151
|
Application of Rho Kinase Inhibitors for the Treatment of Corneal Endothelial Diseases. J Ophthalmol 2017; 2017:2646904. [PMID: 28751979 PMCID: PMC5511675 DOI: 10.1155/2017/2646904] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 06/05/2017] [Indexed: 11/30/2022] Open
Abstract
ROCK (Rho kinase) signaling regulates a wide spectrum of fundamental cellular events and is involved in a variety of pathological conditions. It has therefore attracted research interest as a potential therapeutic target for combating various diseases. We showed that inhibition of ROCK enhances cell proliferation, promotes cell adhesion onto a substrate, and suppresses apoptosis of corneal endothelial cells (CECs). In addition, we reported that a ROCK inhibitor enhances wound healing in the corneal endothelium in animal models and in pilot clinical research. We also demonstrated the usefulness of a ROCK inhibitor as an adjunct drug in tissue engineering therapy as it enhances the engraftment of CECs onto recipient corneas. In 2013, we initiated a clinical trial to test the effectiveness of injection of cultured human CECs into the anterior chamber of patients with corneal endothelial decompensation. This paper reviews the accumulating evidence supporting the potency of ROCK inhibitors in clinical use, both as eye drops and as adjunct drugs in cell-based therapies, for the treatment of corneal endothelial decompensation.
Collapse
|
152
|
In Silico Investigations of Chemical Constituents of Clerodendrum colebrookianum in the Anti-Hypertensive Drug Targets: ROCK, ACE, and PDE5. Interdiscip Sci 2017. [DOI: 10.1007/s12539-017-0243-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
153
|
Meekins LC, Rosado-Adames N, Maddala R, Zhao JJ, Rao PV, Afshari NA. Corneal Endothelial Cell Migration and Proliferation Enhanced by Rho Kinase (ROCK) Inhibitors in In Vitro and In Vivo Models. Invest Ophthalmol Vis Sci 2017; 57:6731-6738. [PMID: 27951595 PMCID: PMC6018452 DOI: 10.1167/iovs.16-20414] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To explore the role of Rho-associated kinases (ROCK) in corneal physiology and regeneration, and the effects of suppressing its activity in stimulating corneal endothelial cell proliferation and migration in vitro and in vivo. Methods Immunohistochemistry was performed to detect RhoA and ROCK-1 and ROCK-2 in human corneal tissue. Adult porcine corneal endothelial cells (CECs) were isolated, grown to confluence, and further characterized. Under the treatment of ROCK inhibitors, changes in the cellular distribution profile of ZO-1 and F-actin were examined by immunofluorescence staining. Corneal endothelial cells migration was evaluated by scratch assay and analyzed with Axiovision software. Cell proliferation was quantified using Click-iT EdU HCS Assay. In vivo, the corneal endothelia of rabbits were surgically injured and H-1152 was topically applied for 10 days. Progress of wound healing was evaluated daily by monitoring corneal edema, inflammation, and thickness using slit-lamp examination, photography, and pachymetry. Rabbits were euthanized and enucleated for further evaluation. Results H-1152 exhibited significant stimulatory effect on CEC migration and proliferation in vitro compared with both untreated and Y-27632–treated cells. Furthermore, topical administration of H-1152 led to marked reduction in corneal edema and formation of multinucleate CECs in vivo suggestive of proliferation associated with healing. Conclusions H-1152 exhibited a better stimulatory effect on CEC migration and proliferation in vitro than Y-27632. Our findings suggest that topical administration of H-1152 promotes healing of injured corneal endothelium in vivo. These results demonstrate the efficacy of ROCK inhibitors as a potential topical therapy for patients with corneal endothelial disease.
Collapse
Affiliation(s)
- Landon C Meekins
- Duke University Eye Center, Duke University Medical Center, Durham, North Carolina, United States
| | - Noel Rosado-Adames
- Duke University Eye Center, Duke University Medical Center, Durham, North Carolina, United States
| | - Rupalatha Maddala
- Duke University Eye Center, Duke University Medical Center, Durham, North Carolina, United States
| | - Jiagang J Zhao
- Shiley Eye Institute, University of California San Diego, La Jolla, California, United States
| | - Ponugoti V Rao
- Duke University Eye Center, Duke University Medical Center, Durham, North Carolina, United States
| | - Natalie A Afshari
- Shiley Eye Institute, University of California San Diego, La Jolla, California, United States
| |
Collapse
|
154
|
Schnoor M, García Ponce A, Vadillo E, Pelayo R, Rossaint J, Zarbock A. Actin dynamics in the regulation of endothelial barrier functions and neutrophil recruitment during endotoxemia and sepsis. Cell Mol Life Sci 2017; 74:1985-1997. [PMID: 28154894 PMCID: PMC11107778 DOI: 10.1007/s00018-016-2449-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 12/19/2016] [Accepted: 12/21/2016] [Indexed: 01/20/2023]
Abstract
Sepsis is a leading cause of death worldwide. Increased vascular permeability is a major hallmark of sepsis. Dynamic alterations in actin fiber formation play an important role in the regulation of endothelial barrier functions and thus vascular permeability. Endothelial integrity requires a delicate balance between the formation of cortical actin filaments that maintain endothelial cell contact stability and the formation of actin stress fibers that generate pulling forces, and thus compromise endothelial cell contact stability. Current research has revealed multiple molecular pathways that regulate actin dynamics and endothelial barrier dysfunction during sepsis. These include intracellular signaling proteins of the small GTPases family (e.g., Rap1, RhoA and Rac1) as well as the molecules that are directly acting on the actomyosin cytoskeleton such as myosin light chain kinase and Rho kinases. Another hallmark of sepsis is an excessive recruitment of neutrophils that also involves changes in the actin cytoskeleton in both endothelial cells and neutrophils. This review focuses on the available evidence about molecules that control actin dynamics and regulate endothelial barrier functions and neutrophil recruitment. We also discuss treatment strategies using pharmaceutical enzyme inhibitors to target excessive vascular permeability and leukocyte recruitment in septic patients.
Collapse
Affiliation(s)
- Michael Schnoor
- Department for Molecular Biomedicine, Centre for Investigation and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Av. IPN 2508, San Pedro Zacatenco, GAM, 07360, Mexico City, Mexico.
| | - Alexander García Ponce
- Department for Molecular Biomedicine, Centre for Investigation and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Av. IPN 2508, San Pedro Zacatenco, GAM, 07360, Mexico City, Mexico
| | - Eduardo Vadillo
- Department for Molecular Biomedicine, Centre for Investigation and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Av. IPN 2508, San Pedro Zacatenco, GAM, 07360, Mexico City, Mexico
| | - Rosana Pelayo
- Oncology Research Unit, National Medical Center, Mexican Institute for Social Security, 06720, Mexico City, Mexico
| | - Jan Rossaint
- Department of Anaesthesiology, Critical Care and Pain Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany
| | - Alexander Zarbock
- Department of Anaesthesiology, Critical Care and Pain Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany.
| |
Collapse
|
155
|
Qudrat A, Mosabbir AA, Truong K. Engineered Proteins Program Mammalian Cells to Target Inflammatory Disease Sites. Cell Chem Biol 2017; 24:703-711.e2. [PMID: 28552580 DOI: 10.1016/j.chembiol.2017.05.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 02/08/2017] [Accepted: 05/03/2017] [Indexed: 12/28/2022]
Abstract
Disease sites in atherosclerosis and cancer feature cell masses (e.g., plaques/tumors), a low pH extracellular microenvironment, and various pro-inflammatory cytokines such as tumor necrosis factor α (TNFα). The ability to engineer a cell to seek TNFα sources allows for targeted therapeutic delivery. To accomplish this, here we introduced a system of proteins: an engineered TNFα chimeric receptor (named TNFR1chi), a previously engineered Ca2+-activated RhoA (named CaRQ), vesicular stomatitis virus glycoprotein G (VSVG), and thymidine kinase. Upon binding TNFα, TNFR1chi generates a Ca2+ signal that in turn activates CaRQ-mediated non-apoptotic blebs that allow migration toward the TNFα source. Next, the addition of VSVG, upon low pH induction, causes membrane fusion of the engineered and TNFα source cells. Finally, after ganciclovir treatment cells undergo death via the thymidine kinase suicide mechanism. Hence, we assembled a system of proteins that forms the basis of engineering a cell to target inflammatory disease sites characterized by TNFα secretion and a low-pH microenvironment.
Collapse
Affiliation(s)
- Anam Qudrat
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street Room 407, Rosebrugh Building, Toronto, ON M5S 3G9, Canada
| | - Abdullah Al Mosabbir
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street Room 407, Rosebrugh Building, Toronto, ON M5S 3G9, Canada
| | - Kevin Truong
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street Room 407, Rosebrugh Building, Toronto, ON M5S 3G9, Canada; Edward S. Rogers Sr. Department of Electrical and Computer Engineering, University of Toronto, 10 King's College Circle, Toronto, ON M5S 3G4, Canada.
| |
Collapse
|
156
|
Yamada H, Yoneda M, Inaguma S, Gosho M, Murasawa Y, Isogai Z, Zako M. A Rho-Associated Kinase Inhibitor Protects Permeability in a Cell Culture Model of Ocular Disease, and Reduces Aqueous Flare in Anterior Uveitis. J Ocul Pharmacol Ther 2017; 33:176-185. [DOI: 10.1089/jop.2016.0085] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
- Hiroshi Yamada
- Department of Ophthalmology, Aichi Medical University, Nagakute, Japan
| | - Masahiko Yoneda
- Department of Biochemistry and Molecular Biology, School of Nursing and Health, Aichi Prefectural University, Nagoya, Japan
| | - Shingo Inaguma
- Department of Pathology, Aichi Medical University, Nagakute, Japan
| | - Masahiko Gosho
- Department of Clinical Trial and Clinical Epidemiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yusuke Murasawa
- Department of Advanced Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Zenzo Isogai
- Department of Advanced Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Masahiro Zako
- Department of Ophthalmology, Aichi Medical University, Nagakute, Japan
- Department of Ophthalmology, Asai Hospital, Seto, Japan
| |
Collapse
|
157
|
Karsy M, Brock A, Guan J, Taussky P, Kalani MYS, Park MS. Neuroprotective strategies and the underlying molecular basis of cerebrovascular stroke. Neurosurg Focus 2017; 42:E3. [DOI: 10.3171/2017.1.focus16522] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Stroke is a leading cause of disability in the US. Although there has been significant progress in the area of medical and surgical thrombolytic technologies, neuroprotective agents to prevent secondary cerebral injury and to minimize disability remain limited. Only limited success has been reported in preclinical and clinical trials evaluating a variety of compounds. In this review, the authors discuss the most up-to-date information regarding the underlying molecular biology of stroke as well as strategies that aim to mitigate this complex signaling cascade. Results of historical research trials involving N-methyl-d-aspartate and α-amino-3-hydroxy-5-methyl-4-isoxazole propionate receptor antagonists, clomethiazole, antioxidants, citicoline, nitric oxide, and immune regulators have laid the groundwork for current progress. In addition, more recent studies involving therapeutic hypothermia, magnesium, albumin, glyburide, uric acid, and a variety of other treatments have provided more options. The use of neuroprotective agents in combination or with existing thrombolytic treatments may be one of many exciting areas of further development. Although past trials of neuroprotective agents in ischemic stroke have been limited, significant insights into mechanisms of stroke, animal models, and trial design have incrementally improved approaches for future therapies.
