AccScience Publishing / AN / Volume 1 / Issue 3 / DOI: 10.36922/an.v1i3.153
ORIGINAL RESEARCH ARTICLE

Emx1-Cre-mediated inactivation of PDK1 prevents plaque deposition in an Alzheimer’s disease-like mouse model

Xiaolian Ye1 Liyang Yao2 Wenhao Chen3 Wenkai Shao2 Yimin Hu3* Bing Zhang1* Guiquan Chen2*
Show Less
1 Department of Radiology, Medical Imaging Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Institute of Medical Imaging and Artificial Intelligence, Jiangsu Key Laboratory of Molecular Medicine, Institute of Brain Science, Nanjing University, Nanjing, Jiangsu Province, 210008, China
2 Model Animal Research Center, Department of Neurology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, 12 Xuefu Avenue, Nanjing, Jiangsu Province, 210061, China
3 Department of Anesthesiology, The Second Affiliated Changzhou People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, 213000, China
Advanced Neurology 2022, 1(3), 153 https://doi.org/10.36922/an.v1i3.153
Submitted: 9 July 2022 | Accepted: 17 October 2022 | Published: 30 November 2022
© 2022 by the Author(s). This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution 4.0 International License ( https://creativecommons.org/licenses/by/4.0/ )
Abstract

BX912, an inhibitor for 3-phosphoinositide-dependent protein kinase 1 (PDK1), has been shown to produce beneficial effects in mouse models of Alzheimer’s disease (AD). To test the hypothesis that early inhibition on PDK1 may prevent neuropathology in a 5×FAD mouse model, we employed a genetic approach to generate a mutant line, termed as Pdk1 cKO/5×FAD, in which PDK1 is inactivated, specifically in the developing cortex of 5×FAD mice through Emx1-Cre-mediated gene recombination. We discovered that the Pdk1 cKO/5×FAD mice exhibited a massive reduction of plaque pathology compared with their 5×FAD littermates. We also demonstrated that gliosis was remarkably attenuated in Pdk1 cKO/5×FAD cortices and amyloid precursor protein levels were significantly lower in Pdk1 cKO/5×FAD cortices compared with 5×FAD littermates. This study suggests that early inhibition on PDK1 may effectively prevent AD-like neuropathology.

Keywords
Alzheimer’s disease
Plaque deposition
PDK1
Neuroinflammation
Amyloid precursor protein
Conditional knockout
Funding
National Natural Science Foundation of China
Natural Key Research and Development Program of China
Natural Science Foundation of Jiangsu
Changzhou Municipal Key Research Program
References
[1]

Li T, Han Y, 2022, Insights on amyloid-related imaging abnormalities from the “Pre-Alzheimer’s Disease Alliance of China”. Adv Neurol, 1: 2.

[2]

Scheltens P, Blennow K, Breteler MM, et al., 2016, Alzheimer’s disease. Lancet, 388: 505–517. 

[3]

Horsburgh K, Holland P, Reimer M, et al., 2011, Axon-glial disruption: The link between vascular disease and Alzheimer’s disease? Biochem Soc Trans, 39: 881–885.

[4]

Frisoni GB, Altomare D, Thal DR, et al., 2022, The probabilistic model of Alzheimer disease: The amyloid hypothesis revised. Nat Rev Neurosci, 23: 53–66. 

[5]

Hardy, J, Selkoe DJ, 2002, The amyloid hypothesis of Alzheimer’s disease: Progress and problems on the road to therapeutics. Science, 297: 353–356.

[6]

Goedert M, Eisenberg DS, Crowther RA, 2017, Propagation of Tau aggregates and neurodegeneration. Ann Rev Neurosci, 40: 189–210.

[7]

Mintun MA, Lo AC, Evans CD, et al., 2021, Donanemab in early Alzheimer’s disease. N Engl J Med, 384: 1691–1704.

[8]

Sevigny J, Chiao P, Bussiere T, et al., 2016, The antibody aducanumab reduces A beta plaques in Alzheimer’s disease. Nature, 537: 50–56.

