The Role of the JAK/STAT Signaling Pathway in the Pathogenesis of Alzheimer’s Disease: New Potential Treatment Target
Abstract
:1. Introduction
2. Alzheimer’s Disease
2.1. Modification of Amyloid and Tau Protein in Alzheimer’s Disease
2.2. The Role of Innate Immunity in Alzheimer’s Disease
2.3. Neuroinflammation
2.4. The Canonical JAK/STAT Signaling Pathway
2.5. Regulation of JAK/STAT Signaling Pathway
2.6. The JAK/STAT Signaling Pathway during Alzheimer’s Disease Progression
2.7. JAK/STAT as a Potential Therapeutic Target in Alzheimer’s Disease
2.7.1. Janus Kinase Inhibitors (Jakinibs)
2.7.2. JAK Inhibitors
2.7.3. STAT Inhibitors
2.7.4. Natural Products and Derivatives
3. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
Abbreviations
AD | Alzheimer’s disease |
ADAMTS13 | Disintegrin and metalloprotease with thrombospondin type I motif, member 13 |
APP | Amyloid protein precursor |
ASOs | Antisense oligonucleotides |
Aβ | Amyloid-β |
BACE1 | β-site amyloid precursor protein cleaving enzyme-1 |
BDNF | Brain-derived neurotrophic factor |
CDK5 | Cyclin-dependent kinase 5 |
CHEIs | Cholinesterase inhibitors |
CSZ | Cilostazol |
IFN | Interferon |
IL-6 | Inteleukin-6 |
IPAD | Intramural Peri-Arterial Drainage |
JAK | Janus kinase |
JAK/STAT | Janus kinase/signal transducer and activator of transcription signaling pathway |
KLF4 | Kruppel-like factor 4 |
MAPK | Mitogen-activated protein kinase |
NFTs | Neurofibrillary tangles |
ODN | Oligonucleotide decoys |
OT | Oxytocin |
PTPs | Protein tyrosine phosphatases |
PUFAs | Polyunsaturated fatty acids |
ROS | Reactive oxygen species |
SOCS | Suppressor of cytokine signaling |
STAT | Signal transducers and activator of transcription |
TLRs | Toll-like receptors |
TNF-α | Tumor necrosis factor-alfa |
TREM2 | Triggering receptors expressed on myeloid cells 2 |
TwX | Twendee X |
TXN | Taxifolin |
References
- Mayeux, R.; Stern, Y. Epidemiology of Alzheimer disease. Cold Spring Harb. Perspect. Med. 2012, 2, a006239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, X.-X.; Tian, Y.; Wang, Z.-T.; Ma, Y.-H.; Tan, L.; Yu, J.-T. The epidemiology of Alzheimer’s disease: Modifiable risk factors and prevention. J. Prev. Alzheimer’s Dis. 2021, 8, 313–321. [Google Scholar] [CrossRef] [PubMed]
- Barber, R.C. The genetics of Alzheimer’s disease. Scientifica 2012, 2012, 246210. [Google Scholar] [CrossRef] [Green Version]
- Jack, C.R.J.; Bennett, D.A.; Blennow, K.; Carrillo, M.C.; Dunn, B.; Haeberlein, S.B.; Holtzman, D.M.; Jagust, W.; Jessen, F.; Karlawish, J.; et al. NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 2018, 14, 535–562. [Google Scholar] [CrossRef] [PubMed]
- Morris, G.P.; Clark, I.A.; Vissel, B. Questions concerning the role of amyloid-β in the definition, aetiology and diagnosis of Alzheimer’s disease. Acta Neuropathol. 2018, 136, 663–689. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pluta, R.; Ułamek, M.; Jabłoński, M. Alzheimer’s mechanisms in ischemic brain degeneration. Anat. Rec. 2009, 292, 1863–1881. [Google Scholar] [CrossRef] [PubMed]
- Salminen, A.; Kauppinen, A.; Kaarniranta, K. Hypoxia/ischemia activate processing of Amyloid Precursor Protein: Impact of vascular dysfunction in the pathogenesis of Alzheimer’s disease. J. Neurochem. 2017, 140, 536–549. [Google Scholar] [CrossRef] [Green Version]
- Pluta, R.; Januszewski, S.; Czuczwar, S.J. Brain ischemia as a prelude to Alzheimer’s disease. Front. Aging Neurosci. 2021, 13, 636653. [Google Scholar] [CrossRef]
- Pluta, R. Brain ischemia as a bridge to Alzheimer’s disease. Neural Regen. Res. 2022, 17, 791–792. [Google Scholar] [CrossRef]
