Skip to main content

Advertisement

Log in

Genetic Ablation of Hematopoietic Cell Kinase Accelerates Alzheimer’s Disease–Like Neuropathology in Tg2576 Mice

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

Microglial dysregulation, pertaining to impairment in phagocytosis, clearance and containment of amyloid-β (Aβ), and activation of neuroinflammation, has been posited to contribute to the pathogenesis of Alzheimer’s disease (AD). Detailed cellular mechanisms that are disrupted during the disease course to display such impairment in microglia, however, remain largely undetermined. We hypothesize that loss of hematopoietic cell kinase (HCK), a phagocytosis-regulating member of the Src family tyrosine kinases that mediate signals from triggering receptor expressed on myeloid cells 2 and other immunoreceptors, impairs microglial homeostasis and Aβ clearance, leading to the accelerated buildup of Aβ pathology and cognitive decline during the early stage of neuropathological development. To elucidate the pivotal role of HCK in AD, we generated a constitutive knockout of HCK in the Tg2576 mouse model of AD. We found that HCK deficiency accelerated cognitive decline along with elevated Aβ level and plaque burden, attenuated microglial Aβ phagocytosis, induced iNOS expression in microglial clusters, and reduced pre-synaptic protein at the hippocampal regions. Our findings substantiate that HCK plays a prominent role in regulating microglial neuroprotective functions and attenuating early AD neuropathology.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Lane C, Hardy J, Schott J (2018) Alzheimer’s disease. Eur J Neurol 25:59–70. https://doi.org/10.1016/j.med.2019.03.012

    Article  CAS  PubMed  Google Scholar 

  2. Hansen DV, Hanson JE, Sheng M (2018) Microglia in Alzheimer’s disease. J Cell Biol 217:459–472. https://doi.org/10.1083/jcb.201709069

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Colonna M, Butovsky O (2017) Microglia function in the central nervous system during health and neurodegeneration. Annu Rev Immunol 35:441–468. https://doi.org/10.1146/annurev-immunol-051116-052358

    Article  CAS  PubMed  Google Scholar 

  4. Efthymiou AG, Goate AM (2017) Late onset Alzheimer’s disease genetics implicates microglial pathways in disease risk. Mol Neurodegener 12:43. https://doi.org/10.1186/s13024-017-0184-x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Sims R, van der Lee SJ, Naj AC et al (2017) Rare coding variants in PLCG2, ABI3, and TREM2 implicate microglial-mediated innate immunity in Alzheimer’s disease. Nat Genet 49:1373–1384. https://doi.org/10.1038/ng.3916

  6. Sala Frigerio C, Wolfs L, Fattorelli N et al (2019) The major risk factors for Alzheimer’s disease: age, sex, and genes modulate the microglia response to Aβ plaques. Cell Rep 27:1293–1306. https://doi.org/10.1016/j.celrep.2019.03.099

  7. Lim SL, Tran DN, Zumkehr J, Chen C, Ghiaar S, Kieu Z, Villanueva E, Gallup V et al (2018) Inhibition of hematopoietic cell kinase dysregulates microglial function and accelerates early stage Alzheimer’s disease-like neuropathology. Glia 66:2700–2718. https://doi.org/10.1002/glia.23522

  8. Wang Y, Ulland TK, Ulrich JD et al (2016) TREM2-mediated early microglial response limits diffusion and toxicity of amyloid plaques. J Exp Med 213:667–675. https://doi.org/10.1084/jem.20151948

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Heneka MT, Kummer MP, Stutz A, Delekate A, Schwartz S, Vieira-Saecker A, Griep A, Axt D et al (2013) NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature 493:674–678. https://doi.org/10.1038/nature11729.NLRP3

  10. Xu L, He D, Bai Y (2016) Microglia-mediated inflammation and neurodegenerative disease. Mol Neurobiol 53:6709–6715. https://doi.org/10.1007/s12035-015-9593-4

