo. Eight deaths occurred: two in the placebo group, four in the fasudil 30 mg group, and two in the fasudil 60 mg group. The most common serious adverse events were respiratory failure (seven events), gastrostomy (five events), pneumonia (four events), and dysphagia (four events). No serious adverse events or deaths were attributed to study treatment. Adverse events, which were mainly related to disease progression, occurred in 139 participants in the placebo group, 108 in the fasudil 30 mg group, and 105 in the fasudil 60 mg group. INTERPRETATION Fasudil was well tolerated and safe in people with amyotrophic lateral sclerosis. The effect of fasudil on efficacy outcomes should be explored in larger clinical trials with a longer treatment duration, oral administration, and potentially higher dose of the trial drug. FUNDING Framework of the E-Rare Joint Transnational Call 2016 "Clinical research for new therapeutic uses of already existing molecules (repurposing) in rare diseases".
Collapse
Affiliation(s)
- Jan C Koch
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Andreas Leha
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
| | - Helen Bidner
- Münchner Studienzentrum, Technical University Munich, School of Medicine and Health, Munich, Germany
| | - Isabell Cordts
- Department of Neurology, Klinikum rechts der Isar, Technical University Munich, School of Medicine and Health, Munich, Germany
| | | | - René Günther
- Department of Neurology, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany; German Centre for Neurodegenerative Diseases, Site Dresden, Dresden, Germany
| | - Daniel Zeller
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Nathalie Braun
- Neuromuscular Diseases Unit/ALS Clinic, Cantonal Hospital St Gallen, St Gallen, Switzerland
| | - Moritz Metelmann
- Department of Neurology, University Hospital Leipzig, Leipzig, Germany
| | - Philippe Corcia
- Centre de Référence Maladie Rare (CRMR) SLA et les Autres Maladies du Neurone Moteur (FILSLAN), Tours, France; Faculté de Médecine, INSERM U1253, "iBrain Imaging Brain and Neuropsychiatry" Université François-Rabelais de Tours, Tours, France
| | - Elisa De La Cruz
- ALS centre, CHU Gui de Chauliac, Univ Montpellier, INM, INSERM, Montpellier, France
| | - Patrick Weydt
- Department for Neuromuscular Disorders, University Hospital Bonn, Bonn, Germany; German Centre for Neurodegenerative Diseases, Site Bonn, Bonn, Germany
| | - Thomas Meyer
- Department of Neurology, Center for ALS and Other Motor Neuron Disorders, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Julian Großkreutz
- Department of Neurology, Jena University Hospital, Jena, Germany; Precision Neurology of Neuromuscular and Motoneuron Diseases, University of Lübeck, Lübeck, Germany
| | - Marie-Hélène Soriani
- ALS Reference Centre, Pasteur 2 Hospital, CHU de Nice, Université Côte d'Azur, UMR2CA, Nice, France
| | - Shahram Attarian
- Neuromuscular Disease and ALS Reference Center, Timone University Hospital, Aix-Marseille University, CHU Timone, Marseille, France
| | - Jochen H Weishaupt
- Division for Neurodegenerative Diseases, Neurology Department, Mannheim Center for Translational Medicine, University Medicine Mannheim, Heidelberg University, Mannheim, Germany
| | - Ute Weyen
- Department of Neurology, Ruhr-University Bochum, BG-Kliniken Bergmannsheil, Bochum, Germany
| | - Josua Kuttler
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Mary-Louise Rogers
- MND&NR Lab, FHMRI, College of Medicine and Public health, Flinders University, Bedford Park, Adelaide, SA, Australia
| | - Emily Feneberg
- Department of Neurology, Klinikum rechts der Isar, Technical University Munich, School of Medicine and Health, Munich, Germany
| | - Marcus Deschauer
- Department of Neurology, Klinikum rechts der Isar, Technical University Munich, School of Medicine and Health, Munich, Germany
| | - Christoph Neuwirth
- Neuromuscular Diseases Unit/ALS Clinic, Cantonal Hospital St Gallen, St Gallen, Switzerland
| | - Joanne Wuu
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Albert C Ludolph
- Department of Neurology, Ulm University, Ulm, Germany; German Center for Neurodegenerative Diseases, Site Ulm, Ulm, Germany
| | - Jens Schmidt
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany; Department of Neurology and Pain Treatment, Neuromuscular Center, Center for Translational Medicine, Immanuel University Hospital Rüdersdorf, Brandenburg Medical School Theodor Fontane, Rüdersdorf bei Berlin, Berlin, Germany; Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Rüdersdorf bei Berlin, Berlin, Germany
| | - Yvonne Remane
- Central Pharmacy, Leipzig University Medical Center, Leipzig, Germany
| | - William Camu
- ALS centre, CHU Gui de Chauliac, Univ Montpellier, INM, INSERM, Montpellier, France
| | - Tim Friede
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
| | - Michael Benatar
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Markus Weber
- Neuromuscular Diseases Unit/ALS Clinic, Cantonal Hospital St Gallen, St Gallen, Switzerland
| | - Paul Lingor
- Department of Neurology, Klinikum rechts der Isar, Technical University Munich, School of Medicine and Health, Munich, Germany; German Center for Neurodegenerative Diseases, Site Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
8
|
Mulder IA, Abbinanti M, Woller SA, Ruschel J, Coutinho JM, de Vries HE, van Bavel E, Rosen K, McKerracher L, Ayata C. The novel ROCK2 selective inhibitor NRL-1049 preserves the blood-brain barrier after acute injury. J Cereb Blood Flow Metab 2024; 44:1238-1252. [PMID: 38833563 PMCID: PMC11542141 DOI: 10.1177/0271678x241238845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/13/2024] [Accepted: 02/19/2024] [Indexed: 06/06/2024]
Abstract
Endothelial blood-brain barrier (BBB) dysfunction is critical in the pathophysiology of brain injury. Rho-associated protein kinase (ROCK) activation disrupts BBB integrity in the injured brain. We aimed to test the efficacy of a novel ROCK2 inhibitor in preserving the BBB after acute brain injury. We characterized the molecular structure and pharmacodynamic and pharmacokinetic properties of a novel selective ROCK2 inhibitor, NRL-1049, and its first metabolite, 1-hydroxy-NRL-1049 (referred to as NRL-2017 hereon) and tested the efficacy of NRL-1049 on the BBB integrity in rodent models of acute brain injury. Our data show that NRL-1049 and NRL-2017 both inhibit ROCK activity and are 44-fold and 17-fold more selective towards ROCK2 than ROCK1, respectively. When tested in a mouse model of cortical cryoinjury, NRL-1049 significantly attenuated the increase in water content. Interestingly, 60% of the mice in the vehicle arm developed seizures within 2 hours after cryoinjury versus none in the NRL-1049 arm. In spontaneously hypertensive rats, NRL-1049 attenuated the dramatic surge in Evans Blue extravasation compared with the vehicle arm after transient middle cerebral artery occlusion. Hemorrhagic transformation was also reduced. We show that NRL-1049, a selective ROCK2 inhibitor, is a promising drug candidate to preserve the BBB after brain injury.
Collapse
Affiliation(s)
- Inge A Mulder
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Biomedical Engineering and Physics, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, the Netherlands
- Amsterdam Neurosciences, Neurovascular Disorders, Amsterdam, the Netherlands
| | | | | | | | - Jonathan M Coutinho
- Amsterdam Neurosciences, Neurovascular Disorders, Amsterdam, the Netherlands
- Department of Neurology, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
| | - Helga E de Vries
- Amsterdam Neurosciences, Neurovascular Disorders, Amsterdam, the Netherlands
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit, Amsterdam, the Netherlands
| | - Ed van Bavel
- Department of Biomedical Engineering and Physics, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, the Netherlands
- Amsterdam Neurosciences, Neurovascular Disorders, Amsterdam, the Netherlands
| | | | - Lisa McKerracher
- BioAxone BioSciences Inc, Boston, MA, USA
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Cenk Ayata
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Stroke Service, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Sajib MS, Zahra FT, Lamprou M, Akwii RG, Park JH, Osorio M, Tullar P, Doci CL, Zhang C, Huveneers S, Van Buul JD, Wang MH, Markiewski MM, Srivastava SK, Zheng Y, Gutkind JS, Hu J, Bickel U, Maeda DY, Zebala JA, Lionakis MS, Trasti S, Mikelis CM. Tumor-induced endothelial RhoA activation mediates tumor cell transendothelial migration and metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.22.614304. [PMID: 39372784 PMCID: PMC11451620 DOI: 10.1101/2024.09.22.614304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
The endothelial barrier plays an active role in transendothelial tumor cell migration during metastasis, however, the endothelial regulatory elements of this step remain obscure. Here we show that endothelial RhoA activation is a determining factor during this process. Breast tumor cell-induced endothelial RhoA activation is the combined outcome of paracrine IL-8-dependent and cell-to-cell contact β 1 integrin-mediated mechanisms, with elements of this pathway correlating with clinical data. Endothelial-specific RhoA blockade or in vivo deficiency inhibited the transendothelial migration and metastatic potential of human breast tumor and three murine syngeneic tumor cell lines, similar to the pharmacological blockade of the downstream RhoA pathway. These findings highlight endothelial RhoA as a potent, universal target in the tumor microenvironment for anti-metastatic treatment of solid tumors.
Collapse
|
10
|
Zhao M, Wang J, Liu G, Li S, Ding Y, Ji X, Zhao W. Multi-Target and Multi-Phase Adjunctive Cerebral Protection for Acute Ischemic Stroke in the Reperfusion Era. Biomolecules 2024; 14:1181. [PMID: 39334947 PMCID: PMC11429592 DOI: 10.3390/biom14091181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/25/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Stroke remains the leading cause of death and disability in some countries, predominantly attributed to acute ischemic stroke (AIS). While intravenous thrombolysis and endovascular thrombectomy are widely acknowledged as effective treatments for AIS, boasting a high recanalization rate, there is a significant discrepancy between the success of revascularization and the mediocre clinical outcomes observed among patients with AIS. It is now increasingly understood that the implementation of effective cerebral protection strategies, serving as adjunctive treatments to reperfusion, can potentially improve the outcomes of AIS patients following recanalization therapy. Herein, we reviewed several promising cerebral protective methods that have the potential to slow down infarct growth and protect ischemic penumbra. We dissect the underlying reasons for the mismatch between high recanalization rates and moderate prognosis and introduce a novel concept of "multi-target and multi-phase adjunctive cerebral protection" to guide our search for neuroprotective agents that can be administered alongside recanalization therapy.
Collapse
Affiliation(s)
- Min Zhao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Jing Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Guiyou Liu
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Sijie Li
- Department of Emergency, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Xunming Ji
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Wenbo Zhao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| |
Collapse
|
11
|
Islam MM, Gaska I, Oshinowo O, Otumala A, Shekhar S, Au Yong N, Myers DR. Single-pericyte nanomechanics measured by contraction cytometry. APL Bioeng 2024; 8:036109. [PMID: 39131206 PMCID: PMC11316606 DOI: 10.1063/5.0213761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 07/02/2024] [Indexed: 08/13/2024] Open
Abstract
Pericytes line the microvasculature throughout the body and play a key role in regulating blood flow by constricting and dilating vessels. However, the biophysical mechanisms through which pericytes transduce microenvironmental chemical and mechanical cues to mediate vessel diameter, thereby impacting oxygen and nutrient delivery, remain largely unknown. This knowledge gap is clinically relevant as numerous diseases are associated with the aberrant contraction of pericytes, which are unusually susceptible to injury. Here, we report the development of a high-throughput hydrogel-based pericyte contraction cytometer that quantifies single-cell contraction forces from murine and human pericytes in different microvascular microenvironments and in the presence of competing vasoconstricting and vasodilating stimuli. We further show that murine pericyte survival in hypoxia is mediated by the mechanical microenvironment and that, paradoxically, pre-treating pericytes to reduce contraction increases hypoxic cell death. Moreover, using the contraction cytometer as a drug-screening tool, we found that cofilin-1 could be applied extracellularly to release murine pericytes from hypoxia-induced contractile rigor mortis and, therefore, may represent a novel approach for mitigating the long-lasting decrease in blood flow that occurs after hypoxic injury.
Collapse
Affiliation(s)
| | - Ignas Gaska
- Departments of Physics, Cell Biology and Biochemistry, Emory University, Atlanta, Georgia 30322, USA
| | | | | | - Shashank Shekhar
- Departments of Physics, Cell Biology and Biochemistry, Emory University, Atlanta, Georgia 30322, USA
| | | | - David R. Myers
- Author to whom correspondence should be addressed:. Tel.: 404-727-0401
| |
Collapse
|
12
|
Góral I, Wichur T, Sługocka E, Godyń J, Szałaj N, Zaręba P, Głuch-Lutwin M, Mordyl B, Panek D, Więckowska A. Connecting GSK-3β Inhibitory Activity with IKK-β or ROCK-1 Inhibition to Target Tau Aggregation and Neuroinflammation in Alzheimer's Disease-Discovery, In Vitro and In Cellulo Activity of Thiazole-Based Inhibitors. Molecules 2024; 29:2616. [PMID: 38893493 PMCID: PMC11173485 DOI: 10.3390/molecules29112616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/26/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
GSK-3β, IKK-β, and ROCK-1 kinases are implicated in the pathomechanism of Alzheimer's disease due to their involvement in the misfolding and accumulation of amyloid β (Aβ) and tau proteins, as well as inflammatory processes. Among these kinases, GSK-3β plays the most crucial role. In this study, we present compound 62, a novel, remarkably potent, competitive GSK-3β inhibitor (IC50 = 8 nM, Ki = 2 nM) that also exhibits additional ROCK-1 inhibitory activity (IC50 = 2.3 µM) and demonstrates anti-inflammatory and neuroprotective properties. Compound 62 effectively suppresses the production of nitric oxide (NO) and pro-inflammatory cytokines in the lipopolysaccharide-induced model of inflammation in the microglial BV-2 cell line. Furthermore, it shows neuroprotective effects in an okadaic-acid-induced tau hyperphosphorylation cell model of neurodegeneration. The compound also demonstrates the potential for further development, characterized by its chemical and metabolic stability in mouse microsomes and fair solubility.