- Owen, K.L.; Brockwell, N.K.; Parker, B.S. JAK-STAT signaling: A double-edged sword of immune regulation and cancer progression. Cancers 2019, 11, 2002. [Google Scholar] [CrossRef]
- Hu, X.; Li, J.; Fu, M.; Zhao, X.; Wang, W. The JAK/STAT signaling pathway: From bench to clinic. Signal Transduct. Target. Ther. 2021, 6, 402. [Google Scholar] [CrossRef]
- Yu, X.; Li, X. microRNA-1906 protects cerebral ischemic injury through activating Janus kinase 2/signal transducer and activator of transcription 3 pathway in rats. Neuroreport 2020, 31, 871–878. [Google Scholar] [CrossRef]
- Dong, Y.; Hu, C.; Huang, C.; Gao, J.; Niu, W.; Wang, D.; Wang, Y.; Niu, C. Interleukin-22 plays a protective role by regulating the JAK2-STAT3 pathway to improve inflammation, oxidative stress, and neuronal apoptosis following cerebral ischemia-reperfusion injury. Mediat. Inflamm. 2021, 2021, 6621296. [Google Scholar] [CrossRef]
- Fan, L.; Zhou, L. AG490 protects cerebral ischemia/reperfusion injury via inhibiting the JAK2/3 signaling pathway. Brain Behav. 2021, 11, e01911. [Google Scholar] [CrossRef] [PubMed]
- Planas, A.M.; Gorina, R.; Chamorro, A. Signalling pathways mediating inflammatory responses in brain ischaemia. Biochem. Soc. Trans. 2006, 34, 1267–1270. [Google Scholar] [CrossRef] [PubMed]
- Lu, J.; Wang, J.; Yu, L.; Cui, R.; Zhang, Y.; Ding, H.; Yan, G. Shaoyao-Gancao decoction promoted microglia M2 polarization via the IL-13-mediated JAK2/STAT6 pathway to alleviate cerebral ischemia-reperfusion injury. Mediat. Inflamm. 2022, 2022, 1707122. [Google Scholar] [CrossRef]
- Nevado-Holgado, A.J.; Ribe, E.; Thei, L.; Furlong, L.; Mayer, M.-A.; Quan, J.; Richardson, J.C.; Cavanagh, J.; Consortium, N.; Lovestone, S. Genetic and real-world clinical data, combined with empirical validation, nominate Jak/Stat signaling as a target for Alzheimer’s disease therapeutic development. Cells 2019, 8, 425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, L.; Liu, Y.; Wang, Y.; Li, J.; Liu, N. Azeliragon ameliorates Alzheimer’s disease via the Janus tyrosine kinase and signal transducer and activator of transcription signaling pathway. Clinics 2021, 76, e2348. [Google Scholar] [CrossRef] [PubMed]
- Saharinen, P.; Takaluoma, K.; Silvennoinen, O. Regulation of the Jak2 tyrosine kinase by its pseudokinase domain. Mol. Cell. Biol. 2000, 20, 3387–3395. [Google Scholar] [CrossRef]
- Vickers, J.C.; Mitew, S.; Woodhouse, A.; Fernandez-Martos, C.M.; Kirkcaldie, M.T.; Canty, A.J.; McCormack, G.H.; King, A.E. Defining the earliest pathological changes of Alzheimer’s disease. Curr. Alzheimer Res. 2016, 13, 281–287. [Google Scholar] [CrossRef] [PubMed]
- Nussbaum, R.L. Genome-wide association studies, Alzheimer disease, and understudied populations. JAMA 2013, 309, 1527–1528. [Google Scholar] [CrossRef] [PubMed]
- Mahley, R.W.; Weisgraber, K.H.; Huang, Y. Apolipoprotein E4: A causative factor and therapeutic target in neuropathology, including Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2006, 103, 5644–5651. [Google Scholar] [CrossRef] [Green Version]
- Reitz, C.; Mayeux, R. Alzheimer disease: Epidemiology, diagnostic criteria, risk factors and biomarkers. Biochem. Pharmacol. 2014, 88, 640–651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wen, Y.; Yang, S.-H.; Liu, R.; Perez, E.J.; Brun-Zinkernagel, A.M.; Koulen, P.; Simpkins, J.W. Cdk5 is involved in NFT-like tauopathy induced by transient cerebral ischemia in female rats. Biochim. Biophys. Acta—Mol. Basis Dis. 2007, 1772, 473–483. [Google Scholar] [CrossRef] [PubMed]