Collapse
|
158
|
Muhamed I, Chowdhury F, Maruthamuthu V. Biophysical Tools to Study Cellular Mechanotransduction. Bioengineering (Basel) 2017; 4:E12. [PMID: 28952491 PMCID: PMC5590431 DOI: 10.3390/bioengineering4010012] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 01/30/2017] [Accepted: 02/02/2017] [Indexed: 01/25/2023] Open
Abstract
The cell membrane is the interface that volumetrically isolates cellular components from the cell's environment. Proteins embedded within and on the membrane have varied biological functions: reception of external biochemical signals, as membrane channels, amplification and regulation of chemical signals through secondary messenger molecules, controlled exocytosis, endocytosis, phagocytosis, organized recruitment and sequestration of cytosolic complex proteins, cell division processes, organization of the cytoskeleton and more. The membrane's bioelectrical role is enabled by the physiologically controlled release and accumulation of electrochemical potential modulating molecules across the membrane through specialized ion channels (e.g., Na⁺, Ca2+, K⁺ channels). The membrane's biomechanical functions include sensing external forces and/or the rigidity of the external environment through force transmission, specific conformational changes and/or signaling through mechanoreceptors (e.g., platelet endothelial cell adhesion molecule (PECAM), vascular endothelial (VE)-cadherin, epithelial (E)-cadherin, integrin) embedded in the membrane. Certain mechanical stimulations through specific receptor complexes induce electrical and/or chemical impulses in cells and propagate across cells and tissues. These biomechanical sensory and biochemical responses have profound implications in normal physiology and disease. Here, we discuss the tools that facilitate the understanding of mechanosensitive adhesion receptors. This article is structured to provide a broad biochemical and mechanobiology background to introduce a freshman mechano-biologist to the field of mechanotransduction, with deeper study enabled by many of the references cited herein.
Collapse
Affiliation(s)
- Ismaeel Muhamed
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA.
| | - Farhan Chowdhury
- Department of Mechanical Engineering and Energy Processes, Southern Illinois University Carbondale, Carbondale, IL 62901, USA.
| | - Venkat Maruthamuthu
- Department of Mechanical and Aerospace Engineering, Old Dominion University, Norfolk, VA 23529, USA.
| |
Collapse
|
159
|
Dekmak A, Mantash S, Shaito A, Toutonji A, Ramadan N, Ghazale H, Kassem N, Darwish H, Zibara K. Stem cells and combination therapy for the treatment of traumatic brain injury. Behav Brain Res 2016; 340:49-62. [PMID: 28043902 DOI: 10.1016/j.bbr.2016.12.039] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 10/30/2016] [Accepted: 12/29/2016] [Indexed: 12/15/2022]
Abstract
TBI is a nondegenerative, noncongenital insult to the brain from an external mechanical force; for instance a violent blow in a car accident. It is a complex injury with a broad spectrum of symptoms and has become a major cause of death and disability in addition to being a burden on public health and societies worldwide. As such, finding a therapy for TBI has become a major health concern for many countries, which has led to the emergence of many monotherapies that have shown promising effects in animal models of TBI, but have not yet proven any significant efficacy in clinical trials. In this paper, we will review existing and novel TBI treatment options. We will first shed light on the complex pathophysiology and molecular mechanisms of this disorder, understanding of which is a necessity for launching any treatment option. We will then review most of the currently available treatments for TBI including the recent approaches in the field of stem cell therapy as an optimal solution to treat TBI. Therapy using endogenous stem cells will be reviewed, followed by therapies utilizing exogenous stem cells from embryonic, induced pluripotent, mesenchymal, and neural origin. Combination therapy is also discussed as an emergent novel approach to treat TBI. Two approaches are highlighted, an approach concerning growth factors and another using ROCK inhibitors. These approaches are highlighted with regard to their benefits in minimizing the outcomes of TBI. Finally, we focus on the consequent improvements in motor and cognitive functions after stem cell therapy. Overall, this review will cover existing treatment options and recent advancements in TBI therapy, with a focus on the potential application of these strategies as a solution to improve the functional outcomes of TBI.
Collapse
Affiliation(s)
- AmiraSan Dekmak
- ER045, Laboratory of Stem Cells, Faculty of Sciences, DSST, PRASE, Lebanese University, Beirut, Lebanon
| | - Sarah Mantash
- ER045, Laboratory of Stem Cells, Faculty of Sciences, DSST, PRASE, Lebanese University, Beirut, Lebanon; Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Abdullah Shaito
- Department of Biological and Chemical Sciences, Lebanese International University, Beirut, Lebanon
| | - Amer Toutonji
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Naify Ramadan
- ER045, Laboratory of Stem Cells, Faculty of Sciences, DSST, PRASE, Lebanese University, Beirut, Lebanon; Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Hussein Ghazale
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Nouhad Kassem
- ER045, Laboratory of Stem Cells, Faculty of Sciences, DSST, PRASE, Lebanese University, Beirut, Lebanon
| | - Hala Darwish
- Faculty of Medicine, Hariri School of Nursing, American University of Beirut, Beirut, Lebanon
| | - Kazem Zibara
- ER045, Laboratory of Stem Cells, Faculty of Sciences, DSST, PRASE, Lebanese University, Beirut, Lebanon; Laboratory of Cardiovascular Diseases and Stem Cells, Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon.
| |
Collapse
|
160
|
Shenkar R, Shi C, Austin C, Moore T, Lightle R, Cao Y, Zhang L, Wu M, Zeineddine HA, Girard R, McDonald DA, Rorrer A, Gallione C, Pytel P, Liao JK, Marchuk DA, Awad IA. RhoA Kinase Inhibition With Fasudil Versus Simvastatin in Murine Models of Cerebral Cavernous Malformations. Stroke 2016; 48:187-194. [PMID: 27879448 DOI: 10.1161/strokeaha.116.015013] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/12/2016] [Accepted: 10/14/2016] [Indexed: 01/12/2023]
Abstract
BACKGROUND AND PURPOSE We sought to compare the effect of chronic treatment with commonly tolerated doses of Fasudil, a specific RhoA kinase (ROCK) inhibitor, and simvastatin (with pleiotropic effects including ROCK inhibition) on cerebral cavernous malformation (CCM) genesis and maturation in 2 models that recapitulate the human disease. METHODS Two heterozygous murine models, Ccm1+/-Msh2-/- and Ccm2+/-Trp53-/-, were treated from weaning to 4 to 5 months of age with Fasudil (100 mg/kg per day), simvastatin (40 mg/kg per day) or with placebo. Mouse brains were blindly assessed for CCM lesion burden, nonheme iron deposition (as a quantitative measure of chronic lesional hemorrhage), and ROCK activity. RESULTS Fasudil, but not simvastatin, significantly decreased mature CCM lesion burden in Ccm1+/-Msh2-/- mice, and in meta-analysis of both models combined, when compared with mice receiving placebo. Fasudil and simvastatin both significantly decreased the integrated iron density per mature lesion area in Ccm1+/-Msh2-/- mice, and in both models combined, compared with mice given placebo. ROCK activity in mature lesions of Ccm1+/-Msh2-/- mice was similar with both treatments. Fasudil, but not simvastatin, improved survival in Ccm1+/-Msh2-/- mice. Fasudil and simvastatin treatment did not affect survival or lesion development significantly in Ccm2+/-Trp53-/- mice alone, and Fasudil benefit seemed limited to males. CONCLUSIONS ROCK inhibitor Fasudil was more efficacious than simvastatin in improving survival and blunting the development of mature CCM lesions. Both drugs significantly decreased chronic hemorrhage in CCM lesions. These findings justify the development of ROCK inhibitors and the clinical testing of commonly used statin agents in CCM.
Collapse
Affiliation(s)
- Robert Shenkar
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Changbin Shi
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Cecilia Austin
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Thomas Moore
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Rhonda Lightle
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Ying Cao
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Lingjiao Zhang
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Meijing Wu
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Hussein A Zeineddine
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Romuald Girard
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - David A McDonald
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Autumn Rorrer
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Carol Gallione
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Peter Pytel
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - James K Liao
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Douglas A Marchuk
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Issam A Awad
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk).
| |
Collapse
|
161
|
Thompson JM, Nguyen QH, Singh M, Pavesic MW, Nesterenko I, Nelson LJ, Liao AC, Razorenova OV. Rho-associated kinase 1 inhibition is synthetically lethal with von Hippel-Lindau deficiency in clear cell renal cell carcinoma. Oncogene 2016; 36:1080-1089. [PMID: 27841867 PMCID: PMC5323317 DOI: 10.1038/onc.2016.272] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 06/06/2016] [Accepted: 06/20/2016] [Indexed: 12/18/2022]
Abstract
Clear Cell Renal Cell Carcinoma (CC-RCC) is the most lethal of all genitourinary cancers. The functional loss of the von Hippel-Lindau (VHL) gene occurs in 90% of CC-RCC, driving cancer progression. The objective of this study was to identify chemical compounds that are synthetically lethal with VHL deficiency in CC-RCC. An annotated chemical library, the Library of Pharmacologically Active Compounds (LOPAC), was screened in parallel on VHL-deficient RCC4 cells and RCC4VHL cells with re-introduced VHL cDNA. The ROCK inhibitor, Y-27632, was identified and validated for selective targeting of VHL-deficient CC-RCC in multiple genetic backgrounds by clonogenic assays. Downregulation of ROCK1 by siRNA selectively reduced the colony forming ability of VHL-deficient CC-RCC, thus mimicking the effect of Y-27632 treatment, whereas downregulation of ROCK2 had no effect. In addition, two other ROCK inhibitors, RKI 1447 and GSK 429286, selectively targeted VHL-deficient CC-RCC. CC-RCC treatment with ROCK inhibitors is cytotoxic and cytostatic based on BrdU assay, Propidium Iodide (PI) staining, and growth curves; and blocks cell migration based on transwell assay. Importantly, knockdown of Hypoxia Inducible Factor (HIF) β in the VHL-deficient CC-RCC had a protective effect against Y-27632 treatment, mimicking VHL reintroduction. On the other hand, CC-RCCVHL cells were sensitized to Y-27632 treatment in hypoxia (2% O2). These results suggest that synthetic lethality between ROCK inhibition and VHL deficiency is dependent on HIF activation. Moreover, HIF1α or HIF2α overexpression in CC-RCCVHL cells is sufficient to sensitize them to ROCK inhibition. Finally, Y-27632 treatment inhibited growth of subcutaneous 786-OT1 CC-RCC tumors in mice. Thus, ROCK inhibitors represent potential therapeutics for VHL-deficient CC-RCC.
Collapse
Affiliation(s)
- J M Thompson
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| | - Q H Nguyen
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| | - M Singh
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| | - M W Pavesic
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| | - I Nesterenko
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| | - L J Nelson
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| | - A C Liao
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| | - O V Razorenova
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
162
|
Pellegrino PR, Schiller AM, Haack KKV, Zucker IH. Central Angiotensin-II Increases Blood Pressure and Sympathetic Outflow via Rho Kinase Activation in Conscious Rabbits. Hypertension 2016; 68:1271-1280. [PMID: 27672026 DOI: 10.1161/hypertensionaha.116.07792] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 08/25/2016] [Indexed: 01/08/2023]
Abstract
Elevated sympathetic tone and activation of the renin-angiotensin system are pathophysiologic hallmarks of hypertension, and the interactions between these systems are particularly deleterious. The importance of Rho kinase as a mediator of the effects of angiotensin-II (AngII) in the periphery is clear, but the role of Rho kinase in sympathoexcitation caused by central AngII is not well established. We hypothesized that AngII mediates its effects in the brain by the activation of the RhoA/Rho kinase pathway. Chronically instrumented, conscious rabbits received the following intracerebroventricular infusion treatments for 2 weeks via osmotic minipump: AngII, Rho kinase inhibitor Fasudil, AngII plus Fasudil, or a vehicle control. AngII increased mean arterial pressure over the course of the infusion, and this effect was prevented by the coadministration of Fasudil. AngII increased cardiac and vascular sympathetic outflow as quantified by the heart rate response to metoprolol and the depressor effect of hexamethonium; coadministration of Fasudil abolished both of these effects. AngII increased baseline renal sympathetic nerve activity in conscious animals and impaired baroreflex control of sympathetic nerve activity; again Fasudil coinfusion prevented these effects. Each of these end points showed a statistically significant interaction between AngII and Fasudil. Quantitative immunofluorescence of brain slices confirmed that Rho kinase activity was increased by AngII and decreased by Fasudil. Taken together, these data indicate that hypertension, elevated sympathetic outflow, and baroreflex dysfunction caused by central AngII are mediated by Rho kinase activation and suggest that Rho kinase inhibition may be an important therapeutic target in sympathoexcitatory cardiovascular diseases.
