1
|
López-Hidalgo R, Ballestín R, Lorenzo L, Sánchez-Martí S, Blasco-Ibáñez JM, Crespo C, Nacher J, Varea E. Early chronic fasudil treatment rescues hippocampal alterations in the Ts65Dn model for down syndrome. Neurochem Int 2024; 174:105679. [PMID: 38309665 DOI: 10.1016/j.neuint.2024.105679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/05/2024]
Abstract
Down syndrome (DS) is the most common genetic disorder associated with intellectual disability. To study this syndrome, several mouse models have been developed. Among the most common is the Ts65Dn model, which mimics most of the alterations observed in DS. Ts65Dn mice, as humans with DS, show defects in the structure, density, and distribution of dendritic spines in the cerebral cortex and hippocampus. Fasudil is a potent inhibitor of the RhoA kinase pathway, which is involved in the formation and stabilization of dendritic spines. Our study analysed the effect of early chronic fasudil treatment on the alterations observed in the hippocampus of the Ts65Dn model. We observed that treating Ts65Dn mice with fasudil induced an increase in neural plasticity in the hippocampus: there was an increment in the expression of PSA-NCAM and BDNF, in the dendritic branching and spine density of granule neurons, as well as in cell proliferation and neurogenesis in the subgranular zone. Finally, the treatment reduced the unbalance between excitation and inhibition present in this model. Overall, early chronic treatment with fasudil increases cell plasticity and eliminates differences with euploid animals.
Collapse
Affiliation(s)
- Rosa López-Hidalgo
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
| | - Raúl Ballestín
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
| | - Lorena Lorenzo
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
| | - Sandra Sánchez-Martí
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
| | - José Miguel Blasco-Ibáñez
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
| | - Carlos Crespo
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
| | - Juan Nacher
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain; CIBERSAM, Spanish National Network for Research in Mental Health, Madrid, Spain; Institute of research of the Clinic Hospital from Valencia (INCLIVA), Valencia, Spain
| | - Emilio Varea
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain.
| |
Collapse
|
2
|
Sato K, Nakagawa S, Morofuji Y, Matsunaga Y, Fujimoto T, Watanabe D, Izumo T, Niwa M, Walter FR, Vigh JP, Santa-Maria AR, Deli MA, Matsuo T. Effects of fasudil on blood-brain barrier integrity. Fluids Barriers CNS 2022; 19:43. [PMID: 35659272 PMCID: PMC9166508 DOI: 10.1186/s12987-022-00336-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 05/04/2022] [Indexed: 11/17/2022] Open
Abstract
Background Cerebral infarction accounts for 85% of all stroke cases. Even in an era of rapid and effective recanalization using an intravascular approach, the majority of patients have poor functional outcomes. Thus, there is an urgent need for the development of therapeutic agents to treat acute ischemic stroke. We evaluated the effect of fasudil, a Rho kinase inhibitor, on blood brain barrier (BBB) functions under normoxia or oxygen–glucose deprivation (OGD) conditions using a primary cell-based in vitro BBB model. Methods BBB models from rat primary cultures (brain capillary endothelial cells, astrocytes, and pericytes) were subjected to either normoxia or 6 h OGD/24 h reoxygenation. To assess the effects of fasudil on BBB functions, we evaluated real time impedance, transendothelial electrical resistance (TEER), sodium fluorescein permeability, and tight junction protein expression using western blotting. Lastly, to understand the observed protective mechanism on BBB functions by fasudil we examined the role of cyclooxygenase-2 and thromboxane A2 receptor agonist U-46619 in BBB-forming cells. Results We found that treatment with 0.3–30 µM of fasudil increased cellular impedance. Fasudil enhanced barrier properties in a concentration-dependent manner, as measured by an increased (TEER) and decreased permeability. Fasudil also increased the expression of tight junction protein claudin-5. Reductions in TEER and increased permeability were observed after OGD/reoxygenation exposure in mono- and co-culture models. The improvement in BBB integrity by fasudil was confirmed in both of the models, but was significantly higher in the co-culture than in the monoculture model. Treatment with U-46619 did not show significant changes in TEER in the monoculture model, whereas it showed a significant reduction in TEER in the co-culture model. Fasudil significantly improved the U-46619-induced TEER reduction in the co-culture models. Pericytes and astrocytes have opposite effects on endothelial cells and may contribute to endothelial injury in hyperacute ischemic stroke. Overall, fasudil protects the integrity of BBB both by a direct protective effect on endothelial cells and by a pathway mediated via pericytes and astrocytes. Conclusions Our findings suggest that fasudil is a BBB-protective agent against acute ischemic stroke. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00336-w.
Collapse
Affiliation(s)
- Kei Sato
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Shinsuke Nakagawa
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Yoichi Morofuji
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan.
| | - Yuki Matsunaga
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Takashi Fujimoto
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Daisuke Watanabe
- BBB Laboratory, PharmaCo-Cell Company Ltd, Nagasaki, 852-8135, Japan
| | - Tsuyoshi Izumo
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Masami Niwa
- BBB Laboratory, PharmaCo-Cell Company Ltd, Nagasaki, 852-8135, Japan
| | - Fruzsina R Walter
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Szeged, 6726, Hungary
| | - Judit P Vigh
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Szeged, 6726, Hungary
| | - Ana Raquel Santa-Maria
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Szeged, 6726, Hungary.,Wyss Institute for Biologically Inspired Engineering at Harvard University, 3 Blackfan Circle, Boston, MA, 02115, USA
| | - Maria A Deli
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Szeged, 6726, Hungary
| | - Takayuki Matsuo
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| |
Collapse
|
3
|
Zhang Z, Liu M, Zheng Y. Role of Rho GTPases in stem cell regulation. Biochem Soc Trans 2021; 49:2941-2955. [PMID: 34854916 PMCID: PMC9008577 DOI: 10.1042/bst20211071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 02/05/2023]
Abstract
The future of regenerative medicine relies on our understanding of stem cells which are essential for tissue/organ generation and regeneration to maintain and/or restore tissue homeostasis. Rho family GTPases are known regulators of a wide variety of cellular processes related to cytoskeletal dynamics, polarity and gene transcription. In the last decade, major new advances have been made in understanding the regulatory role and mechanism of Rho GTPases in self-renewal, differentiation, migration, and lineage specification in tissue-specific signaling mechanisms in various stem cell types to regulate embryonic development, adult tissue homeostasis, and tissue regeneration upon stress or damage. Importantly, implication of Rho GTPases and their upstream regulators or downstream effectors in the transformation, migration, invasion and tumorigenesis of diverse cancer stem cells highlights the potential of Rho GTPase targeting in cancer therapy. In this review, we discuss recent evidence of Rho GTPase signaling in the regulation of embryonic stem cells, multiple somatic stem cells, and cancer stem cells. We propose promising areas where Rho GTPase pathways may serve as useful targets for stem cell manipulation and related future therapies.
Collapse
Affiliation(s)
- Zheng Zhang
- Division of Experimental Hematology and Cancer Biology, Children’s Hospital Medical Center, University of Cincinnati, 3333 Burnet Avenue, Cincinnati, OH 45229, U.S.A
| | - Ming Liu
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Children’s Hospital Medical Center, University of Cincinnati, 3333 Burnet Avenue, Cincinnati, OH 45229, U.S.A
| |
Collapse
|
4
|
Bryniarska-Kubiak N, Kubiak A, Lekka M, Basta-Kaim A. The emerging role of mechanical and topographical factors in the development and treatment of nervous system disorders: dark and light sides of the force. Pharmacol Rep 2021; 73:1626-1641. [PMID: 34390472 PMCID: PMC8599311 DOI: 10.1007/s43440-021-00315-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 12/14/2022]
Abstract
Nervous system diseases are the subject of intensive research due to their association with high mortality rates and their potential to cause irreversible disability. Most studies focus on targeting the biological factors related to disease pathogenesis, e.g. use of recombinant activator of plasminogen in the treatment of stroke. Nevertheless, multiple diseases such as Parkinson’s disease and Alzheimer’s disease still lack successful treatment. Recently, evidence has indicated that physical factors such as the mechanical properties of cells and tissue and topography play a crucial role in homeostasis as well as disease progression. This review aims to depict these factors’ roles in the progression of nervous system diseases and consequently discusses the possibility of new therapeutic approaches. The literature is reviewed to provide a deeper understanding of the roles played by physical factors in nervous system disease development to aid in the design of promising new treatment approaches.
Collapse
Affiliation(s)
- Natalia Bryniarska-Kubiak
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland.
| | - Andrzej Kubiak
- Department of Biophysical Microstructures, Institute of Nuclear Physics, Polish Academy of Sciences, 31342, Kraków, Poland
| | - Małgorzata Lekka
- Department of Biophysical Microstructures, Institute of Nuclear Physics, Polish Academy of Sciences, 31342, Kraków, Poland
| | - Agnieszka Basta-Kaim
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland.
| |
Collapse
|
5
|
Hepatogenic Potential and Liver Regeneration Effect of Human Liver-derived Mesenchymal-Like Stem Cells. Cells 2020; 9:cells9061521. [PMID: 32580448 PMCID: PMC7348751 DOI: 10.3390/cells9061521] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/18/2020] [Accepted: 06/20/2020] [Indexed: 12/22/2022] Open
Abstract
Human liver-derived stem cells (hLD-SCs) have been proposed as a possible resource for stem cell therapy in patients with irreversible liver diseases. However, it is not known whether liver resident hLD-SCs can differentiate toward a hepatic fate better than mesenchymal stem cells (MSCs) obtained from other origins. In this study, we compared the differentiation ability and regeneration potency of hLD-SCs with those of human umbilical cord matrix-derived stem cells (hUC-MSCs) by inducing hepatic differentiation. Undifferentiated hLD-SCs expressed relatively high levels of endoderm-related markers (GATA4 and FOXA1). During directed hepatic differentiation supported by two small molecules (Fasudil and 5-azacytidine), hLD-SCs presented more advanced mitochondrial respiration compared to hUC-MSCs. Moreover, hLD-SCs featured higher numbers of hepatic progenitor cell markers on day 14 of differentiation (CPM and CD133) and matured into hepatocyte-like cells by day 7 through 21 with increased hepatocyte markers (ALB, HNF4A, and AFP). During in vivo cell transplantation, hLD-SCs migrated into the liver of ischemia-reperfusion injury-induced mice within 2 h and relieved liver injury. In the thioacetamide (TAA)-induced liver injury mouse model, transplanted hLD-SCs trafficked into the liver and spontaneously matured into hepatocyte-like cells within 14 days. These results collectively suggest that hLD-SCs hold greater hepatogenic potential, and hepatic differentiation-induced hLD-SCs may be a promising source of stem cells for liver regeneration.