- Cheung, Z.H.; Gong, K.; Ip, N.Y. Cyclin-dependent kinase 5 supports neuronal survival through phosphorylation of Bcl-2. J. Neurosci. 2008, 28, 4872–4877. [Google Scholar] [CrossRef] [Green Version]
- Mushtaq, G.; Greig, N.H.; Anwar, F.; Al-Abbasi, F.A.; Zamzami, M.A.; Al-Talhi, H.A.; Kamal, M.A. Neuroprotective mechanisms mediated by CDK5 inhibition. Curr. Pharm. Des. 2016, 22, 527–534. [Google Scholar] [CrossRef] [Green Version]
- Kivipelto, M.; Helkala, E.-L.; Hänninen, T.; Laakso, M.P.; Hallikainen, M.; Alhainen, K.; Soininen, H.; Tuomilehto, J.; Nissinen, A. Midlife vascular risk factors and late-life mild cognitive impairment. Neurology 2001, 56, 1683–1689. [Google Scholar] [CrossRef]
- Swan, G.E.; DeCarli, C.; Miller, B.L.; Reed, T.; Wolf, P.A.; Jack, L.M.; Carmelli, D. Association of midlife blood pressure to late-life cognitive decline and brain morphology. Neurology 1998, 51, 986–993. [Google Scholar] [CrossRef]
- Serrano-Pozo, A.; Frosch, M.P.; Masliah, E.; Hyman, B.T. Neuropathological alterations in Alzheimer disease. Cold Spring Harb. Perspect. Med. 2011, 1, a006189. [Google Scholar] [CrossRef] [Green Version]
- Viola, K.L.; Klein, W.L. Amyloid beta oligomers in Alzheimer’s disease pathogenesis, treatment, and diagnosis. Acta Neuropathol. 2015, 129, 183–206. [Google Scholar] [CrossRef]
- Perl, D.P. Neuropathology of Alzheimer’s disease. Mt. Sinai J. Med. 2010, 77, 32–42. [Google Scholar] [CrossRef] [PubMed]
- Kang, J.; Lemaire, H.G.; Unterbeck, A.; Salbaum, J.M.; Masters, C.L.; Grzeschik, K.H.; Multhaup, G.; Beyreuther, K.; Müller-Hill, B. The precursor of Alzheimer’s disease amyloid A4 protein resembles a cell-surface receptor. Nature 1987, 325, 733–736. [Google Scholar] [CrossRef] [PubMed]
- McCarter, J.F.; Liebscher, S.; Bachhuber, T.; Abou-Ajram, C.; Hübener, M.; Hyman, B.T.; Haass, C.; Meyer-Luehmann, M. Clustering of plaques contributes to plaque growth in a mouse model of Alzheimer’s disease. Acta Neuropathol. 2013, 126, 179–188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haroutunian, V.; Schnaider-Beeri, M.; Schmeidler, J.; Wysocki, M.; Purohit, D.P.; Perl, D.P.; Libow, L.S.; Lesser, G.T.; Maroukian, M.; Grossman, H.T. Role of the neuropathology of Alzheimer disease in dementia in the oldest-old. Arch. Neurol. 2008, 65, 1211–1217. [Google Scholar] [CrossRef] [Green Version]
- Wiśniewski, H.M.; Narang, H.K.; Terry, R.D. Neurofibrillary tangles of paired helical filaments. J. Neurol. Sci. 1976, 27, 173–181. [Google Scholar] [CrossRef]
- Lee, V.M.; Balin, B.J.; Otvos, L.J.; Trojanowski, J.Q. A68: A major subunit of paired helical filaments and derivatized forms of normal Tau. Science 1991, 251, 675–678. [Google Scholar] [CrossRef]
- Bierer, L.M.; Hof, P.R.; Purohit, D.P.; Carlin, L.; Schmeidler, J.; Davis, K.L.; Perl, D.P. Neocortical neurofibrillary tangles correlate with dementia severity in Alzheimer’s disease. Arch. Neurol. 1995, 52, 81–88. [Google Scholar] [CrossRef]
- Funk, K.E.; Mrak, R.E.; Kuret, J. Granulovacuolar degeneration (GVD) bodies of Alzheimer’s disease (AD) resemble late-stage autophagic organelles. Neuropathol. Appl. Neurobiol. 2011, 37, 295–306. [Google Scholar] [CrossRef] [Green Version]
- Okamoto, K.; Hirai, S.; Iizuka, T.; Yanagisawa, T.; Watanabe, M. Reexamination of granulovacuolar degeneration. Acta Neuropathol. 1991, 82, 340–345. [Google Scholar] [CrossRef]
- Ball, M.J.; Lo, P. Granulovacuolar degeneration in the ageing brain and in dementia. J. Neuropathol. Exp. Neurol. 1977, 36, 474–487. [Google Scholar] [CrossRef]
- Chen, X.-Q.; Mobley, W.C. Alzheimer disease pathogenesis: Insights from molecular and cellular biology studies of oligomeric Aβ and tau species. Front. Neurosci. 2019, 13, 659. [Google Scholar] [CrossRef] [PubMed]