Collapse
Affiliation(s)
- Peter R Pellegrino
- From the Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (P.R.P., A.M.S., I.H.Z.); U.S. Army Institute of Surgical Research, Fort Sam Houston, TX (A.M.S.); and Department of Pediatrics, Emory University School of Medicine, Atlanta, GA (K.K.V.H.)
| | - Alicia M Schiller
- From the Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (P.R.P., A.M.S., I.H.Z.); U.S. Army Institute of Surgical Research, Fort Sam Houston, TX (A.M.S.); and Department of Pediatrics, Emory University School of Medicine, Atlanta, GA (K.K.V.H.)
| | - Karla K V Haack
- From the Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (P.R.P., A.M.S., I.H.Z.); U.S. Army Institute of Surgical Research, Fort Sam Houston, TX (A.M.S.); and Department of Pediatrics, Emory University School of Medicine, Atlanta, GA (K.K.V.H.)
| | - Irving H Zucker
- From the Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (P.R.P., A.M.S., I.H.Z.); U.S. Army Institute of Surgical Research, Fort Sam Houston, TX (A.M.S.); and Department of Pediatrics, Emory University School of Medicine, Atlanta, GA (K.K.V.H.).
| |
Collapse
|
163
|
Barabutis N, Verin A, Catravas JD. Regulation of pulmonary endothelial barrier function by kinases. Am J Physiol Lung Cell Mol Physiol 2016; 311:L832-L845. [PMID: 27663990 DOI: 10.1152/ajplung.00233.2016] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 09/15/2016] [Indexed: 12/15/2022] Open
Abstract
The pulmonary endothelium is the target of continuous physiological and pathological stimuli that affect its crucial barrier function. The regulation, defense, and repair of endothelial barrier function require complex biochemical processes. This review examines the role of endothelial phosphorylating enzymes, kinases, a class with profound, interdigitating influences on endothelial permeability and lung function.
Collapse
Affiliation(s)
- Nektarios Barabutis
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia
| | - Alexander Verin
- Vascular Biology Center, Augusta University, Augusta, Georgia; and
| | - John D Catravas
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia, .,School of Medical Diagnostic and Translational Sciences, College of Health Sciences, Old Dominion University, Norfolk, Virginia
| |
Collapse
|
164
|
Prasanna G, Li B, Mogi M, Rice DS. Pharmacology of novel intraocular pressure-lowering targets that enhance conventional outflow facility: Pitfalls, promises and what lies ahead? Eur J Pharmacol 2016; 787:47-56. [DOI: 10.1016/j.ejphar.2016.03.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 02/15/2016] [Accepted: 03/01/2016] [Indexed: 12/18/2022]
|
165
|
Lusthaus JA, Goldberg I. Investigational and experimental drugs for intraocular pressure reduction in ocular hypertension and glaucoma. Expert Opin Investig Drugs 2016; 25:1201-8. [DOI: 10.1080/13543784.2016.1223042] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
166
|
Jia XF, Ye F, Wang YB, Feng DX. ROCK inhibition enhances neurite outgrowth in neural stem cells by upregulating YAP expression in vitro. Neural Regen Res 2016; 11:983-7. [PMID: 27482229 PMCID: PMC4962598 DOI: 10.4103/1673-5374.184499] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Spontaneous axonal regeneration of neurons does not occur after spinal cord injury because of inhibition by myelin and other inhibitory factors. Studies have demonstrated that blocking the Rho/Rho-kinase (ROCK) pathway can promote neurite outgrowth in spinal cord injury models. In the present study, we investigated neurite outgrowth and neuronal differentiation in neural stem cells from the mouse subventricular zone after inhibition of ROCK in vitro. Inhibition of ROCK with Y-27632 increased neurite length, enhanced neuronal differentiation, and upregulated the expression of two major signaling pathway effectors, phospho-Akt and phospho-mitogen-activated protein kinase, and the Hippo pathway effector YAP. These results suggest that inhibition of ROCK mediates neurite outgrowth in neural stem cells by activating the Hippo signaling pathway.
Collapse
Affiliation(s)
- Xu-Feng Jia
- Department of Orthopedic Surgery, Jianyang People's Hospital of Sichuan Province, Jianyang, Sichuan Province, China
| | - Fei Ye
- Department of Spine Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Yan-Bo Wang
- Department of Spine Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Da-Xiong Feng
- Department of Spine Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| |
Collapse
|
167
|
Abstract
Hypertensive cardiac remodeling is characterized by left ventricular hypertrophy and interstitial fibrosis, which can lead to heart failure with preserved ejection fraction. The Rho-associated coiled-coil containing kinases (ROCKs) are members of the serine/threonine protein kinase family, which mediates the downstream effects of the small GTP-binding protein RhoA. There are 2 isoforms: ROCK1 and ROCK2. They have different functions in different types of cells and tissues. There is growing evidence that ROCKs contribute to the development of cardiovascular diseases, including cardiac fibrosis, hypertrophy, and subsequent heart failure. Recent experimental studies using ROCK inhibitors, such as fasudil, have shown the benefits of ROCK inhibition in cardiac remodeling. Mice lacking each ROCK isoform also exhibit reduced myocardial fibrosis in a variety of pathological models of cardiac remodeling. Indeed, clinical studies with fasudil have suggested that ROCKs could be potential novel therapeutic targets for cardiovascular diseases. In this review, we summarize the current understanding of the roles of ROCKs in the development of cardiac fibrosis and hypertrophy and discuss their therapeutic potential for deleterious cardiac remodeling. (Circ J 2016; 80: 1491-1498).
Collapse
Affiliation(s)
- Toru Shimizu
- Section of Cardiology, Department of Medicine, University of Chicago
| | | |
Collapse
|
168
|
Skaria T, Burgener J, Bachli E, Schoedon G. IL-4 Causes Hyperpermeability of Vascular Endothelial Cells through Wnt5A Signaling. PLoS One 2016; 11:e0156002. [PMID: 27214384 PMCID: PMC4877093 DOI: 10.1371/journal.pone.0156002] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 05/06/2016] [Indexed: 11/18/2022] Open
Abstract
Microvascular leakage due to endothelial barrier dysfunction is a prominent feature of T helper 2 (Th2) cytokine mediated allergic inflammation. Interleukin-4 (IL-4) is a potent Th2 cytokine, known to impair the barrier function of endothelial cells. However, the effectors mediating IL-4 induced cytoskeleton remodeling and consequent endothelial barrier dysfunction remain poorly defined. Here we have used whole genome transcriptome profiling and gene ontology analyses to identify the genes and processes regulated by IL-4 signaling in human coronary artery endothelial cells (HCAEC). The study revealed Wnt5A as an effector that can mediate actin cytoskeleton remodeling in IL-4 activated HCAEC through the regulation of LIM kinase (LIMK) and Cofilin (CFL). Following IL-4 treatment, LIMK and CFL were phosphorylated, thereby indicating the possibility of actin stress fiber formation. Imaging of actin showed the formation of stress fibers in IL-4 treated live HCAEC. Stress fiber formation was notably decreased in the presence of Wnt inhibitory factor 1 (WIF1). Non-invasive impedance measurements demonstrated that IL-4 increased the permeability and impaired the barrier function of HCAEC monolayers. Silencing Wnt5A significantly reduced permeability and improved the barrier function of HCAEC monolayers upon IL-4 treatment. Our study identifies Wnt5A as a novel marker of IL-4 activated vascular endothelium and demonstrates a critical role for Wnt5A in mediating IL-4 induced endothelial barrier dysfunction. Wnt5A could be a potential therapeutic target for reducing microvascular leakage and edema formation in Th2 driven inflammatory diseases.
Collapse
Affiliation(s)
- Tom Skaria
- Inflammation Research Unit, Department of Medicine, Division of Internal Medicine, University Hospital Zürich, Zürich, Switzerland
| | - Julia Burgener
- Inflammation Research Unit, Department of Medicine, Division of Internal Medicine, University Hospital Zürich, Zürich, Switzerland
| | - Esther Bachli
- Department of Medicine, Uster Hospital, Uster, Switzerland
| | - Gabriele Schoedon
- Inflammation Research Unit, Department of Medicine, Division of Internal Medicine, University Hospital Zürich, Zürich, Switzerland
- * E-mail:
| |
Collapse
|
169
|
Hu C, Sun L, Xiao L, Han Y, Fu X, Xiong X, Xu X, Liu Y, Yang S, Liu F, Kanwar YS. Insights into the Mechanisms Involved in the Expression and Regulation of Extracellular Matrix Proteins in Diabetic Nephropathy. Curr Med Chem 2016; 22:2858-70. [PMID: 26119175 DOI: 10.2174/0929867322666150625095407] [Citation(s) in RCA: 156] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 06/15/2015] [Accepted: 06/24/2015] [Indexed: 02/06/2023]
Abstract
Diabetic Nephropathy (DN) is believed to be a major microvascular complication of diabetes. The hallmark of DN includes deposition of Extracellular Matrix (ECM) proteins, such as, collagen, laminin and fibronectin in the mesangium and renal tubulo-interstitium of the glomerulus and basement membranes. Such an increased expression of ECM leads to glomerular and tubular basement membranes thickening and increase of mesangial matrix, ultimately resulting in glomerulosclerosis and tubulointerstitial fibrosis. The characteristic morphologic glomerular mesangial lesion has been described as Kimmelstiel-Wilson nodule, and the process at times is referred to as diabetic nodular glomerulosclerosis. Thus, the accumulation of ECM proteins plays a critical role in the development of DN. The relevant mechanism(s) involved in the increased ECM expression and their regulation in the kidney in diabetic state has been extensively investigated and documented in the literature. Nevertheless, there are certain other mechanisms that may yet be conclusively defined. Recent studies demonstrated that some of the new signaling pathways or molecules including, Notch, Wnt, mTOR, TLRs and small GTPase may play a pivotal role in the modulation of ECM regulation and expression in DN. Such modulation could be operational for instance Notch through Notch1/Jagged1 signaling, Wnt by Wnt/β- catenin pathway and mTOR via PI3-K/Akt/mTOR signaling pathways. All these pathways may be critical in the modulation of ECM expression and tubulo-interstitial fibrosis. In addition, TLRs, mainly the TLR2 and TLR4, by TLR2- dependent and TGF-β-dependent conduits, may modulate ECM expression and generate a fibrogenic response. Small GTPase like Rho, Ras and Rab family by targeting relevant genes may also influence the accumulation of ECM proteins and renal fibrosis in hyperglycemic states. This review summarizes the recent information about the role and mechanisms by which these molecules and signaling pathways regulate ECM synthesis and its expression in high glucose ambience in vitro and in vivo states. The understanding of such signaling pathways and the molecules that influence expression, secretion and amassing of ECM may aid in developing strategies for the amelioration of diabetic nephropathy.
Collapse
Affiliation(s)
| | - L Sun
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
170
|
Topical delivery of a Rho-kinase inhibitor to the cornea via mucoadhesive film. Eur J Pharm Sci 2016; 91:256-64. [PMID: 27196964 DOI: 10.1016/j.ejps.2016.05.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 04/29/2016] [Accepted: 05/14/2016] [Indexed: 01/21/2023]
Abstract
The application of inhibitors of the Rho kinase pathway (ROCK inhibitors) to the surface of the eye in the form of eyedrops has beneficial effects which aid the recovery of diseased or injured endothelial cells that line the inner surface of the cornea. The aim of this study was to test the plausibility of delivering a selective ROCK inhibitor, Y-27632, to the cornea using a thin polymeric film. Mucoadhesive polymeric thin films were prepared incorporating Y-27632 and diffusional release into PBS was determined. Topical ocular delivery from the applied film was investigated using freshly excised porcine eyes and eyedrops of equivalent concentration acted as comparators; after 24h the formulations were removed and the corneas extracted. Drug-loaded thin polymeric films, with high clarity and pliability were produced. ROCK inhibitor Y-27632 was weakly retained within the film, with release attaining equilibrium after 1h. This in turn facilitated its rapid ocular delivery, and an approximately three-fold greater penetration of Y-27632 into cryoprobe-treated corneas was observed from the thin film (p<0.01) compared to eyedrops. These findings support the further development of ROCK inhibitor delivery to the cornea via release from thin mucoadhesive films to treat vision loss cause by corneal endothelial dysfunction.