Collapse
|
6
|
Mulherkar S, Tolias KF. RhoA-ROCK Signaling as a Therapeutic Target in Traumatic Brain Injury. Cells 2020; 9:E245. [PMID: 31963704 PMCID: PMC7016605 DOI: 10.3390/cells9010245] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/09/2020] [Accepted: 01/16/2020] [Indexed: 12/19/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability worldwide. TBIs, which range in severity from mild to severe, occur when a traumatic event, such as a fall, a traffic accident, or a blow, causes the brain to move rapidly within the skull, resulting in damage. Long-term consequences of TBI can include motor and cognitive deficits and emotional disturbances that result in a reduced quality of life and work productivity. Recovery from TBI can be challenging due to a lack of effective treatment options for repairing TBI-induced neural damage and alleviating functional impairments. Central nervous system (CNS) injury and disease are known to induce the activation of the small GTPase RhoA and its downstream effector Rho kinase (ROCK). Activation of this signaling pathway promotes cell death and the retraction and loss of neural processes and synapses, which mediate information flow and storage in the brain. Thus, inhibiting RhoA-ROCK signaling has emerged as a promising approach for treating CNS disorders. In this review, we discuss targeting the RhoA-ROCK pathway as a therapeutic strategy for treating TBI and summarize the recent advances in the development of RhoA-ROCK inhibitors.
Collapse
Affiliation(s)
- Shalaka Mulherkar
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Kimberley F. Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA;
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
7
|
Tang Y, Han L, Bai X, Liang X, Zhao J, Huang F, Wang J. Intranasal Delivery of Bone Marrow Stromal Cells Preconditioned with Fasudil to Treat a Mouse Model of Parkinson's Disease. Neuropsychiatr Dis Treat 2020; 16:249-262. [PMID: 32158210 PMCID: PMC6986408 DOI: 10.2147/ndt.s238646] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 01/13/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Stem cell transplantation is a promising strategy with great potential to treat Parkinson's disease (PD). Nevertheless, improving the cell delivery route and optimising implanted cells are necessary to increase the therapeutic effect. Herein, we investigated whether intranasal delivery of bone marrow stromal cells (BMSCs) has beneficial effects in a PD mouse model and whether the therapeutic potential of BMSCs could be enhanced by preconditioning with fasudil. METHODS A PD mouse model was developed by intraperitoneally administering 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Mice were treated intranasally with phosphate buffered saline (PBS), BMSCs, or BMSCs preconditioned with fasudil. One month later, the effects of BMSC treatment were analysed. RESULTS Our study showed that fasudil could accelerate the proliferation of BMSCs and promote brain-derived neurotrophic factor (BDNF) secretion in vitro. Intranasally administered BMSCs were capable of surviving and migrating in the brain. Intranasal delivery of BMSCs preconditioned with fasudil significantly improved motor function and reduced dopaminergic neuron loss in substantia nigra; treatment with BMSCs and PBS resulted in similar outcomes. Preconditioning with fasudil inhibited the activation and aggregation of microglia, suppressed immune response, and reinforced BDNF secretion in MPTP-PD mice significantly more than treatment with BMSCs alone. CONCLUSION The present study demonstrates that intranasally administering BMSCs preconditioned with fasudil is a promising cell-based therapy for PD.
Collapse
Affiliation(s)
- Yilin Tang
- Department of Neurology and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China
| | - Linlin Han
- Department of Neurology and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China
| | - Xiaochen Bai
- Department of Neurology and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China.,The State Key Laboratory of Medical Neurobiology, The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Xiaoniu Liang
- Department of Neurology and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China
| | - Jue Zhao
- Department of Neurology and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China
| | - Fang Huang
- The State Key Laboratory of Medical Neurobiology, The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Jian Wang
- Department of Neurology and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China
| |
Collapse
|
8
|
Torres-Cuevas I, Corral-Debrinski M, Gressens P. Brain oxidative damage in murine models of neonatal hypoxia/ischemia and reoxygenation. Free Radic Biol Med 2019; 142:3-15. [PMID: 31226400 DOI: 10.1016/j.freeradbiomed.2019.06.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/26/2019] [Accepted: 06/10/2019] [Indexed: 02/08/2023]
Abstract
The brain is one of the main organs affected by hypoxia and reoxygenation in the neonatal period and one of the most vulnerable to oxidative stress. Hypoxia/ischemia and reoxygenation leads to impairment of neurogenesis, disruption of cortical migration, mitochondrial damage and neuroinflammation. The extent of the injury depends on the clinical manifestation in the affected regions. Preterm newborns are highly vulnerable, and they exhibit severe clinical manifestations such as intraventricular hemorrhage (IVH), retinopathy of prematurity (ROP) and diffuse white matter injury (DWMI) among others. In the neonatal period, the accumulation of high levels of reactive oxygen species exacerbated by the immature antioxidant defense systems in represents cellular threats that, if they exceed or bypass physiological counteracting mechanisms, are responsible of significant neuronal damage. Several experimental models in mice mimic the consequences of perinatal asphyxia and the use of oxygen in the reanimation process that produce brain injury. The aim of this review is to highlight brain damage associated with oxidative stress in different murine models of hypoxia/ischemia and reoxygenation.
Collapse
Affiliation(s)
| | | | - Pierre Gressens
- INSERM UMR1141, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
9
|
Brockmann C, Corkhill C, Jaroslawska E, Dege S, Brockmann T, Kociok N, Joussen AM. Systemic Rho-kinase inhibition using fasudil in mice with oxygen-induced retinopathy. Graefes Arch Clin Exp Ophthalmol 2019; 257:1699-1708. [PMID: 31152312 DOI: 10.1007/s00417-019-04365-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 04/28/2019] [Accepted: 05/18/2019] [Indexed: 11/30/2022] Open
Abstract
PURPOSE To investigate the influence of the selective Rho-kinase (ROCK) inhibitor, fasudil, on the mRNA level of proinflammatory factors and the retinal vascular development in mice with oxygen-induced retinopathy (OIR). METHODS C57BL/6J mice underwent standard protocol for OIR induction from postnatal days 7 to 12. Subsequently, they received a daily intraperitoneal injection of fasudil or sodium chloride from P12 to P16. Analyses were performed using vascular staining on retinal flat mounts, RNA expression by qPCR, and immunohistochemistry on paraffin sections. RESULTS On retinal flat mounts, the proportion of avascular area and tuft formation did not differ between the fasudil and NaCl group. Immunohistochemical staining revealed a less intense staining with inflammatory markers after fasudil. Nevertheless, there were no differences on RNA level between the two groups. CONCLUSIONS In conclusion, our findings support that daily systemic application of fasudil does not decrease retinal neovascularization in rodents with oxygen-induced retinopathy. The results of our study together with the controversial results on the effects of different ROCK inhibitors from the literature makes it apparent that effects of ROCK inhibition are more complex, and further studies are necessary to analyze its potential therapeutic effects.
Collapse
Affiliation(s)
- Claudia Brockmann
- Corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Department of Ophthalmology, Charité - Universitätsmedizin Berlin, Augustenburger Platz, 113353, Berlin, Germany. .,Berlin Institute of Health (BIH), Berlin, Germany.
| | - Caitlin Corkhill
- Corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Department of Ophthalmology, Charité - Universitätsmedizin Berlin, Augustenburger Platz, 113353, Berlin, Germany
| | - Elzbieta Jaroslawska
- Corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Department of Ophthalmology, Charité - Universitätsmedizin Berlin, Augustenburger Platz, 113353, Berlin, Germany.,Department of Ophthalmology, Medical University of Lublin, Lublin, Poland
| | - Sabrina Dege
- Corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Department of Ophthalmology, Charité - Universitätsmedizin Berlin, Augustenburger Platz, 113353, Berlin, Germany
| | - Tobias Brockmann
- Corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Department of Ophthalmology, Charité - Universitätsmedizin Berlin, Augustenburger Platz, 113353, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Norbert Kociok
- Corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Department of Ophthalmology, Charité - Universitätsmedizin Berlin, Augustenburger Platz, 113353, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Antonia M Joussen
- Corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Department of Ophthalmology, Charité - Universitätsmedizin Berlin, Augustenburger Platz, 113353, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
10
|
Wang L, Xu J, Guo D, Zhou X, Jiang W, Wang J, Tang J, Zou Y, Bi M, Li Q. Fasudil alleviates brain damage in rats after carbon monoxide poisoning through regulating neurite outgrowth inhibitor/oligodendrocytemyelin glycoprotein signalling pathway. Basic Clin Pharmacol Toxicol 2019; 125:152-165. [PMID: 30916885 DOI: 10.1111/bcpt.13233] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 02/07/2019] [Indexed: 12/21/2022]
Abstract
Carbon monoxide (CO) poisoning can lead to many serious neurological symptoms. Currently, there are no effective therapies for CO poisoning. In this study, rats exposed to CO received hyperbaric oxygen therapy, and those in the Fasudil group were given additional Fasudil injection once daily. We found that the escape latency in CO poisoning group (CO group) was significantly prolonged, the T1 /Ttotal was obviously decreased, and the mean escape time and the active escape latency were notably extended compared with those in normal control group (NC group, P < 0.05). After administration of Fasudil, the escape latency was significantly shortened, T1 /Ttotal was gradually increased as compared with CO group (>1 week, P < 0.05). Ultrastructural damage of neurons and blood-brain barrier of rats was serious in CO group, while the structural and functional integrity of neuron and mitochondria maintained relatively well in Fasudil group. Moreover, we also noted that the expressions of neurite outgrowth inhibitor (Nogo), oligodendrocyte-myelin glycoprotein (OMgp) and Rock in brain tissue were significantly increased in CO group, and the elevated levels of the three proteins were still observed at 2 months after CO poisoning. Fasudil markedly reduced their expressions compared with those of CO group (P < 0.05). In summary, the activation of Nogo-OMgp/Rho signalling pathway is associated with brain injury in rats with CO poisoning. Fasudil can efficiently down-regulate the expressions of Nogo, OMgp and Rock proteins, paving a way for the treatment of acute brain damage after CO poisoning.