- Spears, W.; Furgerson, M.; Sweetnam, J.M.; Evans, P.; Gearing, M.; Fechheimer, M.; Furukawa, R. Hirano bodies differentially modulate cell death induced by tau and the amyloid precursor protein intracellular domain. BMC Neurosci. 2014, 15, 74. [Google Scholar] [CrossRef] [Green Version]
- Lima-Filho, R.A.S.; Oliveira, M.M. A role for cellular prion protein in late-onset alzheimer’s disease: Evidence from preclinical studies. J. Neurosci. 2018, 38, 2146–2148. [Google Scholar] [CrossRef] [Green Version]
- Masliah, E.; Terry, R. The role of synaptic proteins in the pathogenesis of disorders of the central nervous system. Brain Pathol. 1993, 3, 77–85. [Google Scholar] [CrossRef] [PubMed]
- Wang, M.-M.; Miao, D.; Cao, X.-P.; Tan, L.; Tan, L. Innate immune activation in Alzheimer’s disease. Ann. Transl. Med. 2018, 6, 177. [Google Scholar] [CrossRef] [PubMed]
- Benarroch, E.E. Microglia: Multiple roles in surveillance, circuit shaping, and response to injury. Neurology 2013, 81, 1079–1088. [Google Scholar] [CrossRef]
- Kettenmann, H.; Hanisch, U.-K.; Noda, M.; Verkhratsky, A. Physiology of microglia. Physiol. Rev. 2011, 91, 461–553. [Google Scholar] [CrossRef]
- Parkhurst, C.N.; Yang, G.; Ninan, I.; Savas, J.N.; Yates, J.R., 3rd; Lafaille, J.J.; Hempstead, B.L.; Littman, D.R.; Gan, W.-B. Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell 2013, 155, 1596–1609. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chao, M. V Neurotrophins and their receptors: A convergence point for many signalling pathways. Nat. Rev. Neurosci. 2003, 4, 299–309. [Google Scholar] [CrossRef]
- Rauskolb, S.; Zagrebelsky, M.; Dreznjak, A.; Deogracias, R.; Matsumoto, T.; Wiese, S.; Erne, B.; Sendtner, M.; Schaeren-Wiemers, N.; Korte, M.; et al. Global deprivation of brain-derived neurotrophic factor in the CNS reveals an area specific requirement for dendritic growth. J. Neurosci. 2010, 30, 1739–1749. [Google Scholar] [CrossRef]
- Morris, G.P.; Clark, I.A.; Zinn, R.; Vissel, B. Microglia: A new frontier for synaptic plasticity, learning and memory, and neurodegenerative disease research. Neurobiol. Learn. Mem. 2013, 105, 40–53. [Google Scholar] [CrossRef] [PubMed]
- Jiang, T.; Yu, J.-T.; Zhu, X.-C.; Tan, L. TREM2 in Alzheimer’s disease. Mol. Neurobiol. 2013, 48, 180–185. [Google Scholar] [CrossRef] [PubMed]
- Heneka, M.T.; Kummer, M.P.; Latz, E. Innate immune activation in neurodegenerative disease. Nat. Rev. Immunol. 2014, 14, 463–477. [Google Scholar] [CrossRef]
- Jiang, T.; Tan, L.; Zhu, X.-C.; Zhang, Q.-Q.; Cao, L.; Tan, M.-S.; Gu, L.-Z.; Wang, H.-F.; Ding, Z.-Z.; Zhang, Y.-D.; et al. Upregulation of TREM2 ameliorates neuropathology and rescues spatial cognitive impairment in a transgenic mouse model of Alzheimer’s disease. Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol. 2014, 39, 2949–2962. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Crocker, P.R.; Redelinghuys, P. Siglecs as positive and negative regulators of the immune system. Biochem. Soc. Trans. 2008, 36, 1467–1471. [Google Scholar] [CrossRef]
- Yuan, Q.; Chu, C.; Jia, J. Association studies of 19 candidate SNPs with sporadic Alzheimer’s disease in the North Chinese Han population. Neurol. Sci. 2012, 33, 1021–1028. [Google Scholar] [CrossRef]
- Jiang, T.; Yu, J.-T.; Hu, N.; Tan, M.-S.; Zhu, X.-C.; Tan, L. CD33 in Alzheimer’s disease. Mol. Neurobiol. 2014, 49, 529–535. [Google Scholar] [CrossRef] [PubMed]
