Amyotrophic lateral sclerosis (ALS), or Lou Gehrig's disease, is an adult-onset neurodegenerative disease that affects upper and lower motor neurons. The key clinical features in ALS patients include muscle wasting, and progressive loss of spinal motor neurons and upper motor neurons and their axons in the lateral columns of the spinal cord. The past 10 years have witnessed a tremendous expansion in the molecular mechanisms of this devastating disease thanks to the discoveries of genetic mutations that are causally linked to both familial ALS (FALS) and sporadic ALS (SALS). Characterizations of these ALS disease genes suggest that dysfunctions in protein homeostasis via the ubiquitin-proteasome pathways (proteostasis) might contribute to the pathogenesis and disease progression in ALS. Consistent with the genetic data, a key pathological feature in FALS and SALS is accumulation of misfolded proteins in motor neurons, which disrupts normal neuronal functions, including axonal transport, mitochondrial bioenergetics, gene expression, and synaptic connectivity. Persistent accumulation of misfolded proteins eventually triggers endoplasmic reticulum (ER) stress-induced cell death, which leads to neurodegeneration through mechanisms that are poorly understood. To investigate how misfolded proteins promote neurodegeneration, we have focused on the IRE1?-ASK1-JNK pathway because of its role in ER stress-induced cell death. Our results showed that a highly conserved kinase HIPK2 (homeodomain interacting protein kinase 2) acts downstream of IRE1?-ASK1 and upstream of JNK to promote ER stress-mediated cell death. Using proteomics and phospho-peptide mapping, we showed that ER stress activates HIPK2 by promoting phosphorylation on specific Serine and Threonine residues within the kinase domain of HIPK2. Mutagenesis analyses confirmed that phosphorylation of these amino acids in HIPK2 is prerequisite to promote ER stress-mediated neuronal cell death. To investigate the role of HIPK2 in ALS, we have developed phospho-HIPK2-specific antibodies and showed that HIPK2 activation can be detected in the spinal cord of SOD1G93A mice before symptom onset. Furthermore, the level of phospho-HIPK2 positively correlates with disease progression in SOD1G93A mice, suggesting that HIPK2 activation may directly promote neurodegeneration. In support of this idea, loss of HIPK2 significantly protects cultured spinal motor neurons from SOD1G93A-induced cell death. Given the implications of ER stress in both FALS and SALS, our results support the hypotheses that (1) HIPK2 is an essential target in the downstream of IRE1? pathway that promotes ER stress-induced neurodegeneration, and (2) HIPK2 activation may have broader implications in the pathogenesis of, and in the therapeutics for, both FALS and SALS.

Public Health Relevance

Neurodegenerative diseases represent a major challenge in the health care for the civilian and Veteran populations. In particular, there are concerns that exposure to chemicals or injury during combat may increase the propensity of neurodegeneration in Veterans. ALS is a fatal neurodegenerative disease characterized by rapidly progressive loss of motor neurons in brain and spinal cord. By analyzing ALS mouse models and a large number of tissue samples from FALS and SALS patients, we show that misfolded proteins activate a highly conserved protein kinase, called HIPK2, which serves as a molecular switch to promote neuronal cell death, leading to hypothesize that HIPK2 is a feasible therapeutic target for ALS. To test this, we will (1) determine if inhibition of HIPK2 can mitigate neurodegeneration and prolong survival in preclinical models of ALS, and (2) characterize the broader role of HIPK2 as a therapeutic target and a biomarker for ALS.

Agency
National Institute of Health (NIH)
Institute
Veterans Affairs (VA)
Type
Non-HHS Research Projects (I01)
Project #
5I01BX001108-07
Application #
9553446
Study Section
Neurobiology E (NURE)
Project Start
2011-07-01
Project End
2020-06-30
Budget Start
2018-07-01
Budget End
2019-06-30
Support Year
7
Fiscal Year
2018
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
Shang, Yulei; Zhang, Jiasheng; Huang, Eric J (2018) HIPK2-Mediated Transcriptional Control of NMDA Receptor Subunit Expression Regulates Neuronal Survival and Cell Death. J Neurosci 38:4006-4019
Ward, Michael E; Chen, Robert; Huang, Hsin-Yi et al. (2017) Individuals with progranulin haploinsufficiency exhibit features of neuronal ceroid lipofuscinosis. Sci Transl Med 9:
Lombardi, Laura M; Zaghlula, Manar; Sztainberg, Yehezkel et al. (2017) An RNA interference screen identifies druggable regulators of MeCP2 stability. Sci Transl Med 9:
Le Pichon, Claire E; Meilandt, William J; Dominguez, Sara et al. (2017) Loss of dual leucine zipper kinase signaling is protective in animal models of neurodegenerative disease. Sci Transl Med 9:
Celona, Barbara; Dollen, John von; Vatsavayai, Sarat C et al. (2017) Suppression of C9orf72 RNA repeat-induced neurotoxicity by the ALS-associated RNA-binding protein Zfp106. Elife 6:
Lui, Hansen; Zhang, Jiasheng; Makinson, Stefanie R et al. (2016) Progranulin Deficiency Promotes Circuit-Specific Synaptic Pruning by Microglia via Complement Activation. Cell 165:921-35
Luo, Sarah X; Timbang, Leah; Kim, Jae-Ick et al. (2016) TGF-? Signaling in Dopaminergic Neurons Regulates Dendritic Growth, Excitatory-Inhibitory Synaptic Balance, and Reversal Learning. Cell Rep 17:3233-3245
Luo, Sarah X; Huang, Eric J (2016) Dopaminergic Neurons and Brain Reward Pathways: From Neurogenesis to Circuit Assembly. Am J Pathol 186:478-88
Shang, Yulei; Huang, Eric J (2016) Mechanisms of FUS mutations in familial amyotrophic lateral sclerosis. Brain Res 1647:65-78
Lee, Sebum; Shang, Yulei; Redmond, Stephanie A et al. (2016) Activation of HIPK2 Promotes ER Stress-Mediated Neurodegeneration in Amyotrophic Lateral Sclerosis. Neuron 91:41-55

Showing the most recent 10 out of 14 publications