Collapse
Affiliation(s)
- Izabella Góral
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (I.G.); (T.W.); (E.S.); (J.G.); (N.S.); (P.Z.); (D.P.)
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, 16 Lazarza St., 31-530 Krakow, Poland
| | - Tomasz Wichur
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (I.G.); (T.W.); (E.S.); (J.G.); (N.S.); (P.Z.); (D.P.)
| | - Emilia Sługocka
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (I.G.); (T.W.); (E.S.); (J.G.); (N.S.); (P.Z.); (D.P.)
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, 16 Lazarza St., 31-530 Krakow, Poland
| | - Justyna Godyń
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (I.G.); (T.W.); (E.S.); (J.G.); (N.S.); (P.Z.); (D.P.)
| | - Natalia Szałaj
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (I.G.); (T.W.); (E.S.); (J.G.); (N.S.); (P.Z.); (D.P.)
| | - Paula Zaręba
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (I.G.); (T.W.); (E.S.); (J.G.); (N.S.); (P.Z.); (D.P.)
| | - Monika Głuch-Lutwin
- Department of Pharmacobiology, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (M.G.-L.); (B.M.)
| | - Barbara Mordyl
- Department of Pharmacobiology, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (M.G.-L.); (B.M.)
| | - Dawid Panek
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (I.G.); (T.W.); (E.S.); (J.G.); (N.S.); (P.Z.); (D.P.)
| | - Anna Więckowska
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (I.G.); (T.W.); (E.S.); (J.G.); (N.S.); (P.Z.); (D.P.)
| |
Collapse
|
13
|
Pinoșanu EA, Pîrșcoveanu D, Albu CV, Burada E, Pîrvu A, Surugiu R, Sandu RE, Serb AF. Rhoa/ROCK, mTOR and Secretome-Based Treatments for Ischemic Stroke: New Perspectives. Curr Issues Mol Biol 2024; 46:3484-3501. [PMID: 38666949 PMCID: PMC11049286 DOI: 10.3390/cimb46040219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Ischemic stroke triggers a complex cascade of cellular and molecular events leading to neuronal damage and tissue injury. This review explores the potential therapeutic avenues targeting cellular signaling pathways implicated in stroke pathophysiology. Specifically, it focuses on the articles that highlight the roles of RhoA/ROCK and mTOR signaling pathways in ischemic brain injury and their therapeutic implications. The RhoA/ROCK pathway modulates various cellular processes, including cytoskeletal dynamics and inflammation, while mTOR signaling regulates cell growth, proliferation, and autophagy. Preclinical studies have demonstrated the neuroprotective effects of targeting these pathways in stroke models, offering insights into potential treatment strategies. However, challenges such as off-target effects and the need for tissue-specific targeting remain. Furthermore, emerging evidence suggests the therapeutic potential of MSC secretome in stroke treatment, highlighting the importance of exploring alternative approaches. Future research directions include elucidating the precise mechanisms of action, optimizing treatment protocols, and translating preclinical findings into clinical practice for improved stroke outcomes.
Collapse
Affiliation(s)
- Elena Anca Pinoșanu
- Department of Neurology, University of Medicine and Pharmacy of Craiova, St. Petru Rares, No. 2-4, 200433 Craiova, Romania; (E.A.P.); (D.P.); (C.V.A.)
- Doctoral School, University of Medicine and Pharmacy of Craiova, St. Petru Rares, No. 2-4, 200433 Craiova, Romania
| | - Denisa Pîrșcoveanu
- Department of Neurology, University of Medicine and Pharmacy of Craiova, St. Petru Rares, No. 2-4, 200433 Craiova, Romania; (E.A.P.); (D.P.); (C.V.A.)
| | - Carmen Valeria Albu
- Department of Neurology, University of Medicine and Pharmacy of Craiova, St. Petru Rares, No. 2-4, 200433 Craiova, Romania; (E.A.P.); (D.P.); (C.V.A.)
| | - Emilia Burada
- Department of Physiology, University of Medicine and Pharmacy of Craiova, St. Petru Rares, No. 2-4, 200433 Craiova, Romania;
| | - Andrei Pîrvu
- Dolj County Regional Centre of Medical Genetics, Clinical Emergency County Hospital Craiova, St. Tabaci, No. 1, 200642 Craiova, Romania;
| | - Roxana Surugiu
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, St. Petru Rares, No. 2-4, 200433 Craiova, Romania;
| | - Raluca Elena Sandu
- Department of Neurology, University of Medicine and Pharmacy of Craiova, St. Petru Rares, No. 2-4, 200433 Craiova, Romania; (E.A.P.); (D.P.); (C.V.A.)
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, St. Petru Rares, No. 2-4, 200433 Craiova, Romania;
| | - Alina Florina Serb
- Department of Biochemistry and Pharmacology, Biochemistry Discipline, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq., No. 2, 300041 Timisoara, Romania;
| |
Collapse
|
14
|
Wolff AW, Peine J, Höfler J, Zurek G, Hemker C, Lingor P. SAFE-ROCK: A Phase I Trial of an Oral Application of the ROCK Inhibitor Fasudil to Assess Bioavailability, Safety, and Tolerability in Healthy Participants. CNS Drugs 2024; 38:291-302. [PMID: 38416402 PMCID: PMC10980656 DOI: 10.1007/s40263-024-01070-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/09/2024] [Indexed: 02/29/2024]
Abstract
BACKGROUND The intravenous (IV) formulation of Rho-kinase (ROCK) inhibitor fasudil has been approved for the treatment of subarachnoid haemorrhage since 1995. Additionally, fasudil has shown promising preclinical results for various chronic diseases, including neurodegenerative diseases such as amyotrophic lateral sclerosis, Parkinson's disease, and dementia, in which long-term intravenous (IV) administration might not be suitable. OBJECTIVE The objective of this study was to assess the absolute bioavailability of oral, in comparison to IV, application of the approved formulation of fasudil (ERIL®) and to evaluate the safety and tolerability of the oral application of fasudil. METHODS This was a phase I, single-center, open-label, randomized, two period cross-over clinical trial in healthy women and men. By applying a cross-over design, each subject served as their own control. Two treatments were investigated, separated by a wash out phase of at least 3 days. Oral fasudil was administered once on day 1 to assess pharmacokinetics and three times on day 2, at an interval of 8 ± 1 h, to assess safety and gastrointestinal tolerability. For pharmacometrics of IV fasudil, it was administered once on day 1. Plasma profiles of fasudil and its active metabolite hydroxyfasudil after oral or IV administration were measured by liquid chromatography electrospray tandem mass spectrometry. Tolerability was assessed as proportion of subjects without significant drug intolerance, and safety was assessed by the proportion of subjects without clinical or laboratory treatment-associated serious adverse events. Gastrointestinal safety was assessed by applying the gastrointestinal symptom rating scale (GSRS). RESULTS Fourteen subjects aged 30-70 years were included in this trial. After oral administration, fasudil concentrations in blood were mostly very low [1.4 g/L; coefficient of variation (CV) 41.0%]. After IV application, the peak concentration was 100.6 µg/L (CV 74.2%); however, a high variance in peak concentrations were assessed for both treatments. The maximal concentrations of hydroxyfasudil in blood were similar after oral and IV treatment [111.6 µg/L (CV 24.1%) and 108.4 µg/L (CV 19.7%), respectively]. Exposure of hydroxyfasudil (assessed as AUC0-tz) differed between both treatments, with 449 µg × h/L after IV treatment and 309 µg × h/L after oral treatment. Therefore, the absolute bioavailability of hydroxyfasudil after the oral treatment was approximately 69% of the IV treatment. No serious adverse events (SAEs) occurred during this trial, and good tolerability of oral fasudil (90 mg/day) was documented. CONCLUSIONS Oral fasudil was generally well tolerated in the studied population, and no safety concerns were identified. However, systemic bioavailability of oral hydroxyfasudil corresponded to 69%, and dose adjustments need to considered. The results presented here lay grounds for future trials of fasudil in chronic diseases, which require an oral long-term application. This trial was registered with EudraCT (no. 2019-001805-26).
Collapse
Affiliation(s)
- Andreas W Wolff
- Clinical Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Jörg Peine
- Institute for Clinical Research, AtoZ-CRO GmbH, Overath, Germany
| | | | | | - Claus Hemker
- CTC North GmbH & Co. KG at the University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Paul Lingor
- Clinical Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
15
|
Wolff AW, Bidner H, Remane Y, Zimmer J, Aarsland D, Rascol O, Wyse RK, Hapfelmeier A, Lingor P. Protocol for a randomized, placebo-controlled, double-blind phase IIa study of the safety, tolerability, and symptomatic efficacy of the ROCK-inhibitor Fasudil in patients with Parkinson's disease (ROCK-PD). Front Aging Neurosci 2024; 16:1308577. [PMID: 38419648 PMCID: PMC10899319 DOI: 10.3389/fnagi.2024.1308577] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 02/01/2024] [Indexed: 03/02/2024] Open
Abstract
Background The Rho-kinase (ROCK) inhibitor Fasudil has shown symptomatic and disease-modifying effects in Parkinson's disease (PD) models in vitro and in vivo. In Japan, Fasudil has been approved for the treatment of subarachnoid haemorrhage since 1995 and shows a favourable safety profile. Objectives/design To investigate the safety, tolerability, and symptomatic efficacy of ROCK-inhibitor Fasudil in comparison to placebo in a randomized, national, multicenter, double-blind phase IIa study in patients with PD. Methods/analysis We plan to include 75 patients with at least 'probable' PD (MDS criteria), Hoehn and Yahr stages 1-3, and age 30-80 years in 13 German study sites. Patients must be non-fluctuating and their response to PD medication must have been stable for 6 weeks. Patients will be randomly allocated to treatment with the oral investigational medicinal product (IMP) containing either Fasudil in two dosages, or placebo, for a total of 22 days. As primary analysis, non-inferiority of low/high dose of Fasudil on the combined endpoint consisting of occurrence of intolerance and/or treatment-related serious adverse events (SAEs) over 22 days will be assessed in a sequential order, starting with the lower dose. Secondary endpoints will include tolerability alone over 22 days and occurrence of treatment-related SAEs (SARs) over 22 and 50 days and will be compared on group level. Additional secondary endpoints include efficacy on motor and non-motor symptoms, measured on established scales, and will be assessed at several timepoints. Biomaterial will be collected to determine pharmacokinetics of Fasudil and its active metabolite, and to evaluate biomarkers of neurodegeneration. Ethics/registration/discussion After positive evaluation by the competent authority and the ethics committee, patient recruitment started in the 3rd quarter of 2023. ROCK-PD is registered with Eudra-CT (2021-003879-34) and clinicaltrials.gov (NCT05931575). Results of this trial can pave way for conducting extended-duration studies assessing both symptomatic efficacy and disease-modifying properties of Fasudil.
Collapse
Affiliation(s)
- Andreas W Wolff
- Department of Neurology, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Helen Bidner
- Münchner Studienzentrum (MSZ), School of Medicine, Technical University of Munich, Munich, Germany
| | - Yvonne Remane
- Department of Clinical Pharmacy and Drug Safety Center, Leipzig University, Leipzig, Germany
| | - Janine Zimmer
- Department of Clinical Pharmacy and Drug Safety Center, Leipzig University, Leipzig, Germany
| | - Dag Aarsland
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- Centre for Age-Related Research, Stavanger University Hospital, Stavanger, Norway
| | - Olivier Rascol
- Clinical Investigation Center CIC1436, Departments of Clinical Pharmacology and Neurosciences, University of Toulouse 3, University Hospital of Toulouse, INSERM, Toulouse, France
| | | | - Alexander Hapfelmeier
- Institute of AI and Informatics in Medicine, School of Medicine, Technical University of Munich, Munich, Germany
- Institute of General Practice and Health Services Research, School of Medicine, Technical University of Munich, Munich, Germany
| | - Paul Lingor
- Department of Neurology, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
16
|
Yasuda Y, Wang L, Chitano P, Seow CY. Rho-Kinase Inhibition of Active Force and Passive Tension in Airway Smooth Muscle: A Strategy for Treating Airway Hyperresponsiveness in Asthma. BIOLOGY 2024; 13:115. [PMID: 38392332 PMCID: PMC10886476 DOI: 10.3390/biology13020115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/05/2024] [Accepted: 02/09/2024] [Indexed: 02/24/2024]
Abstract
Rho-kinase inhibitors have been identified as a class of potential drugs for treating asthma because of their ability to reduce airway inflammation and active force in airway smooth muscle (ASM). Past research has revealed that, besides the effect on the ASM's force generation, rho-kinase (ROCK) also regulates actin filament formation and filament network architecture and integrity, thus affecting ASM's cytoskeletal stiffness. The present review is not a comprehensive examination of the roles played by ROCK in regulating ASM function but is specifically focused on passive tension, which is partially determined by the cytoskeletal stiffness of ASM. Understanding the molecular basis for maintaining active force and passive tension in ASM by ROCK will allow us to determine the suitability of ROCK inhibitors and its downstream enzymes as a class of drugs in treating airway hyperresponsiveness seen in asthma. Because clinical trials using ROCK inhibitors in the treatment of asthma have yet to be conducted, the present review focuses on the in vitro effects of ROCK inhibitors on ASM's mechanical properties which include active force generation, relaxation, and passive stiffness. The review provides justification for future clinical trials in the treatment of asthma using ROCK inhibitors alone and in combination with other pharmacological and mechanical interventions.