- Bradshaw, E.M.; Chibnik, L.B.; Keenan, B.T.; Ottoboni, L.; Raj, T.; Tang, A.; Rosenkrantz, L.L.; Imboywa, S.; Lee, M.; Von Korff, A.; et al. CD33 Alzheimer’s disease locus: Altered monocyte function and amyloid biology. Nat. Neurosci. 2013, 16, 848–850. [Google Scholar] [CrossRef] [PubMed]
- Drouin-Ouellet, J.; Cicchetti, F. Inflammation and neurodegeneration: The story “retolled”. Trends Pharmacol. Sci. 2012, 33, 542–551. [Google Scholar] [CrossRef]
- Su, F.; Bai, F.; Zhou, H.; Zhang, Z. Microglial toll-like receptors and Alzheimer’s disease. Brain. Behav. Immun. 2016, 52, 187–198. [Google Scholar] [CrossRef]
- Doi, Y.; Mizuno, T.; Maki, Y.; Jin, S.; Mizoguchi, H.; Ikeyama, M.; Doi, M.; Michikawa, M.; Takeuchi, H.; Suzumura, A. Microglia activated with the toll-like receptor 9 ligand CpG attenuate oligomeric amyloid {beta} neurotoxicity in in vitro and in vivo models of Alzheimer’s disease. Am. J. Pathol. 2009, 175, 2121–2132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Scholtzova, H.; Chianchiano, P.; Pan, J.; Sun, Y.; Goñi, F.; Mehta, P.D.; Wisniewski, T. Amyloid β and Tau Alzheimer’s disease related pathology is reduced by Toll-like receptor 9 stimulation. Acta Neuropathol. Commun. 2014, 2, 101. [Google Scholar] [CrossRef] [Green Version]
- Liu, C.; Cui, G.; Zhu, M.; Kang, X.; Guo, H. Neuroinflammation in Alzheimer’s disease: Chemokines produced by astrocytes and chemokine receptors. Int. J. Clin. Exp. Pathol. 2014, 7, 8342–8355. [Google Scholar] [PubMed]
- Heneka, M.T.; Carson, M.J.; El Khoury, J.; Landreth, G.E.; Brosseron, F.; Feinstein, D.L.; Jacobs, A.H.; Wyss-Coray, T.; Vitorica, J.; Ransohoff, R.M.; et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015, 14, 388–405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Latta, C.H.; Brothers, H.M.; Wilcock, D.M. Neuroinflammation in Alzheimer’s disease; a source of heterogeneity and target for personalized therapy. Neuroscience 2015, 302, 103–111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Phillips, E.C.; Croft, C.L.; Kurbatskaya, K.; O’Neill, M.J.; Hutton, M.L.; Hanger, D.P.; Garwood, C.J.; Noble, W. Astrocytes and neuroinflammation in Alzheimer’s disease. Biochem. Soc. Trans. 2014, 42, 1321–1325. [Google Scholar] [CrossRef]
- Sawikr, Y.; Yarla, N.S.; Peluso, I.; Kamal, M.A.; Aliev, G.; Bishayee, A. Neuroinflammation in Alzheimer’s disease: The preventive and therapeutic potential of polyphenolic nutraceuticals. Adv. Protein Chem. Struct. Biol. 2017, 108, 33–57. [Google Scholar] [CrossRef]
- Fu, X.Y.; Kessler, D.S.; Veals, S.A.; Levy, D.E.; Darnell, J.E.J. ISGF3, the transcriptional activator induced by interferon alpha, consists of multiple interacting polypeptide chains. Proc. Natl. Acad. Sci. USA 1990, 87, 8555–8559. [Google Scholar] [CrossRef] [Green Version]
- Müller, M.; Briscoe, J.; Laxton, C.; Guschin, D.; Ziemiecki, A.; Silvennoinen, O.; Harpur, A.G.; Barbieri, G.; Witthuhn, B.A.; Schindler, C. The protein tyrosine kinase JAK1 complements defects in interferon-alpha/beta and -gamma signal transduction. Nature 1993, 366, 129–135. [Google Scholar] [CrossRef]
- Lai, K.S.; Jin, Y.; Graham, D.K.; Witthuhn, B.A.; Ihle, J.N.; Liu, E.T. A kinase-deficient splice variant of the human JAK3 is expressed in hematopoietic and epithelial cancer cells. J. Biol. Chem. 1995, 270, 25028–25036. [Google Scholar] [CrossRef]
- Nicolas, C.S.; Amici, M.; Bortolotto, Z.A.; Doherty, A.; Csaba, Z.; Fafouri, A.; Dournaud, P.; Gressens, P.; Collingridge, G.L.; Peineau, S. The role of JAK-STAT signaling within the CNS. JAK-STAT 2013, 2, e22925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- O’Shea, J.J.; Schwartz, D.M.; Villarino, A.V.; Gadina, M.; McInnes, I.B.; Laurence, A. The JAK-STAT pathway: Impact on human disease and therapeutic intervention. Annu. Rev. Med. 2015, 66, 311–328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shuai, K.; Liu, B. Regulation of JAK-STAT signalling in the immune system. Nat. Rev. Immunol. 2003, 3, 900–911. [Google Scholar] [CrossRef] [PubMed]