Collapse
|
171
|
Skaria T, Bachli E, Schoedon G. Wnt5A/Ryk signaling critically affects barrier function in human vascular endothelial cells. Cell Adh Migr 2016; 11:24-38. [PMID: 27159116 DOI: 10.1080/19336918.2016.1178449] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Satisfactory therapeutic strategies for septic shock are still missing. Previously we found elevated levels of Wnt5A in patients with severe sepsis and septic shock. Wnt5A is released by activated macrophages but knowledge of its effects in the vascular system remains scant. Here we investigate the response of human coronary artery endothelial cells (HCAEC) to Wnt5A. We used a genome-wide differential expression approach to define novel targets regulated by Wnt5A. Gene ontology analysis of expression profiles revealed clusters of genes involved in actin cytoskeleton remodeling as the predominant targets of Wnt5A. Wnt5A targeted Rho-associated protein serine/threonine kinase (ROCK), leading to phosphorylation of LIM kinase-2 (LIMK2) and inactivation of the actin depolymerization factor cofilin-1 (CFL1). Functional experiments recording cytoskeletal rearrangements in living cells showed that Wnt5A enhanced stress fiber formation as a consequence of reduced actin depolymerization. The antagonist Wnt inhibitory factor 1 (WIF1) that specifically interferes with the WIF domain of Ryk receptors prevented actin polymerization. Wnt5A disrupted β-catenin and VE-cadherin adherens junctions forming inter-endothelial gaps. Functional experiments targeting the endothelial monolayer integrity and live recording of trans-endothelial resistance revealed enhanced permeability of Wnt5A-treated HCAEC. Ryk silencing completely prevented Wnt5A-induced endothelial hyperpermeability. Wnt5A decreased wound healing capacity of HCAEC monolayers; this was restored by the ROCK inhibitor Y-27632. Here we show that Wnt5A acts on the vascular endothelium causing enhanced permeability through Ryk interaction and downstream ROCK/LIMK2/CFL1 signaling. Wnt5A/Ryk signaling might provide novel therapeutic strategies to prevent capillary leakage in systemic inflammation and septic shock.
Collapse
Affiliation(s)
- Tom Skaria
- a Inflammation Research Unit, Division of Internal Medicine, University Hospital Zürich , Zürich , Switzerland
| | - Esther Bachli
- b Department of Medicine , Uster Hospital , Uster , Switzerland
| | - Gabriele Schoedon
- a Inflammation Research Unit, Division of Internal Medicine, University Hospital Zürich , Zürich , Switzerland
| |
Collapse
|
172
|
Phosphodiesterase inhibitor KMUP-3 displays cardioprotection via protein kinase G and increases cardiac output via G-protein-coupled receptor agonist activity and Ca(2+) sensitization. Kaohsiung J Med Sci 2016; 32:55-67. [PMID: 26944323 DOI: 10.1016/j.kjms.2016.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 01/08/2016] [Accepted: 01/11/2016] [Indexed: 11/20/2022] Open
Abstract
KMUP-3 (7-{2-[4-(4-nitrobenzene) piperazinyl]ethyl}-1, 3-dimethylxanthine) displays cardioprotection and increases cardiac output, and is suggested to increase cardiac performance and improve myocardial infarction. To determine whether KMUP-3 improves outcomes in hypoperfused myocardium by inducing Ca(2+) sensitization to oppose protein kinase (PK)G-mediated Ca(2+) blockade, we measured left ventricular systolic blood pressure, maximal rates of pressure development, mean arterial pressure and heart rate in rats, and measured contractility and expression of PKs/RhoA/Rho kinase (ROCK)II in beating guinea pig left atria. Hemodynamic changes induced by KMUP-3 (0.5-3.0 mg/kg, intravenously) were inhibited by Y27632 [(R)-(+)-trans-4-1-aminoethyl)-N-(4-Pyridyl) cyclohexane carboxamide] and ketanserin (1 mg/kg, intravenously). In electrically stimulated left guinea pig atria, positive inotropy induced by KMUP-3 (0.1-100μM) was inhibited by the endothelial NO synthase (eNOS) inhibitors N-nitro-l-arginine methyl ester (L-NAME) and 7-nitroindazole, cyclic AMP antagonist SQ22536 [9-(terahydro-2-furanyl)-9H-purin-6-amine], soluble guanylyl cyclase (sGC) antagonist ODQ (1H-[1,2,4] oxadiazolo[4,3-a] quinoxalin-1-one), RhoA inhibitor C3 exoenzyme, β-blocker propranolol, 5-hydroxytryptamine 2A antagonist ketanserin, ROCK inhibitor Y27632 and KMUP-1 (7-{2-[4-(2-chlorobenzene) piperazinyl]ethyl}-1, 3-dimethylxanthine) at 10μM. Western blotting assays indicated that KMUP-3 (0.1-10μM) increased PKA, RhoA/ROCKII, and PKC translocation and CIP-17 (an endogenous 17-kDa inhibitory protein) activation. In spontaneous right atria, KMUP-3 induced negative chronotropy that was blunted by 7-nitroindazole and atropine. In neonatal myocytes, L-NAME inhibited KMUP-3-induced eNOS phosphorylation and RhoA/ROCK activation. In H9c2 cells, Y-27632 (50μM) and PKG antagonist KT5823 [2,3,9,10,11,12-hexahydro-10R- methoxy-2,9-dimethyl-1-oxo-9S,12R-epoxy-1H-diindolo(1,2,3-fg:3',2',1'-kl) pyrrolo(3,4-i)(1,6)benzodiazocine-10-carboxylic acid, methyl ester] (3μM) reversed KMUP-3 (1-100μM)-induced Ca(2+)-entry blockade. GPCR agonist activity of KMUP-3 appeared opposed to KMUP-1, and increased cardiac output via Ca(2+) sensitization, and displayed cardioprotection via cyclic GMP/PKG-mediated myocardial preconditioning in animal studies.
Collapse
|
173
|
Bai X, Dee R, Mangum KD, Mack CP, Taylor JM. RhoA signaling and blood pressure: The consequence of failing to “Tone it Down”. World J Hypertens 2016; 6:18-35. [DOI: 10.5494/wjh.v6.i1.18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/24/2015] [Accepted: 01/22/2016] [Indexed: 02/06/2023] Open
Abstract
Uncontrolled high blood pressure is a major risk factor for heart attack, stroke, and kidney failure and contributes to an estimated 25% of deaths worldwide. Despite numerous treatment options, estimates project that reasonable blood pressure (BP) control is achieved in only about half of hypertensive patients. Improvements in the detection and management of hypertension will undoubtedly be accomplished through a better understanding of the complex etiology of this disease and a more comprehensive inventory of the genes and genetic variants that influence BP regulation. Recent studies (primarily in pre-clinical models) indicate that the small GTPase RhoA and its downstream target, Rho kinase, play an important role in regulating BP homeostasis. Herein, we summarize the underlying mechanisms and highlight signaling pathways and regulators that impart tight spatial-temporal control of RhoA activity. We also discuss known allelic variations in the RhoA pathway and consider how these polymorphisms may affect genetic risk for hypertension and its clinical manifestations. Finally, we summarize the current (albeit limited) clinical data on the efficacy of targeting the RhoA pathway in hypertensive patients.
Collapse
|
174
|
Zhu L, Yang Y, Lu X. The selectivity and promiscuity of brain-neuroregenerative inhibitors between ROCK1 and ROCK2 isoforms: An integration of SB-QSSR modelling, QM/MM analysis and in vitro kinase assay. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2016; 27:47-65. [PMID: 26854727 DOI: 10.1080/1062936x.2015.1132765] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The Rho-associated kinases (ROCKs) have long been recognized as an attractive therapeutic target for various neurological diseases; selective inhibition of ROCK1 and ROCK2 isoforms would result in distinct biological effects on neurogenesis, neuroplasticity and neuroregeneration after brain surgery and traumatic brain injury. However, the discovery and design of isoform-selective inhibitors remain a great challenge due to the high conservation and similarity between the kinase domains of ROCK1 and ROCK2. Here, a structure-based quantitative structure-selectivity relationship (SB-QSSR) approach was used to correlate experimentally measured selectivity with the difference in inhibitor binding to the two kinase isoforms. The resulting regression models were examined rigorously through both internal cross-validation and external blind validation; a nonlinear predictor was found to have high fitting stability and strong generalization ability, which was then employed to perform virtual screening against a structurally diverse, drug-like compound library. Consequently, five and seven hits were identified as promising candidates of 1-o-2 and 2-o-1 selective inhibitors, respectively, from which seven purchasable compounds were tested in vitro using a standard kinase assay protocol to determine their inhibitory activity against and selectivity between ROCK1 and ROCK2. The structural basis, energetic property and biological implication underlying inhibitor selectivity and promiscuity were also investigated systematically using a hybrid quantum mechanics/molecular mechanics (QM/MM) scheme.
Collapse
Affiliation(s)
- L Zhu
- a Department of Neurosurgery , People's Hospital affiliated to Jiangsu University , Zhenjiang , China
| | - Y Yang
- a Department of Neurosurgery , People's Hospital affiliated to Jiangsu University , Zhenjiang , China
| | - X Lu
- a Department of Neurosurgery , People's Hospital affiliated to Jiangsu University , Zhenjiang , China
| |
Collapse
|
175
|
Change in vascular smooth muscle response to 5-HT due to short- or long-term endothelial denudation of the bovine digital vein. Vet J 2016; 207:154-159. [DOI: 10.1016/j.tvjl.2015.10.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 09/27/2015] [Accepted: 10/04/2015] [Indexed: 11/19/2022]
|
176
|
Yamanaka O, Kitano-Izutani A, Tomoyose K, Reinach PS. Pathobiology of wound healing after glaucoma filtration surgery. BMC Ophthalmol 2015; 15 Suppl 1:157. [PMID: 26818010 PMCID: PMC4895697 DOI: 10.1186/s12886-015-0134-8] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Conjunctival and subconjunctival fibrogenesis and inflammation are sight compromising side effects that can occur subsequent to glaucoma filtration surgery. Despite initial declines in intraocular pressure resulting from increasing aqueous outflow, one of the activated responses includes marshalling of proinflammatory and pro-fibrogenic cytokine mediator entrance into the aqueous through a sclerostomy window and their release by local cells, as well as infiltrating activated immune cells. These changes induce dysregulated inflammation, edema and extracellular matrix remodeling, which occlude outflow facility. A number of therapeutic approaches are being taken to offset declines in outflow facility since the current procedure of inhibiting fibrosis with either mitomycin C (MMC) or 5-fluorouracil (5-FU) injection is nonselective. One of them entails developing a new strategy for reducing fibrosis induced by wound healing responses including myofibroblast transdifferentiation and extracellular matrix remodeling in tissue surrounding surgically created shunts. The success of this endeavor is predicated on having a good understanding of conjunctival wound healing pathobiology. In this review, we discuss the roles of inappropriately activated growth factor and cytokine receptor linked signaling cascades inducing conjunctival fibrosis/scarring during post-glaucoma surgery wound healing. Such insight may identify drug targets for blocking fibrogenic signaling and excessive fibrosis which reduces rises in outflow facility resulting from glaucoma filtration surgery.