Collapse
Affiliation(s)
- Yuto Yasuda
- Centre for Heart Lung Innovation, St. Paul's Hospital, Providence Health Care, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Lu Wang
- Centre for Heart Lung Innovation, St. Paul's Hospital, Providence Health Care, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Pasquale Chitano
- Centre for Heart Lung Innovation, St. Paul's Hospital, Providence Health Care, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Chun Y Seow
- Centre for Heart Lung Innovation, St. Paul's Hospital, Providence Health Care, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| |
Collapse
|
17
|
Huang Y, Wang S, Zhang X, Yang C, Wang S, Cheng H, Ke A, Gao C, Guo K. Identification of Fasudil as a collaborator to promote the anti-tumor effect of lenvatinib in hepatocellular carcinoma by inhibiting GLI2-mediated hedgehog signaling pathway. Pharmacol Res 2024; 200:107082. [PMID: 38280440 DOI: 10.1016/j.phrs.2024.107082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/16/2024] [Accepted: 01/23/2024] [Indexed: 01/29/2024]
Abstract
Lenvatinib is a frontline tyrosine kinase inhibitor for patients with advanced hepatocellular carcinoma (HCC). However, just 25% of patients benefit from the treatment, and acquired resistance always develops. To date, there are neither effective medications to combat lenvatinib resistance nor accurate markers that might predict how well a patient would respond to the lenvatinib treatment. Thus, novel strategies to recognize and deal with lenvatinib resistance are desperately needed. In the current study, a robust Lenvatinib Resistance index (LRi) model to predict lenvatinib response status in HCC was first established. Subsequently, five candidate drugs (Mercaptopurine, AACOCF3, NU1025, Fasudil, and Exisulind) that were capable of reversing lenvatinib resistance signature were initially selected by performing the connectivity map (CMap) analysis, and fasudil finally stood out by conducting a series of cellular functional assays in vitro and xenograft mouse model. Transcriptomics revealed that the co-administration of lenvatinib and fasudil overcame lenvatinib resistance by remodeling the hedgehog signaling pathway. Mechanistically, the feedback activation of EGFR by lenvatinib led to the activation of the GLI2-ABCC1 pathway, which supported the HCC cell's survival and proliferation. Notably, co-administration of lenvatinib and fasudil significantly inhibited IHH, the upstream switch of the hedgehog pathway, to counteract GLI2 activation and finally enhance the effectiveness of lenvatinib. These findings elucidated a novel EGFR-mediated mechanism of lenvatinib resistance and provided a practical approach to overcoming drug resistance in HCC through meaningful drug repurposing strategies.
Collapse
Affiliation(s)
- Yilan Huang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion Ministry of Education, Shanghai, China
| | - Siwei Wang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion Ministry of Education, Shanghai, China; Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaojun Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Yang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sikai Wang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion Ministry of Education, Shanghai, China
| | - Hongxia Cheng
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion Ministry of Education, Shanghai, China
| | - Aiwu Ke
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion Ministry of Education, Shanghai, China.
| | - Chao Gao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion Ministry of Education, Shanghai, China.
| | - Kun Guo
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion Ministry of Education, Shanghai, China.
| |
Collapse
|
18
|
Bonnevie ED, Scanzello CR, Mauck RL. Modulating mechanobiology as a therapeutic target for synovial fibrosis to restore joint lubrication. Osteoarthritis Cartilage 2024; 32:41-51. [PMID: 37866546 PMCID: PMC10880438 DOI: 10.1016/j.joca.2023.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 09/10/2023] [Accepted: 09/12/2023] [Indexed: 10/24/2023]
Abstract
OBJECTIVES Fibroses are disorders linked to persistence of myofibroblasts due to biochemical (e.g., Transforming growth factor-β) and biophysical cues (e.g., a stiff microenvironment). In the context of osteoarthritis, fibrotic changes in the joint-lining synovium have been linked with disease progression. The objective of this study was to probe synovial fibroblast mechanobiology and how essential functions (i.e., lubrication) are altered in fibrotic environments. DESIGN Both ex vivo and in vitro synovium models were assessed for fibrotic and lubrication biomarkers to better understand the role of mechanobiology and lubrication. Additionally, in vitro, work on small molecules targeting mechanobiology was assessed. RESULTS Our results indicated that modulating mechanobiology could rescue the fibrotic phenotype instigated by stiffening microenvironment that resulted in altered lubricant expression. A small molecule therapeutic, fasudil, blocked ROCK-mediated contractility and this inhibition of the fibrotic mechano-response of synovial fibroblasts restored proper lubrication function, providing insight into mechanisms of disease progression as well as a new avenue for therapeutic development. CONCLUSION This study identifies synovial fibrosis as a condition that potentially has joint-wide deficits through inhibiting lubrication. Additionally, modulating mechanobiology (i.e., ROCK-mediated contractility) may pose a potential target for small molecule therapies that can be delivered to the joint space. CLASSIFICATION Applied Biological Sciences.
Collapse
Affiliation(s)
- Edward D Bonnevie
- Translational Musculoskeletal Research Center, CMC VA Medical Center, United States; McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, United States.
| | - Carla R Scanzello
- Translational Musculoskeletal Research Center, CMC VA Medical Center, United States; Division of Rheumatology, Perelman School of Medicine, University of Pennsylvania, United States
| | - Robert L Mauck
- Translational Musculoskeletal Research Center, CMC VA Medical Center, United States; McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, United States; Department of Bioengineering, University of Pennsylvania, United States.
| |
Collapse
|
19
|
Lu W, Wen J. H 2S-RhoA/ROCK Pathway and Glial Cells in Axonal Remyelination After Ischemic Stroke. Mol Neurobiol 2023; 60:5493-5504. [PMID: 37322287 DOI: 10.1007/s12035-023-03422-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 06/03/2023] [Indexed: 06/17/2023]
Abstract
Ischemic stroke is one of the main reasons of disability and death. Stroke-induced functional deficits are mainly due to the secondary degeneration of the white matter characterized by axonal demyelination and injury of axon-glial integrity. Enhancement of the axonal regeneration and remyelination could promote the neural functional recovery. However, cerebral ischemia-induced activation of RhoA/Rho kinase (ROCK) pathway plays a crucial and harmful role in the process of axonal recovery and regeneration. Inhibition of this pathway could promote the axonal regeneration and remyelination. In addition, hydrogen sulfide (H2S) has the significant neuroprotective role during the recovery of ischemic stroke via inhibiting the inflammatory response and oxidative stress, regulating astrocyte function, promoting the differentiation of endogenous oligodendrocyte precursor cells (OPCs) to mature oligodendrocyte. Among all of these effects, promoting the formation of mature oligodendrocyte is a crucial part of axonal regeneration and remyelination. Furthermore, numerous studies have uncovered the crosstalk between astrocytes and oligodendrocyte, microglial cells and oligodendrocyte in the axonal remyelination following ischemic stroke. The purpose of this review was to discuss the relationship among H2S, RhoA/ROCK pathway, astrocytes, and microglial cells in the axonal remyelination following ischemic stroke to reveal new strategies for preventing and treating this devastating disease.
Collapse
Affiliation(s)
- Weizhuo Lu
- Medical Branch, Hefei Technology College, Hefei, China
| | - Jiyue Wen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
20
|
Lu W, Chen Z, Wen J. The role of RhoA/ROCK pathway in the ischemic stroke-induced neuroinflammation. Biomed Pharmacother 2023; 165:115141. [PMID: 37437375 DOI: 10.1016/j.biopha.2023.115141] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/03/2023] [Accepted: 07/07/2023] [Indexed: 07/14/2023] Open
Abstract
It is widely known that ischemic stroke is the prominent cause of death and disability. To date, neuroinflammation following ischemic stroke represents a complex event, which is an essential process and affects the prognosis of both experimental stroke animals and stroke patients. Intense neuroinflammation occurring during the acute phase of stroke contributes to neuronal injury, BBB breakdown, and worse neurological outcomes. Inhibition of neuroinflammation may be a promising target in the development of new therapeutic strategies. RhoA is a small GTPase protein that activates a downstream effector, ROCK. The up-regulation of RhoA/ROCK pathway possesses important roles in promoting the neuroinflammation and mediating brain injury. In addition, nuclear factor-kappa B (NF-κB) is another vital regulator of ischemic stroke-induced neuroinflammation through regulating the functions of microglial cells and astrocytes. After stroke onset, the microglial cells and astrocytes are activated and undergo the morphological and functional changes, thereby deeply participate in a complicated neuroinflammation cascade. In this review, we focused on the relationship among RhoA/ROCK pathway, NF-κB and glial cells in the neuroinflammation following ischemic stroke to reveal new strategies for preventing the intense neuroinflammation.
Collapse
Affiliation(s)
- Weizhuo Lu
- Medical Branch, Hefei Technology College, Hefei, China
| | - Zhiwu Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| | - Jiyue Wen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
21
|
Chen B, Jin W. A comprehensive review of stroke-related signaling pathways and treatment in western medicine and traditional Chinese medicine. Front Neurosci 2023; 17:1200061. [PMID: 37351420 PMCID: PMC10282194 DOI: 10.3389/fnins.2023.1200061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/19/2023] [Indexed: 06/24/2023] Open
Abstract
This review provides insight into the complex network of signaling pathways and mechanisms involved in stroke pathophysiology. It summarizes the historical progress of stroke-related signaling pathways, identifying potential interactions between them and emphasizing that stroke is a complex network disease. Of particular interest are the Hippo signaling pathway and ferroptosis signaling pathway, which remain understudied areas of research, and are therefore a focus of the review. The involvement of multiple signaling pathways, including Sonic Hedgehog (SHH), nuclear factor erythroid 2-related factor 2 (Nrf2)/antioxidant response element (ARE), hypoxia-inducible factor-1α (HIF-1α), PI3K/AKT, JAK/STAT, and AMPK in pathophysiological mechanisms such as oxidative stress and apoptosis, highlights the complexity of stroke. The review also delves into the details of traditional Chinese medicine (TCM) therapies such as Rehmanniae and Astragalus, providing an analysis of the recent status of western medicine in the treatment of stroke and the advantages and disadvantages of TCM and western medicine in stroke treatment. The review proposes that since stroke is a network disease, TCM has the potential and advantages of a multi-target and multi-pathway mechanism of action in the treatment of stroke. Therefore, it is suggested that future research should explore more treasures of TCM and develop new therapies from the perspective of stroke as a network disease.
Collapse
Affiliation(s)
- Binhao Chen
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Weifeng Jin
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
22
|
Glotfelty EJ, Tovar-y-Romo LB, Hsueh SC, Tweedie D, Li Y, Harvey BK, Hoffer BJ, Karlsson TE, Olson L, Greig NH. The RhoA-ROCK1/ROCK2 Pathway Exacerbates Inflammatory Signaling in Immortalized and Primary Microglia. Cells 2023; 12:1367. [PMID: 37408199 PMCID: PMC10216802 DOI: 10.3390/cells12101367] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 07/07/2023] Open
Abstract
Neuroinflammation is a unifying factor among all acute central nervous system (CNS) injuries and chronic neurodegenerative disorders. Here, we used immortalized microglial (IMG) cells and primary microglia (PMg) to understand the roles of the GTPase Ras homolog gene family member A (RhoA) and its downstream targets Rho-associated coiled-coil-containing protein kinases 1 and 2 (ROCK1 and ROCK2) in neuroinflammation. We used a pan-kinase inhibitor (Y27632) and a ROCK1- and ROCK2-specific inhibitor (RKI1447) to mitigate a lipopolysaccharide (LPS) challenge. In both the IMG cells and PMg, each drug significantly inhibited pro-inflammatory protein production detected in media (TNF-α, IL-6, KC/GRO, and IL-12p70). In the IMG cells, this resulted from the inhibition of NF-κB nuclear translocation and the blocking of neuroinflammatory gene transcription (iNOS, TNF-α, and IL-6). Additionally, we demonstrated the ability of both compounds to block the dephosphorylation and activation of cofilin. In the IMG cells, RhoA activation with Nogo-P4 or narciclasine (Narc) exacerbated the inflammatory response to the LPS challenge. We utilized a siRNA approach to differentiate ROCK1 and ROCK2 activity during the LPS challenges and showed that the blockade of both proteins may mediate the anti-inflammatory effects of Y27632 and RKI1447. Using previously published data, we show that genes in the RhoA/ROCK signaling cascade are highly upregulated in the neurodegenerative microglia (MGnD) from APP/PS-1 transgenic Alzheimer's disease (AD) mice. In addition to illuminating the specific roles of RhoA/ROCK signaling in neuroinflammation, we demonstrate the utility of using IMG cells as a model for primary microglia in cellular studies.