- Hilton, D.J. Negative regulators of cytokine signal transduction. Cell. Mol. Life Sci. 1999, 55, 1568–1577. [Google Scholar] [CrossRef] [PubMed]
- Greenhalgh, C.J.; Hilton, D.J. Negative regulation of cytokine signaling. J. Leukoc. Biol. 2001, 70, 348–356. [Google Scholar] [CrossRef]
- Starr, R.; Willson, T.A.; Viney, E.M.; Murray, L.J.; Rayner, J.R.; Jenkins, B.J.; Gonda, T.J.; Alexander, W.S.; Metcalf, D.; Nicola, N.A.; et al. A family of cytokine-inducible inhibitors of signalling. Nature 1997, 387, 917–921. [Google Scholar] [CrossRef]
- Nicholson, S.E.; Willson, T.A.; Farley, A.; Starr, R.; Zhang, J.-G.; Baca, M.; Alexander, W.S.; Metcalf, D.; Hilton, D.J.; Nicola, N.A. Mutational analyses of the SOCS proteins suggest a dual domain requirement but distinct mechanisms for inhibition of LIF and IL-6 signal transduction. EMBO J. 1999, 18, 375–385. [Google Scholar] [CrossRef]
- Yoshimura, A. The CIS family: Negative regulators of JAK-STAT signaling. Cytokine Growth Factor Rev. 1998, 9, 197–204. [Google Scholar] [CrossRef]
- Penninger, J.M.; Irie-Sasaki, J.; Sasaki, T.; Oliveira-dos-Santos, A.J. CD45: New jobs for an old acquaintance. Nat. Immunol. 2001, 2, 389–396. [Google Scholar] [CrossRef]
- Irie-Sasaki, J.; Sasaki, T.; Matsumoto, W.; Opavsky, A.; Cheng, M.; Welstead, G.; Griffiths, E.; Krawczyk, C.; Richardson, C.D.; Aitken, K.; et al. CD45 is a JAK phosphatase and negatively regulates cytokine receptor signalling. Nature 2001, 409, 349–354. [Google Scholar] [CrossRef]
- Cheng, A.; Uetani, N.; Simoncic, P.D.; Chaubey, V.P.; Lee-Loy, A.; McGlade, C.J.; Kennedy, B.P.; Tremblay, M.L. Attenuation of leptin action and regulation of obesity by protein tyrosine phosphatase 1B. Dev. Cell 2002, 2, 497–503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mowen, K.A.; Tang, J.; Zhu, W.; Schurter, B.T.; Shuai, K.; Herschman, H.R.; David, M. Arginine methylation of STAT1 modulates IFNalpha/beta-induced transcription. Cell 2001, 104, 731–741. [Google Scholar] [CrossRef] [PubMed]
- Liu, B.; Liao, J.; Rao, X.; Kushner, S.A.; Chung, C.D.; Chang, D.D.; Shuai, K. Inhibition of Stat1-mediated gene activation by PIAS1. Proc. Natl. Acad. Sci. USA 1998, 95, 10626–10631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shuai, K. Modulation of STAT signaling by STAT-interacting proteins. Oncogene 2000, 19, 2638–2644. [Google Scholar] [CrossRef] [Green Version]
- Rycyzyn, M.A.; Clevenger, C. V The intranuclear prolactin/cyclophilin B complex as a transcriptional inducer. Proc. Natl. Acad. Sci. USA 2002, 99, 6790–6795. [Google Scholar] [CrossRef] [Green Version]
- Chen, Y.; Wen, R.; Yang, S.; Schuman, J.; Zhang, E.E.; Yi, T.; Feng, G.-S.; Wang, D. Identification of Shp-2 as a Stat5A phosphatase. J. Biol. Chem. 2003, 278, 16520–16527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aoki, N.; Matsuda, T. A cytosolic protein-tyrosine phosphatase PTP1B specifically dephosphorylates and deactivates prolactin-activated STAT5a and STAT5b. J. Biol. Chem. 2000, 275, 39718–39726. [Google Scholar] [CrossRef] [Green Version]
- ten Hoeve, J.; de Jesus Ibarra-Sanchez, M.; Fu, Y.; Zhu, W.; Tremblay, M.; David, M.; Shuai, K. Identification of a nuclear Stat1 protein tyrosine phosphatase. Mol. Cell. Biol. 2002, 22, 5662–5668. [Google Scholar] [CrossRef] [Green Version]