    Article  CAS  PubMed  Google Scholar 

  11. Mackenzie IR, Hao CMD (1995) Role of microglia in senile plaque formation. Neurobiol Aging 16:797–804. https://doi.org/10.1016/0197-4580(95)00092-S

    Article  CAS  PubMed  Google Scholar 

  12. Spangenberg E, Severson PL, Hohsfield LA, Crapser J, Zhang J, Burton EA, Zhang Y, Spevak W et al (2019) Sustained microglial depletion with CSF1R inhibitor impairs parenchymal plaque development in an Alzheimer’s disease model. Nat Commun 10:3758. https://doi.org/10.1038/s41467-019-11674-z

  13. Asai H, Ikezu S, Tsunoda S, et al (2015) Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat Neurosci 18:1584–1593. https://doi.org/10.1038/nn.4132

  14. Spangenberg EE, Lee RJ, Najafi AR, Rice RA, Elmore MR, Blurton-Jones M, West BL, Green KN (2016) Eliminating microglia in Alzheimer’s mice prevents neuronal loss without modulating amyloid-b pathology. Brain 139:1265–1281. https://doi.org/10.1093/brain/aww016

    Article  PubMed  PubMed Central  Google Scholar 

  15. Bie B, Wu J, Foss JF, Naguib M (2019) Activation of mGluR1 mediates C1q-dependent microglial phagocytosis of glutamatergic synapses in Alzheimer’s rodent models. Mol Neurobiol 56:5568–5585. https://doi.org/10.1007/s12035-019-1467-8

    Article  CAS  PubMed  Google Scholar 

  16. Dagher NN, Najafi AR, Kayala KMN, Elmore MR, White TE, Medeiros R, West BL, Green KN (2015) Colony-stimulating factor 1 receptor inhibition prevents microglial plaque association and improves cognition in 3xTg-AD mice. J Neuroinflammation 12:139. https://doi.org/10.1186/s12974-015-0366-9

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Konishi H, Kiyama H (2018) Microglial TREM2/DAP12 signaling: a double-edged sword in neural diseases. Front Cell Neurosci 12:1–14. https://doi.org/10.3389/fncel.2018.00206

    Article  CAS  Google Scholar 

  18. Lee S-H, Suk K (2018) Kinase-based taming of brain microglia toward disease-modifying therapy. Front Cell Neurosci 12:1–12. https://doi.org/10.3389/fncel.2018.00474

    Article  CAS  Google Scholar 

  19. Guiet R, Poincloux R, Castandet J, Marois L, Labrousse A, Le Cabec V, Maridonneau-Parini I (2008) Hematopoietic cell kinase (Hck) isoforms and phagocyte duties - from signaling and actin reorganization to migration and phagocytosis. Eur J Cell Biol 87:527–542. https://doi.org/10.1016/j.ejcb.2008.03.008

  20. Lowell CA, Soriano P, Varmus HE (1994) Functional overlap in the src gene family: Inactivation of hck and fgr impairs natural immunity. Genes Dev 8:387–398. https://doi.org/10.1101/gad.8.4.387

    Article  CAS  PubMed  Google Scholar 

  21. Zhang B, Gaiteri C, Bodea L-G, Wang Z, McElwee J, Podtelezhnikov AA, Zhang C, Xie T et al (2013) Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer’s disease. Cell 153:707–720. https://doi.org/10.1016/j.cell.2013.03.030

  22. Ayata P, Badimon A, Strasburger HJ, Duff MK, Montgomery SE, Loh YE, Ebert A, Pimenova AA et al (2018) Epigenetic regulation of brain region-specific microglia clearance activity. Nat Neurosci 21:1049–1060. https://doi.org/10.1038/s41593-018-0192-3

  23. Choucair N, Laporte V, Levy R, Arnold AS, Gies JP, Poindron P, Lombard Y (2006) Phagocytic functions of microglial cells in the central nervous system and their importance in two neurodegenerative diseases: multiple sclerosis and Alzheimer’s disease. Cent Eur J Biol 1:463–493. https://doi.org/10.2478/s11535-006-0038-y