[9]

Peng Y, Hu Y, Xu S, et al., 2012, L-3-n-butylphthalide reduces tau phosphorylation and improves cognitive deficits in AbetaPP/PS1-Alzheimer’s transgenic mice. J Alzheimers Dis, 29: 379–391. 

[10]

Peng Y, Sun J, Hon S, et al., 2010, L-3-n-butylphthalide improves cognitive impairment and reduces amyloid-beta in a transgenic model of Alzheimer’s disease. J Neurosci, 30: 8180–8189.

[11]

Chen G, Chen KS, Kobayashi D, et al., 2007, Active-amyloid immunization restores spatial learning in PDAPP mice displaying very low levels of beta-amyloid. J Neurosci, 27: 2654–2662.

[12]

Schenk D, Barbour R, Dunn W, et al., 1999, Immunization with amyloid-attenuates Alzheimer-disease-like pathology in the PDAPP mouse. Nature, 400: 173–177.

[13]

Yao YL, Wang YX, Yang FC, et al., 2022, Targeting AKT and CK2 represents a novel therapeutic strategy for SMO constitutive activation-driven medulloblastoma. CNS Neurosci Ther, 28: 1033–1044.

[14]

Wang J, Yu Z, Tao Y, et al., 2021, A novel palmitic acid hydroxy stearic acid (5-PAHSA) plays a neuroprotective role by inhibiting phosphorylation of the m-TOR-ULK1 pathway and regulating autophagy. CNS Neurosci Ther, 27: 484–496. 

[15]

Wang H, Liu M, Zou G, et al., 2021, Deletion of PDK1 in oligodendrocyte lineage cells causes white matter abnormality and myelination defect in the central nervous system. Neurobiol Dis, 148: 105212.

[16]

Wang H, Liu M, Ye Z, et al., 2021, Akt regulates Sox10 expression to control oligodendrocyte differentiation via phosphorylating FoxO1. J Neurosci, 41: 8163–8180.

[17]

Ma Y, Xu X, Li C, et al., 2021, Induced neural progenitor cell-derived extracellular vesicles promote neural progenitor cell survival via extracellular signal-regulated kinase pathway. CNS Neurosci Ther, 27: 1605–1609.

[18]

Fedder-Semmes KN, Appel B, 2021, The Akt-mTOR pathway drives myelin sheath growth by regulating Cap-dependent translation. J Neurosci, 41: 8532. 

[19]

Bonet IJ, Khomula EV, Araldi D, et al., 2021, PI3K/AKT signaling in high molecular weight hyaluronan (HMWH)- induced anti-hyperalgesia and reversal of nociceptor sensitization. J Neurosci, 41: 8414.

[20]

Zhao XF, Liao Y, Alam MM, et al., 2020, Microglial mTOR is neuronal protective and antiepileptogenic in the pilocarpine model of temporal lobe epilepsy. J Neurosci, 40: 7593.

[21]

Fruman DA, Chiu H, Hopkins BD, et al., 2017, The PI3K pathway in human disease. Cell, 170: 605–635.

[22]

Pietri M, Dakowski C, Hannaoui S, et al., 2013, PDK1 decreases TACE-mediated alpha-secretase activity and promotes disease progression in prion and Alzheimer’s diseases. Nat Med, 19: 1124–1131.

[23]

Pei JJ, Khatoon S, An WL, et al., 2003, Role of protein kinase B in Alzheimer’s neurofibrillary pathology. Acta Neuropathol, 105: 381–392.

[24]

Mathys H, Davila-Velderrain J, Peng Z, et al., 2019, Single-cell transcriptomic analysis of Alzheimer’s disease. Nature, 570: 332–337.

[25]

Yang S, Pascual-Guiral S, Ponce R, et al., 2018, Reducing the levels of Akt activation by PDK1 knock-in mutation protects neuronal cultures against synthetic smyloid-beta peptides. Front Aging Neurosci, 9, 435.