- Costa-Pereira, A.P.; Tininini, S.; Strobl, B.; Alonzi, T.; Schlaak, J.F.; Is’harc, H.; Gesualdo, I.; Newman, S.J.; Kerr, I.M.; Poli, V. Mutational switch of an IL-6 response to an interferon-γ-like response. Proc. Natl. Acad. Sci. USA 2002, 99, 8043–8047. [Google Scholar] [CrossRef] [Green Version]
- Zhang, B.; Gaiteri, C.; Bodea, L.-G.; Wang, Z.; McElwee, J.; Podtelezhnikov, A.A.; Zhang, C.; Xie, T.; Tran, L.; Dobrin, R.; et al. Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer’s disease. Cell 2013, 153, 707–720. [Google Scholar] [CrossRef]
- Chiba, T.; Yamada, M.; Aiso, S. Targeting the JAK2/STAT3 axis in Alzheimer’s disease. Expert Opin. Ther. Targets 2009, 13, 1155–1167. [Google Scholar] [CrossRef] [PubMed]
- Chiba, T.; Yamada, M.; Sasabe, J.; Terashita, K.; Shimoda, M.; Matsuoka, M.; Aiso, S. Amyloid-beta causes memory impairment by disturbing the JAK2/STAT3 axis in hippocampal neurons. Mol. Psychiatry 2009, 14, 206–222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alvarez, A.; Cacabelos, R.; Sanpedro, C.; García-Fantini, M.; Aleixandre, M. Serum TNF-alpha levels are increased and correlate negatively with free IGF-I in Alzheimer disease. Neurobiol. Aging 2007, 28, 533–536. [Google Scholar] [CrossRef] [PubMed]
- Li, W.; Liu, H.; Yu, M.; Zhang, X.; Zhang, Y.; Liu, H.; Wilson, J.X.; Huang, G. Folic acid alters methylation profile of JAK-STAT and long-term depression signaling pathways in Alzheimer’s disease models. Mol. Neurobiol. 2016, 53, 6548–6556. [Google Scholar] [CrossRef]
- Bakulski, K.M.; Dolinoy, D.C.; Sartor, M.A.; Paulson, H.L.; Konen, J.R.; Lieberman, A.P.; Albin, R.L.; Hu, H.; Rozek, L.S. Genome-wide DNA methylation differences between late-onset Alzheimer’s disease and cognitively normal controls in human frontal cortex. J. Alzheimers Dis. 2012, 29, 571–588. [Google Scholar] [CrossRef] [Green Version]
- Kalaria, R.N. The role of cerebral ischemia in Alzheimer’s disease. Neurobiol. Aging 2000, 21, 321–330. [Google Scholar] [CrossRef]
- Chiu, Y.-J.; Lin, C.-H.; Lee, M.-C.; Hsieh-Li, H.M.; Chen, C.-M.; Wu, Y.-R.; Chang, K.-H.; Lee-Chen, G.-J. Formulated Chinese medicine Shaoyao Gancao Tang reduces NLRP1 and NLRP3 in Alzheimer’s disease cell and mouse models for neuroprotection and cognitive improvement. Aging 2021, 13, 15620–15637. [Google Scholar] [CrossRef]
- Schwartz, D.M.; Kanno, Y.; Villarino, A.; Ward, M.; Gadina, M.; O’Shea, J.J. JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nat. Rev. Drug Discov. 2017, 16, 843–862. [Google Scholar] [CrossRef]
- Cheng, Z.; Zou, X.; Jin, Y.; Gao, S.; Lv, J.; Li, B.; Cui, R. The role of KLF (4) in Alzheimer’s disease. Front. Cell. Neurosci. 2018, 12, 325. [Google Scholar] [CrossRef] [Green Version]
- Yui, D.; Nishida, Y.; Nishina, T.; Mogushi, K.; Tajiri, M.; Ishibashi, S.; Ajioka, I.; Ishikawa, K.; Mizusawa, H.; Murayama, S.; et al. Enhanced phospholipase A2 group 3 expression by oxidative stress decreases the insulin-degrading enzyme. PLoS ONE 2015, 10, e0143518. [Google Scholar] [CrossRef]
- Cui, D.-M.; Zeng, T.; Ren, J.; Wang, K.; Jin, Y.; Zhou, L.; Gao, L. KLF4 knockdown attenuates TBI-induced neuronal damage through p53 and JAK-STAT3 signaling. CNS Neurosci. Ther. 2017, 23, 106–118. [Google Scholar] [CrossRef] [PubMed]
- Qin, S.; Zou, Y.; Zhang, C.-L. Cross-talk between KLF4 and STAT3 regulates axon regeneration. Nat. Commun. 2013, 4, 2633. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, J.; Wang, X.; Yi, X.; Wang, Y.; Liu, Q.; Ge, R. Induction of KLF4 contributes to the neurotoxicity of MPP + in M17 cells: A new implication in Parkinson’s disease. J. Mol. Neurosci. 2013, 51, 109–117. [Google Scholar] [CrossRef]
