Accumulating evidence suggests that apoptosis is one of the mechanisms responsible for motor-neuron degeneration in amyotrophic lateral sclerosis (ALS). Specifically, a recent study showed that neuronal deletion of both Bax and Bak in SOD1G93A mice (ALS model) not only halted neuronal loss, but also prevented axonal degeneration and NMJ denervation, and extended survival. Thus, these results suggest that the deletion of both Bax and Bak might conferred a stronger resistance to apoptotic stimuli than deletion of Bax alone, given that a redundant apoptotic pathway can be activated in cells. Thus, these results once again point to the mitochondrial apoptotic pathway as a major contributor to the pathogenesis of ALS and indicate that further investigations, possibly targeting ?other relevant pathways? regulating mitochondrial apoptosis, are needed to clarify this issue. In this regard, our recent study revealed a very interesting finding that knockout of the novel mitochondrial proapoptotic protein PSAP (presenilin-associated protein) in SOD1G93A mice greatly improved motor function, increased lifespan, and protected NMJ from denervation in these mice. Specifically, PSAP was shown to induce apoptosis in a Bax- and Bak-independent manner, strongly suggesting that the PSAP pathway is one of the most sought after ?other relevant pathways? that contribute to neurodegeneration in ALS. Thus, our novel findings may open a new avenue for studying the pathogenesis of ALS. We also found that PSAP functions downstream of death receptor DR6, which is implicated in ALS recently, and that knockdown of PSAP protected axons from degeneration caused by NGF withdrawal. These novel findings led to our hypothesis that DR6-PSAP represents a novel pathway mediating the pathogenic effect of SOD1G93A in ALS. This innovative hypothesis will be tested by the following two objectives:
Aim 1 : Determine the role of PSAP in mediating the pathogenic effects of SOD1G93A using a novel PSAP-knockout mouse model. 1-a, Determine the effect of PSAP knockout on ALS onset and survival of SOD1G93A mice. 1-b, Determine the effect of PSAP knockout on motor neuron survival and protection of neuromuscular junction integrity in SOD1G93A mice.
Aim 2 : Determine the mechanism by which PSAP mediates the pathogenic effect of SOD1G93A in ALS using a cultured cell system. 2-a, Determine a novel DR6-PSAP pathway that mediates the neurotoxicity of SOD1G93A in ALS. 2-b, Determine the role of PSAP in DR6 signaling and PTP opening. Significance: The novel finding that knockout of the apoptotic mitochondrial protein PSAP greatly improved motor function and inreased lifespan in ALS SOD1G93A mice provides a new avenue for studying the mechanism underlying ALS pathogenesis. Specifically, the finding that knockout of PSAP improved walking ability and prevented incontinence in SOD1G93A mice raises the possibility that understanding the molecular mechanism by which PSAP contributes to the pathogenesis of ALS may lead to the identification of a novel therapeutic target and development of methods of treatment to improve the quality of life of human patients.

Public Health Relevance

Amyotrophic lateral sclerosis (ALS) is a fatal adult-onset neurodegenerative disease characterized by the selective death of motor neurons and axon degeneration leading to progressive paralysis with no cure or effective treatment available today. The objective of this proposal is to determine the pathogenic role of a novel molecule, PSAP, in neurodegeneration in ALS. The long-term goal is to reveal new targets for therapeutic intervention and development of new prevention strategies of the disease.

Agency
National Institute of Health (NIH)
Institute
National Institute of Neurological Disorders and Stroke (NINDS)
Type
Exploratory/Developmental Grants (R21)
Project #
1R21NS095256-01A1
Application #
9182525
Study Section
Neural Oxidative Metabolism and Death Study Section (NOMD)
Program Officer
Gubitz, Amelie
Project Start
2016-09-01
Project End
2018-08-31
Budget Start
2016-09-01
Budget End
2017-08-31
Support Year
1
Fiscal Year
2016
Total Cost
Indirect Cost
Name
University of Tennessee Knoxville
Department
Other Basic Sciences
Type
Graduate Schools
DUNS #
003387891
City
Knoxville
State
TN
Country
United States
Zip Code
37996
Dou, Quanlin; Hao, Feng; Sun, Longsheng et al. (2017) CRE and SRE mediate LPA-induced CCN1 transcription in mouse aortic smooth muscle cells. Can J Physiol Pharmacol 95:275-280
Hu, Chen; Xu, Junjie; Zeng, Linlin et al. (2017) Pen-2 and Presenilin are Sufficient to Catalyze Notch Processing. J Alzheimers Dis 56:1263-1269
Dong, Yunzhou; Wu, Yong; Cui, Mei-Zhen et al. (2017) Lysophosphatidic Acid Triggers Apoptosis in HeLa Cells through the Upregulation of Tumor Necrosis Factor Receptor Superfamily Member 21. Mediators Inflamm 2017:2754756
An, Dong; Hao, Feng; Zhang, Fuqiang et al. (2017) CD14 is a key mediator of both lysophosphatidic acid and lipopolysaccharide induction of foam cell formation. J Biol Chem 292:14391-14400
An, Dong; Hao, Feng; Hu, Chen et al. (2017) JNK1 Mediates Lipopolysaccharide-Induced CD14 and SR-AI Expression and Macrophage Foam Cell Formation. Front Physiol 8:1075
Hao, Feng; Zhang, Fuqiang; Wu, Daniel Dongwei et al. (2016) Lysophosphatidic acid-induced vascular neointimal formation in mouse carotid arteries is mediated by the matricellular protein CCN1/Cyr61. Am J Physiol Cell Physiol 311:C975-C984
Hu, Chen; Zeng, Linlin; Li, Ting et al. (2016) Nicastrin is required for amyloid precursor protein (APP) but not Notch processing, while anterior pharynx-defective 1 is dispensable for processing of both APP and Notch. J Neurochem 136:1246-1258