Collapse
Affiliation(s)
- Li Wang
- Department of Neurology, Qianfoshan Hospital Affiliated to Shandong University, Jinan Shandong, China.,Department of Integration of Chinese and Western Medicine, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai Shandong, China
| | - Jianghua Xu
- Department of neurology, Yantai YEDA Hospital, Yantai Shandong, China
| | - Dadong Guo
- Eye Institute of Shandong University of Traditional Chinese Medicine, Jinan Shandong, China
| | - Xudong Zhou
- The First Affiliated Hospital of Shandong, University of Traditional Chinese Medicine, Jinan Shandong, China
| | - Wenwen Jiang
- Department of Integration of Chinese and Western Medicine, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai Shandong, China
| | - Jinglin Wang
- Emergency Centre, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai Shandong, China
| | - Jiyou Tang
- Department of Neurology, Qianfoshan Hospital Affiliated to Shandong University, Jinan Shandong, China
| | - Yong Zou
- Department of Integration of Chinese and Western Medicine, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai Shandong, China
| | - Mingjun Bi
- Emergency Centre, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai Shandong, China
| | - Qin Li
- Department of Integration of Chinese and Western Medicine, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai Shandong, China
| |
Collapse
|
11
|
Hu Y, Li X, Huang G, Wang J, Lu W. Fasudil may induce the differentiation of bone marrow mesenchymal stem cells into neuron‑like cells via the Wnt/β‑catenin pathway. Mol Med Rep 2019; 19:3095-3104. [PMID: 30816472 PMCID: PMC6423592 DOI: 10.3892/mmr.2019.9978] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 02/18/2019] [Indexed: 01/27/2023] Open
Abstract
Bone mesenchymal stem cells (MSCs) are an excellent donor graft source due to their potential for self-renewal and multidirectional differentiation. However, it is difficult to obtain high quality MSCs and to induce them to differentiate into neuron-like cells. Fasudil, a Rho kinase inhibitor, exhibits therapeutic potential in spinal cord injuries and stroke. The present study investigated the effect of fasudil on the differentiation of MSCs into neuron-like cells. MSCs were obtained from rat femur marrow, expanded in culture medium, and used at the third passage for subsequent experiments. MSCs were pre-induced with 10 ng/ml basic fibroblast growth factor (bFGF) for 24 h, which was followed by induction with fasudil. A control untreated group and a group treated with fasudil + XAV939, a Wnt/β-catenin pathway inhibitor, were also used in the present study. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR), western blot analysis and immunofluorescence staining were performed in order to detect neuron-specific markers, including neuron-specific enolase (NSE), nestin and neurofilament-M (NF-M). Following induction with fasudil, neuron-like cell morphology was observed. In the fasudil + XAV939 and control groups, no obvious changes in cell shape were observed. The results of RT-qPCR, western blot analysis and immunofluorescence staining indicated that expression of the neuron-specific markers NSE, nestin and NF-M was detected in the fasudil group. The differentiation of MSCs into neuron-like cells induced by fasudil was eliminated when the Wnt/β-catenin pathway was inhibited. The present study demonstrated that fasudil may induce MSCs to differentiate into neuron-like cells, however further studies are required to determine the specific mechanisms involved in the effect of fasudil on the Wnt/β-catenin pathway. In addition, further research is required to examine the functional characteristics of the induced neuron-like cells, in order to establish their suitability for clinical treatments in the future.
Collapse
Affiliation(s)
- Yahui Hu
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Xin Li
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Guowei Huang
- Department of Nutrition and Food Hygiene, School of Public Health, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Jizuo Wang
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Wei Lu
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| |
Collapse
|
12
|
Wang J, Sui RX, Miao Q, Wang Q, Song LJ, Yu JZ, Li YH, Xiao BG, Ma CG. Hydroxyfasudil alleviates demyelination through the inhibition of MOG antibody and microglia activation in cuprizone mouse model. Clin Immunol 2019; 201:35-47. [PMID: 30660624 DOI: 10.1016/j.clim.2019.01.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 01/14/2019] [Accepted: 01/14/2019] [Indexed: 02/06/2023]
Abstract
Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the central nervous system characterized by oligodendrocyte loss and progressive neurodegeneration. The cuprizone (CPZ)-induced demyelination is widely used to investigate the demyelination/remyelination. Here, we explored the therapeutic effects of Hydroxyfasudil (HF), an active metabolite of Fasudil, in CPZ model. HF improved behavioral abnormality and reduced myelin damage in the corpus callosum. Splenic atrophy and myelin oligodendrocyte glycoprotein (MOG) antibody were observed in CPZ model, which were partially restored and obviously inhibited by HF, therefore reducing pathogenic binding of MOG antibody to oligodendrocytes. HF inhibited the percentages of CD4+IL-17+ T cells from splenocytes and infiltration of CD4+ T cells and CD68+ macrophages in the brain. HF also declined microglia-mediated neuroinflammation, and promoted the production of astrocyte-derived brain derived neurotrophic factor (BDNF) and regeneration of NG2+ oligodendrocyte precursor cells. These results provide potent evidence for the therapeutic effects of HF in CPZ-induced demyelination.
Collapse
Affiliation(s)
- Jing Wang
- Shanxi Medical University, Taiyuan030001, China
| | - Ruo-Xuan Sui
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Traditional Chinese Medicine, Taiyuan030024, China
| | - Qiang Miao
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Traditional Chinese Medicine, Taiyuan030024, China
| | - Qing Wang
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Traditional Chinese Medicine, Taiyuan030024, China
| | - Li-Juan Song
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Traditional Chinese Medicine, Taiyuan030024, China
| | - Jie-Zhong Yu
- Institute of Brain Science, Shanxi Datong University, Datong037009, China
| | - Yan-Hua Li
- Institute of Brain Science, Shanxi Datong University, Datong037009, China
| | - Bao-Guo Xiao
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200025, China.
| | - Cun-Gen Ma
- Shanxi Medical University, Taiyuan030001, China; The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Traditional Chinese Medicine, Taiyuan030024, China; Institute of Brain Science, Shanxi Datong University, Datong037009, China.
| |
Collapse
|
13
|
Zibara K, Ballout N, Mondello S, Karnib N, Ramadan N, Omais S, Nabbouh A, Caliz D, Clavijo A, Hu Z, Ghanem N, Gajavelli S, Kobeissy F. Combination of drug and stem cells neurotherapy: Potential interventions in neurotrauma and traumatic brain injury. Neuropharmacology 2018; 145:177-198. [PMID: 30267729 DOI: 10.1016/j.neuropharm.2018.09.032] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 09/17/2018] [Accepted: 09/21/2018] [Indexed: 12/12/2022]
Abstract
Traumatic brain injury (TBI) has been recognized as one of the major public health issues that leads to devastating neurological disability. As a consequence of primary and secondary injury phases, neuronal loss following brain trauma leads to pathophysiological alterations on the molecular and cellular levels that severely impact the neuropsycho-behavioral and motor outcomes. Thus, to mitigate the neuropathological sequelae post-TBI such as cerebral edema, inflammation and neural degeneration, several neurotherapeutic options have been investigated including drug intervention, stem cell use and combinational therapies. These treatments aim to ameliorate cellular degeneration, motor decline, cognitive and behavioral deficits. Recently, the use of neural stem cells (NSCs) coupled with selective drug therapy has emerged as an alternative treatment option for neural regeneration and behavioral rehabilitation post-neural injury. Given their neuroprotective abilities, NSC-based neurotherapy has been widely investigated and well-reported in numerous disease models, notably in trauma studies. In this review, we will elaborate on current updates in cell replacement therapy in the area of neurotrauma. In addition, we will discuss novel combination drug therapy treatments that have been investigated in conjunction with stem cells to overcome the limitations associated with stem cell transplantation. Understanding the regenerative capacities of stem cell and drug combination therapy will help improve functional recovery and brain repair post-TBI. This article is part of the Special Issue entitled "Novel Treatments for Traumatic Brain Injury".
Collapse
Affiliation(s)
- Kazem Zibara
- ER045, Laboratory of Stem Cells, PRASE, Lebanese University, Beirut, Lebanon; Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Nissrine Ballout
- ER045, Laboratory of Stem Cells, PRASE, Lebanese University, Beirut, Lebanon
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Nabil Karnib
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon
| | - Naify Ramadan
- Department of Women's and Children's Health (KBH), Division of Clinical Pediatrics, Karolinska Institute, Sweden
| | - Saad Omais
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Ali Nabbouh
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon
| | - Daniela Caliz
- Lois Pope LIFE Center, Neurosurgery, University of Miami, 33136, Miami, FL, USA
| | - Angelica Clavijo
- Lois Pope LIFE Center, Neurosurgery, University of Miami, 33136, Miami, FL, USA
| | - Zhen Hu
- Lois Pope LIFE Center, Neurosurgery, University of Miami, 33136, Miami, FL, USA
| | - Noël Ghanem
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Shyam Gajavelli
- Lois Pope LIFE Center, Neurosurgery, University of Miami, 33136, Miami, FL, USA.