    Article  CAS  Google Scholar 

  24. Krady JK, Basu A, Levison SW, Milner RJ (2002) Differential expression of protein tyrosine kinase genes during microglial activation. Glia 40:11–24. https://doi.org/10.1002/glia.10101

    Article  PubMed  Google Scholar 

  25. Castillo E, Leon J, Mazzei G, Abolhassani N, Haruyama N, Saito T, Saido T, Hokama M et al (2017) Comparative profiling of cortical gene expression in Alzheimer’s disease patients and mouse models demonstrates a link between amyloidosis and neuroinflammation. Sci Rep 7:1–16. https://doi.org/10.1038/s41598-017-17999-3

  26. Bonham LW, Sirkis DW, Yokoyama JS (2019) The transcriptional landscape of microglial genes in aging and neurodegenerative disease. Front Immunol 10:1–17. https://doi.org/10.3389/fimmu.2019.01170

    Article  CAS  Google Scholar 

  27. Hsiao K, Chapman P, Nilsen S et al (1996) Correlative memory deficits, Abeta elevation, and amyloid plaques in transgenic mice. Science (80- ) 274:99–102. https://doi.org/10.1126/science.274.5284.99

    Article  CAS  Google Scholar 

  28. Rodriguez-Ortiz CJ, Hoshino H, Cheng D et al (2013) Neuronal-specific overexpression of a mutant valosin-containing protein associated with IBMPFD promotes aberrant ubiquitin and TDP-43 accumulation and cognitive dysfunction in transgenic mice. Am J Pathol 183:504–515. https://doi.org/10.1016/j.ajpath.2013.04.014

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Zumkehr J, Rodriguez-Ortiz CJ, Cheng D, Kieu Z, Wai T, Hawkins C, Kilian J, Lim SL et al (2015) Ceftriaxone ameliorates tau pathology and cognitive decline via restoration of glial glutamate transporter in a mouse model of Alzheimer’s disease. Neurobiol Aging 36:2260–2271. https://doi.org/10.1016/j.neurobiolaging.2015.04.005

  30. Kitazawa M, Cheng D, Tsukamoto MR, Koike MA, Wes PD, Vasilevko V, Cribbs DH, LaFerla F (2011) Blocking IL-1 signaling rescues cognition, attenuates tau pathology, and restores neuronal β-catenin pathway function in an Alzheimer’s disease model. J Immunol 187:6539–6549. https://doi.org/10.4049/jimmunol.1100620

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Ulrich JD, Finn MB, Wang Y, Shen A, Mahan TE, Jiang H, Stewart FR, Piccio L et al (2014) Altered microglial response to Abeta plaques in APPPS1-21 mice heterozygous for TREM2. Mol Neurodegener 9:20. https://doi.org/10.1186/1750-1326-9-20

  32. Marsh SE, Abud EM, Lakatos A, Karimzadeh A, Yeung ST, Davtyan H, Fote GM, Lau L et al (2016) The adaptive immune system restrains Alzheimer’s disease pathogenesis by modulating microglial function. Proc Natl Acad Sci 113:E1316–E1325. https://doi.org/10.1073/pnas.1525466113

  33. Terry RD, Masliah E, Salmon DP et al (1991) Physical basis of cognitive alterations in Alzheimer’s disease: synapse loss is the major correlate of cognitive impairment. Ann Neurol 30:572–580. https://doi.org/10.1002/ana.410300410

    Article  CAS  PubMed  Google Scholar 

  34. DeKosky ST, Scheff SW (1990) Synapse loss in frontal cortex biopsies in Alzheimer’s disease: correlation with cognitive severity. Ann Neurol 27:457–464. https://doi.org/10.1002/ana.410270502