Collapse
Affiliation(s)
- Elliot J. Glotfelty
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD 21224, USA
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Luis B. Tovar-y-Romo
- Division of Neuroscience, Institute of Cellular Physiology, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Shih-Chang Hsueh
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - David Tweedie
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Yazhou Li
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Brandon K. Harvey
- Molecular Mechanisms of Cellular Stress and Inflammation Unit, Integrative Neuroscience Department, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Barry J. Hoffer
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Tobias E. Karlsson
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Lars Olson
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Nigel H. Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD 21224, USA
| |
Collapse
|
23
|
Hou HT, Wang ZQ, Wang J, Yang Q, He GW. Antispastic Effect of Fasudil and Cocktail of Fasudil and Nitroglycerin in Internal Thoracic Artery. Ann Thorac Surg 2023; 115:1152-1161. [PMID: 35172179 DOI: 10.1016/j.athoracsur.2021.11.079] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/20/2021] [Accepted: 11/29/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Spasm of arterial grafts in coronary artery bypass grafting is a clinical problem and can occasionally be lethal. Perioperative spasm in the internal thoracic artery (ITA) and coronary arteries occurs in 0.43% of patients. This study aimed to investigate the antispastic effect of a RhoA/Rho-kinase (Rho-associated coiled-coil-containing protein kinase [ROCK]) inhibitor (fasudil) with and without nitroglycerin in combination in the ITA. METHODS Isolated human ITA rings taken from 68 patients who were undergoing coronary bypass were studied in a myograph. Cumulative concentration-relaxation curves for fasudil (-9 to -3.5 log M) were established in the ITA, which was precontracted with potassium chloride or U46619. The inhibitory effect of fasudil (-6.3 or -5.3 log M) or fasudil in combination with nitroglycerin were also tested. The ROCK2 protein was measured by Western blot. RESULTS Fasudil caused similar relaxation in ITA rings contracted by potassium chloride or U46619. Pretreatment with -5.3 log M fasudil significantly depressed contraction induced by potassium chloride (P = .004 vs control; P = .017 vs -6.3 log M) and U46619 (P = .010 vs control; P = .041 vs. -6.3 log M). Fasudil in combination with nitroglycerin had more effect and more rapid and sustained relaxation than either vasodilator alone. Fasudil caused a decrease of ROCK2 protein content (P = .014). CONCLUSIONS Fasudil fully relaxes some vasoconstrictor-induced contraction and decreases ROCK2 protein content in the ITA. The combination of fasudil and nitroglycerin has a superior effect than either vasodilator alone. The new cocktail solution composed of fasudil and nitroglycerin (pH 7.4) has effective antispastic action and may prove to be a new antispastic method for arterial conduits during coronary bypass surgery.
Collapse
Affiliation(s)
- Hai-Tao Hou
- Institute of Cardiovascular Diseases and Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Tianjin University and Chinese Academy of Medical Sciences, Tianjin, China
| | - Zheng-Qing Wang
- Institute of Cardiovascular Diseases and Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Tianjin University and Chinese Academy of Medical Sciences, Tianjin, China
| | - Jun Wang
- Institute of Cardiovascular Diseases and Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Tianjin University and Chinese Academy of Medical Sciences, Tianjin, China
| | - Qin Yang
- Institute of Cardiovascular Diseases and Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Tianjin University and Chinese Academy of Medical Sciences, Tianjin, China
| | - Guo-Wei He
- Institute of Cardiovascular Diseases and Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Tianjin University and Chinese Academy of Medical Sciences, Tianjin, China; Drug Research and Development Center, Wannan Medical College, Wuhu, Anhui, China; Department of Surgery, Oregon Health and Science University, Portland, Oregon.
| |
Collapse
|
24
|
Thapa K, Shivam K, Khan H, Kaur A, Dua K, Singh S, Singh TG. Emerging Targets for Modulation of Immune Response and Inflammation in Stroke. Neurochem Res 2023; 48:1663-1690. [PMID: 36763312 DOI: 10.1007/s11064-023-03875-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/18/2023] [Accepted: 01/24/2023] [Indexed: 02/11/2023]
Abstract
The inflammatory and immunological responses play a significant role after stroke. The innate immune activation stimulated by microglia during stroke results in the migration of macrophages and lymphocytes into the brain and are responsible for tissue damage. The immune response and inflammation following stroke have no defined targets, and the intricacies of the immunological and inflammatory processes are only partially understood. Innate immune cells enter the brain and meninges during the acute phase, which can cause ischemia damage. Activation of systemic immunity is caused by danger signals sent into the bloodstream by injured brain cells, which is followed by a significant immunodepression that encourages life-threatening infections. Neuropsychiatric sequelae, a major source of post-stroke morbidity, may be induced by an adaptive immune response that is initiated by antigen presentation during the chronic period and is directed against the brain. Thus, the current review discusses the role of immune response and inflammation in stroke pathogenesis, their role in the progression of injury during the stroke, and the emerging targets for the modulation of the mechanism of immune response and inflammation that may have possible therapeutic benefits against stroke.
Collapse
Affiliation(s)
- Komal Thapa
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.,School of Pharmacy, Chitkara University, Rajpura, Himachal Pradesh, 174103, India
| | - Kumar Shivam
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Amarjot Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia.,Faculty of Health, Australian Research Centre in Complementary & Integrative Medicine, University of Technology Sydney, Ultimo, 2007, Australia
| | - Sachin Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar - Delhi G.T. Road, Phagwara, Punjab, 144411, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
25
|
Proietti R, Giordani AS, Lorenzo CA. ROCK (RhoA/Rho Kinase) Activation in Atrial Fibrillation: Molecular Pathways and Clinical Implications. Curr Cardiol Rev 2023; 19:e171122210986. [PMID: 36625201 PMCID: PMC10280999 DOI: 10.2174/1573403x19666221117092951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 11/07/2022] [Accepted: 11/07/2022] [Indexed: 12/15/2022] Open
Abstract
Among the complex mechanisms of AF pathogenesis, intracellular calcium overload and oxidative stress play a major role, both triggered by inflammatory processes. The additional basic event taking place in AF is atrial fibrotic remodeling, again triggered by oxidative stress, which is determined by connexins rearrangement and differentiation of fibroblasts into active collagensecreting myofibroblasts. RhoA/ROCK system is the final pathway of a wide spectrum of molecular effectors such as Angiotensin II, platelet-derived growth factor, connective tissue growth factor and transforming growth factor β, that overall determine calcium dysregulation and pro-fibrotic remodeling. Both in experimental and clinical studies, RhoA/ROCK activation has been linked to superoxide ion production, fibrotic remodeling and connexins rearrangement, with important consequences for AF pathogenesis. ROCK pathway inhibition may therefore be a therapeutic or preventive target for special AF subgroups of patients.
Collapse
Affiliation(s)
- Riccardo Proietti
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, United Kingdom
| | - Andrea S. Giordani
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Calò A. Lorenzo
- Department of Medicine (DIMED), Nephrology, Dialysis and Transplantation Unit, University of Padua and Azienda Ospedale Università di Padova, Padua, Italy
| |
Collapse
|
26
|
Eren F, Yilmaz SE. Neuroprotective approach in acute ischemic stroke: A systematic review of clinical and experimental studies. Brain Circ 2022; 8:172-179. [PMID: 37181847 PMCID: PMC10167855 DOI: 10.4103/bc.bc_52_22] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/01/2022] [Accepted: 09/08/2022] [Indexed: 12/14/2022] Open
Abstract
Ischemic stroke is a disease with worldwide economic and social negative effects. It is a serious disease with high disability and mortality. Ionic imbalance, excitotoxicity, oxidative stress, and inflammation are induced during and after ischemic stroke. Cellular dysfunction, apoptosis, and necrosis are activated directly or indirectly mechanisms. The studies about neuroprotection in neurodegenerative diseases have increased in recent years. Data about the mechanisms of progressive molecular improvement in the brain tissue are increasing in acute ischemic stroke. Based on these data, preclinical and clinical studies on new neuroprotective treatments are being designed. An effective neuroprotective strategy can prolong the indication period of recanalization treatments in the acute stage of ischemic stroke. In addition, it can reduce neuronal necrosis and protect the brain against ischemia-related reperfusion injury. The current review has evaluated the recent clinical and experimental studies. The molecular mechanism of each of the neuroprotective strategies is also summarized. This review may help develop future strategies for combination treatment to protect the cerebral tissue from ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Fettah Eren
- Department of Neurology, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Sueda Ecem Yilmaz
- Department of Neurology, School of Medicine, Selcuk University, Konya, Turkey
| |
Collapse
|
27
|
Mani S, Jindal D, Chopra H, Jha SK, Singh SK, Ashraf GM, Kamal M, Iqbal D, Chellappan DK, Dey A, Dewanjee S, Singh KK, Ojha S, Singh I, Gautam RK, Jha NK. ROCK2 Inhibition: A Futuristic Approach for the Management of Alzheimer's Disease. Neurosci Biobehav Rev 2022; 142:104871. [PMID: 36122738 DOI: 10.1016/j.neubiorev.2022.104871] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/30/2022] [Accepted: 09/12/2022] [Indexed: 12/06/2022]
Abstract
Neurons depend on mitochondrial functions for membrane excitability, neurotransmission, and plasticity.Mitochondrialdynamicsare important for neural cell maintenance. To maintain mitochondrial homeostasis, lysosomes remove dysfunctionalmitochondria through mitophagy. Mitophagy promotes mitochondrial turnover and prevents the accumulation of dysfunctional mitochondria. In many neurodegenerative diseases (NDDs), including Alzheimer's disease (AD), mitophagy is disrupted in neurons.Mitophagy is regulated by several proteins; recently,Rho-associated coiled-coil containing protein kinase 2 (ROCK2) has been suggested to negatively regulate the Parkin-dependent mitophagy pathway.Thus, ROCK2inhibitionmay bea promising therapyfor NDDs. This review summarizesthe mitophagy pathway, the role of ROCK2in Parkin-dependentmitophagyregulation,and mitophagy impairment in the pathology of AD. We further discuss different ROCK inhibitors (synthetic drugs, natural compounds,and genetherapy-based approaches)and examine their effects on triggering neuronal growth and neuroprotection in AD and other NDDs. This comprehensive overview of the role of ROCK in mitophagy inhibition provides a possible explanation for the significance of ROCK inhibitors in the therapeutic management of AD and other NDDs.
Collapse
Affiliation(s)
- Shalini Mani
- Centre for Emerging Disease, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India.
| | - Divya Jindal
- Centre for Emerging Disease, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India
| | - Hitesh Chopra
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida, Uttar Pradesh 201310, India; Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, 140413, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | | | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Danish Iqbal
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, India
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Keshav K Singh
- Department of Genetics, UAB School of Medicine, The University of Alabama at Birmingham
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Inderbir Singh
- MM School of Pharmacy, MM University, Sadopur-Ambala -134007, India
| | - Rupesh K Gautam
- MM School of Pharmacy, MM University, Sadopur-Ambala -134007, India.
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida, Uttar Pradesh 201310, India; Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, 140413, India.
| |
Collapse
|
28
|
Sheikh AM, Yano S, Mitaki S, Tabassum S, Yamaguchi S, Nagai A. Rho-Kinase inhibition decreases focal cerebral ischemia-induced glial activation in rats. J Cent Nerv Syst Dis 2022; 14:11795735221123910. [PMID: 36106069 PMCID: PMC9465613 DOI: 10.1177/11795735221123910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 08/05/2022] [Indexed: 11/17/2022] Open
Abstract
Background Rho-kinase inhibition in a rat middle cerebral artery occlusion (MCAO) model
is reported to improve neurological functions and decrease infarction
size. Objective The objective of this study is to investigate the underlying mechanisms of
such improvement by evaluating the effects of Rho-kinase inhibition on
astrocytes and microglial accumulation and activation in this condition. Methods Adult male Sprague-Dawley (SD) rats were used to generate the MCAO model,
which received an I.P injection of a chemical Rho-kinase inhibitor (Fasudil-
5 mg/kg/day) or vehicle (PBS) for 2 and 4 days. Results Fasudil treatment significantly decreased the stroke volumes and water
content in the lesion areas, as revealed by MRI. Immunostaining and Western
blotting results demonstrated that Fasudil significantly decreased the
levels of Aquaporin-4, a water channel protein. The number of
GFAP+ astrocytes and Iba-1+ macrophage/microglia
was decreased in the lesion areas. Proinflammatory transcription factor
NF-κB protein levels were decreased in the Fasudil group 2 days after MCAO.
Also, proinflammatory mediators including TNF-α, IL-1β, and iNOS levels were
decreased. In vitro migration study using a human microglial cell line
(HMO6) confirmed the inhibitory effects of Fasudil on the process. Fasudil
also decreased combined IL-1β and IFNγ-induced NF-κB nuclear translocation
in HMO6. Moreover, Fasudil transiently decreased combined IL-1β and
IFNγ-induced iNOS, TNFα, and IL-1β mRNA levels in HMO6. Conclusion Our study demonstrates the inhibitory effects of Rho-kinase on NF-κB-mediated
glial activation and cerebral edema, which might be a promising therapeutic
target in acute cerebral ischemia conditions.
