Astrocytes respond to all forms of CNS insult by a process commonly referred as reactive astrogliosis characterized as hypertrophy of cellular processes and upregulation of intermediated filament proteins such as glial fibrillary acidic protein (GFAP). Compelling evidence has indicated that reactive astrogliosis is not simply an all-or-none phenomenon but, rather, a finely tuned continuum of molecular, cellular, and functional changes that range from subtle alternations in gene expression to scar formation. Recent research findings have also suggested that reactive astrogliosis may exert both beneficial and detrimental effects in a temporal dependent manner regulated by specific molecular signaling cascades. Increasing evidence has indicated that p38 plays a role in many other biological functions at the CNS. The activation of p38 signaling releases pro-inflammatory cytokines that are known to be involved in various CNS disorders including ischemic stroke, Alzheimer's disease, Parkinson's disease, multiple sclerosis, neuropathic pain and depression. The role of p38 MAPK in peripheral immune and inflammatory response has been extensive studied. However, the action of p38 in astrogliosis, the major component of neuroinflammatory response in CNS, has largely unknown. Our preliminary study demonstrated an activation of p38 signaling in reactive astrogliosis in the glial scar area in a mouse ischemic stroke model. Our laboratory has established a novel transgenic mouse line (hGFAP Cre/p38 loxP) in which the hGFAP promoter-driven Cre transgene deletes the floxed p38?ene and a downregulation of GFAP expression was found in primary astrocytes derived from the transgenic mice. Consistently, attenuation of reactive astrogliosis was observed upon p38 inhibition in primary astrocyte cultures. Reduction of ischemic stroke induced astrogliosis was observed in the hGFAP cre / p38 loxp mice as compared with wild type littermates. As Cre-mediated recombination is irreversible, embryonic expression in neural precursor cells directed by the GFAP promoter have resulted in substantial Cre-mediated recombination in mature neurons as well as glial cell in all GFAP-Cre mice characterized to date. In addition, the constitutive p38 knockout will not be able to study the role of p38 in astrogliosis in a temporal dependent manner. Therefore, there is a clear need of an inducible astrocyte specific p38 knockout mouse line. In this application, we proposed to establish an inducible astrocyte specific p38 knockout mouse line, characterize the inducible gene recombination and p38 knockout in the established transgenic mouse line, and determine the spatial and temporal dependent action of p38 knockout on glial scar formation and functional recovery after ischemic stroke.
Specific Aim 1 is to establish the inducible astrocyte specific p38 knockout, hGFAP Cre-ERT2 / p38 loxP, mouse line.
Specific Aim 2 is to determine the spatial and temporal dependent action of p38 knockout on glial scar formation and functional recovery after ischemic stroke using the established hGFAP Cre-ERT2 / p38 loxP mouse line. We predict that, in the established hGFAP Cre-ERT2 / p38 loxP mouse line, tamoxifen inducible gene recombination will be achieved predominantly in astrocyte with a minimal in the neural stem cell at subventricular zone. We expect that induction of p38 MAPK knockout in astrocyte after ischemic stroke attenuates glial scar formation, ameliorates the inhibitory action of glial scar on axonal sprouting and synaptogenesis, and enhances functional recovery.

Public Health Relevance

Astrocytes respond to all forms of CNS insult by a process commonly referred as reactive astrogliosis which may exert both beneficial and detrimental effects in a context-dependent manner regulated by specific molecular signaling cascades. In this application, we proposed to establish an inducible astrocyte specific p38 knockout mouse line, characterize the inducible gene recombination and p38 knockout in the established transgenic mouse line, and determine the spatial and temporal dependent action of p38 knockout on glial scar formation and functional recovery after ischemic stroke. The proposed study will develop a novel mouse line to study the role of p38 signaling in reactive astrogliosis i term of various neurological disorders including ischemic stroke.

Agency
National Institute of Health (NIH)
Institute
National Institute of Neurological Disorders and Stroke (NINDS)
Type
Exploratory/Developmental Grants (R21)
Project #
1R21NS087209-01A1
Application #
8822444
Study Section
Brain Injury and Neurovascular Pathologies Study Section (BINP)
Program Officer
Bosetti, Francesca
Project Start
2014-09-15
Project End
2016-08-31
Budget Start
2014-09-15
Budget End
2015-08-31
Support Year
1
Fiscal Year
2014
Total Cost
Indirect Cost
Name
University of North Texas
Department
Pharmacology
Type
Graduate Schools
DUNS #
City
Fort Worth
State
TX
Country
United States
Zip Code
76107
Yang, Shao-Hua; Lou, Min; Luo, Benyan et al. (2018) Precision Medicine for Ischemic Stroke, Let Us Move Beyond Time Is Brain. Transl Stroke Res 9:93-95
Li, Wenjun; Roy Choudhury, Gourav; Winters, Ali et al. (2018) Hyperglycemia Alters Astrocyte Metabolism and Inhibits Astrocyte Proliferation. Aging Dis 9:674-684
Lin, Shao-Peng; Li, Wenjun; Winters, Ali et al. (2018) Artemisinin Prevents Glutamate-Induced Neuronal Cell Death Via Akt Pathway Activation. Front Cell Neurosci 12:108
Zhang, Hongxia; Sun, Fen; Wang, Jixian et al. (2017) Combining Injectable Plasma Scaffold with Mesenchymal Stem/Stromal Cells for Repairing Infarct Cavity after Ischemic Stroke. Aging Dis 8:203-214
Cai, Bin; Li, Wenjun; Mao, XiaoOu et al. (2016) Neuroglobin Overexpression Inhibits AMPK Signaling and Promotes Cell Anabolism. Mol Neurobiol 53:1254-65
Roy Choudhury, Gourav; Winters, Ali; Rich, Ryan M et al. (2015) Methylene blue protects astrocytes against glucose oxygen deprivation by improving cellular respiration. PLoS One 10:e0123096
Xie, Luokun; Yang, Shao-Hua (2015) Interaction of astrocytes and T cells in physiological and pathological conditions. Brain Res 1623:63-73
He, Weiliang; Liu, Ran; Yang, Shao-Hua et al. (2015) Chemotherapeutic effect of tamoxifen on temozolomide-resistant gliomas. Anticancer Drugs 26:293-300
Li, W; Huang, R; Chen, Z et al. (2014) PTEN degradation after ischemic stroke: a double-edged sword. Neuroscience 274:153-61