    Article  CAS  PubMed  Google Scholar 

  35. Braak H, Braak E, Bohl J (1993) Staging of Alzheimer-related cortical destruction. Eur Neurol 33:403–408. https://doi.org/10.1159/000116984

    Article  CAS  PubMed  Google Scholar 

  36. Lanz TA, Carter DB, Merchant KM (2003) Dendritic spine loss in the hippocampus of young PDAPP and Tg2576 mice and its prevention by the ApoE2 genotype. Neurobiol Dis 13:246–253. https://doi.org/10.1016/S0969-9961(03)00079-2

    Article  CAS  PubMed  Google Scholar 

  37. Cole SL, Vassar R (2007) The Alzheimer’s disease Beta-secretase enzyme, BACE1. Mol Neurodegener 2:22. https://doi.org/10.1186/1750-1326-2-22

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Itagaki S, McGeer PL, Akiyama H, Zhu S, Selkoe D (1989) Relationship of microglia and astrocytes to amyloid deposits of Alzheimer disease. J Neuroimmunol 24:173–182. https://doi.org/10.1016/0165-5728(89)90115-X

    Article  CAS  PubMed  Google Scholar 

  39. Wegiel J, Wisniewski H (1990) The complex of microglial cells and amyloid star in three-dimensional reconstruction. Ann Neurol 81:116–124. https://doi.org/10.1007/bf00334499

    Article  CAS  Google Scholar 

  40. Medeiros R, Kitazawa M, Passos GF, Baglietto-Vargas D, Cheng D, Cribbs DH, LaFerla F (2013) Aspirin-triggered lipoxin A4 stimulates alternative activation of microglia and reduces Alzheimer disease-like pathology in mice. Am J Pathol 182:1780–1789. https://doi.org/10.1016/j.ajpath.2013.01.051

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Sasaki A, Shoji M, Harigaya Y, Kawarabayashi T, Ikeda M, Naito M, Matsubara E, Abe K et al (2002) Amyloid cored plaques in Tg2576 transgenic mice are characterized by giant plaques, slightly activated microglia, and the lack of paired helical filament-typed, dystrophic neurites. Virchows Arch 441:358–367. https://doi.org/10.1007/s00428-002-0643-8

  42. Condello C, Yuan P, Schain A, Grutzendler J (2015) Microglia constitute a barrier that prevents neurotoxic protofibrillar Aβ42 hotspots around plaques. Nat Commun 6:6176. https://doi.org/10.1038/ncomms7176

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Yuan P, Condello C, Keene CD et al (2016) TREM2 haplodeficiency in mice and humans impairs the microglia barrier function leading to decreased amyloid compaction and severe axonal dystrophy. Neuron 90:724–739. https://doi.org/10.1016/j.neuron.2016.05.003

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Jansen IE, Savage JE, Watanabe K, Bryois J, Williams DM, Steinberg S, Sealock J, Karlsson IK et al (2019) Genome-wide meta-analysis identifies new loci and functional pathways influencing Alzheimer’s disease risk. Nat Genet 51:404–413. https://doi.org/10.1038/s41588-018-0311-9

  45. Malm TM, Jay TR, Landreth GE (2015) The evolving biology of microglia in Alzheimer’s disease. Neurotherapeutics 12:81–93. https://doi.org/10.1007/s13311-014-0316-8

    Article  CAS  PubMed  Google Scholar 

  46. Mosher KI, Wyss-Coray T (2014) Microglial dysfunction in brain aging and Alzheimer’s disease. Biochem Pharmacol 88:594–604. https://doi.org/10.1016/j.bcp.2014.01.008

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Wood JG, Zinsmeister P (1991) Tyrosine phosphorylation systems in Alzheimer’s disease pathology. Neurosci Lett 121:12–16. https://doi.org/10.1016/0304-3940(91)90637-9