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon; Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Department of Emergency Medicine, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
14
|
Yin H, Chen L, Yang B, Bardelang D, Wang C, Lee SMY, Wang R. Fluorescence enhancement and pK a shift of a rho kinase inhibitor by a synthetic receptor. Org Biomol Chem 2018; 15:4336-4343. [PMID: 28470298 DOI: 10.1039/c7ob00547d] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Fasudil (FSD), a selective rho kinase (ROCK) inhibitor, was found to form 1 : 1 host-guest inclusion complexes with a synthetic macrocyclic receptor, cucurbit[7]uril (CB[7]), in aqueous solutions, as evidenced by 1H NMR, photoluminescence and UV-visible spectroscopic titrations, isothermal titration calorimetry (ITC) titration, and electrospray ionization (ESI) mass spectrometry, as well as density functional theory (DFT) molecular modeling. Upon encapsulation, whereas the UV-vis absorbance of FSD experienced a moderate decrease and bathochromic shift, the fluorescence intensity of FSD at 354 nm was dramatically enhanced for up to 69-fold at neutral pH, which could potentially be applied in fluorescent tracking of the drug delivery and release. More interestingly, the binding affinity (Ka = (4.28 ± 0.21) × 106 M-1), of FSD-CB[7] complexes under acidic conditions (pH = 2.0), is approximately three orders of magnitude higher than that (2.2∼6.6 × 103 M-1) under neutral pH conditions (pH = 7.0). Accordingly, UV-visible spectroscopic titration of the free and complexed FSD under various pH conditions has demonstrated that the encapsulation of FSD by CB[7] shifted the pKa of the isoquinoline-N upward from 3.05 to 5.96 (ΔpKa of 2.91). The significantly higher binding affinity of the complexes under acidic conditions may be applied in developing the "enteric" formulation of FSD. Furthermore, our in vitro study of the bioactivity of FSD in the absence and presence of CB[7] on a neural cell line, SH-SY5Y, showed that the complexation preserved the drug's pro-neurite efficacy. Thus this discovery may lead to a fluorescence-trackable, orally administered enteric formulation of rho kinase inhibitors that are stable under gastric conditions, without compromising bioactivity of the drugs.
Collapse
Affiliation(s)
- Hang Yin
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, China.
| | | | | | | | | | | | | |
Collapse
|
15
|
Nizamudeen ZA, Chakrabarti L, Sottile V. Exposure to the ROCK inhibitor fasudil promotes gliogenesis of neural stem cells in vitro. Stem Cell Res 2018; 28:75-86. [DOI: 10.1016/j.scr.2018.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/30/2018] [Accepted: 02/02/2018] [Indexed: 12/27/2022] Open
|
16
|
Judge RA, Vasudevan A, Scott VE, Simler GH, Pratt SD, Namovic MT, Putman CB, Aguirre A, Stoll VS, Mamo M, Swann SI, Cassar SC, Faltynek CR, Kage KL, Boyce-Rustay JM, Hobson AD. Design of Aminobenzothiazole Inhibitors of Rho Kinases 1 and 2 by Using Protein Kinase A as a Structure Surrogate. Chembiochem 2018; 19:613-621. [PMID: 29314498 DOI: 10.1002/cbic.201700547] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Indexed: 11/11/2022]
Abstract
We describe the design, synthesis, and structure-activity relationships (SARs) of a series of 2-aminobenzothiazole inhibitors of Rho kinases (ROCKs) 1 and 2, which were optimized to low nanomolar potencies by use of protein kinase A (PKA) as a structure surrogate to guide compound design. A subset of these molecules also showed robust activity in a cell-based myosin phosphatase assay and in a mechanical hyperalgesia in vivo pain model.
Collapse
Affiliation(s)
- Russell A Judge
- AbbVie, Inc., 1 North Waukegan Road, North Chicago, IL, 60064, USA
| | - Anil Vasudevan
- AbbVie, Inc., 1 North Waukegan Road, North Chicago, IL, 60064, USA
| | - Victoria E Scott
- AbbVie, Inc., 1 North Waukegan Road, North Chicago, IL, 60064, USA
| | - Gricelda H Simler
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, USA
| | - Steve D Pratt
- AbbVie, Inc., 1 North Waukegan Road, North Chicago, IL, 60064, USA
| | - Marian T Namovic
- AbbVie, Inc., 1 North Waukegan Road, North Chicago, IL, 60064, USA
| | - C Brent Putman
- AbbVie, Inc., 1 North Waukegan Road, North Chicago, IL, 60064, USA
| | - Ana Aguirre
- AbbVie, Inc., 1 North Waukegan Road, North Chicago, IL, 60064, USA
| | - Vincent S Stoll
- AbbVie, Inc., 1 North Waukegan Road, North Chicago, IL, 60064, USA
| | - Mulugeta Mamo
- AbbVie, Inc., 1 North Waukegan Road, North Chicago, IL, 60064, USA.,Current address: Novartis Institute for Biomedical Research, 4560 Horton Street, Emeryville, CA, 94608, USA
| | - Steven I Swann
- AbbVie, Inc., 1 North Waukegan Road, North Chicago, IL, 60064, USA.,Current address: Takeda Pharmaceuticals, 10410 Science Center Drive, San Diego, CA, 92121, USA
| | - Steven C Cassar
- AbbVie, Inc., 1 North Waukegan Road, North Chicago, IL, 60064, USA
| | | | - Karen L Kage
- AbbVie, Inc., 1 North Waukegan Road, North Chicago, IL, 60064, USA.,Current address: Altor Bioscience, 2810 North Commerce Parkway, Miramar, FL, 33025, USA
| | - Janel M Boyce-Rustay
- AbbVie, Inc., 1 North Waukegan Road, North Chicago, IL, 60064, USA.,Current address: Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Adrian D Hobson
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, USA
| |
Collapse
|
17
|
Saal K, Galter D, Roeber S, Bähr M, Tönges L, Lingor P. Altered Expression of Growth Associated Protein-43 and Rho Kinase in Human Patients with Parkinson's Disease. Brain Pathol 2017; 27:13-25. [PMID: 26748453 PMCID: PMC8029215 DOI: 10.1111/bpa.12346] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 12/14/2015] [Indexed: 12/21/2022] Open
Abstract
Causative treatment strategies for Parkinson's disease (PD) will have to address multiple underlying pathomechanisms to attenuate neurodegeneration. Additionally, the intrinsic regenerative capacity of the central nervous system is also an important factor contributing to restoration. Extracellular cues can limit sprouting and regrowth of adult neurons, but even aged neurons have a low intrinsic regeneration capacity. Whether this capacity has been lost or if growth inhibitory cues are increased during PD progression has not been resolved yet. In this study, we assessed the regenerative potential in the nigrostriatal system in post-mortem brain sections of PD patients compared to age-matched and young controls. Investigation of the expression pattern of the regeneration-associated protein GAP-43 suggested a lower regenerative capacity in nigral dopaminergic neurons of PD patients. Furthermore, the increase in protein expression of the growth-inhibitory protein ROCK2 in astrocytes and a similar trend in microglia, suggests an important role for ROCK2 in glial PD pathology, which is initiated already in normal aging. Considering the role of astro- and microglia in PD pathogenesis as well as beneficial effects of ROCK inhibition on neuronal survival and regeneration in neurodegenerative disease models, our data strengthens the importance of the ROCK pathway as a therapeutic target in PD.
Collapse
Affiliation(s)
- Kim‐Ann Saal
- Department of NeurologyUniversity Medicine GöttingenGöttingenGermany
| | - Dagmar Galter
- Department of NeuroscienceKarolinska InstituteStockholmSweden
| | - Sigrun Roeber
- Department of NeuropathologyLudwig‐Maximilians‐UniversityMunichGermany
| | - Mathias Bähr
- Department of NeurologyUniversity Medicine GöttingenGöttingenGermany
- DFG‐Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB)GöttingenGermany
| | - Lars Tönges
- Department of NeurologyUniversity Medicine GöttingenGöttingenGermany
- DFG‐Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB)GöttingenGermany
| | - Paul Lingor
- Department of NeurologyUniversity Medicine GöttingenGöttingenGermany
- DFG‐Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB)GöttingenGermany
| |
Collapse
|
18
|
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: 3.6] [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
|
19
|
The fate of medications evaluated for ischemic stroke pharmacotherapy over the period 1995-2015. Acta Pharm Sin B 2016; 6:522-530. [PMID: 27818918 PMCID: PMC5071630 DOI: 10.1016/j.apsb.2016.06.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 05/26/2016] [Accepted: 05/31/2016] [Indexed: 01/26/2023] Open
Abstract
Stroke is a brain damage caused by a loss of blood supply to a portion of the brain, which requires prompt and effective treatment. The current pharmacotherapy for ischemic stroke primarily relies on thrombolysis using recombinant tissue plasminogen activators (rt-PAs) to breakdown blood clots. Neuroprotective agents that inhibit excitatory neurotransmitters are also used to treat ischemic stroke but have failed to translate into clinical benefits. This poses a major challenge in biomedical research to understand what causes the progressive brain cell death after stroke and how to develop an effective pharmacotherapy for stroke. This brief review analyzes the fate of about 430 potentially useful stroke medications over the period 1995–2015 and describes in detail those that successfully reached the market. Hopefully, the information from this analysis will shed light on how future stroke research can improve stroke drug discovery.