Collapse
Affiliation(s)
- Osamu Yamanaka
- Department of Ophthalmology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-0012, Japan.
| | - Ai Kitano-Izutani
- Department of Ophthalmology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-0012, Japan.
| | - Katsuo Tomoyose
- Department of Ophthalmology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-0012, Japan.
| | - Peter S Reinach
- Departments of Ophthalmology and Optometry Wenzhou Medical University, Wenzhou, 325027, People's Republic of China.
| |
Collapse
|
177
|
Wang S, Chen C, Su K, Zha D, Liang W, Hillebrands JL, Goor HV, Ding G. Angiotensin II induces reorganization of the actin cytoskeleton and myosin light-chain phosphorylation in podocytes through rho/ROCK-signaling pathway. Ren Fail 2015; 38:268-75. [PMID: 26652313 DOI: 10.3109/0886022x.2015.1117896] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
AIMS In the present study, we have evaluated the effect of angiotensin II (Ang II) on actin cytoskeleton reorganization and myosin light-chain (MLC) phosphorylation in podocytes to demonstrate whether the Rho/Rho-associated coiled kinase (ROCK) pathway is involved podocyte injury. METHODS Eighteen male Sprague-Dawley rats were divided into three groups and treated with Ang II, saline or telmisartan. Morphological changes were studied at 28 days after treatment. Immunohistochemistry and Western blotting were used to determine the renal expression of p-MLC and ROCK2. Cultured podocytes were treated with Ang II (10(-7 )M) with or without Rho-kinase inhibitor (Y27632, 10(-6 )M) for variable time periods. F-actin was visualized with fluorescein isothiocyanate (FITC)-conjugated phalloidin or tetraethyl rhodamine isothiocyanate (TRITC)-conjugated phalloidin. p-MLC expression was evaluated by immunofluorescence and Western blot. The activation of Rho/ROCK was evaluated by Western blot. RESULTS The expression of p-MLC in glomeruli increased significantly in rats treated with Ang II when compared to the control rats as shown by Western blot (p < 0.05). In cultured podocytes, Rho A and ROCK2 increased after incubation with Ang II. Ang II increased the expression of ROCK2, which was accompanied with altered morphology, redistribution of actin and increased phosphorylation of MLC. The distribution of actin changed to a large extent, although overall quantitative differences were not observed. Addition of Y-27632 to podocytes treated with Ang II could ameliorate F-actin cytoskeleton remodeling and the increment in p-MLC expression. CONCLUSION Ang II-induced podocyte cytoskeleton protein expression changing through the RhoA/ROCK2 p-MLC/F-actin pathway.
Collapse
Affiliation(s)
- Siyuan Wang
- a Department of Nephrology , Renmin Hospital of Wuhan University , Wuhan , Hubei , China.,b Department of General , Tongji Medical College, Huazhong University of Science & Technology, The Central Hospital of Wuhan , Wuhan , Hubei , China
| | - Cheng Chen
- a Department of Nephrology , Renmin Hospital of Wuhan University , Wuhan , Hubei , China.,c Department of Pathology and Medical Biology , University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| | - Ke Su
- a Department of Nephrology , Renmin Hospital of Wuhan University , Wuhan , Hubei , China
| | - Dongqing Zha
- a Department of Nephrology , Renmin Hospital of Wuhan University , Wuhan , Hubei , China
| | - Wei Liang
- a Department of Nephrology , Renmin Hospital of Wuhan University , Wuhan , Hubei , China
| | - J L Hillebrands
- c Department of Pathology and Medical Biology , University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| | - Harry van Goor
- c Department of Pathology and Medical Biology , University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| | - Guohua Ding
- a Department of Nephrology , Renmin Hospital of Wuhan University , Wuhan , Hubei , China
| |
Collapse
|
178
|
Agarwal A, Parriott J, Demirel S, Argo C, Sepah YJ, Do DV, Nguyen QD. Nonbiological pharmacotherapies for the treatment of diabetic macular edema. Expert Opin Pharmacother 2015; 16:2625-35. [PMID: 26523670 DOI: 10.1517/14656566.2015.1100172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION During the past decade, there have been significant advances in the pharmacotherapies for the treatment of diabetic macular edema (DME). Among the presently available treatment options, anti-vascular endothelial growth factors (anti-VEGF) agents are the most favored agents due to their efficacy and safety. The index review focuses on nonbiological therapies that have entered in phase 3 clinical trials for DME. AREAS COVERED An extensive review of the literature was performed to identify various nonbiological immunotherapies i.e., drugs other than '-mAbs' (monoclonal antibodies including anti-VEGF agents), '-mibs' (proteasome inhibitors), '-NAbs' (nanoparticle albumin-bound), and '-nibs' (small molecule inhibitor/tyrosine kinase inhibitors), among others. Extended-release low-dose corticosteroid devices have been recently approved for the treatment of DME. Other compounds such as non-steroidal anti-inflammatory drugs, antibody mimetic proteins, nonbiological growth factor inhibitors, and inhibitors of protein kinase C have been described. EXPERT OPINION A number of therapies are under development for the pharmacological management of DME. Due to the rising healthcare costs associated with anti-VEGF agents, a number of alternate treatment options have been explored recently. Some of these agents have reached phase 3 in clinical trials and appear to have a promising role in the management of DME. As further research is conducted, the role of each individual agent will become more defined, alone or in combination therapy.
Collapse
Affiliation(s)
- Aniruddha Agarwal
- a Ocular Imaging Research and Reading Center (OIRRC) , Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center , Omaha , NE , USA
| | - Jacob Parriott
- b College of Medicine , University of Nebraska , Omaha , NE , USA
| | - Sibel Demirel
- a Ocular Imaging Research and Reading Center (OIRRC) , Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center , Omaha , NE , USA.,c Department of Ophthalmology , Ankara University Faculty of Medicine , Ankara , Turkey
| | - Colby Argo
- b College of Medicine , University of Nebraska , Omaha , NE , USA
| | - Yasir Jamal Sepah
- a Ocular Imaging Research and Reading Center (OIRRC) , Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center , Omaha , NE , USA
| | - Diana V Do
- a Ocular Imaging Research and Reading Center (OIRRC) , Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center , Omaha , NE , USA
| | - Quan Dong Nguyen
- a Ocular Imaging Research and Reading Center (OIRRC) , Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center , Omaha , NE , USA
| |
Collapse
|
179
|
Lee W, Miyagawa Y, Long C, Zhang M, Cooper DKC, Hara H. Effect of Rho-kinase Inhibitor, Y27632, on Porcine Corneal Endothelial Cell Culture, Inflammation and Immune Regulation. Ocul Immunol Inflamm 2015; 24:579-93. [PMID: 26471144 DOI: 10.3109/09273948.2015.1056534] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE To investigate the effect of the Rho-kinase inhibitor, Y27632, on pig corneal endothelial cell (pCEC) culture, and on inflammation and immune regulation of the responses of human cells to pCECs. METHODS pCECs were cultured with/without Y27632 to assess cell proliferation and in vitro wound healing assay. The level of MCP-1 and VEGF in pCECs stimulated with human TNF-α were measured. Proliferation of human PBMCs stimulated with pCECs, and cytokine production in human T cells, and monocyte migration after stimulation were investigated. RESULTS Y27632 promoted pCEC proliferation, prevented pCEC death, and enhanced in vitro wound healing. After stimulation, there were significantly lower levels of MCP-1 and VEGF measured in pCECs cultured with Y27632, and significantly reduced human PBMC proliferation, cytokine production, and monocyte migration. CONCLUSIONS The application of the Rho-kinase inhibitor will be beneficial when culturing pCECs, and may provide a novel therapy to reduce inflammation after corneal xenotransplantation.
Collapse
Affiliation(s)
- Whayoung Lee
- a Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh , Pittsburgh , Pennsylvania , USA
| | - Yuko Miyagawa
- a Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh , Pittsburgh , Pennsylvania , USA
| | - Cassandra Long
- a Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh , Pittsburgh , Pennsylvania , USA
| | - Matthew Zhang
- a Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh , Pittsburgh , Pennsylvania , USA
| | - David K C Cooper
- a Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh , Pittsburgh , Pennsylvania , USA
| | - Hidetaka Hara
- a Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh , Pittsburgh , Pennsylvania , USA
| |
Collapse
|
180
|
Schumacher FR, Siew K, Zhang J, Johnson C, Wood N, Cleary SE, Al Maskari RS, Ferryman JT, Hardege I, Yasmin, Figg NL, Enchev R, Knebel A, O'Shaughnessy KM, Kurz T. Characterisation of the Cullin-3 mutation that causes a severe form of familial hypertension and hyperkalaemia. EMBO Mol Med 2015; 7:1285-1306. [PMID: 26286618 PMCID: PMC4604684 DOI: 10.15252/emmm.201505444] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 07/17/2015] [Accepted: 07/21/2015] [Indexed: 02/05/2023] Open
Abstract
Deletion of exon 9 from Cullin-3 (CUL3, residues 403-459: CUL3(Δ403-459)) causes pseudohypoaldosteronism type IIE (PHA2E), a severe form of familial hyperkalaemia and hypertension (FHHt). CUL3 binds the RING protein RBX1 and various substrate adaptors to form Cullin-RING-ubiquitin-ligase complexes. Bound to KLHL3, CUL3-RBX1 ubiquitylates WNK kinases, promoting their ubiquitin-mediated proteasomal degradation. Since WNK kinases activate Na/Cl co-transporters to promote salt retention, CUL3 regulates blood pressure. Mutations in both KLHL3 and WNK kinases cause PHA2 by disrupting Cullin-RING-ligase formation. We report here that the PHA2E mutant, CUL3(Δ403-459), is severely compromised in its ability to ubiquitylate WNKs, possibly due to altered structural flexibility. Instead, CUL3(Δ403-459) auto-ubiquitylates and loses interaction with two important Cullin regulators: the COP9-signalosome and CAND1. A novel knock-in mouse model of CUL3(WT) (/Δ403-459) closely recapitulates the human PHA2E phenotype. These mice also show changes in the arterial pulse waveform, suggesting a vascular contribution to their hypertension not reported in previous FHHt models. These findings may explain the severity of the FHHt phenotype caused by CUL3 mutations compared to those reported in KLHL3 or WNK kinases.
Collapse
Affiliation(s)
- Frances-Rose Schumacher
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee, UK
| | - Keith Siew
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK
| | - Jinwei Zhang
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee, UK
| | - Clare Johnson
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee, UK
| | - Nicola Wood
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee, UK
| | - Sarah E Cleary
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK
| | - Raya S Al Maskari
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK
| | - James T Ferryman
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK
| | - Iris Hardege
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK
| | - Yasmin
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK
| | - Nichola L Figg
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Axel Knebel
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee, UK
| | - Kevin M O'Shaughnessy
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK
| | - Thimo Kurz
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee, UK
| |
Collapse
|
181
|
Zhang X, Zhang T, Gao F, Li Q, Shen C, Li Y, Li W, Zhang X. Fasudil, a Rho‑kinase inhibitor, prevents intima‑media thickening in a partially ligated carotid artery mouse model: Effects of fasudil in flow‑induced vascular remodeling. Mol Med Rep 2015; 12:7317-25. [PMID: 26458725 PMCID: PMC4626179 DOI: 10.3892/mmr.2015.4409] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 05/19/2015] [Indexed: 11/06/2022] Open
Abstract
Vascular remodeling in response to hemodynamic alterations is a physiological process that requires coordinated signaling between endothelial, inflammatory and vascular smooth muscle cells (VSMCs). Extensive experimental and clinical studies have indicated the critical role of the Ras homolog gene family, member A/Rho‑associated kinase (ROCK) signaling pathway in the pathogenesis of cardiovascular disease, where ROCK activation has been demonstrated to promote inflammation and remodeling through inducing the expression of proinflammatory cytokines and adhesion molecules in endothelial cells and VSMCs. However, the role of ROCK in flow‑induced vascular remodeling has not been fully defined. The current study aimed to investigate the effect of the ROCK signaling pathway in flow‑induced vascular remodeling by comparing the responses to partial carotid artery ligation in mice treated with fasudil (a ROCK inhibitor) and untreated mice. Intima‑media thickness and neointima formation were evaluated by morphology. VSMC proliferation and inflammation of the vessel wall were assessed by immunohistochemistry. In addition, the expression levels of ROCK and the downstream effectors of ROCK, myosin light chain (MLC) and phosphorylated‑MLC (p‑MLC), were quantified by western blot analysis. Following a reduction in blood flow, ROCK1 and p‑MLC expression increased in the untreated left common carotid arteries (LCA). Fasudil‑treated mice developed a significantly smaller intima‑media thickness compared with the untreated mice. Quantitative immunohistochemistry of the fasudil‑treated LCA indicated that there was a reduction in proliferation when compared with untreated vessels. There were fewer CD45+ cells observed in the fasudil‑treated LCA compared with the untreated LCA. In conclusion, the expression of ROCK was enhanced in flow‑induced carotid artery remodeling and ROCK inhibition as a result of fasudil treatment may attenuate flow‑induced carotid artery remodeling.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of Vascular Surgery, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Tao Zhang
- Department of Vascular Surgery, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Fu Gao
- Department of Vascular Surgery, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Qingle Li
- Department of Vascular Surgery, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Chenyang Shen
- Department of Vascular Surgery, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Yankui Li
- Department of General Surgery, The Second Affiliated Hospital, Tianjin Medical University, Tianjin 300000, P.R. China
| | - Wei Li
- Department of Vascular Surgery, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Xiaoming Zhang
- Department of Vascular Surgery, Peking University People's Hospital, Beijing 100044, P.R. China
| |
Collapse
|
182
|
Van de Velde S, De Groef L, Stalmans I, Moons L, Van Hove I. Towards axonal regeneration and neuroprotection in glaucoma: Rho kinase inhibitors as promising therapeutics. Prog Neurobiol 2015; 131:105-19. [PMID: 26093354 DOI: 10.1016/j.pneurobio.2015.06.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 06/08/2015] [Accepted: 06/09/2015] [Indexed: 11/27/2022]
Abstract
Due to a prolonged life expectancy worldwide, the incidence of age-related neurodegenerative disorders such as glaucoma is increasing. Glaucoma is the second cause of blindness, resulting from a slow and progressive loss of retinal ganglion cells (RGCs) and their axons. Up to now, intraocular pressure (IOP) reduction is the only treatment modality by which ophthalmologists attempt to control disease progression. However, not all patients benefit from this therapy, and the pathophysiology of glaucoma is not always associated with an elevated IOP. These limitations, together with the multifactorial etiology of glaucoma, urge the pressing medical need for novel and alternative treatment strategies. Such new therapies should focus on preventing or retarding RGC death, but also on repair of injured axons, to ultimately preserve or improve structural and functional connectivity. In this respect, Rho-associated coiled-coil forming protein kinase (ROCK) inhibitors hold a promising potential to become very prominent drugs for future glaucoma treatment. Their field of action in the eye does not seem to be restricted to IOP reduction by targeting the trabecular meshwork or improving filtration surgery outcome. Indeed, over the past years, important progress has been made in elucidating their ability to improve ocular blood flow, to prevent RGC death/increase RGC survival and to retard axonal degeneration or induce proper axonal regeneration. Within this review, we aim to highlight the currently known capacity of ROCK inhibition to promote neuroprotection and regeneration in several in vitro, ex vivo and in vivo experimental glaucoma models.