    Article  CAS  PubMed  Google Scholar 

  48. Dhawan G, Floden AM, Combs CK (2012) Amyloid-β oligomers stimulate microglia through a tyrosine kinase dependent mechanism. Neurobiol Aging 33:2247–2261. https://doi.org/10.1016/j.neurobiolaging.2011.10.027.Amyloid-

    Article  CAS  PubMed  Google Scholar 

  49. Dhawan G, Combs CK (2012) Inhibition of Src kinase activity attenuates amyloid associated microgliosis in a murine model of Alzheimer’s disease. J Neuroinflammation 9:117. https://doi.org/10.1186/1742-2094-9-117

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Schweig JE, Yao H, Beaulieu-Abdelahad D, Ait-Ghezala G, Mouzon B, Crawford F, Mullan M, Paris D (2017) Alzheimer’s disease pathological lesions activate the spleen tyrosine kinase. Acta Neuropathol Commun 5:69. https://doi.org/10.1186/s40478-017-0472-2

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Satoh J-I, Tabunoki H, Ishida T et al (2012) Phosphorylated Syk expression is enhanced in Nasu-Hakola disease brains. Neuropathology 32:149–157. https://doi.org/10.1111/j.1440-1789.2011.01256.x

    Article  PubMed  Google Scholar 

  52. Combs CK, Karlo JC, Kao SC, Landreth GE (2001) Beta-amyloid stimulation of microglia and monocytes results in TNFalpha-dependent expression of inducible nitric oxide synthase and neuronal apoptosis. J Neurosci 21:1179–1188. https://doi.org/10.1523/JNEUROSCI.21-04-01179.2001

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Sondag CM, Dhawan G, Combs CK (2009) Beta amyloid oligomers and fibrils stimulate differential activation of primary microglia. J Neuroinflammation 6:1. https://doi.org/10.1186/1742-2094-6-1

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Fan Z, Brooks DJ, Okello A, Edison P (2017) An early and late peak in microglial activation in Alzheimer’s disease trajectory. Brain 40:792–803. https://doi.org/10.1093/brain/aww349

    Article  Google Scholar 

  55. Jiang T, Wan Y, Zhang YD, Zhou JS, Gao Q, Zhu XC, Shi JQ, Lu H et al (2017) TREM2 overexpression has no improvement on neuropathology and cognitive impairment in aging APPswe/PS1dE9 mice. Mol Neurobiol 54:855–865. https://doi.org/10.1007/s12035-016-9704-x

  56. Mucke L, Masliah E, Yu G-Q, Mallory M, Rockenstein EM, Tatsuno G, Hu K, Kholodenko D et al (2000) High-level neuronal expression of Aβ1–42 in wild-type human amyloid protein precursor transgenic mice: synaptotoxicity without plaque formation. J Neurosci 20:4050–4058. https://doi.org/10.1523/JNEUROSCI.20-11-04050.2000

  57. King DL, Arendash GW (2002) Behavioral characterization of the Tg2576 transgenic model of Alzheimer’s disease through 19 months. Physiol Behav 75:627–642. https://doi.org/10.1016/S0031-9384(02)00639-X

    Article  CAS  PubMed  Google Scholar 

  58. Kobayashi DT, Chen KS (2005) Behavioral phenotypes of amyloid-based genetically modified mouse models of Alzheimer’s disease. Genes Brain Behav 4:173–196. https://doi.org/10.1111/j.1601-183X.2005.00124.x

    Article  CAS  PubMed  Google Scholar 

  59. Taglialatela G, Hogan D, Zhang W-R, Dineley KT (2009) Intermediate- and long-term recognition memory deficits in Tg2576 mice are reversed with acute calcineurin inhibition. Behav Brain Res 200:95–99. https://doi.org/10.1016/j.bbr.2008.12.034

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Farr SA, Erickson MA, Niehoff ML, Banks WAMJ (2014) Central and peripheral administration of antisense oligonucleotide targeting amyloid-β protein precursor improves learning and memory and reduces neuroinflammatory cytokines in Tg2576 (AβPPswe) mice. J Alzheimers Dis 40:1005–1016. https://doi.org/10.3233/JAD-131883.Central