Collapse
Key Words
- ADP, adenosine diphosphate
- AMPA, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
- ASIC1a, acid-sensing ion channel 1a
- BDNF, brain-derived neurotrophic factor
- CFDA, the China Food and Drug Administration
- CNTF, ciliary neurotrophic factor
- GDNF, glial cell line–derived neurotrophic factor
- Ion channel
- Ischemic stroke
- MHRA, Medicine and Healthcare Products Regulatory Agency
- NBP, butylphthalide/3-n-butylphthalide
- NGF, nerve growth factor
- NMDA, N-methyl-D-aspartate
- Neuroprotective agent
- Non-NMDA mechanism
- TCM, traditional Chinese medicine
- TRP, transient receptor potential
- TRPC, transient receptor potential canonical
- TRPM, transient receptor potential melastatin
- TRPV, transient receptor potential vanilloid
- Thrombosis
- Traditional Chinese medicine
- iGluRs, ionotropic glutamate receptors
- rt-Pas, recombinant tissue plasminogen activators
Collapse
|
20
|
Synergistic and Superimposed Effect of Bone Marrow-Derived Mesenchymal Stem Cells Combined with Fasudil in Experimental Autoimmune Encephalomyelitis. J Mol Neurosci 2016; 60:486-497. [PMID: 27573128 DOI: 10.1007/s12031-016-0819-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 08/17/2016] [Indexed: 12/23/2022]
Abstract
Bone marrow-derived mesenchymal stem cells (MSCs) are the ideal transplanted cells of cellular therapy for promoting neuroprotection and neurorestoration. However, the optimization of transplanted cells and the improvement of microenvironment around implanted cells are still two critical challenges for enhancing therapeutic effect. In the current study, we observed the therapeutic potential of MSCs combined with Fasudil in mouse model of experimental autoimmune encephalomyelitis (EAE) and explored possible mechanisms of action. The results clearly show that combined intervention of MSCs and Fasudil further reduced the severity of EAE compared with MSCs or Fasudil alone, indicating a synergistic and superimposed effect in treating EAE. The addition of Fasudil inhibited MSC-induced inflammatory signaling TLR-4/MyD88 and inflammatory molecule IFN-γ, IL-1β, and TNF-α but did not convert M1 microglia to M2 phenotype. The delivery of MSCs enhanced the expression of glial cell-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF) compared with that of Fasudil. Importantly, combined intervention of MSCs and Fasudil further increased the expression of BDNF and GDNF compared with the delivery of MSCs alone, indicating that combined intervention of MSCs and Fasudil synergistically contributes to the expression of neurotrophic factors which should be related to the expression of increased galactocerebroside (GalC) compared with mice treated with Fasudil and MSCs alone. However, a lot of investigation is warranted to further elucidate the cross talk of MSCs and Fasudil in the therapeutic potential of EAE/multiple sclerosis.
Collapse
|
21
|
Ding J, Wang J, Li QY, Yu JZ, Ma CG, Wang X, Lu CZ, Xiao BG. Neuroprotection and CD131/GDNF/AKT Pathway of Carbamylated Erythropoietin in Hypoxic Neurons. Mol Neurobiol 2016; 54:5051-5060. [PMID: 27541284 DOI: 10.1007/s12035-016-0022-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Accepted: 08/01/2016] [Indexed: 12/14/2022]
Abstract
Carbamylated erythropoietin (CEPO), an EPO derivative, is attracting widespread interest due to neuroprotective effects without erythropoiesis. However, little is known about molecular mechanisms behind CEPO-mediated neuroprotection. In primary neurons with oxygen-glucose deprivation (OGD) and mice with hypoxia-reoxygenation, the neuroprotection and possible molecular mechanism of CEPO were performed by immunohistochemistry and immunocytochemistry, Western blot, RT-PCR, and ELISA. The comparisons were analyzed by ANOVA followed by unpaired two-tailed Student's t test. Both CEPO and EPO showed the neuroprotective effects in OGD model and hypoxic brain. CEPO did not trigger JAK-2 but activated AKT through glial cell line-derived neurotrophic factor (GDNF). It has been shown that CEPO acts upon a heteroreceptor complex comprising both the EPO receptor and the common β receptor subunit (βcR, also known as CD131). The blockage of CD131 reduced CEPO-mediated GDNF production, while GFR receptor blockage and GDNF neutralization inhibited CEPO-induced neurogenesis. Addition of GDNF to cultured neurons increased phosphorylation of AKT. CEPO protects neurons possible through the CD131/GDNF/AKT pathway.
Collapse
Affiliation(s)
- Jing Ding
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Wang
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qin-Ying Li
- Institute of Neurology, Huashan Hospital, Institute of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, 12 Middle Wulumuqi Road, Shanghai, 200040, China
| | - Jie-Zhong Yu
- Institute of Brain Science, Shanxi Datong University, Shanxi, 037009, China
| | - Cun-Gen Ma
- Institute of Brain Science, Shanxi Datong University, Shanxi, 037009, China
| | - Xin Wang
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chuan-Zhen Lu
- Institute of Neurology, Huashan Hospital, Institute of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, 12 Middle Wulumuqi Road, Shanghai, 200040, China
| | - Bao-Guo Xiao
- Institute of Neurology, Huashan Hospital, Institute of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, 12 Middle Wulumuqi Road, Shanghai, 200040, China.
| |
Collapse
|
22
|
Role of JAK-STAT pathway in reducing cardiomyocytes hypoxia/reoxygenation injury induced by S1P postconditioning. Eur J Pharmacol 2016; 784:129-36. [PMID: 27215146 DOI: 10.1016/j.ejphar.2016.05.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 05/19/2016] [Accepted: 05/19/2016] [Indexed: 01/01/2023]
Abstract
This experiment was designed to explore the protection of sphingosine1-phosphate (S1P) postconditioning on rat myocardial cells injured by hypoxia/reoxygenation acting via the Janus kinase-signal transducer and activator of transcription (JAK-STAT) signal pathway. The data showed that S1P could significantly increase cell viability, lower the rate of apoptosis, decrease the content of lactate dehydrogenase (LDH) and caspase3 activity in the culture medium, increase the activity of total superoxide dismutase (T-SOD) and manganese superoxide dismutase (Mn-SOD), reduce the loss of mitochondrial membrane potential and the fluorescence intensity of intracellular calcium, as well as increase the phosphorylation of JAK2 and STAT3 in comparison with the H/R group. When the JAK inhibitor AG490 or the STAT inhibitor stattic were added, the effects of S1P were inhibited. Our date shows that S1P protects H9c2 cells from hypoxia/reoxygenation injury and that the protection by S1P was inhibited by AG490 and stattic. Therefore S1P protects H9c2 cells against hypoxia/reoxygenation injury via the JAK-STAT pathway.
Collapse
|
23
|
Bye N, Christie KJ, Turbic A, Basrai HS, Turnley AM. Rho kinase inhibition following traumatic brain injury in mice promotes functional improvement and acute neuron survival but has little effect on neurogenesis, glial responses or neuroinflammation. Exp Neurol 2016; 279:86-95. [DOI: 10.1016/j.expneurol.2016.02.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/08/2016] [Accepted: 02/15/2016] [Indexed: 12/27/2022]
|
24
|
Hou XL, Chen Y, Yin H, Duan WG. Combination of fasudil and celecoxib promotes the recovery of injured spinal cord in rats better than celecoxib or fasudil alone. Neural Regen Res 2016; 10:1836-40. [PMID: 26807121 PMCID: PMC4705798 DOI: 10.4103/1673-5374.170314] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Resistance mechanisms of rho-associated kinase (ROCK) inhibitors are associated with the enhanced expression of cyclooxygenase-2 (COX-2). The therapeutic effects of ROCK on nervous system diseases might be enhanced by COX-2 inhibitors. This study investigated the synergistic effect of the combined use of the ROCK inhibitor fasudil and a COX-2 inhibitor celecoxib on spinal cord injury in a rat model established by transecting the right half of the spinal cord at T11. Rat models were orally administrated with celecoxib (20 mg/kg) and/or intramuscularly with fasudil (10 mg/kg) for 2 weeks. Results demonstrated that the combined use of celecoxib and fasudil significantly decreased COX-2 and Rho kinase II expression surrounding the lesion site in rats with spinal cord injury, improved the pathomorphology of the injured spinal cord, and promoted the recovery of motor function. Moreover, the effects of the drug combination were better than celecoxib or fasudil alone. This study demonstrated that the combined use of fasudil and celecoxib synergistically enhanced the functional recovery of injured spinal cord in rats.
Collapse
Affiliation(s)
- Xiao-Lin Hou
- Key Laboratory of Molecular Biology for Sinomedicine, Yunnan Key Laboratory for Enthnomedicine, Kunming, Yunnan Province, China
| | - Yan Chen
- Key Laboratory of Molecular Biology for Sinomedicine, Yunnan Key Laboratory for Enthnomedicine, Kunming, Yunnan Province, China
| | - Hua Yin
- Key Laboratory of Molecular Biology for Sinomedicine, Yunnan Key Laboratory for Enthnomedicine, Kunming, Yunnan Province, China
| | - Wei-Gang Duan
- Key Laboratory of Molecular Biology for Sinomedicine, Yunnan Key Laboratory for Enthnomedicine, Kunming, Yunnan Province, China; Initiative Team of Microenvironment, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan Province, China
| |
Collapse
|
25
|
Xin YL, Yu JZ, Yang XW, Liu CY, Li YH, Feng L, Chai Z, Yang WF, Wang Q, Jiang WJ, Zhang GX, Xiao BG, Ma CG. FSD-C10: A more promising novel ROCK inhibitor than Fasudil for treatment of CNS autoimmunity. Biosci Rep 2015; 35:e00247. [PMID: 26223433 PMCID: PMC4721545 DOI: 10.1042/bsr20150032] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 07/07/2015] [Accepted: 07/21/2015] [Indexed: 12/14/2022] Open
Abstract
Rho-Rho kinase (Rho-ROCK) triggers an intracellular signalling cascade that regulates cell survival, death, adhesion, migration, neurite outgrowth and retraction and influences the generation and development of several neurological disorders. Although Fasudil, a ROCK inhibitor, effectively suppressed encephalomyelitis (EAE), certain side effects may limit its clinical use. A novel and efficient ROCK inhibitor, FSD-C10, has been explored. In the present study, we present chemical synthesis and structure of FSD-C10, as well as the relationship between compound concentration and ROCK inhibition. We compared the inhibitory efficiency of ROCKI and ROCK II, the cell cytotoxicity, neurite outgrowth and dendritic formation, neurotrophic factors and vasodilation between Fasudil and FSD-C10. The results demonstrated that FSD-C10, like Fasudil, induced neurite outgrowth of neurons and dendritic formation of BV-2 microglia and enhanced the production of neurotrophic factor brain-derived neurotrophic factor (BDNF), glial cell line-derived neurotrophic factor (GDNF) and neurotrophin-3 (NT-3). However, the cell cytotoxicity and vasodilation of FSD-C10 were relatively small compared with Fasudil. Although Fasudil inhibited both ROCK I and ROCK II, FSD-C10 more selectively suppressed ROCK II, but not ROCK I, which may be related to vasodilation insensitivity and animal mortality. Thus, FSD-C10 may be a safer and more promising novel ROCK inhibitor than Fasudil for the treatment of several neurological disorders.