Collapse
Affiliation(s)
- Sarah Van de Velde
- Laboratory of Ophthalmology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Lies De Groef
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Leuven, Belgium
| | - Ingeborg Stalmans
- Laboratory of Ophthalmology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Leuven, Belgium.
| | - Inge Van Hove
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Leuven, Belgium
| |
Collapse
|
183
|
Van de Velde S, Van Bergen T, Vandewalle E, Kindt N, Castermans K, Moons L, Stalmans I. Rho kinase inhibitor AMA0526 improves surgical outcome in a rabbit model of glaucoma filtration surgery. PROGRESS IN BRAIN RESEARCH 2015; 220:283-97. [PMID: 26497796 DOI: 10.1016/bs.pbr.2015.04.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
PURPOSE First, to elucidate the effect of Rho kinase inhibitor, AMA0526, on Human Tenon Fibroblast (HTF) proliferation and transdifferentiation to myofibroblasts. Second, the effects of ROCK inhibition on the wound healing process and surgical outcome were investigated in a rabbit model of glaucoma filtration surgery. METHODS After exposure of HTF to AMA0526 (0.1-25 μM), a water-soluble tetrazolium salt-1 assay and caspase 3/7 activity assay were used to assess its effect on cell proliferation and to elucidate any toxic effects, respectively. Immunohistochemistry of α-smooth muscle actin expression was used to investigate fibroblast-to-myofibroblast differentiation induced by transforming growth factor-beta 1 (TGF-β1) in the presence or absence of the ROCK inhibitor. The effect of topical treatment was studied in a rabbit model of glaucoma filtration surgery. Treatment outcome was studied by performing intraocular pressure (IOP) measurements and clinical investigation of the bleb area and survival. Immunohistological analysis for inflammation (CD45), angiogenesis (CD31), and collagen I was performed at day 8, 14, and 30 after surgery (n=5/time point). Separate control groups treated with vehicle were used as control. RESULTS In vitro results showed that AMA0526 dose dependently inhibited proliferation of HTF (P<0.05) without the induction of caspase 3/7 activity. Incubation of HTF with the AMA0526 inhibited TGF-β1 induced fibroblast-to-myofibroblast differentiation. In the rabbit model, topical treatment significantly improved surgical outcome. Compared to vehicle-treated eyes, AMA0526 resulted in increased bleb area (P<0.0001) and prolonged survival (P=0.0025). IOP remained significantly lower throughout the course of the experiment in the AMA0526 group (P<0.0001). Histological evaluation revealed that blebs treated with the ROCK inhibitor were characterized by reduced inflammation, angiogenesis, and collagen deposition at the site of filtration surgery (P<0.05). CONCLUSIONS AMA0526 had profound effects on HTF proliferation and myofibroblast transition and improved glaucoma filtration surgery outcome by interfering at different levels of the wound healing process. Therefore, these data indicate that ROCK inhibitors may be considered as more physiological agents which specifically target the wound healing process to improve the outcome of glaucoma surgery.
Collapse
Affiliation(s)
| | | | - Evelien Vandewalle
- Laboratory of Ophthalmology, KU Leuven, Leuven, Belgium; Department of Ophthalmology, University Hospitals Leuven (UZ Leuven), Leuven, Belgium
| | - Nele Kindt
- Amakem Therapeutics, Diepenbeek, Belgium
| | | | - Lieve Moons
- Research Group of Neural Circuit Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Ingeborg Stalmans
- Laboratory of Ophthalmology, KU Leuven, Leuven, Belgium; Department of Ophthalmology, University Hospitals Leuven (UZ Leuven), Leuven, Belgium.
| |
Collapse
|
184
|
Van de Velde S, Van Bergen T, Vandewalle E, Moons L, Stalmans I. Modulation of wound healing in glaucoma surgery. PROGRESS IN BRAIN RESEARCH 2015; 221:319-40. [PMID: 26518085 DOI: 10.1016/bs.pbr.2015.05.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Glaucoma is a neurodegenerative disease and is the second most important cause of irreversible blindness. Filtration surgery remains the most effective therapy to reduce intraocular pressure in glaucoma patients. The main determinant of long-term surgical success is the healing response. Excessive postoperative wound healing with subsequent fibrosis may lead to obstruction of the created channel which frequently results in early surgical failure and consequent progression of visual field loss. Preoperative use of antimitotics, such as mitomycin-C and 5-fluorouracyl, effectively improves surgery outcome. However, the use of these nonspecific antiproliferative agents can be associated with severe side effects. This review provides an overview of the most important efforts that have been made to explore novel, more specific, and safer agents to prevent glaucoma filtration failure and improve surgery outcome.
Collapse
Affiliation(s)
- Sarah Van de Velde
- Department of Neurosciences, Laboratory of Ophthalmology, KU Leuven, Leuven, Belgium
| | - Tine Van Bergen
- Department of Neurosciences, Laboratory of Ophthalmology, KU Leuven, Leuven, Belgium
| | - Evelien Vandewalle
- Department of Neurosciences, Laboratory of Ophthalmology, KU Leuven, Leuven, Belgium; Department of Ophthalmology, University Hospitals Leuven (UZ Leuven), Leuven, Belgium
| | - Lieve Moons
- Research Group of Neural Circuit Development and Regeneration, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Leuven, Belgium
| | - Ingeborg Stalmans
- Department of Neurosciences, Laboratory of Ophthalmology, KU Leuven, Leuven, Belgium; Department of Ophthalmology, University Hospitals Leuven (UZ Leuven), Leuven, Belgium.
| |
Collapse
|
185
|
Li YH, Yu JZ, Xin YL, Feng L, Chai Z, Liu JC, Zhang HZ, Zhang GX, Xiao BG, Ma CG. Protective effect of a novel Rho kinase inhibitor WAR-5 in experimental autoimmune encephalomyelitis by modulating inflammatory response and neurotrophic factors. Exp Mol Pathol 2015; 99:220-8. [PMID: 26112093 DOI: 10.1016/j.yexmp.2015.06.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 06/19/2015] [Indexed: 11/19/2022]
Abstract
The Rho-kinase (ROCK) inhibitor Fasudil has proven beneficial in experimental autoimmune encephalomyelitis (EAE). Given the small safety window of Fasudil, we are looking for novel ROCK inhibitors, which have similar or stronger effect on EAE with greater safety. In this study, we report that WAR-5, a Y-27632 derivative, alleviates the clinical symptoms, attenuates myelin damage and reduces CNS inflammatory responses in EAE C57BL/6 mice at an extent similar to Fasudil, while exhibits less vasodilator and adverse reaction in vivo. WAR-5 inhibits ROCK activity, and selectively suppresses the expression of ROCK II in spleen, brain and spinal cord of EAE mice, especially in spinal cord, accompanied by decreased expression of Nogo. WAR-5 also regulates the imbalance of Th1/Th17 T cells and regulatory T cells, inhibits inflammatory microenvironment induced with NF-κB-IL-1β pathway. Importantly, WAR-5 converts M1 toward M2 microglia/macrophages that are positively correlated with BDNF and NT-3 production. Taken together, WAR-5 exhibits therapeutic potential in EAE by more selectively inhibits ROCK II, with a greater safety than Fasudil, and is worthy of further clinical study to clarify its clinical value.
Collapse
MESH Headings
- 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives
- 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology
- Amides/pharmacology
- Aminopyridines/pharmacology
- Animals
- Cytokines/metabolism
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/prevention & control
- Enzyme-Linked Immunosorbent Assay
- Female
- Immunoblotting
- Immunoenzyme Techniques
- Inflammation/drug therapy
- Inflammation/immunology
- Inflammation/metabolism
- Macrophages/drug effects
- Macrophages/immunology
- Macrophages/metabolism
- Mice
- Mice, Inbred C57BL
- Microglia/drug effects
- Microglia/immunology
- Microglia/metabolism
- Nerve Growth Factors/metabolism
- Piperidines/pharmacology
- Protein Kinase Inhibitors/pharmacology
- Pyridines/pharmacology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- rho-Associated Kinases/antagonists & inhibitors
- rho-Associated Kinases/metabolism
Collapse
Affiliation(s)
- Yan-hua Li
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong 037009, China
| | - Jie-zhong Yu
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong 037009, China
| | - Yan-le Xin
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong 037009, China
| | - Ling Feng
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong 037009, China
| | - Zhi Chai
- "2011" Collaborative Innovation Center/Research Center of Neurobiology, Shanxi University of Traditional Chinese Medicine, Taiyuan 030619, China
| | - Jian-chun Liu
- "2011" Collaborative Innovation Center/Research Center of Neurobiology, Shanxi University of Traditional Chinese Medicine, Taiyuan 030619, China
| | - Hong-zhen Zhang
- "2011" Collaborative Innovation Center/Research Center of Neurobiology, Shanxi University of Traditional Chinese Medicine, Taiyuan 030619, China
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Bao-guo Xiao
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong 037009, China; Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200025, China.
| | - Cun-gen Ma
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong 037009, China; "2011" Collaborative Innovation Center/Research Center of Neurobiology, Shanxi University of Traditional Chinese Medicine, Taiyuan 030619, China.