Collapse
Affiliation(s)
- Abdullah Md Sheikh
- Department of Laboratory Medicine, Shimane University Faculty of Medicine, Izumo, Japan
| | - Shozo Yano
- Department of Laboratory Medicine, Shimane University Faculty of Medicine, Izumo, Japan
| | - Shingo Mitaki
- Department of Neurology, Shimane University Faculty of Medicine, Izumo, Japan
| | - Shatera Tabassum
- Department of Laboratory Medicine, Shimane University Faculty of Medicine, Izumo, Japan
| | | | - Atsushi Nagai
- Department of Neurology, Shimane University Faculty of Medicine, Izumo, Japan
| |
Collapse
|
29
|
Aykan SA, Xie H, Zheng Y, Chung DY, Kura S, Han Lai J, Erdogan TD, Morais A, Tamim I, Yagmur D, Ishikawa H, Arai K, Abbas Yaseen M, Boas DA, Sakadzic S, Ayata C. Rho-Kinase Inhibition Improves the Outcome of Focal Subcortical White Matter Lesions. Stroke 2022; 53:2369-2376. [PMID: 35656825 DOI: 10.1161/strokeaha.121.037358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Subcortical white matter lesions are exceedingly common in cerebral small vessel disease and lead to significant cumulative disability without an available treatment. Here, we tested a rho-kinase inhibitor on functional recovery after focal white matter injury. METHODS A focal corpus callosum lesion was induced by stereotactic injection of N5-(1-iminoethyl)-L-ornithine in mice. Fasudil (10 mg/kg) or vehicle was administered daily for 2 weeks, starting one day after lesion induction. Resting-state functional connectivity and grid walk performance were studied longitudinally, and lesion volumes were determined at one month. RESULTS Resting-state interhemispheric functional connectivity significantly recovered between days 1 and 14 in the fasudil group (P<0.001), despite worse initial connectivity loss than vehicle before treatment onset. Grid walk test revealed an increased number of foot faults in the vehicle group compared with baseline, which persisted for at least 4 weeks. In contrast, the fasudil arm did not show an increase in foot faults and had smaller lesions at 4 weeks. Immunohistochemical examination of reactive astrocytosis, synaptic density, and mature oligodendrocytes did not reveal a significant difference between treatment arms. CONCLUSIONS These data show that delayed fasudil posttreatment improves functional outcomes after a focal subcortical white matter lesion in mice. Future work will aim to elucidate the mechanisms.
Collapse
Affiliation(s)
- Sanem A Aykan
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Charlestown (S.A.A., H.X., Y.Z., D.Y.C., J.H.L., T.D.E., A.M., I.T., D.Y., C.A.)
| | - Hongyu Xie
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Charlestown (S.A.A., H.X., Y.Z., D.Y.C., J.H.L., T.D.E., A.M., I.T., D.Y., C.A.).,Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai, China (H.X.)
| | - Yi Zheng
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Charlestown (S.A.A., H.X., Y.Z., D.Y.C., J.H.L., T.D.E., A.M., I.T., D.Y., C.A.)
| | - David Y Chung
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Charlestown (S.A.A., H.X., Y.Z., D.Y.C., J.H.L., T.D.E., A.M., I.T., D.Y., C.A.).,Stroke Service, Department of Neurology, Massachusetts General Hospital, Charlestown, MA. (C.A., D.Y.C.)
| | - Sreekanth Kura
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, MA (S.K., D.A.B.)
| | - James Han Lai
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Charlestown (S.A.A., H.X., Y.Z., D.Y.C., J.H.L., T.D.E., A.M., I.T., D.Y., C.A.)
| | - Taylan D Erdogan
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Charlestown (S.A.A., H.X., Y.Z., D.Y.C., J.H.L., T.D.E., A.M., I.T., D.Y., C.A.)
| | - Andreia Morais
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Charlestown (S.A.A., H.X., Y.Z., D.Y.C., J.H.L., T.D.E., A.M., I.T., D.Y., C.A.)
| | - Isra Tamim
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Charlestown (S.A.A., H.X., Y.Z., D.Y.C., J.H.L., T.D.E., A.M., I.T., D.Y., C.A.)
| | - Damla Yagmur
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Charlestown (S.A.A., H.X., Y.Z., D.Y.C., J.H.L., T.D.E., A.M., I.T., D.Y., C.A.)
| | - Hidehiro Ishikawa
- Neuroprotection Research Laboratory, Department of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown. (H.I., K.A.)
| | - Ken Arai
- Neuroprotection Research Laboratory, Department of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown. (H.I., K.A.)
| | - M Abbas Yaseen
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA. (D.A.B., M.A.Y., S.S.)
| | - David A Boas
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, MA (S.K., D.A.B.).,Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA. (D.A.B., M.A.Y., S.S.)
| | - Sava Sakadzic
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA. (D.A.B., M.A.Y., S.S.)
| | - Cenk Ayata
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Charlestown (S.A.A., H.X., Y.Z., D.Y.C., J.H.L., T.D.E., A.M., I.T., D.Y., C.A.).,Stroke Service, Department of Neurology, Massachusetts General Hospital, Charlestown, MA. (C.A., D.Y.C.)
| |
Collapse
|
30
|
Sato K, Nakagawa S, Morofuji Y, Matsunaga Y, Fujimoto T, Watanabe D, Izumo T, Niwa M, Walter FR, Vigh JP, Santa-Maria AR, Deli MA, Matsuo T. Effects of fasudil on blood-brain barrier integrity. Fluids Barriers CNS 2022; 19:43. [PMID: 35659272 PMCID: PMC9166508 DOI: 10.1186/s12987-022-00336-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 05/04/2022] [Indexed: 11/17/2022] Open
Abstract
Background Cerebral infarction accounts for 85% of all stroke cases. Even in an era of rapid and effective recanalization using an intravascular approach, the majority of patients have poor functional outcomes. Thus, there is an urgent need for the development of therapeutic agents to treat acute ischemic stroke. We evaluated the effect of fasudil, a Rho kinase inhibitor, on blood brain barrier (BBB) functions under normoxia or oxygen–glucose deprivation (OGD) conditions using a primary cell-based in vitro BBB model. Methods BBB models from rat primary cultures (brain capillary endothelial cells, astrocytes, and pericytes) were subjected to either normoxia or 6 h OGD/24 h reoxygenation. To assess the effects of fasudil on BBB functions, we evaluated real time impedance, transendothelial electrical resistance (TEER), sodium fluorescein permeability, and tight junction protein expression using western blotting. Lastly, to understand the observed protective mechanism on BBB functions by fasudil we examined the role of cyclooxygenase-2 and thromboxane A2 receptor agonist U-46619 in BBB-forming cells. Results We found that treatment with 0.3–30 µM of fasudil increased cellular impedance. Fasudil enhanced barrier properties in a concentration-dependent manner, as measured by an increased (TEER) and decreased permeability. Fasudil also increased the expression of tight junction protein claudin-5. Reductions in TEER and increased permeability were observed after OGD/reoxygenation exposure in mono- and co-culture models. The improvement in BBB integrity by fasudil was confirmed in both of the models, but was significantly higher in the co-culture than in the monoculture model. Treatment with U-46619 did not show significant changes in TEER in the monoculture model, whereas it showed a significant reduction in TEER in the co-culture model. Fasudil significantly improved the U-46619-induced TEER reduction in the co-culture models. Pericytes and astrocytes have opposite effects on endothelial cells and may contribute to endothelial injury in hyperacute ischemic stroke. Overall, fasudil protects the integrity of BBB both by a direct protective effect on endothelial cells and by a pathway mediated via pericytes and astrocytes. Conclusions Our findings suggest that fasudil is a BBB-protective agent against acute ischemic stroke. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00336-w.
Collapse
Affiliation(s)
- Kei Sato
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Shinsuke Nakagawa
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Yoichi Morofuji
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan.
| | - Yuki Matsunaga
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Takashi Fujimoto
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Daisuke Watanabe
- BBB Laboratory, PharmaCo-Cell Company Ltd, Nagasaki, 852-8135, Japan
| | - Tsuyoshi Izumo
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Masami Niwa
- BBB Laboratory, PharmaCo-Cell Company Ltd, Nagasaki, 852-8135, Japan
| | - Fruzsina R Walter
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Szeged, 6726, Hungary
| | - Judit P Vigh
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Szeged, 6726, Hungary
| | - Ana Raquel Santa-Maria
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Szeged, 6726, Hungary.,Wyss Institute for Biologically Inspired Engineering at Harvard University, 3 Blackfan Circle, Boston, MA, 02115, USA
| | - Maria A Deli
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Szeged, 6726, Hungary
| | - Takayuki Matsuo
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| |
Collapse
|
31
|
Racine ML, Terwoord JD, Ketelhut NB, Bachman NP, Richards JC, Luckasen GJ, Dinenno FA. Rho-kinase inhibition improves haemodynamic responses and circulating ATP during hypoxia and moderate intensity handgrip exercise in healthy older adults. J Physiol 2022; 600:3265-3285. [PMID: 35575293 PMCID: PMC9288513 DOI: 10.1113/jp282730] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 05/09/2022] [Indexed: 11/21/2022] Open
Abstract
Abstract Skeletal muscle haemodynamics and circulating adenosine triphosphate (ATP) responses during hypoxia and exercise are blunted in older (OA) vs. young (YA) adults, which may be associated with impaired red blood cell (RBC) ATP release. Rho‐kinase inhibition improves deoxygenation‐induced ATP release from OA isolated RBCs. We tested the hypothesis that Rho‐kinase inhibition (via fasudil) in vivo would improve local haemodynamic and ATP responses during hypoxia and exercise in OA. Healthy YA (25 ± 3 years; n = 12) and OA (65 ± 5 years; n = 13) participated in a randomized, double‐blind, placebo‐controlled, crossover study on two days (≥5 days between visits). A forearm deep venous catheter was used to administer saline/fasudil and sample venous plasma ATP ([ATP]V). Forearm vascular conductance (FVC) and [ATP]V were measured at rest, during isocapnic hypoxia (80% SpO2), and during graded rhythmic handgrip exercise that was similar between groups (5, 15 and 25% maximum voluntary contraction (MVC)). Isolated RBC ATP release was measured during normoxia/hypoxia. With saline, ΔFVC was lower (P < 0.05) in OA vs. YA during hypoxia (∼60%) and during 15 and 25% MVC (∼25–30%), and these impairments were abolished with fasudil. Similarly, [ATP]V and ATP effluent responses from normoxia to hypoxia and rest to 25% MVC were lower in OA vs. YA and improved with fasudil (P < 0.05). Isolated RBC ATP release during hypoxia was impaired in OA vs. YA (∼75%; P < 0.05), which tended to improve with fasudil in OA (P = 0.082). These data suggest Rho‐kinase inhibition improves haemodynamic responses to hypoxia and moderate intensity exercise in OA, which may be due in part to improved circulating ATP.
![]() Key points Skeletal muscle blood flow responses to hypoxia and exercise are impaired with age. Blunted increases in circulating ATP, a vasodilator, in older adults may contribute to age‐related impairments in haemodynamics. Red blood cells (RBCs) are a primary source of circulating ATP, and treating isolated RBCs with a Rho‐kinase inhibitor improves age‐related impairments in deoxygenation‐induced RBC ATP release. In this study, treating healthy older adults systemically with the Rho‐kinase inhibitor fasudil improved blood flow and circulating ATP responses during hypoxia and moderate intensity handgrip exercise compared to young adults, and also tended to improve isolated RBC ATP release. Improved blood flow regulation with fasudil was also associated with increased skeletal muscle oxygen delivery during hypoxia and exercise in older adults. This is the first study to demonstrate that Rho‐kinase inhibition can significantly improve age‐related impairments in haemodynamic and circulating ATP responses to physiological stimuli, which may have therapeutic implications.
Collapse
Affiliation(s)
- Matthew L Racine
- Human Cardiovascular Physiology Laboratory, Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, 80523, USA
| | - Janée D Terwoord
- Human Cardiovascular Physiology Laboratory, Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, 80523, USA
| | - Nathaniel B Ketelhut
- Human Cardiovascular Physiology Laboratory, Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, 80523, USA
| | - Nate P Bachman
- Human Cardiovascular Physiology Laboratory, Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, 80523, USA
| | - Jennifer C Richards
- Human Cardiovascular Physiology Laboratory, Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, 80523, USA
| | - Gary J Luckasen
- Medical Center of the Rockies, University of Colorado Health, Loveland, CO, 80538, USA
| | - Frank A Dinenno
- Human Cardiovascular Physiology Laboratory, Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, 80523, USA
| |
Collapse
|
32
|
Emfietzoglou M, Terentes-Printzios D, Kotronias RA, Marin F, Montalto C, De Maria GL, Banning AP. The spectrum and systemic associations of microvascular dysfunction in the heart and other organs. NATURE CARDIOVASCULAR RESEARCH 2022; 1:298-311. [PMID: 39196132 DOI: 10.1038/s44161-022-00045-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 03/02/2022] [Indexed: 08/29/2024]
Abstract
Microvascular dysfunction (MVD) contributes to several conditions that increase morbidity and mortality, including ischemic heart disease, heart failure, dementia, chronic kidney disease and hypertension. Consequently, MVD imposes a substantial burden on healthcare systems worldwide. In comparison to macrovascular dysfunction, MVD has been incompletely investigated, and it remains uncertain whether MVD in an organ constitutes a distinct pathology or a manifestation of a systemic disorder. Here, we summarize and appraise the techniques that are used to diagnose MVD. We review the disorders of the heart, brain and kidneys in which the role of MVD has been highlighted and summarize evidence hinting at a systemic or multi-organ nature of MVD. Finally, we discuss the benefits and limitations of implementing MVD testing in clinical practice with a focus on new interventions that are beginning to emerge.