[26]

Stein TD, Johnson, JA, 2002, Lack of neurodegeneration in transgenic mice overexpressing mutant amyloid precursor protein is associated with increased levels of transthyretin and the activation of cell survival pathways. J Neurosci, 22: 7380.

[27]

Lawlor MA, Mora A, Ashby PR, et al., 2002, Essential role of PDK1 in regulating cell size and development in mice. EMBO J, 21: 3728–3738.

[28]

Xu C, Yu L, Hou J, et al., 2017, Conditional deletion of PDK1 in the forebrain causes neuron loss and increased apoptosis during cortical development. Front Cell Neurosci, 11: 330. 

[29]

Wei YJ, Han XN, Zhao CJ, 2020, PDK1 regulates the survival of the developing cortical interneurons. Mol Brain, 13: 65.

[30]

Oakley H, Cole SL, Logan S, et al., 2006, Intraneuronal-amyloid aggregates, neurodegeneration, and neuron loss in transgenic mice with five familial Alzheimer’s disease mutations: Potential factors in amyloid plaque formation. J Neurosci, 26: 10129–10140.

[31]

Liu T, Zhu X, Huang C, et al., 2022, ERK inhibition reduces neuronal death and ameliorates inflammatory responses in forebrain-specific Ppp2cα knockout mice. FASEB J, e222515.

[32]

Huang C, Liu T, Wang Q, et al., 2020, Loss of PP2A disrupts the retention of radial glial progenitors in the telencephalic niche to impair the generation for late-born neurons during cortical development. Cereb Cortex, 30: 4183–4196.

[33]

Cheng S, Liu T, Hu, Y, et al., 2019, Conditional inactivation of Pen-2 in the developing neocortex leads to rapid switch of apical progenitors to basal progenitors. J Neurosci, 39: 2195– 2207.

[34]

Englund C, Fink A, Lau C, et al., 2005, Pax6, Tbr2, and Tbr1 are expressed sequentially by radial glia, intermediate progenitor cells, and postmitotic neurons in developing neocortex. J Neurosci, 25: 247–251.

[35]

Wu J, Shao C, Ye X, et al., 2021, In vivo brain imaging of amyloid-aggregates in Alzheimer’s disease with a near-infrared fluorescent probe. ACS Sensors, 6: 863–870. 

[36]

Ma X, Wang Y, Hua J, et al., 2020, Abeta-sheet-targeted theranostic agent for diagnosing and preventing aggregation of pathogenic peptides in Alzheimer’s disease. Sci CHINA Chem, 63: 73–82.

[37]

Ye X, Chen L, Wang H, et al., 2022, Genetic inhibition of PDK1 robustly reduces plaque deposition and ameliorates gliosis in the 5×FAD mouse model of Alzheimer’s disease. Neuropathol Appl Neurobiol, 48: e12839.

[38]

Han X, Wei Y, Ba R, et al., 2021, PDK1 regulates the lengthening of G1 phase to balance RGC proliferation and differentiation during cortical neurogenesis. Cereb Cortex, 32: 3488–3500. 

[39]

Han X, Wei Y, Wu X, et al., 2020, PDK1 regulates transition period of apical progenitors to basal progenitors by controlling asymmetric cell division. Cereb Cortex, 30: 406–420.

[40]

Wang L, Cheng S, Yin Z, et al., 2015, Conditional inactivation of Akt three isoforms causes tau hyperphosphorylation in the brain. Mol Neurodegener, 10: 33.

[41]

Li QQ, Chen J, Hu P, et al., 2022, Enhancing GluN2A-type NMDA receptors impairs long-term synaptic plasticity and learning and memory. Mol Psychiatry: s41380-022-01579-7.

[42]

Peng SX, Wang YY, Zhang M, et al., 2021, SNP rs10420324 in the AMPA receptor auxiliary subunit TARP γ-8 regulates the susceptibility to antisocial personality disorder. Sci Rep, 11: 11997.

[43]

Xia Y, Zhang Y, Xu M, et al., 2022, Presenilin enhancer2 is crucial for the transition of apical progenitors into neurons but into not basal progenitors in the developing hippocampus. Development, 149: dev200272.