Collapse
Affiliation(s)
- Yan-Le Xin
- 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
| | - Xin-Wang Yang
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong 037009, China
| | - Chun-Yun Liu
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong 037009, China
| | - Yan-Hua Li
- 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
| | - Wan-Fang Yang
- "2011" Collaborative Innovation Center/Research Center of Neurobiology, Shanxi University of Traditional Chinese Medicine, Taiyuan 030619, China
| | - Qing Wang
- "2011" Collaborative Innovation Center/Research Center of Neurobiology, Shanxi University of Traditional Chinese Medicine, Taiyuan 030619, China
| | - Wei-Jia Jiang
- "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, U.S.A
| | - Bao-Guo Xiao
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200040, 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
|
26
|
O'Shea RD, Lau CL, Zulaziz N, Maclean FL, Nisbet DR, Horne MK, Beart PM. Transcriptomic analysis and 3D bioengineering of astrocytes indicate ROCK inhibition produces cytotrophic astrogliosis. Front Neurosci 2015; 9:50. [PMID: 25750613 PMCID: PMC4335181 DOI: 10.3389/fnins.2015.00050] [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] [Received: 09/26/2014] [Accepted: 02/04/2015] [Indexed: 01/07/2023] Open
Abstract
Astrocytes provide trophic, structural and metabolic support to neurons, and are considered genuine targets in regenerative neurobiology, as their phenotype arbitrates brain integrity during injury. Inhibitors of Rho kinase (ROCK) cause stellation of cultured 2D astrocytes, increased L-glutamate transport, augmented G-actin, and elevated expression of BDNF and anti-oxidant genes. Here we further explored the signposts of a cytotrophic, “healthy” phenotype by data-mining of our astrocytic transcriptome in the presence of Fasudil. Gene expression profiles of motor and autophagic cellular cascades and inflammatory/angiogenic responses were all inhibited, favoring adoption of an anti-migratory phenotype. Like ROCK inhibition, tissue engineered bioscaffolds can influence the extracellular matrix. We built upon our evidence that astrocytes maintained on 3D poly-ε-caprolactone (PCL) electrospun scaffolds adopt a cytotrophic phenotype similar to that produced by Fasudil. Using these procedures, employing mature 3D cultured astrocytes, Fasudil (100 μM) or Y27632 (30 μM) added for the last 72 h of culture altered arborization, which featured numerous additional minor processes as shown by GFAP and AHNAK immunolabelling. Both ROCK inhibitors decreased F-actin, but increased G-actin labeling, indicative of disassembly of actin stress fibers. ROCK inhibitors provide additional beneficial effects for bioengineered 3D astrocytes, including enlargement of the overall arbor. Potentially, the combined strategy of bio-compatible scaffolds with ROCK inhibition offers unique advantages for the management of glial scarring. Overall these data emphasize that manipulation of the astrocyte phenotype to achieve a “healthy biology” offers new hope for the management of inflammation in neuropathologies.
Collapse
Affiliation(s)
- Ross D O'Shea
- Department of Physiology, Anatomy and Microbiology, La Trobe University Bundoora, VIC, Australia
| | - Chew L Lau
- Florey Institute of Neuroscience and Mental Health, University of Melbourne Parkville, VIC, Australia
| | - Natasha Zulaziz
- Department of Physiology, Anatomy and Microbiology, La Trobe University Bundoora, VIC, Australia
| | - Francesca L Maclean
- Research School of Engineering, The Australian National University Canberra, ACT, Australia
| | - David R Nisbet
- Research School of Engineering, The Australian National University Canberra, ACT, Australia
| | - Malcolm K Horne
- Florey Institute of Neuroscience and Mental Health, University of Melbourne Parkville, VIC, Australia ; Department of Neurology, St. Vincent's Hospital Fitzroy, VIC, Australia
| | - Philip M Beart
- Florey Institute of Neuroscience and Mental Health, University of Melbourne Parkville, VIC, Australia
| |
Collapse
|
27
|
Huang W, Li Y, Lin Y, Ye X, Zang D. Effects of leukemia inhibitory factor and basic fibroblast growth factor on free radicals and endogenous stem cell proliferation in a mouse model of cerebral infarction. Neural Regen Res 2015; 7:1469-74. [PMID: 25657681 PMCID: PMC4308777 DOI: 10.3969/j.issn.1673-5374.2012.19.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Accepted: 04/23/2012] [Indexed: 11/24/2022] Open
Abstract
The present study established a mouse model of cerebral infarction by middle cerebral artery occlusion, and monitored the effect of 25 μg/kg leukemia inhibitory factor and (or) basic fibroblast growth factor administration 2 hours after model establishment. Results showed that following administration, the number of endogenous neural stem cells in the infarct area significantly increased, malondialdehyde content in brain tissue homogenates significantly decreased, nitric oxide content, glutathione peroxidase and superoxide dismutase activity significantly elevated, and mouse motor function significantly improved as confirmed by the rotarod and bar grab tests. In particular, the effect of leukemia inhibitory factor in combination with basic fibroblast growth factor was the most significant. Results indicate that leukemia inhibitory factor and basic fibroblast growth factor can improve the microenvironment after cerebral infarction by altering free radical levels, improving the quantity of endogenous neural stem cells, and promoting neurological function of mice with cerebral infarction.
Collapse
Affiliation(s)
- Weihui Huang
- Department of Neurology, Tianjin First Center Hospital, Tianjin Medical University, Tianjin 300192, China
| | - Yadan Li
- Department of Neurology, Tianjin First Center Hospital, Tianjin Medical University, Tianjin 300192, China
| | - Yufeng Lin
- Department of Neurology, Tianjin First Center Hospital, Tianjin Medical University, Tianjin 300192, China
| | - Xue Ye
- Department of Neurology, Tianjin First Center Hospital, Tianjin Medical University, Tianjin 300192, China
| | - Dawei Zang
- Department of Neurology, Tianjin First Center Hospital, Tianjin Medical University, Tianjin 300192, China
| |
Collapse
|
28
|
Abstract
Rho kinase (ROCK) is a major downstream effector of the small GTPase RhoA. ROCK family, consisting of ROCK1 and ROCK2, plays central roles in the organization of actin cytoskeleton and is involved in a wide range of fundamental cellular functions, such as contraction, adhesion, migration, proliferation, and apoptosis. Due to the discovery of effective inhibitors, such as fasudil and Y27632, the biological roles of ROCK have been extensively explored with particular attention on the cardiovascular system. In many preclinical models of cardiovascular diseases, including vasospasm, arteriosclerosis, hypertension, pulmonary hypertension, stroke, ischemia-reperfusion injury, and heart failure, ROCK inhibitors have shown a remarkable efficacy in reducing vascular smooth muscle cell hypercontraction, endothelial dysfunction, inflammatory cell recruitment, vascular remodeling, and cardiac remodeling. Moreover, fasudil has been used in the clinical trials of several cardiovascular diseases. The continuing utilization of available pharmacological inhibitors and the development of more potent or isoform-selective inhibitors in ROCK signaling research and in treating human diseases are escalating. In this review, we discuss the recent molecular, cellular, animal, and clinical studies with a focus on the current understanding of ROCK signaling in cardiovascular physiology and diseases. We particularly note that emerging evidence suggests that selective targeting ROCK isoform based on the disease pathophysiology may represent a novel therapeutic approach for the disease treatment including cardiovascular diseases.
Collapse
|
29
|
Exploration on Mechanism of a New Type of Melatonin Receptor Agonist Neu-p11 in Hypoxia–Reoxygenation Injury of Myocardial Cells. Cell Biochem Biophys 2014; 70:999-1003. [DOI: 10.1007/s12013-014-0009-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
30
|
Gibson CL, Srivastava K, Sprigg N, Bath PMW, Bayraktutan U. Inhibition of Rho-kinase protects cerebral barrier from ischaemia-evoked injury through modulations of endothelial cell oxidative stress and tight junctions. J Neurochem 2014; 129:816-26. [PMID: 24528233 DOI: 10.1111/jnc.12681] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 01/20/2014] [Accepted: 02/07/2014] [Indexed: 12/22/2022]
Abstract
Ischaemic strokes evoke blood-brain barrier (BBB) disruption and oedema formation through a series of mechanisms involving Rho-kinase activation. Using an animal model of human focal cerebral ischaemia, this study assessed and confirmed the therapeutic potential of Rho-kinase inhibition during the acute phase of stroke by displaying significantly improved functional outcome and reduced cerebral lesion and oedema volumes in fasudil- versus vehicle-treated animals. Analyses of ipsilateral and contralateral brain samples obtained from mice treated with vehicle or fasudil at the onset of reperfusion plus 4 h post-ischaemia or 4 h post-ischaemia alone revealed these benefits to be independent of changes in the activity and expressions of oxidative stress- and tight junction-related parameters. However, closer scrutiny of the same parameters in brain microvascular endothelial cells subjected to oxygen-glucose deprivation ± reperfusion revealed marked increases in prooxidant NADPH oxidase enzyme activity, superoxide anion release and in expressions of antioxidant enzyme catalase and tight junction protein claudin-5. Cotreatment of cells with Y-27632 prevented all of these changes and protected in vitro barrier integrity and function. These findings suggest that inhibition of Rho-kinase after acute ischaemic attacks improves cerebral integrity and function through regulation of endothelial cell oxidative stress and reorganization of intercellular junctions. Inhibition of Rho-kinase (ROCK) activity in a mouse model of human ischaemic stroke significantly improved functional outcome while reducing cerebral lesion and oedema volumes compared to vehicle-treated counterparts. Studies conducted with brain microvascular endothelial cells exposed to OGD ± R in the presence of Y-27632 revealed restoration of intercellular junctions and suppression of prooxidant NADPH oxidase activity as important factors in ROCK inhibition-mediated BBB protection.