Collapse
Affiliation(s)
| | | | | | - Federico Marin
- Oxford Heart Centre, Oxford University Hospitals, Oxford, UK
| | | | | | | |
Collapse
|
33
|
Wang H, Fang F, Chen S, Jing X, Zhuang Y, Xie Y. Dual efficacy of Fasudil at improvement of survival and reinnervation of flap through RhoA/ROCK/PI3K/Akt pathway. Int Wound J 2022; 19:2000-2011. [PMID: 35315211 PMCID: PMC9705174 DOI: 10.1111/iwj.13800] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 03/14/2022] [Indexed: 11/27/2022] Open
Abstract
Fasudil is reported to be effective at protecting against ischaemic diseases, and at augmenting axon growth. In this study, we aim to evaluate its efficacy in promoting flap survival and reinnervation. Ninety-two Institute of Cancer Research (ICR) mice were used and divided into the control, Fasudil, LY294002, Fasudil+LY294002 groups, receiving a daily intraperitoneal injection of normal saline, Fasudil (10 mg/kg), LY294002 (5 mg/kg), and Fasudil (10 mg/kg) + LY294002 (5 mg/kg), respectively. On days 0 and 5, the blood perfusion and diameter of the iliolumbar artery in the pedicle of the flaps in the four groups were evaluated using laser speckling contrast imaging (LSCI). On day 5, the flaps were photographed and the necrosis rate of the flaps was calculated using Photoshop CS6. In addition, tissues were harvested from the flaps and divided into two parts. One part underwent routine cryosection and immunofluorescent staining using the antibody against CD31 for evaluation of the microvascular density in the four groups. In the other part, the expression of RhoA, ROCK1+2, p-CPI-17, p-MYPT, p-PTEN, p-PI3K, p-Akt, and vascular endothelial growth factor (VEGF) within the flaps were determined using western blotting. Moreover, at days 0, 7, 15, and 30 after flap surgery, the axons within the flaps were evaluated using immunofluorescent staining with the antibody against Neurofilament-200. It turned out that the necrosis rate was (24.4 ± 7.7)%, (5.2 ± 1.6)%, (29.8 ± 4.2)%, and (30.9 ± 7.1)%, respectively, in the control, Fasudil, LY294002, LY294002+Fasudil groups. There was a significant reduction in the necrosis rate of the flaps in the Fasudil group (P < .001). The LSCI and immunofluorescent staining demonstrated that Fasudil could significantly expand the diameter of the iliolumbar artery in the pedicle, boost the overall blood perfusion, and increase the microvascular density of the flaps in the Fasudil group (P < .05), which could all be abolished by PI3K inhibitor LY294002. On day 5, the expression of p-CPI-17, p-MYPT, and p-PTEN were downregulated, whereas pPI3K, p-Akt, and VEGF were upregulated in the Fasudil group (P < .001). As for reinnervation, Neurofilament-200 fluorescent staining revealed that at days 15 and 30 after flap harvest, only in the Fasudil group could new axons be observed. It can be concluded that Fasudil could simultaneously improve the survival and axon growth after flap harvest, a dual efficacy achieved by inhibition of the RhoA/ROCK pathway, which in turn activates /PI3K/AKT pathway.
Collapse
Affiliation(s)
- Hai Wang
- Orthopedic Department, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Fang Fang
- Department of pharmacology, Fujian Medical University, Fuzhou, China
| | - Shaofeng Chen
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Institute of Clinical Applied Anatomy, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xing Jing
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Institute of Clinical Applied Anatomy, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yuehong Zhuang
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Institute of Clinical Applied Anatomy, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yun Xie
- Orthopedic Department, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
34
|
Freitas F, Attwell D. Pericyte-mediated constriction of renal capillaries evokes no-reflow and kidney injury following ischaemia. eLife 2022; 11:74211. [PMID: 35285797 PMCID: PMC8947765 DOI: 10.7554/elife.74211] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 03/09/2022] [Indexed: 12/12/2022] Open
Abstract
Acute kidney injury is common, with ~13 million cases and 1.7 million deaths/year worldwide. A major cause is renal ischaemia, typically following cardiac surgery, renal transplant or severe haemorrhage. We examined the cause of the sustained reduction in renal blood flow ('no-reflow'), which exacerbates kidney injury even after an initial cause of compromised blood supply is removed. Adult male Sprague-Dawley rats, or NG2-dsRed male mice were used in this study. After 60 min kidney ischaemia and 30-60 min reperfusion, renal blood flow remained reduced, especially in the medulla, and kidney tubule damage was detected as Kim-1 expression. Constriction of the medullary descending vasa recta and cortical peritubular capillaries occurred near pericyte somata, and led to capillary blockages, yet glomerular arterioles and perfusion were unaffected, implying that the long-lasting decrease of renal blood flow contributing to kidney damage was generated by pericytes. Blocking Rho kinase to decrease pericyte contractility from the start of reperfusion increased the post-ischaemic diameter of the descending vasa recta capillaries at pericytes, reduced the percentage of capillaries that remained blocked, increased medullary blood flow and reduced kidney injury. Thus, post-ischaemic renal no-reflow, contributing to acute kidney injury, reflects pericytes constricting the descending vasa recta and peritubular capillaries. Pericytes are therefore an important therapeutic target for treating acute kidney injury.
Collapse
Affiliation(s)
- Felipe Freitas
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| |
Collapse
|
35
|
Wang Q, Zhao SF, Xiao X, Liu YN, Wang XL, Du YX. Combined Treatment of Bone Marrow Mesenchymal Stem Cells and Fasudil Promotes Neurovascular Remodeling and Neurological Function Recovery in Ischemic Stroke. Appl Biochem Biotechnol 2022; 194:801-812. [PMID: 34542824 DOI: 10.1007/s12010-021-03679-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/08/2021] [Indexed: 10/20/2022]
Abstract
Stroke remains a highly deadly and disabling disease with limited treatment tragedies due to the limitations of available treatments; novel therapies for stroke are needed. In this article, the synergistic results of dual bone marrow mesenchymal stem cells (BMSC) and fasudil treatment in rat models of ischemic stroke still require further identification. Sprague-Dawley rats were used to construct the middle cerebral artery, occlusion models. BMSCs were incubated with fasudil, and MTT was performed to evaluate cell proliferation. The rats were treated with fasudil + BMSC, BMSC, fasudil, and saline. Blood samples were collected for complete blood count analysis and measurement of serum TNF-α levels. The neurological functions were evaluated. After the rats were sacrificed, immunohistochemical staining and TTC staining was performed. Fasudil promoted the proliferation of BMSCs and induced their differentiation into neuron-like cells. BMSCs increased the proportion of neutrophils; nevertheless, fasudil counteracted the neutrophil increase. The TUJ-1/MAP2/VIII factor expression in the fasudil + BMSC group was significantly higher than that in the other groups. The number of GFAP-positive cells decreased in the fasudil + BMSC and BMSC alone groups. The infarct volume in the fasudil + BMSC and BMSC alone groups was significantly lower than in the fasudil alone and control groups. Both BMSCs and fasudil exert neurorestorative effects in rat models of cerebral ischemia. Fasudil neutralizes the pro-inflammatory effects of BMSCs, while BMSCs and fasudil together had synergistic effects promoting neurovascular remodeling and neurological function recovery in stroke. A combination of BMSCs and fasudil provides a promising method for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Qian Wang
- Department of Neurology, The Second Hospital of Dalian Medical University, 467 Zhongshan Road, Shahekou District, Dalian, 116000, Liaoning, China.
| | - Shu-Fang Zhao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, 45 Changchun Road, Xicheng District, Beijing, 100053, China.
| | - Xia Xiao
- Department of Emergency Intensive Care Unit, The Second Hospital of Dalian Medical University, 467 Zhongshan Road, Shahekou District, Dalian, 116000, Liaoning, China
| | - Ya-Nan Liu
- Department of Geriatrics, The Second Hospital of Dalian Medical University, 467 Zhongshan Road, Shahekou District, Dalian, 116000, Liaoning, China
| | - Xiu-Li Wang
- Department of Histoembryology, College of Basic Medical Sciences, Dalian Medical University, 9 West Section, Lvshunnan Road, Lvshunkou District, Dalian, 116044, Liaoning, China
| | - Yun-Xia Du
- Department of Neurology, The Second Hospital of Dalian Medical University, 467 Zhongshan Road, Shahekou District, Dalian, 116000, Liaoning, China
| |
Collapse
|
36
|
Mohamed NA, Marei I, Crovella S, Abou-Saleh H. Recent Developments in Nanomaterials-Based Drug Delivery and Upgrading Treatment of Cardiovascular Diseases. Int J Mol Sci 2022; 23:1404. [PMID: 35163328 PMCID: PMC8836006 DOI: 10.3390/ijms23031404] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/18/2022] [Accepted: 01/21/2022] [Indexed: 01/27/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading causes of morbidity and mortality worldwide. However, despite the recent developments in the management of CVDs, the early and long outcomes vary considerably in patients, especially with the current challenges facing the detection and treatment of CVDs. This disparity is due to a lack of advanced diagnostic tools and targeted therapies, requiring innovative and alternative methods. Nanotechnology offers the opportunity to use nanomaterials in improving health and controlling diseases. Notably, nanotechnologies have recognized potential applicability in managing chronic diseases in the past few years, especially cancer and CVDs. Of particular interest is the use of nanoparticles as drug carriers to increase the pharmaco-efficacy and safety of conventional therapies. Different strategies have been proposed to use nanoparticles as drug carriers in CVDs; however, controversies regarding the selection of nanomaterials and nanoformulation are slowing their clinical translation. Therefore, this review focuses on nanotechnology for drug delivery and the application of nanomedicine in CVDs.
Collapse
Affiliation(s)
- Nura A. Mohamed
- Biological Science Program, Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Isra Marei
- Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, London SW7 2AZ, UK;
- Department of Pharmacology, Weill Cornell Medicine in Qatar, Doha P.O. Box 24144, Qatar
| | - Sergio Crovella
- Biological Science Program, Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Haissam Abou-Saleh
- Biological Science Program, Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar;
- Biomedical Research Center (BRC), Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
37
|
Shi J, Wei L. Rho Kinases in Embryonic Development and Stem Cell Research. Arch Immunol Ther Exp (Warsz) 2022; 70:4. [PMID: 35043239 PMCID: PMC8766376 DOI: 10.1007/s00005-022-00642-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022]
Abstract
The Rho-associated coiled-coil containing kinases (ROCKs or Rho kinases) belong to the AGC (PKA/PKG/PKC) family of serine/threonine kinases and are major downstream effectors of small GTPase RhoA, a key regulator of actin-cytoskeleton reorganization. The ROCK family contains two members, ROCK1 and ROCK2, which share 65% overall identity and 92% identity in kinase domain. ROCK1 and ROCK2 were assumed to be functionally redundant, based largely on their major common activators, their high degree kinase domain homology, and study results from overexpression with kinase constructs or chemical inhibitors. ROCK signaling research has expanded to all areas of biology and medicine since its discovery in 1996. The rapid advance is befitting ROCK’s versatile functions in modulating various cell behavior, such as contraction, adhesion, migration, proliferation, polarity, cytokinesis, and differentiation. The rapid advance is noticeably driven by an extensive linking with clinical medicine, including cardiovascular abnormalities, aberrant immune responsive, and cancer development and metastasis. The rapid advance during the past decade is further powered by novel biotechnologies including CRISPR-Cas and single cell omics. Current consensus, derived mainly from gene targeting and RNA interference approaches, is that the two ROCK isoforms have overlapping and distinct cellular, physiological and pathophysiology roles. In this review, we present an overview of the milestone discoveries in ROCK research. We then focus on the current understanding of ROCK signaling in embryonic development, current research status using knockout and knockin mouse models, and stem cell research.