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Guillot-Sestier MV, Doty KR, Gate D, Rodriguez J Jr, Leung BP, Rezai-Zadeh KTT (2015) Il10 deficiency rebalances innate immunity to mitigate Alzheimer-like pathology. Neuron 85:534–548. https://doi.org/10.1016/j.neuron.2014.12.068

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Jay TR, Hirsch XAM, Broihier XML et al (2017) Disease progression-dependent effects of TREM2 deficiency in a mouse model of Alzheimer’s disease. J Neurosci 37:637–647. https://doi.org/10.1523/JNEUROSCI.2110-16.2017

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Nizami S, Di Daniel E, Roberts HH, Cowley SA (2019) Microglial inflammation and phagocytosis in Alzheimer’s disease: potential therapeutic targets. Br J Pharmacol 176:3515–3532. https://doi.org/10.1111/bph.14618

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgments

We thank the following people for their kind contribution and assistance: Dr. Clifford Lowell (University of California, San Francisco) for the HCK-KO mice; Dr. David H. Cribbs (University of California, Irvine) for the Tg2576 mice; Dr. David Baglietto-Vargas (University of California, Irvine) for his technical support on Confocal Microscopy; and Dr. Samuel E. Marsh (University of California, Irvine) for his guidance on Imaris analysis.

Authorships

All protein extraction, MSD, western blot analysis, and statistical analyses were conducted by SLL. Guidance on microdissection and immunostaining were provided by JZ. Animal husbandry and genotyping were done by SLL, ZK, EV, and CC, and tissue collection was conducted by SLL, ZK, JZ, and CJRO. Behavioral tests were designed by CJRO and executed by CJRO and SLL. NOR and OIP scoring was done by SLL and CC. Tissue sectioning was done by ZK, CC, and SG, while immunostaining, imaging, and analyses were done by SLL, DNT, CC, and VG. Experiments were conceived and designed by MK and SLL. Manuscript was written by SLL and critically reviewed by MK. All authors read and approved the final manuscript.

Funding

This study was supported by the National Institute of Health/National Institute of Environmental Health Sciences (R01 ES024331) and Alzheimer’s Association (NIRG-12-242598 and AARF-16-440554).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Masashi Kitazawa.

Ethics declarations

All animal studies performed were approved by the University of California Institutional Animal Care and Use Committee and were in accordance with Federal guidelines.

Conflict of Interest

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic Supplementary Material

ESM 1

(PDF 1052 kb)

Fig. S1 Knocking out HCK in Tg2576 mice did not modulate the recognition index (RI) of NOR and OIP nor the motor skills in MWM test. (a-d) WT, HCK-KO, Tg2576 and Tg/HCK-KO mice of 10-12 months were subjected to a battery of behavioral tests, namely NOR (a), OIP (b), and MWM (c-d). There were no significant differences in the RI of NOR (a) and OIP (b) tests between the genotypes during the 1, 3 and 5 mins of the experiments. Data are expressed as mean ± SEM with n = 7-22. All mouse genotypes completed the 1 wk MWM test with no apparent differences in the total distance moved (c) or in the swim speed (d). Data are expressed as mean ± SEM with n = 9-13

Fig. S2 Knocking out HCK in Tg2576 mice moderately modulated the intensity of PSD95 in mouse hippocampus.(a) Representative images of PSD95 (post-synaptic protein marker) at the DG, CA1 and CA3 regions of the hippocampus of WT, HCK-KO, Tg2576 and Tg/HCK-KO mice (11-13 months old). Scale bar, 50 µm. (b) Quantitative analyses of % PSD95 intensities in WT, HCK-KO, Tg2576 and Tg/HCK-KO mice taken relative to that of HCK-KO mice revealed lowest, but not significant, protein expression in Tg/HCK-KO mice among all genotypes. Data are expressed as mean ± SEM from three sections per mouse with n = 7-9. (c) Representative immunoblots of synaptophysin and PSD95 were shown. Tubulin was probed as loading control. (d) Quantitative analysis of synaptophysin and PSD95 band intensities after normalized to that of tubulin did not show significant differences between genotypes. Data are expressed as mean ± SEM with n = 5-8 per genotype