Collapse
Affiliation(s)
- Claire L Gibson
- School of Psychology, University of Leicester, Leicester, UK
| | | | | | | | | |
Collapse
|
31
|
Bolognin S, Lorenzetto E, Diana G, Buffelli M. The potential role of rho GTPases in Alzheimer's disease pathogenesis. Mol Neurobiol 2014; 50:406-22. [PMID: 24452387 DOI: 10.1007/s12035-014-8637-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 01/02/2014] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is characterized by a wide loss of synapses and dendritic spines. Despite extensive efforts, the molecular mechanisms driving this detrimental alteration have not yet been determined. Among the factors potentially mediating this loss of neuronal connectivity, the contribution of Rho GTPases is of particular interest. This family of proteins is classically considered a key regulator of actin cytoskeleton remodeling and dendritic spine maintenance, but new insights into the complex dynamics of its regulation have recently determined how its signaling cascade is still largely unknown, both in physiological and pathological conditions. Here, we review the growing evidence supporting the potential involvement of Rho GTPases in spine loss, which is a unanimously recognized hallmark of early AD pathogenesis. We also discuss some new insights into Rho GTPase signaling framework that might explain several controversial results that have been published. The study of the connection between AD and Rho GTPases represents a quite unchartered avenue that holds therapeutic potential.
Collapse
Affiliation(s)
- Silvia Bolognin
- Department of Neurological and Movement Sciences, Section of Physiology, University of Verona, Strada le Grazie 8, 37134, Verona, Italy,
| | | | | | | |
Collapse
|
32
|
Schuster A, Klotz M, Schwab T, Lilischkis R, Schneider A, Schäfer KH. Granulocyte-colony stimulating factor: a new player for the enteric nervous system. Cell Tissue Res 2013; 355:35-48. [PMID: 24253464 DOI: 10.1007/s00441-013-1744-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 10/10/2013] [Indexed: 11/27/2022]
Abstract
The enteric nervous system (ENS) controls and modulates gut motility and responds to food intake and to internal and external stimuli such as toxins or inflammation. Its plasticity is maintained throughout life by neural progenitor cells within the enteric stem cell niche. Granulocyte-colony stimulating factor (G-CSF) is known to act not only on cells of the immune system but also on neurons and neural progenitors in the central nervous system (CNS). Here, we demonstrate, for the first time, that G-CSF receptor is present on enteric neurons and progenitors and that G-CSF plays a role in the expansion and differentiation of enteric neural progenitor cells. Cultured mouse ENS-neurospheres show increased expansion with increased G-CSF concentrations, in contrast to CNS-derived spheres. In cultures from differentiated ENS- and CNS-neurospheres, neurite outgrowth density is enhanced depending on the amount of G-CSF in the culture. G-CSF might be an important factor in the regeneration and differentiation of the ENS and might be a useful tool for the investigation and treatment of ENS disorders.
Collapse
Affiliation(s)
- Anne Schuster
- Department of Biotechnology, University of Applied Sciences Kaiserslautern, Amerikastraße 1, 66482, Zweibrücken, Germany
| | | | | | | | | | | |
Collapse
|
33
|
Vadodaria KC, Jessberger S. Maturation and integration of adult born hippocampal neurons: signal convergence onto small Rho GTPases. Front Synaptic Neurosci 2013; 5:4. [PMID: 23986696 PMCID: PMC3752586 DOI: 10.3389/fnsyn.2013.00004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 07/29/2013] [Indexed: 01/28/2023] Open
Abstract
Adult neurogenesis, restricted to specific regions in the mammalian brain, represents one of the most interesting forms of plasticity in the mature nervous system. Adult-born hippocampal neurons play important roles in certain forms of learning and memory, and altered hippocampal neurogenesis has been associated with a number of neuropsychiatric diseases such as major depression and epilepsy. Newborn neurons go through distinct developmental steps, from a dividing neurogenic precursor to a synaptically integrated mature neuron. Previous studies have uncovered several molecular signaling pathways involved in distinct steps of this maturational process. In this context, the small Rho GTPases, Cdc42, Rac1, and RhoA have recently been shown to regulate the morphological and synaptic maturation of adult-born dentate granule cells in vivo. Distinct upstream regulators, including growth factors that modulate maturation and integration of newborn neurons have been shown to also recruit the small Rho GTPases. Here we review recent findings and highlight the possibility that small Rho GTPases may act as central assimilators, downstream of critical input onto adult-born hippocampal neurons contributing to their maturation and integration into the existing dentate gyrus (DG) circuitry.
Collapse
Affiliation(s)
- Krishna C Vadodaria
- Brain Research Institute, University of Zurich Zurich, Switzerland ; Neuroscience Center Zurich, University of Zurich and ETH Zurich Zurich, Switzerland
| | | |
Collapse
|
34
|
Wang HL, Hu SH, Chou AH, Wang SS, Weng YH, Yeh TH. H1152 promotes the degradation of polyglutamine-expanded ataxin-3 or ataxin-7 independently of its ROCK-inhibiting effect and ameliorates mutant ataxin-3-induced neurodegeneration in the SCA3 transgenic mouse. Neuropharmacology 2013; 70:1-11. [DOI: 10.1016/j.neuropharm.2013.01.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 12/14/2012] [Accepted: 01/09/2013] [Indexed: 12/18/2022]
|
35
|
Lemmens R, Jaspers T, Robberecht W, Thijs VN. Modifying expression of EphA4 and its downstream targets improves functional recovery after stroke. Hum Mol Genet 2013; 22:2214-20. [PMID: 23418304 DOI: 10.1093/hmg/ddt073] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Functional recovery after stroke varies greatly between patients, potentially due to differences in gene expression. Several processes like angiogenesis, neurogenesis, axonal reorganization and synaptic plasticity act in concert to restore neurological functions. The ephrin family has known roles in all these processes. EphA4 is the most abundant ephrin receptor in the nervous system. Therefore, we investigated whether EphA4 affects functional recovery from stroke, and evaluated the potential of this receptor as a therapeutic target. Motor recovery after photothrombotic stroke was studied in transgenic mice in which expression of EphA4 was reduced. Furthermore, blocking a downstream target of EphA4, ROCK (Rho-associated kinase), by two different compounds was evaluated in the same model. Motor recovery after photothrombotic stroke was markedly enhanced in transgenic mice with reduced levels of EphA4, whereas infarct sizes were similar compared with non-transgenic controls. Pharmacological inhibition of the EphA4 signaling cascade using two ROCK inhibitors,Y-27632 and fasudil, improved motor function of mice after stroke. Infarct size was comparable in all groups studied, suggesting that the benefit obtained by EphA4 inhibition is not neuroprotective in nature but due to an effect on the mechanisms underlying recovery. Our findings show that reduction of EphA4 improves motor function after experimental stroke and demonstrate that ROCK inhibition is a promising therapeutic strategy to enhance recovery after ischemic stroke.
Collapse
Affiliation(s)
- Robin Lemmens
- Laboratory of Neurobiology, Vesalius Research Center, VIB, Leuven, Belgium.
| | | | | | | |
Collapse
|
36
|
Ding J, Li QY, Yu JZ, Wang X, Lu CZ, Ma CG, Xiao BG. Carbamylated erythropoietin ameliorates hypoxia-induced cognitive and behavioral defects with the generation of choline acetyltransferase-positive neurons. J Neurosci Res 2012; 91:73-82. [PMID: 23074165 DOI: 10.1002/jnr.23124] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 06/28/2012] [Accepted: 07/10/2012] [Indexed: 11/09/2022]
Abstract
Carbamylated erythropoietin (CEPO) is attracting widespread interest because of its neuroprotective effects without influencing erythropoiesis. Here we show that CEPO, unlike EPO, does not stimulate erythropoiesis. Both CEPO and EPO inhibit the death/apoptosis of neurons in the hypoxic model of primary neurons and induce neuron proliferation and differentiation in hypoxic mice. Hypoxic mice show apparent memory deficits at 3 and 30 days after hypoxia. The administration of CEPO/EPO significantly improves cognitive and behavioral defects after hypoxic insults. Further investigation shows that CEPO/EPO induces neuron proliferation and differentiation and promotes the generation of choline acetyltransferase (ChAT)(+) neurons in hypoxic mice. Phosphorylated AKT was colabeled with ChAT(+) neurons and coexpressed in bromodeoxyuridine-positive cells, suggesting that the PI3K/AKT pathway may play a pivotal role in CEPO/EPO-cholinergic neuron generation. These results reveal that CEPO/EPO ameliorates hypoxia-induced cognitive and behavioral defects possibly through the generation of ChAT-positive neurons.
Collapse
Affiliation(s)
- Jing Ding
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
37
|
Keung AJ, de Juan-Pardo EM, Schaffer DV, Kumar S. Rho GTPases mediate the mechanosensitive lineage commitment of neural stem cells. Stem Cells 2012; 29:1886-97. [PMID: 21956892 DOI: 10.1002/stem.746] [Citation(s) in RCA: 166] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Adult neural stem cells (NSCs) play important roles in learning and memory and are negatively impacted by neurological disease. It is known that biochemical and genetic factors regulate self-renewal and differentiation, and it has recently been suggested that mechanical and solid-state cues, such as extracellular matrix (ECM) stiffness, can also regulate the functions of NSCs and other stem cell types. However, relatively little is known of the molecular mechanisms through which stem cells transduce mechanical inputs into fate decisions, the extent to which mechanical inputs instruct fate decisions versus select for or against lineage-committed blast populations, or the in vivo relevance of mechanotransductive signaling molecules in native stem cell niches. Here we demonstrate that ECM-derived mechanical signals act through Rho GTPases to activate the cellular contractility machinery in a key early window during differentiation to regulate NSC lineage commitment. Furthermore, culturing NSCs on increasingly stiff ECMs enhances RhoA and Cdc42 activation, increases NSC stiffness, and suppresses neurogenesis. Likewise, inhibiting RhoA and Cdc42 or downstream regulators of cellular contractility rescues NSCs from stiff matrix- and Rho GTPase-induced neurosuppression. Importantly, Rho GTPase expression and ECM stiffness do not alter proliferation or apoptosis rates indicating that an instructive rather than selective mechanism modulates lineage distributions. Finally, in the adult brain, RhoA activation in hippocampal progenitors suppresses neurogenesis, analogous to its effect in vitro. These results establish Rho GTPase-based mechanotransduction and cellular stiffness as biophysical regulators of NSC fate in vitro and RhoA as an important regulatory protein in the hippocampal stem cell niche.