Collapse
Affiliation(s)
- Jianjian Shi
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of Medicine, Indiana University, 1044 West Walnut Street, R4-370, Indianapolis, IN, 46202-5225, USA.
| | - Lei Wei
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of Medicine, Indiana University, 1044 West Walnut Street, R4-370, Indianapolis, IN, 46202-5225, USA.
| |
Collapse
|
38
|
Li W, Zhang Y, Xu J, Chen J, Gao X. Fasudil prevents neomycin-induced hair cell damage by inhibiting autophagy through the miR-489/NDP52 signaling pathway in HEI-OC1 cells. Exp Ther Med 2021; 23:43. [PMID: 34849158 DOI: 10.3892/etm.2021.10965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 08/11/2021] [Indexed: 11/05/2022] Open
Abstract
Hearing loss is a common sensory disorder that is mainly caused by the loss of hair cells (HCs). Drug-induced deafness, for which there is currently no effective treatment, is mainly caused by the inappropriate use of aminoglycoside antibiotics. Fasudil (Fas), a novel isoquinoline sulfonamide derivative, has exhibited antioxidant abilities in a number of previous studies. The aim of the present study was to investigate the potential effects of Fas against neomycin (Neo)-induced hair cell damage and elucidate the underlying mechanism. Flow cytometry and western blot analysis were used to detect the effects of Fas on cell apoptosis and to determine the expression levels of autophagy-related proteins, LC3B and Beclin 1, induced by Neo. Mitochondrial membrane potential and reactive oxygen species (ROS) levels were detected using fluorescent probes. The effect of Fas on Neo-induced hair cell injury marker, GFP-LC3B, was also examined using the immunofluorescence technique. Fas was found to inhibit Neo-induced mitochondrial autophagy and mitochondrial membrane potential decline, in addition to reducing ROS levels and cell apoptosis caused by Neo treatment. However, Fas failed to inhibit the Neo-induced these above changes in cells with NDP52 overexpression. The putative binding sites of microRNA (miR)-489 on the 3'-untranslated region of nuclear dot protein 52 (NDP52) were predicted using the TargetScan 7.0 online tool, and this association was further verified using a dual-luciferase reporter assay. Moreover, the expression of miR-489 negatively regulated the expression of NDP52. Fas and miR-489 mimic inhibited the Neo-induced mitochondrial autophagy and mitochondrial membrane potential decline, in addition to reducing ROS levels and cell apoptosis. Knockdown of miR-489 expression using a miR-489 inhibitor blocked the inhibitory effects of Fas on the mitochondrial membrane potential, cell apoptosis and ROS production. Therefore, Fas may upregulate the expression of miR-489 to negatively regulate the expression of NDP52 at the post-transcriptional level, which in turn inhibits the activation of mitophagy and cell injury induced by Neo. Thus, Fas may act as a novel therapeutic option in the clinical treatment of hearing loss in the future.
Collapse
Affiliation(s)
- Wei Li
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221006, P.R. China
| | - Yanqiu Zhang
- Department of Otolaryngology Head and Neck Surgery, Xuzhou Cancer Hospital, Xuzhou, Jiangsu 221005, P.R. China
| | - Jifeng Xu
- Department of Otolaryngology Head and Neck Surgery, The First Clinical Medical College of Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Jincan Chen
- Department of Otolaryngology Head and Neck Surgery, The First Clinical Medical College of Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Xia Gao
- Research Institute of Otolaryngology, Gulou Hospital Affiliated to Medical College of Nanjing University, Nanjing, Jiangsu 210008, P.R. China.,Department of Otolaryngology Head and Neck Surgery, Gulou Hospital Affiliated to Medical College of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| |
Collapse
|
39
|
Zhao J, Wang D, Yang WL, Niu J, Wu W. Unconventional Synthetic Process of Fasudil Hydrochloride: Costly Homopiperazine Was Avoided. Org Process Res Dev 2021. [DOI: 10.1021/acs.oprd.1c00211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Jianhong Zhao
- Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Dingding Wang
- Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Wu-Lin Yang
- Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Jinming Niu
- Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Weiting Wu
- Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| |
Collapse
|
40
|
Weber AJ, Adamson AB, Greathouse KM, Andrade JP, Freeman CD, Seo JV, Rae RJ, Walker CK, Herskowitz JH. Conditional deletion of ROCK2 induces anxiety-like behaviors and alters dendritic spine density and morphology on CA1 pyramidal neurons. Mol Brain 2021; 14:169. [PMID: 34794469 PMCID: PMC8600782 DOI: 10.1186/s13041-021-00878-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/04/2021] [Indexed: 12/25/2022] Open
Abstract
Rho-associated kinase isoform 2 (ROCK2) is an attractive drug target for several neurologic disorders. A critical barrier to ROCK2-based research and therapeutics is the lack of a mouse model that enables investigation of ROCK2 with spatial and temporal control of gene expression. To overcome this, we generated ROCK2fl/fl mice. Mice expressing Cre recombinase in forebrain excitatory neurons (CaMKII-Cre) were crossed with ROCK2fl/fl mice (Cre/ROCK2fl/fl), and the contribution of ROCK2 in behavior as well as dendritic spine morphology in the hippocampus, medial prefrontal cortex (mPFC), and basolateral amygdala (BLA) was examined. Cre/ROCK2fl/fl mice spent reduced time in the open arms of the elevated plus maze and increased time in the dark of the light-dark box test compared to littermate controls. These results indicated that Cre/ROCK2fl/fl mice exhibited anxiety-like behaviors. To examine dendritic spine morphology, individual pyramidal neurons in CA1 hippocampus, mPFC, and the BLA were targeted for iontophoretic microinjection of fluorescent dye, followed by high-resolution confocal microscopy and neuronal 3D reconstructions for morphometry analysis. In dorsal CA1, Cre/ROCK2fl/fl mice displayed significantly increased thin spine density on basal dendrites and reduced mean spine head volume across all spine types on apical dendrites. In ventral CA1, Cre/ROCK2fl/fl mice exhibited significantly increased spine length on apical dendrites. Spine density and morphology were comparable in the mPFC and BLA between both genotypes. These findings suggest that neuronal ROCK2 mediates spine density and morphology in a compartmentalized manner among CA1 pyramidal cells, and that in the absence of ROCK2 these mechanisms may contribute to anxiety-like behaviors.
Collapse
Affiliation(s)
- Audrey J Weber
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL, 35294, USA
| | - Ashley B Adamson
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL, 35294, USA
| | - Kelsey M Greathouse
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL, 35294, USA
| | - Julia P Andrade
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL, 35294, USA
| | - Cameron D Freeman
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL, 35294, USA
| | - Jung Vin Seo
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL, 35294, USA
| | - Rosaria J Rae
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL, 35294, USA
| | - Courtney K Walker
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL, 35294, USA
| | - Jeremy H Herskowitz
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL, 35294, USA.
| |
Collapse
|
41
|
DeOre BJ, Tran KA, Andrews AM, Ramirez SH, Galie PA. SARS-CoV-2 Spike Protein Disrupts Blood-Brain Barrier Integrity via RhoA Activation. J Neuroimmune Pharmacol 2021; 16:722-728. [PMID: 34687399 PMCID: PMC8536479 DOI: 10.1007/s11481-021-10029-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 10/06/2021] [Indexed: 12/23/2022]
Abstract
The SARS-CoV-2 spike protein has been shown to disrupt blood–brain barrier (BBB) function, but its pathogenic mechanism of action is unknown. Whether angiotensin converting enzyme 2 (ACE2), the viral binding site for SARS-CoV-2, contributes to the spike protein-induced barrier disruption also remains unclear. Here, a 3D-BBB microfluidic model was used to interrogate mechanisms by which the spike protein may facilitate barrier dysfunction. The spike protein upregulated the expression of ACE2 in response to laminar shear stress. Moreover, interrogating the role of ACE2 showed that knock-down affected endothelial barrier properties. These results identify a possible role of ACE2 in barrier homeostasis. Analysis of RhoA, a key molecule in regulating endothelial cytoskeleton and tight junction complex dynamics, reveals that the spike protein triggers RhoA activation. Inhibition of RhoA with C3 transferase rescues its effect on tight junction disassembly. Overall, these results indicate a possible means by which the engagement of SARS-CoV-2 with ACE2 facilitates disruption of the BBB via RhoA activation. Understanding how SARS-CoV-2 dysregulates the BBB may lead to strategies to prevent the neurological deficits seen in COVID-19 patients.
Collapse
Affiliation(s)
- Brandon J DeOre
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ, USA
| | - Kiet A Tran
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ, USA
| | - Allison M Andrews
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.,The Center for Substance Abuse Research Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Servio H Ramirez
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.,The Center for Substance Abuse Research Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA.,The Shriners Hospitals Pediatric Research Center, Philadelphia, PA, 19140, USA
| | - Peter A Galie
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ, USA.
| |
Collapse
|
42
|
Mulder IA, van Bavel ET, de Vries HE, Coutinho JM. Adjunctive cytoprotective therapies in acute ischemic stroke: a systematic review. Fluids Barriers CNS 2021; 18:46. [PMID: 34666786 PMCID: PMC8524879 DOI: 10.1186/s12987-021-00280-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/29/2021] [Indexed: 01/08/2023] Open
Abstract
With the introduction of endovascular thrombectomy (EVT), a new era for treatment of acute ischemic stroke (AIS) has arrived. However, despite the much larger recanalization rate as compared to thrombolysis alone, final outcome remains far from ideal. This raises the question if some of the previously tested neuroprotective drugs warrant re-evaluation, since these compounds were all tested in studies where large-vessel recanalization was rarely achieved in the acute phase. This review provides an overview of compounds tested in clinical AIS trials and gives insight into which of these drugs warrant a re-evaluation as an add-on therapy for AIS in the era of EVT. A literature search was performed using the search terms "ischemic stroke brain" in title/abstract, and additional filters. After exclusion of papers using pre-defined selection criteria, a total of 89 trials were eligible for review which reported on 56 unique compounds. Trial compounds were divided into 6 categories based on their perceived mode of action: systemic haemodynamics, excitotoxicity, neuro-inflammation, blood-brain barrier and vasogenic edema, oxidative and nitrosative stress, neurogenesis/-regeneration and -recovery. Main trial outcomes and safety issues are summarized and promising compounds for re-evaluation are highlighted. Looking at group effect, drugs intervening with oxidative and nitrosative stress and neurogenesis/-regeneration and -recovery appear to have a favourable safety profile and show the most promising results regarding efficacy. Finally, possible theories behind individual and group effects are discussed and recommendation for promising treatment strategies are described.
Collapse
Affiliation(s)
- I A Mulder
- Department of Biomedical Engineering and Physics, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
| | - E T van Bavel
- Department of Biomedical Engineering and Physics, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - H E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - J M Coutinho
- Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
43
|
Appunni S, Gupta D, Rubens M, Ramamoorthy V, Singh HN, Swarup V. Deregulated Protein Kinases: Friend and Foe in Ischemic Stroke. Mol Neurobiol 2021; 58:6471-6489. [PMID: 34549335 DOI: 10.1007/s12035-021-02563-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/10/2021] [Indexed: 12/20/2022]
Abstract
Ischemic stroke is the third leading cause of mortality worldwide, but its medical management is still limited to the use of thrombolytics as a lifesaving option. Multiple molecular deregulations of the protein kinase family occur during the period of ischemia/reperfusion. However, experimental studies have shown that alterations in the expression of essential protein kinases and their pharmacological modulation can modify the neuropathological milieu and hasten neurophysiological recovery. This review highlights the role of key protein kinase members and their implications in the evolution of stroke pathophysiology. Activation of ROCK-, MAPK-, and GSK-3β-mediated pathways following neuronal ischemia/reperfusion injury in experimental conditions aggravate the neuropathology and delays recovery. Targeting ROCK, MAPK, and GSK-3β will potentially enhance myelin regeneration, improve blood-brain barrier (BBB) function, and suppress inflammation, which ameliorates neuronal survival. Conversely, protein kinases such as PKA, Akt, PKCα, PKCε, Trk, and PERK salvage neurons post-ischemia by mechanisms including enhanced toxin metabolism, restoring BBB integrity, neurotrophic effects, and apoptosis suppression. Certain protein kinases such as ERK1/2, JNK, and AMPK have favourable and unfavourable effects in salvaging ischemia-injured neurons. Targeting multiple protein kinase-mediated pathways simultaneously may improve neuronal recovery post-ischemia.
Collapse
Affiliation(s)
- Sandeep Appunni
- Department of Biochemistry, Government Medical College, Kozhikode, Kerala, India
| | - Deepika Gupta
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | | | | | - Himanshu Narayan Singh
- Department of Systems Biology, Columbia University Irving Medical Centre, New York City, NY, USA.
| | - Vishnu Swarup
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
44
|
Bryniarska-Kubiak N, Kubiak A, Lekka M, Basta-Kaim A. The emerging role of mechanical and topographical factors in the development and treatment of nervous system disorders: dark and light sides of the force. Pharmacol Rep 2021; 73:1626-1641. [PMID: 34390472 PMCID: PMC8599311 DOI: 10.1007/s43440-021-00315-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 12/14/2022]
Abstract
Nervous system diseases are the subject of intensive research due to their association with high mortality rates and their potential to cause irreversible disability. Most studies focus on targeting the biological factors related to disease pathogenesis, e.g. use of recombinant activator of plasminogen in the treatment of stroke. Nevertheless, multiple diseases such as Parkinson’s disease and Alzheimer’s disease still lack successful treatment. Recently, evidence has indicated that physical factors such as the mechanical properties of cells and tissue and topography play a crucial role in homeostasis as well as disease progression. This review aims to depict these factors’ roles in the progression of nervous system diseases and consequently discusses the possibility of new therapeutic approaches. The literature is reviewed to provide a deeper understanding of the roles played by physical factors in nervous system disease development to aid in the design of promising new treatment approaches.