| |
Collapse
|
186
|
Green J, Cao J, Bandarage UK, Gao H, Court J, Marhefka C, Jacobs M, Taslimi P, Newsome D, Nakayama T, Shah S, Rodems S. Design, Synthesis, and Structure–Activity Relationships of Pyridine-Based Rho Kinase (ROCK) Inhibitors. J Med Chem 2015; 58:5028-37. [DOI: 10.1021/acs.jmedchem.5b00424] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Jeremy Green
- Vertex Pharmaceuticals,
Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Jingrong Cao
- Vertex Pharmaceuticals,
Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Upul K. Bandarage
- Vertex Pharmaceuticals,
Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Huai Gao
- Vertex Pharmaceuticals,
Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - John Court
- Vertex Pharmaceuticals,
Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Craig Marhefka
- Vertex Pharmaceuticals,
Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Marc Jacobs
- Vertex Pharmaceuticals,
Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Paul Taslimi
- Vertex Pharmaceuticals,
Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - David Newsome
- Vertex Pharmaceuticals,
Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Tomoko Nakayama
- Vertex Pharmaceuticals,
Inc., 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Sundeep Shah
- Vertex Pharmaceuticals,
Inc., 11010 Torreyana Road, San Diego, California 92121, United States
| | - Steve Rodems
- Vertex Pharmaceuticals,
Inc., 11010 Torreyana Road, San Diego, California 92121, United States
| |
Collapse
|
187
|
Nazarshodeh E, Shiri F, Ghasemi JB. 3D-QSAR and virtual screening studies in identification of new Rho kinase inhibitors with different scaffolds. JOURNAL OF THE IRANIAN CHEMICAL SOCIETY 2015. [DOI: 10.1007/s13738-015-0669-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
188
|
The regulatory roles of ROCK and MRCK kinases in the plasticity of cancer cell migration. Cancer Lett 2015; 361:185-96. [DOI: 10.1016/j.canlet.2015.03.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 03/09/2015] [Accepted: 03/11/2015] [Indexed: 12/29/2022]
|
189
|
Boland S, Bourin A, Alen J, Geraets J, Schroeders P, Castermans K, Kindt N, Boumans N, Panitti L, Fransen S, Vanormelingen J, Stassen JM, Leysen D, Defert O. Design, Synthesis, and Biological Evaluation of Novel, Highly Active Soft ROCK Inhibitors. J Med Chem 2015; 58:4309-24. [DOI: 10.1021/acs.jmedchem.5b00308] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Sandro Boland
- Amakem Therapeutics, Agoralaan
Abis, 3590 Diepenbeek, Belgium
| | - Arnaud Bourin
- Amakem Therapeutics, Agoralaan
Abis, 3590 Diepenbeek, Belgium
| | - Jo Alen
- Amakem Therapeutics, Agoralaan
Abis, 3590 Diepenbeek, Belgium
| | - Jacques Geraets
- Amakem Therapeutics, Agoralaan
Abis, 3590 Diepenbeek, Belgium
| | | | | | - Nele Kindt
- Amakem Therapeutics, Agoralaan
Abis, 3590 Diepenbeek, Belgium
| | - Nicki Boumans
- Amakem Therapeutics, Agoralaan
Abis, 3590 Diepenbeek, Belgium
| | - Laura Panitti
- Amakem Therapeutics, Agoralaan
Abis, 3590 Diepenbeek, Belgium
| | - Silke Fransen
- Amakem Therapeutics, Agoralaan
Abis, 3590 Diepenbeek, Belgium
| | | | | | - Dirk Leysen
- CSD Farmakem, Elvire Boelensstraat
7, 9160 Lokeren, Belgium
| | - Olivier Defert
- Amakem Therapeutics, Agoralaan
Abis, 3590 Diepenbeek, Belgium
| |
Collapse
|
190
|
RNAi profiling of primary human AML cells identifies ROCK1 as a therapeutic target and nominates fasudil as an antileukemic drug. Blood 2015; 125:3760-8. [PMID: 25931586 DOI: 10.1182/blood-2014-07-590646] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 04/22/2015] [Indexed: 01/07/2023] Open
Abstract
Acute myeloid leukemia (AML) is characterized by a marked genetic heterogeneity, which complicates the development of novel therapeutics. The delineation of pathways essential within an individual patient's mutational background might overcome this limitation and facilitate personalized treatment. We report the results of a large-scale lentiviral loss-of-function RNA interference (RNAi) screen in primary leukemic cells. Stringent validation identified 6 genes (BNIPL1, ROCK1, RPS13, STK3, SNX27, WDHD1) whose knockdown impaired growth and viability of the cells. Dependence on these genes was not caused by mutation or overexpression, and although some of the candidates seemed to be rather patient specific, others were essential in cells isolated from other AML patients. In addition to the phenotype observed after ROCK1 knockdown, treatment with the approved ROCK inhibitor fasudil resulted in increased apoptosis and decreased viability of primary AML cells. In contrast to observations in some other malignancies, ROCK1 inhibition did not foster growth of immature malignant progenitors but was toxic to this cell fraction in feeder coculture and xenotransplant experiments, indicating a distinct effect of ROCK1 inhibition on leukemic progenitors. We conclude that large-scale RNAi screens in primary patient-derived cells are feasible and can complement other methods for personalized cancer therapies, such as expression and mutation profiling.
Collapse
|
191
|
Role of L-type Ca(2+) channels, sarcoplasmic reticulum and Rho kinase in rat basilar artery contractile properties in a new model of subarachnoid hemorrhage. Vascul Pharmacol 2015; 72:64-72. [PMID: 25937251 DOI: 10.1016/j.vph.2015.04.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 03/20/2015] [Accepted: 04/22/2015] [Indexed: 11/22/2022]
Abstract
We have previously described that L-type Ca(2+) channels' (LTCCs) activation and metabotropic Ca(2+) release from the sarcoplasmic reticulum (SR) regulate RhoA/Rho kinase (ROCK) activity and sustained arterial contraction. We have investigated whether this signaling pathway can be altered in a new experimental model of subarachnoid hemorrhage (SAH). For this purpose, arterial reactivity was evaluated on days 1 to 5 after surgery. A significant increase of basal tone, measured 4 and 60min after normalization, was observed on day 5 after SAH and at 60min on days 2 and 3 after SAH. This phenomenon was suppressed with LTCCs and ROCK inhibitors. We have also studied arterial rings vasoreactivity in response to high K(+) solutions. Interestingly, there were no significant differences in the phasic component of the high K(+)-induced contraction between sham and SAH groups, whereas a significant increase in the sustained contraction was observed on day 5 after SAH. This latter component was sensitive to fasudil, and selectively reduced by low nifedipine concentration, and phospholipase C and SR-ATPase inhibitors. Therefore, our data suggest that the metabotropic function of LTCCs is potentiated in SAH. Our results could provide a new strategy to optimize the pharmacological treatment of this pathological process.
Collapse
|
192
|
Mikelis CM, Simaan M, Ando K, Fukuhara S, Sakurai A, Amornphimoltham P, Masedunskas A, Weigert R, Chavakis T, Adams RH, Offermanns S, Mochizuki N, Zheng Y, Gutkind JS. RhoA and ROCK mediate histamine-induced vascular leakage and anaphylactic shock. Nat Commun 2015; 6:6725. [PMID: 25857352 PMCID: PMC4394241 DOI: 10.1038/ncomms7725] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 02/22/2015] [Indexed: 01/08/2023] Open
Abstract
Histamine-induced vascular leakage is an integral component of many highly prevalent human diseases, including allergies, asthma, and anaphylaxis. Yet, how histamine induces the disruption of the endothelial barrier is not well defined. By using genetically modified animal models, pharmacologic inhibitors, and a synthetic biology approach, here we show that the small GTPase RhoA mediates histamine-induced vascular leakage. Histamine causes the rapid formation of focal adherens junctions, disrupting the endothelial barrier by acting on H1R Gαq-coupled receptors, which is blunted in endothelial Gαq/11 KO mice. Interfering with RhoA and ROCK function abolishes endothelial permeability, while phospholipase Cβ plays a limited role. Moreover, endothelial-specific RhoA gene deletion prevents vascular leakage and passive cutaneous anaphylaxis in vivo, and ROCK inhibitors protect from lethal systemic anaphylaxis. This study supports a key role for the RhoA signaling circuitry in vascular permeability, thereby identifying novel pharmacological targets for many human diseases characterized by aberrant vascular leakage.
Collapse
Affiliation(s)
- Constantinos M Mikelis
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - May Simaan
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Koji Ando
- Department of Cell Biology, CREST-JST, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565-8565, Japan
| | - Shigetomo Fukuhara
- Department of Cell Biology, CREST-JST, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565-8565, Japan
| | - Atsuko Sakurai
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Panomwat Amornphimoltham
- Intracellular Membrane Trafficking Unit, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Andrius Masedunskas
- Intracellular Membrane Trafficking Unit, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Roberto Weigert
- Intracellular Membrane Trafficking Unit, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry, Faculty of Medicine, Technische Universität Dresden, Dresden 01307, Germany
| | - Ralf H Adams
- 1] Department of Tissue Morphogenesis, Max-Planck Institute for Molecular Biomedicine, Münster D-48149, Germany [2] Faculty of Medicine, University of Münster, Münster 48149, Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max-Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Naoki Mochizuki
- Department of Cell Biology, CREST-JST, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565-8565, Japan
| | - Yi Zheng
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital, University of Cincinnati College of Medicine, Cincinnati, Ohio 45229, USA
| | - J Silvio Gutkind
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
193
|
Lee J, Park S, Roh S. Y-27632, a ROCK inhibitor, delays senescence of putative murine salivary gland stem cells in culture. Arch Oral Biol 2015; 60:875-82. [PMID: 25804560 DOI: 10.1016/j.archoralbio.2015.03.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 01/16/2015] [Accepted: 03/01/2015] [Indexed: 12/25/2022]
Abstract
OBJECTIVE A loss of functional salivary glands often occurs after radiotherapy for head and neck tumour, and causes many problems in oral health. Recently, the use of salispheres, which consist of salivary gland stem cells (SGSCs), has been suggested as therapy for these problems. However, an insufficient number of cells can be obtained and cultured for cell transplantation. In the present study, salispheres were propagated and passaged by suspension culture to acquire a sufficient number of SGSCs for cell therapy. DESIGN The relationship between sphere formation and the degree of cellular senescence was investigated by analysing senescence-associated β-galactosidase activity and the expression of senescence-related markers such as CDKN2A (p16) and p21. Because the sphere formation potential of SGSCs was decreased and the senescence of the cells was increased after passaging subculture, Y-27632, a Rho-associated kinase inhibitor, was used to treat the passaging subculture to aid the proliferation of the cells in culture. RESULTS The number of spheres was higher in the Y-27632 treatment group than in the control group, and the expression of c-Kit, a proliferation marker, was also increased. In addition, the expression of p16 and p21 proteins tended to be lower in the Y-27632 group. CONCLUSION Y-27632 suppresses the expression of senescence-related proteins and enhances cellular proliferation. This study points to the possibility of scaling-up the therapeutic use of SGSCs, which requires a large amount of cells.
Collapse
Affiliation(s)
- Jingu Lee
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul 110 744, Republic of Korea.
| | - Sangkyu Park
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul 110 744, Republic of Korea.
| | - Sangho Roh
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul 110 744, Republic of Korea.
| |
Collapse
|
194
|
Transgenic plants as low-cost platform for chemotherapeutic drugs screening. Int J Mol Sci 2015; 16:2174-86. [PMID: 25608652 PMCID: PMC4307356 DOI: 10.3390/ijms16012174] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 01/09/2015] [Indexed: 01/18/2023] Open
Abstract
In this work we explored the possibility of using genetically modified Arabidopsis thaliana plants as a rapid and low-cost screening tool for evaluating human anticancer drugs action and efficacy. Here, four different inhibitors with a validated anticancer effect in humans and distinct mechanism of action were screened in the plant model for their ability to interfere with the cytoskeletal and endomembrane networks. We used plants expressing a green fluorescent protein (GFP) tagged microtubule-protein (TUA6-GFP), and three soluble GFPs differently sorted to reside in the endoplasmic reticulum (GFPKDEL) or to accumulate in the vacuole through a COPII dependent (AleuGFP) or independent (GFPChi) mechanism. Our results demonstrated that drugs tested alone or in combination differentially influenced the monitored cellular processes including cytoskeletal organization and endomembrane trafficking. In conclusion, we demonstrated that A. thaliana plants are sensitive to the action of human chemotherapeutics and can be used for preliminary screening of drugs efficacy. The cost-effective subcellular imaging in plant cell may contribute to better clarify drugs subcellular targets and their anticancer effects.