Collapse
Affiliation(s)
- Albert J Keung
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, California 94720-3220, USA
| | | | | | | |
Collapse
|
38
|
Lau CL, Perreau VM, Chen MJ, Cate HS, Merlo D, Cheung NS, O'Shea RD, Beart PM. Transcriptomic profiling of astrocytes treated with the Rho kinase inhibitor fasudil reveals cytoskeletal and pro-survival responses. J Cell Physiol 2012; 227:1199-211. [PMID: 21604263 DOI: 10.1002/jcp.22838] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Inhibitors of Rho kinase (ROCK) have potential for management of neurological disorders by inhibition of glial scarring. Since astrocytes play key roles in brain physiology and pathology, we determined changes in the astrocytic transcriptome produced by the ROCK inhibitor Fasudil to obtain mechanistic insights into its beneficial action during brain injury. Cultured murine astrocytes were treated with Fasudil (100 µM) and morphological analyses revealed rapid stellation by 1 h and time-dependent (2-24 h) dissipation of F-actin-labelled stress fibres. Microarray analyses were performed on RNA and the time-course of global gene profiling (2, 6, 12 and 24 h) provided a comprehensive description of transcriptomic changes. Hierarchical clustering of differentially expressed genes and analysis for over-represented gene ontology groups using the DAVID database focused attention on Fasudil-induced changes to major biological processes regulating cellular shape and motility (actin cytoskeleton, axon guidance, transforming growth factor-β (TGFβ) signalling and tight junctions). Bioinformatic analyses of transcriptomic changes revealed how these biological processes contributed to changes in astrocytic motility and cytoskeletal reorganisation. Here genes associated with extracellular matrix were also involved, but unexpected was a subset of alterations (EAAT2, BDNF, anti-oxidant species, metabolic and signalling genes) indicative of adoption by astrocytes of a pro-survival phenotype. Expression profiles of key changes with Fasudil and another ROCK inhibitor Y27632 were validated by real-time PCR. Although effects of ROCK inhibition have been considered to be primarily cytoskeletal via reduction of glial scarring, we demonstrate additional advantageous actions likely to contribute to their ameliorative actions in brain injury.
Collapse
Affiliation(s)
- Chew L Lau
- Florey Neuroscience Institutes, University of Melbourne, Parkville, VIC, Australia
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Tönges L, Koch JC, Bähr M, Lingor P. ROCKing Regeneration: Rho Kinase Inhibition as Molecular Target for Neurorestoration. Front Mol Neurosci 2011; 4:39. [PMID: 22065949 PMCID: PMC3207219 DOI: 10.3389/fnmol.2011.00039] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Accepted: 10/16/2011] [Indexed: 12/31/2022] Open
Abstract
Regenerative failure in the CNS largely depends on pronounced growth inhibitory signaling and reduced cellular survival after a lesion stimulus. One key mediator of growth inhibitory signaling is Rho-associated kinase (ROCK), which has been shown to modulate growth cone stability by regulation of actin dynamics. Recently, there is accumulating evidence the ROCK also plays a deleterious role for cellular survival. In this manuscript we illustrate that ROCK is involved in a variety of intracellular signaling pathways that comprise far more than those involved in neurite growth inhibition alone. Although ROCK function is currently studied in many different disease contexts, our review focuses on neurorestorative approaches in the CNS, especially in models of neurotrauma. Promising strategies to target ROCK by pharmacological small molecule inhibitors and RNAi approaches are evaluated for their outcome on regenerative growth and cellular protection both in preclinical and in clinical studies.
Collapse
Affiliation(s)
- Lars Tönges
- Department of Neurology, University Medicine Göttingen Göttingen, Germany
| | | | | | | |
Collapse
|
40
|
Lau CL, O'Shea RD, Broberg BV, Bischof L, Beart PM. The Rho kinase inhibitor Fasudil up-regulates astrocytic glutamate transport subsequent to actin remodelling in murine cultured astrocytes. Br J Pharmacol 2011; 163:533-45. [PMID: 21309758 DOI: 10.1111/j.1476-5381.2011.01259.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE Glutamate transporters play a major role in maintaining brain homeostasis and the astrocytic transporters, EAAT1 and EAAT2, are functionally dominant. Astrocytic excitatory amino acid transporters (EAATs) play important roles in various neuropathologies wherein astrocytes undergo cytoskeletal changes. Astrocytic plasticity is well documented, but the interface between EAAT function, actin and the astrocytic cytoskeleton is poorly understood. Because Rho kinase (ROCK) is a key determinant of actin polymerization, we investigated the effects of ROCK inhibitors on EAAT activity and astrocytic morphology. EXPERIMENTAL APPROACH The functional activity of glutamate transport was determined in murine cultured astrocytes after exposure to the ROCK inhibitors Fasudil (HA-1077) and Y27632 using biochemical, molecular and morphological approaches. Cytochemical analyses assessed changes in astrocytic morphology, F-/G-actin, and localizations of EAAT1/2. RESULTS Fasudil and Y27632 increased [(3)H]-D-aspartate (D-Asp) uptake into astrocytes, and the action of Fasudil was time-dependent and concentration-related. The rapid stellation of astrocytes (glial fibrillary acidic protein immunocytochemistry) induced by Fasudil was accompanied by reduced phalloidin staining of F-actin and increased V(max) for [(3)H]-D-Asp uptake. Immunoblotting after biotinylation demonstrated that Fasudil increased the expression of EAAT1 and EAAT2 on the cell surface. Immunocytochemistry indicated that Fasudil induced prominent labelling of astrocytic processes by EAAT1/2. CONCLUSION AND IMPLICATIONS These data show for the first time that ROCK plays a major role in determining the cell surface expression of EAAT1/2, providing new evidence for an association between transporter function and astrocytic phenotype. ROCK inhibitors, via the actin cytoskeleton, effect a consequent elevation of glutamate transporter function - this activity profile may contribute to their beneficial actions in neuropathologies.
Collapse
Affiliation(s)
- C L Lau
- Molecular Neuropharmacology, Florey Neuroscience Institutes, Parkville, Australia
| | | | | | | | | |
Collapse
|
41
|
Leong SY, Faux CH, Turbic A, Dixon KJ, Turnley AM. The Rho Kinase Pathway Regulates Mouse Adult Neural Precursor Cell Migration. Stem Cells 2011; 29:332-43. [DOI: 10.1002/stem.577] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
42
|
The evolving landscape of neuroinflammation after neonatal hypoxia-ischemia. ACTA NEUROCHIRURGICA. SUPPLEMENT 2011; 111:93-100. [PMID: 21725737 DOI: 10.1007/978-3-7091-0693-8_15] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Hypoxic-ischemic brain injury remains a leading cause of mortality and morbidity in neonates. The inflammatory response, which is characterized in part by activation of local immune cells, has been implicated as a core component for the progression of damage to the immature brain following hypoxia-ischemia (HI). However, mounting evidence implicates circulating immune cells recruited to the site of damage as orchestrators of neuron-glial interactions and perpetuators of secondary brain injury. This suggests that re-directing our attention from the local inflammatory response toward the molecular mediators believed to link brain-immune cell interactions may be a more effective approach to mitigating the inflammatory sequelae of perinatal HI. In this review, we focus our attention on cyclooxygenase-2, a mediator by which peripheral immune cells may modulate signaling pathways in the brain that lead to a worsened outcome. Additionally, we present an overview of emerging therapeutic modalities that target mechanisms of neuroinflammation in the hypoxic-ischemic neonate.
Collapse
|
43
|
Nunes KP, Rigsby CS, Webb RC. RhoA/Rho-kinase and vascular diseases: what is the link? Cell Mol Life Sci 2010; 67:3823-36. [PMID: 20668910 PMCID: PMC2996825 DOI: 10.1007/s00018-010-0460-1] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2010] [Revised: 07/07/2010] [Accepted: 07/08/2010] [Indexed: 12/29/2022]
Abstract
RhoA/Rho-kinase pathway plays an important role in many pathological conditions. RhoA participates in the regulation of smooth muscle tone and activates many downstream kinases. The best characterized are the serine/threonine kinase isoforms (Rho-kinase or ROCK), ROCKα/ROCK2 and ROCKβ/ROCK1. ROCK is necessary for diverse functions such as local blood flow, arterial/pulmonary blood pressure, airway resistance and intestinal peristalsis. ROCK activation permits actin/myosin interactions and smooth muscle cells contraction by maintaining the activity of myosin light-chain kinase, independently of the free cytosolic calcium level. The sensitization of smooth muscle myofilaments to calcium has been implicated in many pathological states, such as hypertension, diabetes, heart attack, stroke, pulmonary hypertension, erectile dysfunction, and cancer. The focus of this review is on the involvement of RhoA/Rho-kinase in diseases. We will briefly describe the ROCK isoforms and the role of RhoA/Rho-kinase in the vasculature, before exploring the most recent findings regarding this pathway and various diseases.
Collapse
Affiliation(s)
- Kenia Pedrosa Nunes
- Department of Physiology, Medical College of Georgia, Augusta, GA 30912, USA.
| | | | | |
Collapse
|