[44]

Teng X, Hu P, Chen Y, et al., 2022, A novel Lgi1 mutation causes white matter abnormalities and impairs motor coordination in mice. FASEB J, 36: e22212. 

[45]

Hou J, Bi H, Ye Z, et al., 2021, Pen-2 negatively regulates the differentiation of oligodendrocyte precursor cells into astrocytes in the central nervous system. J Neurosci, 41: 4976–4990.

[46]

Bi H, Zhou C, Zhang Y, et al., 2021, Neuron-specific deletion of presenilin enhancer2 causes progressive astrogliosis and age-related neurodegeneration in the cortex independent of the Notch signaling. CNS Neurosci Ther, 27: 174–185.

[47]

Hou J, Bi H, Ge Q, et al., 2022, Heterogeneity analysis of astrocytes following spinal cord injury at single-cell resolution. FASEB J, 36: e22442.

[48]

Saura CA, Chen G, Malkani S, et al., 2005, Conditional inactivation of presenilin 1 prevents amyloid accumulation and temporarily rescues contextual and spatial working memory impairments in amyloid precursor protein transgenic mice. J Neurosci, 25: 6755–6764.

[49]

Lai KO, Liang ZY, Fei EK, et al., 2015, Cyclin-dependent kinase 5 (Cdk5)-dependent phosphorylation of p70 ribosomal S6 kinase 1 (S6K) is required for dendritic spine morphogenesis. J Biol Chem, 290: 14637–14646.

[50]

Ruvinsky I, Meyuhas O, 2006, Ribosomal protein S6 phosphorylation: From protein synthesis to cell size. Trends Biochem Sci, 31: 342–348.

[51]

Hay N, Sonenberg N, 2004, Upstream and downstream of mTOR. Genes Dev, 18: 1926–1945.

[52]

Kimberly WT, LaVoie MJ, Ostaszewski BL, et al., 2003, Gamma-secretase is a membrane protein complex comprised of presenilin, nicastrin, Aph-1, and Pen-2. Proc Natl Acad Sci USA, 100: 6382–6387.

[53]

De Strooper B, Karran E, 2016, The cellular phase of Alzheimer’s disease. Cell, 164: 603–615.

[54]

Anguela XM, High KA, 2019, Entering the modern era of gene therapy. Ann Rev Med, 70: 273–288.

[55]

Mendell JR, Al-Zaidy S, Shell R, et al., 2017, Single-dose gene-replacement therapy for spinal muscular atrophy. N Engl J Med, 377: 1713–1722.

[56]

Thomsen GM, Gowing G, Latter J, et al., 2014, Delayed disease onset and extended survival in the SOD1G93A rat model of amyotrophic lateral sclerosis after suppression of mutant SOD1 in the motor cortex. J Neurosci, 34: 15587.

[57]

McBride JL, Pitzer MR, Boudreau RL, et al., 2011, Preclinical safety of RNAi-mediated HTT suppression in the rhesus macaque as a potential therapy for Huntington’s disease. Mol Ther, 19: 2152–2162.

[58]

Duan YY, Ye T, Qu Z, et al., 2022, Brain-wide Cas9-mediated cleavage of a gene causing familial Alzheimer’s disease alleviates amyloid-related pathologies in mice. Nat Biomed Eng, 6: 168–180.

[59]

Peifer C, Alessi DR, 2008, Small molecule inhibitors of PDK1. ChemMedChem, 3: 1810–1838. 

[60]

Bain J, Plater L, Elliott M, et al., 2007, The selectivity of protein kinase inhibitors: A further update. Biochem J, 408: 297–315.

[61]

Pelletier J, Thomas G, Volarević S, 2018, Ribosome biogenesis in cancer: New players and therapeutic avenues. Nat Rev Cancer, 18: 51–63.

Conflict of interest
The authors have no conflicts of interest to declare.
Share
Back to top
Advanced Neurology, Electronic ISSN: 2810-9619 Published by AccScience Publishing