Collapse
|
195
|
Knipe RS, Tager AM, Liao JK. The Rho kinases: critical mediators of multiple profibrotic processes and rational targets for new therapies for pulmonary fibrosis. Pharmacol Rev 2015; 67:103-17. [PMID: 25395505 PMCID: PMC4279074 DOI: 10.1124/pr.114.009381] [Citation(s) in RCA: 163] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by progressive lung scarring, short median survival, and limited therapeutic options, creating great need for new pharmacologic therapies. IPF is thought to result from repetitive environmental injury to the lung epithelium, in the context of aberrant host wound healing responses. Tissue responses to injury fundamentally involve reorganization of the actin cytoskeleton of participating cells, including epithelial cells, fibroblasts, endothelial cells, and macrophages. Actin filament assembly and actomyosin contraction are directed by the Rho-associated coiled-coil forming protein kinase (ROCK) family of serine/threonine kinases (ROCK1 and ROCK2). As would therefore be expected, lung ROCK activation has been demonstrated in humans with IPF and in animal models of this disease. ROCK inhibitors can prevent fibrosis in these models, and more importantly, induce the regression of already established fibrosis. Here we review ROCK structure and function, upstream activators and downstream targets of ROCKs in pulmonary fibrosis, contributions of ROCKs to profibrotic cellular responses to lung injury, ROCK inhibitors and their efficacy in animal models of pulmonary fibrosis, and potential toxicities of ROCK inhibitors in humans, as well as involvement of ROCKs in fibrosis in other organs. As we discuss, ROCK activation is required for multiple profibrotic responses, in the lung and multiple other organs, suggesting ROCK participation in fundamental pathways that contribute to the pathogenesis of a broad array of fibrotic diseases. Multiple lines of evidence therefore indicate that ROCK inhibition has great potential to be a powerful therapeutic tool in the treatment of fibrosis, both in the lung and beyond.
Collapse
Affiliation(s)
- Rachel S Knipe
- Pulmonary and Critical Care Unit and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts (R.S.K., A.M.T.); and Section of Cardiology, Department of Medicine, University of Chicago, Chicago, Illinois (J.K.L.)
| | - Andrew M Tager
- Pulmonary and Critical Care Unit and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts (R.S.K., A.M.T.); and Section of Cardiology, Department of Medicine, University of Chicago, Chicago, Illinois (J.K.L.)
| | - James K Liao
- Pulmonary and Critical Care Unit and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts (R.S.K., A.M.T.); and Section of Cardiology, Department of Medicine, University of Chicago, Chicago, Illinois (J.K.L.)
| |
Collapse
|
196
|
Lee HS, Kim KS, Lim HS, Choi M, Kim HK, Ahn HY, Shin JC, Joe YA. Priming Wharton's Jelly-Derived Mesenchymal Stromal/Stem Cells With ROCK Inhibitor Improves Recovery in an Intracerebral Hemorrhage Model. J Cell Biochem 2014; 116:310-9. [DOI: 10.1002/jcb.24969] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 08/29/2014] [Indexed: 12/21/2022]
Affiliation(s)
- Hyun-Sun Lee
- Cancer Research Institute and Department of Medical Life Sciences; College of Medicine; The Catholic University of Korea; Seoul 137-701 Korea
| | - Kwang S. Kim
- Cancer Research Institute and Department of Medical Life Sciences; College of Medicine; The Catholic University of Korea; Seoul 137-701 Korea
| | - Hee-Suk Lim
- Cancer Research Institute and Department of Medical Life Sciences; College of Medicine; The Catholic University of Korea; Seoul 137-701 Korea
| | - Moran Choi
- Cancer Research Institute and Department of Medical Life Sciences; College of Medicine; The Catholic University of Korea; Seoul 137-701 Korea
| | - Hyun-Kyung Kim
- Cancer Research Institute and Department of Medical Life Sciences; College of Medicine; The Catholic University of Korea; Seoul 137-701 Korea
| | - Hyun-Young Ahn
- Department of Obstetrics and Gynecology; College of Medicine; The Catholic University of Korea; Seoul 137-701 Korea
| | - Jong-Chul Shin
- Department of Obstetrics and Gynecology; College of Medicine; The Catholic University of Korea; Seoul 137-701 Korea
| | - Young Ae Joe
- Cancer Research Institute and Department of Medical Life Sciences; College of Medicine; The Catholic University of Korea; Seoul 137-701 Korea
| |
Collapse
|
197
|
|
198
|
Zhu YT, Li F, Han B, Tighe S, Zhang S, Chen SY, Liu X, Tseng SCG. Activation of RhoA-ROCK-BMP signaling reprograms adult human corneal endothelial cells. ACTA ACUST UNITED AC 2014; 206:799-811. [PMID: 25202030 PMCID: PMC4164941 DOI: 10.1083/jcb.201404032] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Activation of RhoA-ROCK-BMP signaling reprograms adult human corneal endothelial cells into neural crest–like progenitors, which effectively form corneal endothelial monolayers that may eliminate the need for corneal transplantation. Currently there are limited treatment options for corneal blindness caused by dysfunctional corneal endothelial cells. The primary treatment involves transplantation of healthy donor human corneal endothelial cells, but a global shortage of donor corneas necessitates other options. Conventional tissue approaches for corneal endothelial cells are based on EDTA-trypsin treatment and run the risk of irreversible endothelial mesenchymal transition by activating canonical Wingless-related integration site (Wnt) and TGF-β signaling. Herein, we demonstrate an alternative strategy that avoids disruption of cell–cell junctions and instead activates Ras homologue gene family A (RhoA)–Rho-associated protein kinase (ROCK)–canonical bone morphogenic protein signaling to reprogram adult human corneal endothelial cells to neural crest–like progenitors via activation of the miR302b-Oct4-Sox2-Nanog network. This approach allowed us to engineer eight human corneal endothelial monolayers of transplantable size, with a normal density and phenotype from one corneoscleral rim. Given that a similar signal network also exists in the retinal pigment epithelium, this partial reprogramming approach may have widespread relevance and potential for treating degenerative diseases.
Collapse
Affiliation(s)
- Ying-Ting Zhu
- TissueTech, Inc., Ocular Surface Center, and Ocular Surface Research & Education Foundation, Miami, FL 33173
| | - Fu Li
- Pediatric Research Institute and Department of Pediatric Hematology, Qilu Children's Hospital, Shandong University, Jinan, Shandong 250022, People's Republic of China
| | - Bo Han
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, People's Republic of China
| | - Sean Tighe
- TissueTech, Inc., Ocular Surface Center, and Ocular Surface Research & Education Foundation, Miami, FL 33173
| | - Suzhen Zhang
- TissueTech, Inc., Ocular Surface Center, and Ocular Surface Research & Education Foundation, Miami, FL 33173
| | - Szu-Yu Chen
- TissueTech, Inc., Ocular Surface Center, and Ocular Surface Research & Education Foundation, Miami, FL 33173
| | - Xin Liu
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, People's Republic of China
| | - Scheffer C G Tseng
- TissueTech, Inc., Ocular Surface Center, and Ocular Surface Research & Education Foundation, Miami, FL 33173
| |
Collapse
|
199
|
Croze RH, Buchholz DE, Radeke MJ, Thi WJ, Hu Q, Coffey PJ, Clegg DO. ROCK Inhibition Extends Passage of Pluripotent Stem Cell-Derived Retinal Pigmented Epithelium. Stem Cells Transl Med 2014; 3:1066-78. [PMID: 25069775 PMCID: PMC4149306 DOI: 10.5966/sctm.2014-0079] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 06/04/2014] [Indexed: 01/08/2023] Open
Abstract
Human embryonic stem cells (hESCs) offer a potentially unlimited supply of cells for emerging cell-based therapies. Unfortunately, the process of deriving distinct cell types can be time consuming and expensive. In the developed world, age-related macular degeneration (AMD) is the leading cause of blindness in the elderly, with more than 7.2 million people afflicted in the U.S. alone. Both hESC-derived retinal pigmented epithelium (hESC-RPE) and induced pluripotent stem cell-derived RPE (iPSC-RPE) are being developed for AMD therapies by multiple groups, but their potential for expansion in culture is limited. To attempt to overcome this passage limitation, we examined the involvement of Rho-associated, coiled-coil protein kinase (ROCK) in hESC-RPE and iPSC-RPE culture. We report that inhibiting ROCK1/2 with Y-27632 allows extended passage of hESC-RPE and iPSC-RPE. Microarray analysis suggests that ROCK inhibition could be suppressing an epithelial-to-mesenchymal transition through various pathways. These include inhibition of key ligands of the transforming growth factor-β pathway (TGFB1 and GDF6) and Wnt signaling. Two important processes are affected, allowing for an increase in hESC-RPE expansion. First, ROCK inhibition promotes proliferation by inducing multiple components that are involved in cell cycle progression. Second, ROCK inhibition affects many pathways that could be converging to suppress RPE-to-mesenchymal transition. This allows hESC-RPE to remain functional for an extended but finite period in culture.
Collapse
Affiliation(s)
- Roxanne H Croze
- Center for Stem Cell Biology and Engineering, Center for the Study of Macular Degeneration, Neuroscience Research Institute, and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, California, USA
| | - David E Buchholz
- Center for Stem Cell Biology and Engineering, Center for the Study of Macular Degeneration, Neuroscience Research Institute, and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, California, USA
| | - Monte J Radeke
- Center for Stem Cell Biology and Engineering, Center for the Study of Macular Degeneration, Neuroscience Research Institute, and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, California, USA
| | - William J Thi
- Center for Stem Cell Biology and Engineering, Center for the Study of Macular Degeneration, Neuroscience Research Institute, and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, California, USA
| | - Qirui Hu
- Center for Stem Cell Biology and Engineering, Center for the Study of Macular Degeneration, Neuroscience Research Institute, and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, California, USA
| | - Peter J Coffey
- Center for Stem Cell Biology and Engineering, Center for the Study of Macular Degeneration, Neuroscience Research Institute, and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, California, USA
| | - Dennis O Clegg
- Center for Stem Cell Biology and Engineering, Center for the Study of Macular Degeneration, Neuroscience Research Institute, and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, California, USA
| |
Collapse
|
200
|
Kale VP, Hengst JA, Desai DH, Dick TE, Choe KN, Colledge AL, Takahashi Y, Sung SS, Amin SG, Yun JK. A novel selective multikinase inhibitor of ROCK and MRCK effectively blocks cancer cell migration and invasion. Cancer Lett 2014; 354:299-310. [PMID: 25172415 DOI: 10.1016/j.canlet.2014.08.032] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Revised: 08/15/2014] [Accepted: 08/20/2014] [Indexed: 11/26/2022]
Abstract
Two structurally related protein kinase families, the Rho kinases (ROCK) and the myotonic dystrophy kinase-related Cdc42-binding kinases (MRCK) are required for migration and invasion of cancer cells. We hypothesized that simultaneous targeting of these two kinase families might represent a novel therapeutic strategy to block the migration and invasion of metastatic cancers. To this end, we developed DJ4 as a novel small molecule inhibitor of these kinases. DJ4 potently inhibited activities of ROCK and MRCK in an ATP competitive manner. In cellular functional assays, DJ4 treatment significantly blocked stress fiber formation and inhibited migration and invasion of multiple cancer cell lines in a concentration dependent manner. Our results strongly indicate that DJ4 may be further developed as a novel anti-metastatic chemotherapeutic agent for multiple cancers.
Collapse
Affiliation(s)
- Vijay Pralhad Kale
- Department of Pharmacology, Penn State Hershey College of Medicine, Hershey, PA 17033, USA
| | - Jeremy A Hengst
- Department of Pharmacology, Penn State Hershey College of Medicine, Hershey, PA 17033, USA
| | - Dhimant H Desai
- Department of Pharmacology, Penn State Hershey College of Medicine, Hershey, PA 17033, USA
| | - Taryn E Dick
- Department of Pharmacology, Penn State Hershey College of Medicine, Hershey, PA 17033, USA
| | - Katherine N Choe
- Department of Pharmacology, Penn State Hershey College of Medicine, Hershey, PA 17033, USA
| | - Ashley L Colledge
- Department of Pharmacology, Penn State Hershey College of Medicine, Hershey, PA 17033, USA
| | - Yoshinori Takahashi
- Department of Pharmacology, Penn State Hershey College of Medicine, Hershey, PA 17033, USA
| | - Shen-Shu Sung
- Department of Pharmacology, Penn State Hershey College of Medicine, Hershey, PA 17033, USA
| | - Shantu G Amin
- Department of Pharmacology, Penn State Hershey College of Medicine, Hershey, PA 17033, USA
| | - Jong K Yun
- Department of Pharmacology, Penn State Hershey College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|