Fig. S3 HCK deficiency in Tg2576 mice did not modulate APP production and processing as well as BACE1 activity. (a) Representative immunoblots of full length and CTFs APP expression in hippocampal lysates of WT, HCK-KO, Tg2576 and Tg/HCK-KO mice using 6E10 and CT20 antibodies, respectively. GAPDH was probed as protein loading control. (b) Quantitative analysis of full length (6E10) and CTFs: C83 and C99 (CT20) band intensities after normalized to that of GAPDH. Deleting HCK in Tg2576 mice did not modulate the expression of full length APP nor that of fragments C83 and C99 from that of Tg2576 mice. However, they were highly expressed in the AD mice than in the WT or HCK-KO mice as expected. Data are expressed as mean ± SEM with n = 6-8 per genotype. ** p < 0.01, *** p < 0.001 and **** p < 0.0001 between indicated genotypes. (c) Representative immunoblots of immature (70 kDa) and mature (80 kDa) BACE1 expression in hippocampal lysates of WT, HCK-KO, Tg2576 and Tg/HCK-KO mice. Tubulin was probed as protein loading control. (d) Quantitative analysis of immature and mature BACE1 band intensities after normalized to that of tubulin. There were no apparent differences between the genotypes in both forms of BACE1. Data are expressed as mean ± SEM with n = 5-8 per genotype

Fig. S4 HCK deletion in Tg2576 mice resulted in apparent reduction of Thioflavin-S sphericity and Iba1+ microglia processes length. (a) Representative confocal images of Thioflavin-S plaque (green) clustered by Iba1+ (red) microglia in Tg2576 (left) and Tg/HCK-KO (right) mice (11-13 months old). Scale bar, 20 µm. Knocking out HCK reduced the sphericity/compactness of Thioflavin-S plaques by 24% when compared to Tg2576 mice (b). Quantification of Thioflavin-S plaque volume covered by Iba1+ cells per plaque volume per cell (c) did not show significant differences between the two mouse genotypes. Neither did volumetric and Imaris automated analyses of Iba1+ cells clustering around Thioflavin-S plaques showed significant differences in the number of cells per plaque volume (d), nor % Iba1+ cells clustering Thioflavin-S plaques per plaque volume per cell (e). Nonetheless, there was near significant reduction in the total processes length of Iba1+ microglia per microglia (f), but no drastic alteration in the microglia branching in Tg/HCK-KO mice (g). Plaques were analyzed in the hemibrains (excluding the thalamus) of 7 Tg2576 (n = 76) and 7-9 Tg/HCK-KO mice (n = 80-92). Data are expressed as mean ± SEM from one section per mouse

Fig. S5 HCK deficiency in Tg2576 mice significantly reduced the number of CD11b+ cells in microglial clusters. Clusters of microglial cells positively stained for CD11b revealed significant difference in the number of CD11b+ cells between Tg2576 and Tg/HCK-KO mice. Microglial clusters were analyzed in the hemibrains (excluding the thalamus) of 7 Tg2576 (n = 34) and 11 Tg/HCK-KO mice (n = 67). Data are expressed as mean ± SEM from 1 section per mouse. ** p < 0.01 relative to Tg2576 mice

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lim, S.L., Tran, D.N., Kieu, Z. et al. Genetic Ablation of Hematopoietic Cell Kinase Accelerates Alzheimer’s Disease–Like Neuropathology in Tg2576 Mice. Mol Neurobiol 57, 2447–2460 (2020). https://doi.org/10.1007/s12035-020-01894-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-020-01894-6

Keywords

Navigation