Collapse
Affiliation(s)
- Natalia Bryniarska-Kubiak
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland.
| | - Andrzej Kubiak
- Department of Biophysical Microstructures, Institute of Nuclear Physics, Polish Academy of Sciences, 31342, Kraków, Poland
| | - Małgorzata Lekka
- Department of Biophysical Microstructures, Institute of Nuclear Physics, Polish Academy of Sciences, 31342, Kraków, Poland
| | - Agnieszka Basta-Kaim
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland.
| |
Collapse
|
45
|
Quadir H, Hakobyan K, Gaddam M, Ojinnaka U, Ahmed Z, Kannan A, Mostafa JA. Role of Rho-Associated Protein Kinase Inhibition As Therapeutic Strategy for Parkinson's Disease: Dopaminergic Survival and Enhanced Mitophagy. Cureus 2021; 13:e16973. [PMID: 34377615 PMCID: PMC8349301 DOI: 10.7759/cureus.16973] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 08/07/2021] [Indexed: 12/24/2022] Open
Abstract
The GTP-binding protein, Rho, plays a significant role in the cellular pathology of Parkinson’s disease. The downstream effector of Rho, Rho-associated kinase (ROCK), performs several functions, including microglial inflammatory response and enhanced Parkin-mediated mitophagy. Its inhibition shows neuroprotective effects in carried studies. Parkinson’s disease pathology also rests on incomplete removal of damaged mitochondria, leading to neuronal impairment. ROCK has different isoforms, inhibition of which have been shown to decrease the adverse changes in microglia. There has also been evidence of a decreased release of inflammatory cytokines and a reduction in degradation of dopaminergic neurons on the addition of ROCK inhibitors. Additionally, ROCK inhibitors have recently been shown to increase the activity of hexokinase 2 (HK2), relocating it to mitochondria, and therefore leading to upregulated mitochondrial targeting. Understanding the cellular basis of ROCK activity and its inhibition may help us advance in creating new strategies for the treatment of Parkinson’s disease.
Collapse
Affiliation(s)
- Huma Quadir
- Internal Medicine/Family Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA.,Neurology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Knkush Hakobyan
- Diagnostic Radiology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Mrunanjali Gaddam
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Ugochi Ojinnaka
- Family Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Zubayer Ahmed
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Amudhan Kannan
- Medicine, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, IND.,General Surgery Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Jihan A Mostafa
- Faculty Member, California Institute of Behavioral Neurosciences & Psychology, Fairfield, California, USA
| |
Collapse
|
46
|
Giraldo E, Nebot VJ, Đorđević S, Requejo-Aguilar R, Alastrue-Agudo A, Zagorodko O, Armiñan A, Martinez-Rojas B, Vicent MJ, Moreno-Manzano V. A rationally designed self-immolative linker enhances the synergism between a polymer-rock inhibitor conjugate and neural progenitor cells in the treatment of spinal cord injury. Biomaterials 2021; 276:121052. [PMID: 34388362 DOI: 10.1016/j.biomaterials.2021.121052] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 07/04/2021] [Accepted: 07/24/2021] [Indexed: 10/24/2022]
Abstract
Rho/ROCK signaling induced after spinal cord injury (SCI) contributes to secondary damage by promoting apoptosis, inflammation, and axon growth inhibition. The specific Rho-kinase inhibitor fasudil can contribute to functional regeneration after SCI, although inherent low stability has hampered its use. To improve the therapeutic potential of fasudil, we now describe a family of rationally-designed bioresponsive polymer-fasudil conjugates based on an understanding of the conditions after SCI, such as low pH, enhanced expression of specific proteases, and a reductive environment. Fasudil conjugated to poly-l-glutamate via a self-immolative redox-sensitive linker (PGA-SS-F) displays optimal release kinetics and, consequently, treatment with PGA-SS-F significantly induces neurite elongation and axon growth in dorsal root ganglia explants, spinal cord organotypic cultures, and neural precursor cells (NPCs). The intrathecal administration of PGA-SS-F after SCI in a rat model prevents early apoptosis and induces the expression of axonal growth- and neuroplasticity-associated markers to a higher extent than the free form of fasudil. Moreover, a combination treatment comprising the acute transplantation of NPCs pre-treated with PGA-SS-F leads to enhanced cell engraftment and reduced cyst formation after SCI. In chronic SCI, combinatory treatment increases the preservation of neuronal fibers. Overall, this synergistic combinatorial strategy may represent a potentially efficient clinical approach to SCI treatment.
Collapse
Affiliation(s)
- E Giraldo
- Neuronal and Tissue Regeneration Lab. Prince Felipe Research Institute, Valencia, Spain; Department of Biotechnology. Universitat Politècnica de València, Valencia, Spain
| | - V J Nebot
- Polymer Therapeutics Lab. Prince Felipe Research Institute, Valencia, Spain; PTS S.L., Valencia, Spain
| | - S Đorđević
- Polymer Therapeutics Lab. Prince Felipe Research Institute, Valencia, Spain
| | - R Requejo-Aguilar
- Neuronal and Tissue Regeneration Lab. Prince Felipe Research Institute, Valencia, Spain; Dept. Biochemistry and Molecular Biology, University of Cordoba, Cordoba, Spain. Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Cordoba, Spain
| | - A Alastrue-Agudo
- Neuronal and Tissue Regeneration Lab. Prince Felipe Research Institute, Valencia, Spain
| | - O Zagorodko
- Polymer Therapeutics Lab. Prince Felipe Research Institute, Valencia, Spain
| | - A Armiñan
- Polymer Therapeutics Lab. Prince Felipe Research Institute, Valencia, Spain
| | - B Martinez-Rojas
- Neuronal and Tissue Regeneration Lab. Prince Felipe Research Institute, Valencia, Spain
| | - M J Vicent
- Polymer Therapeutics Lab. Prince Felipe Research Institute, Valencia, Spain.
| | - V Moreno-Manzano
- Neuronal and Tissue Regeneration Lab. Prince Felipe Research Institute, Valencia, Spain.
| |
Collapse
|
47
|
Tanaka T, Mori M, Sekino M, Higashijima U, Takaki M, Yamashita Y, Kakiuchi S, Tashiro M, Morimoto K, Tasaki O, Izumikawa K. Impact of plasma 5-hydroxyindoleacetic acid, a serotonin metabolite, on clinical outcome in septic shock, and its effect on vascular permeability. Sci Rep 2021; 11:14146. [PMID: 34238999 PMCID: PMC8266895 DOI: 10.1038/s41598-021-93649-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/28/2021] [Indexed: 12/18/2022] Open
Abstract
Septic shock is characterized by dysregulated vascular permeability. We hypothesized that the vascular permeability of endothelial cells (ECs) would be regulated by serotonin via serotonin-Rho-associated kinase (ROCK) signaling. We aimed to determine the impact of 5-hydroxyindoleacetic acid (5-HIAA) on septic shock as a novel biomarker. Plasma 5-HIAA levels and disease severity indices were obtained from 47 patients with sepsis. The association between 5-HIAA levels and severity indices was analyzed. Permeability upon serotonin stimulation was determined using human pulmonary microvascular ECs. 5-HIAA were significantly higher in septic shock patients than in patients without shock or healthy controls (p = 0.004). These elevated levels were correlated with severity indexes (SOFA score [p < 0.001], APACHE II [p < 0.001], and PaO2:FiO2 [p = 0.02]), and longitudinally associated with worse clinical outcomes (mechanical ventilation duration [p = 0.009] and ICU duration [p = 0.01]). In the experiment, serotonin increased the permeability of ECs, which was inhibited by the ROCK inhibitor (p < 0.001). Serotonin increases vascular permeability of ECs via ROCK signaling. This suggests a novel mechanism by which serotonin disrupts endothelial barriers via ROCK signaling and causes the pathogenesis of septic shock with a vascular leak. Serotonin serves as a novel biomarker of vascular permeability.
Collapse
Affiliation(s)
- Takeshi Tanaka
- Infection Control and Education Center, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan. .,Department of Infectious Diseases, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan.
| | - Masahiko Mori
- Department of Paediatrics, University of Oxford, Oxford, OX1 3SY, UK
| | - Motohiro Sekino
- Division of Intensive Care, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Ushio Higashijima
- Division of Intensive Care, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Masahiro Takaki
- Department of Infectious Diseases, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Yoshiro Yamashita
- Department of Infectious Diseases, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Satoshi Kakiuchi
- Department of Infectious Diseases, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Masato Tashiro
- Infection Control and Education Center, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan.,Department of Infectious Diseases, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Konosuke Morimoto
- Department of Infectious Diseases, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Osamu Tasaki
- Acute and Critical Care Center, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Koichi Izumikawa
- Infection Control and Education Center, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan.,Department of Infectious Diseases, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| |
Collapse
|
48
|
Li X, Tong J, Liu J, Wang Y. Down-regulation of ROCK2 alleviates ethanol-induced cerebral nerve injury partly by the suppression of the NF-κB signaling pathway. Bioengineered 2021; 11:779-790. [PMID: 32684089 PMCID: PMC8291877 DOI: 10.1080/21655979.2020.1795404] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Chronic alcohol consumption leads to hippocampal neuronal impairment, which related to neuronal death, oxidative stress, and inflammatory response. Rho-associated protein kinase 2 (ROCK2) is a major regulator in the central nervous system injury. However, the effects of ROCK2 in ethanol-induced brain injury have not been explored. In this work, we investigated the neuroprotective effects and the mechanism of ROCK2 inhibition in vivo. Wistar rats were exposed to 37% ethanol for 8 weeks to establish brain injury models. Morris water maze test was performed to evaluate cognitive function, and we found that the down-regulation of ROCK2 reduced the escape latency and increased the passing times and percentage of time spent in the target quadrant of rats. The results of H&E staining and Nissl staining showed that ROCK2 inhibition alleviated the pathological injury induced by ethanol. PI staining and Western blot confirmed that inhibiting ROCK2 attenuated the neuronal death and apoptosis as reflected by the reduced PI-positive neurons and the decreased expression of cleaved-caspase-3 and cleaved-caspase-9. Furthermore, the down-regulation of ROCK2 ameliorated the oxidative stress and inflammatory response induced by ethanol in rats as reflected by the up-regulation of IL-10, SOD, and GSH and reduction of TNF-α, IL-6, and MDA respectively. Additionally, Western blot and EMSA analysis revealed that the down-regulation of ROCK2 suppressed the nuclear transfer of NF-κB p65. In conclusion, our data suggested that ROCK2 inhibition ameliorated ethanol-mediated hippocampal neuronal impairment by anti-apoptotic, anti-inflammatory, anti-oxidative effects at least partially through the suppression of the NF-κB pathway.
Collapse
Affiliation(s)
- Xinguo Li
- Department of Neurosurgery, The First Hospital of China Medical University , Shenyang, People's Republic of China
| | - Jing Tong
- Department of Gastroenterology, The First Hospital of China Medical University , Shenyang, People's Republic of China
| | - Jihui Liu
- Department of Neurosurgery, The First Hospital of China Medical University , Shenyang, People's Republic of China
| | - Yibao Wang
- Department of Neurosurgery, The First Hospital of China Medical University , Shenyang, People's Republic of China
| |
Collapse
|
49
|
Nwafor DC, Brichacek AL, Ali A, Brown CM. Tissue-Nonspecific Alkaline Phosphatase in Central Nervous System Health and Disease: A Focus on Brain Microvascular Endothelial Cells. Int J Mol Sci 2021; 22:5257. [PMID: 34067629 PMCID: PMC8156423 DOI: 10.3390/ijms22105257] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 12/21/2022] Open
Abstract
Tissue-nonspecific alkaline phosphatase (TNAP) is an ectoenzyme bound to the plasma membranes of numerous cells via a glycosylphosphatidylinositol (GPI) moiety. TNAP's function is well-recognized from earlier studies establishing its important role in bone mineralization. TNAP is also highly expressed in cerebral microvessels; however, its function in brain cerebral microvessels is poorly understood. In recent years, few studies have begun to delineate a role for TNAP in brain microvascular endothelial cells (BMECs)-a key component of cerebral microvessels. This review summarizes important information on the role of BMEC TNAP, and its implication in health and disease. Furthermore, we discuss current models and tools that may assist researchers in elucidating the function of TNAP in BMECs.
Collapse
Affiliation(s)
- Divine C. Nwafor
- Department of Neuroscience, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA; (D.C.N.); (A.A.)
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Allison L. Brichacek
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA;
| | - Ahsan Ali
- Department of Neuroscience, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA; (D.C.N.); (A.A.)
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Candice M. Brown
- Department of Neuroscience, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA; (D.C.N.); (A.A.)
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA;
| |
Collapse
|
50
|
Brain capillary pericytes exert a substantial but slow influence on blood flow. Nat Neurosci 2021; 24:633-645. [PMID: 33603231 PMCID: PMC8102366 DOI: 10.1038/s41593-020-00793-2] [Citation(s) in RCA: 219] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 12/23/2020] [Indexed: 01/30/2023]
Abstract
The majority of the brain's vasculature is composed of intricate capillary networks lined by capillary pericytes. However, it remains unclear whether capillary pericytes influence blood flow. Using two-photon microscopy to observe and manipulate brain capillary pericytes in vivo, we find that their optogenetic stimulation decreases lumen diameter and blood flow, but with slower kinetics than similar stimulation of mural cells on upstream pial and precapillary arterioles. This slow vasoconstriction was inhibited by the clinically used vasodilator fasudil, a Rho-kinase inhibitor that blocks contractile machinery. Capillary pericytes were also slower to constrict back to baseline following hypercapnia-induced dilation, and slower to dilate towards baseline following optogenetically induced vasoconstriction. Optical ablation of single capillary pericytes led to sustained local dilation and a doubling of blood cell flux selectively in capillaries lacking pericyte contact. These data indicate that capillary pericytes contribute to basal blood flow resistance and slow modulation of blood flow throughout the brain.
Collapse
|