Ischemic brain injury causes death and long-term disability in patients who suffer cardiac arrest and embolism stroke in the United States and in our VA community. Ischemia-induced neuronal hyperactivity in the affected neurons is a key step in the development of neurodegeneration as it leads to cell death, irreversible loss and reorganization of neuronal circuits, and eventually neuronal deficiency. Immediately after Ischemia, cell membrane depolarization, excess glutamate secretion, overactivity of ionotropic glutamate receptor, and loss of GABA signaling are thought to cause neuronal hyperactivity. GABA-B-Rs (R1 and R2) are members of the family C of the G-protein coupled receptor (GPCR) superfamily, which also includes the extracellular Ca2+- sensing receptor (CaSR). GABA-B-Rs generally function in the form of heterodimer comprised of GABA-B-R1 and GABA-B-R2 subunit that is required for stable cell-surface expression and signaling of the receptor complex to produce inhibitory neuronal input and prevent neuronal overactivity. The GABA-B-Rs and the CaSR are co-expressed in many regions of the brain, including hippocampus. Unlike the GABA-B-R1/R2 heterodimer, the CaSR can function in the form of homodimer to exert excitatory signaling responses -- activation of Ca2+ and non-selective cation channels, stimulation of phospholipase C, increases in [Ca2+]i, and increasing cell excitability in neurons and other cell systems. GABA-B-R1 can heterodimerize with CaSR and suppress the total and cell-surface expression of CaSR protein and its signaling responses in transfected HEK- 293 cells. Contrarily, knocking out GABA-B-R1 gene in hippocampal neurons up-regulates CaSR expression. We further observed CaSR overexpression in ischemic hippocampal neurons, which showed reduced GABA- B-R1 expression, suggesting a counteracting interaction between the CaSR and GABA-B-R1 expression. The increased expression and activity of CaSR could contribute to the hyperactivity of the ischemic neurons via its own excitatory actions and/or its ability to interfere with the formation of GABA-B-R1/R2 heterodimers, therefore reducing GABA responses. In supporting the latter notion, we found that in the HippCaSR-KO mice, which have their CaSR genes deleted in hippocampal neurons, ischemia no longer inhibited the GABA-B-R1 expression or caused cell death. We hypothesize that ischemia-induced CaSR overexpression causes neuronal hyperactivity and cell death by stimulating CaSR-mediated signaling responses and by inhibiting GABA-B-R1 expression via stoichiometric competition for binding to GABA-B-R2, and that blocking the expression or activity of CaSR together with enhancement of GABA-B-R signaling are required for optimal neuroprotection against ischemic injuries. Our proposal will (1) determine whether deleting CaSR gene or blocking CaSR activity by specific antagonists (or calcilytics) protects against the ischemia-induced neuronal injury and blunts the inhibitory effect of ischemia on the expression, trafficking, and dimerization of GABA-B-R1/R2 and (2) determine whether sustaining GABA-B-R1 expression is required for neuroprotection against cerebral ischemia and whether a combined therapy with calcilytics and GABA-B-R1 agonists further enhances neuroprotection against ischemia-induced brain injury. The successful completion of the study will establish a novel pathway that produces ischemia-induced neuronal injury and will develop a new therapy for treating diseases due to ischemic brain injury.

Public Health Relevance

Ischemic brain injury produces focal or generalized neuronal hyperexcitability and Ca2+ overload in the cytoplasm of the affected neurons that lead to cell death, tissue scarring (gliosis), reorganization of neuronal networks, and eventually neuronal deficiency and long-term disability. We found that down-regulation of the GABA B receptors (GABA-B-Rs) and overexpression of the extracellular calcium-sensing receptor (CaSR) play a critical role in promoting neuronal hyperexcitability and Ca2+ overload in ischemic brains. This proposal will utilize novel mouse models and neuron cultures to determine whether the CaSR overexpression causes neuronal injury by activating intrinsic excitatory responses and by suppressing the inhibitory GABA signaling. Successful completion of this project will help us learn how to prevent neurodegeneration due to ischemic brain injury caused by cardiac arrest and stroke and more importantly design novel therapies for the injuries.

Agency
National Institute of Health (NIH)
Institute
Veterans Affairs (VA)
Type
Non-HHS Research Projects (I01)
Project #
1I01BX001960-01A1
Application #
8543213
Study Section
Neurobiology C (NURC)
Project Start
2013-04-01
Project End
2017-03-31
Budget Start
2013-04-01
Budget End
2014-03-31
Support Year
1
Fiscal Year
2013
Total Cost
Indirect Cost
Name
Veterans Affairs Medical Center San Francisco
Department
Type
DUNS #
078763885
City
San Francisco
State
CA
Country
United States
Zip Code
94121
Hu, Diane P; Ferro, Federico; Yang, Frank et al. (2017) Cartilage to bone transformation during fracture healing is coordinated by the invading vasculature and induction of the core pluripotency genes. Development 144:221-234
Santa Maria, Christian; Cheng, Zhiqiang; Li, Alfred et al. (2016) Interplay between CaSR and PTH1R signaling in skeletal development and osteoanabolism. Semin Cell Dev Biol 49:11-23
Zhang, M; Wang, H; Zhang, J et al. (2016) Unilateral anterior crossbite induces aberrant mineral deposition in degenerative temporomandibular cartilage in rats. Osteoarthritis Cartilage 24:921-31
Kim, Wonnam; Takyar, Farzin M; Swan, Karena et al. (2016) Calcium-Sensing Receptor Promotes Breast Cancer by Stimulating Intracrine Actions of Parathyroid Hormone-Related Protein. Cancer Res 76:5348-60
Schepelmann, M; Yarova, P L; Lopez-Fernandez, I et al. (2016) The vascular Ca2+-sensing receptor regulates blood vessel tone and blood pressure. Am J Physiol Cell Physiol 310:C193-204
Graca, J A Z; Schepelmann, M; Brennan, S C et al. (2016) Comparative expression of the extracellular calcium-sensing receptor in the mouse, rat, and human kidney. Am J Physiol Renal Physiol 310:F518-33
Al-Dujaili, Saja A; Koh, Amy J; Dang, Ming et al. (2016) Calcium Sensing Receptor Function Supports Osteoblast Survival and Acts as a Co-Factor in PTH Anabolic Actions in Bone. J Cell Biochem 117:1556-67
Bikle, Daniel D; Tahimic, Candice; Chang, Wenhan et al. (2015) Role of IGF-I signaling in muscle bone interactions. Bone 80:79-88
Yang, T; Zhang, J; Cao, Y et al. (2015) Wnt5a/Ror2 mediates temporomandibular joint subchondral bone remodeling. J Dent Res 94:803-12
Wang, Tao; Wang, Yongmei; Menendez, Alicia et al. (2015) Osteoblast-Specific Loss of IGF1R Signaling Results in Impaired Endochondral Bone Formation During Fracture Healing. J Bone Miner Res 30:1572-84

Showing the most recent 10 out of 21 publications