- Ohnesorge, N.; Viemann, D.; Schmidt, N.; Czymai, T.; Spiering, D.; Schmolke, M.; Ludwig, S.; Roth, J.; Goebeler, M.; Schmidt, M. Erk5 activation elicits a vasoprotective endothelial phenotype via induction of Kruppel-like factor 4 (KLF4). J. Biol. Chem. 2010, 285, 26199–26210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, H.; Xi, X.; Zhao, B.; Su, Z.; Wang, Z. KLF4 protects brain microvascular endothelial cells from ischemic stroke induced apoptosis by transcriptionally activating MALAT1. Biochem. Biophys. Res. Commun. 2018, 495, 2376–2382. [Google Scholar] [CrossRef] [PubMed]
- Rubio-Perez, J.M.; Morillas-Ruiz, J.M. A review: Inflammatory process in Alzheimer’s disease, role of cytokines. Sci. World J. 2012, 2012, 756357. [Google Scholar] [CrossRef] [PubMed]
- Peters, D.G.; Connor, J.R.; Meadowcroft, M.D. The relationship between iron dyshomeostasis and amyloidogenesis in Alzheimer’s disease: Two sides of the same coin. Neurobiol. Dis. 2015, 81, 49–65. [Google Scholar] [CrossRef] [Green Version]
- Tronel, C.; Rochefort, G.Y.; Arlicot, N.; Bodard, S.; Chalon, S.; Antier, D. Oxidative stress is related to the deleterious effects of heme oxygenase-1 in an in vivo neuroinflammatory rat model. Oxid. Med. Cell. Longev. 2013, 2013, 264935. [Google Scholar] [CrossRef] [Green Version]
- Burel, S.A.; Han, S.-R.; Lee, H.-S.; Norris, D.A.; Lee, B.-S.; Machemer, T.; Park, S.-Y.; Zhou, T.; He, G.; Kim, Y.; et al. Preclinical evaluation of the toxicological effects of a novel constrained ethyl modified antisense compound targeting signal transducer and activator of transcription 3 in mice and cynomolgus monkeys. Nucleic Acid Ther. 2013, 23, 213–227. [Google Scholar] [CrossRef]
- Taylor, P.C. Clinical efficacy of launched JAK inhibitors in rheumatoid arthritis. Rheumatology 2019, 58, i17–i26. [Google Scholar] [CrossRef]
- Miklossy, G.; Hilliard, T.S.; Turkson, J. Therapeutic modulators of STAT signalling for human diseases. Nat. Rev. Drug Discov. 2013, 12, 611–629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Turkson, J. STAT proteins as novel targets for cancer drug discovery. Expert Opin. Ther. Targets 2004, 8, 409–422. [Google Scholar] [CrossRef]
- Siddiquee, K.A.Z.; Gunning, P.T.; Glenn, M.; Katt, W.P.; Zhang, S.; Schrock, C.; Sebti, S.M.; Jove, R.; Hamilton, A.D.; Turkson, J. An oxazole-based small-molecule STAT3 inhibitor modulates STAT3 stability and processing and induces antitumor cell effects. ACS Chem. Biol. 2007, 2, 787–798. [Google Scholar] [CrossRef] [PubMed]
- Schust, J.; Sperl, B.; Hollis, A.; Mayer, T.U.; Berg, T. Stattic: A small-molecule inhibitor of STAT3 activation and dimerization. Chem. Biol. 2006, 13, 1235–1242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hückel, M.; Schurigt, U.; Wagner, A.H.; Stöckigt, R.; Petrow, P.K.; Thoss, K.; Gajda, M.; Henzgen, S.; Hecker, M.; Bräuer, R. Attenuation of murine antigen-induced arthritis by treatment with a decoy oligodeoxynucleotide inhibiting signal transducer and activator of transcription-1 (STAT-1). Arthritis Res. Ther. 2005, 8, R17. [Google Scholar] [CrossRef] [Green Version]
- Shen, J.; Li, R.; Li, G. Inhibitory effects of decoy-ODN targeting activated STAT3 on human glioma growth in vivo. In Vivo 2009, 23, 237–243. [Google Scholar]
- Sen, M.; Joyce, S.; Panahandeh, M.; Li, C.; Thomas, S.M.; Maxwell, J.; Wang, L.; Gooding, W.E.; Johnson, D.E.; Grandis, J.R. Targeting STAT3 abrogates EGFR inhibitor resistance in cancer. Clin. Cancer Res. 2012, 18, 4986–4996. [Google Scholar] [CrossRef] [Green Version]
- Niu, G.; Wright, K.L.; Huang, M.; Song, L.; Haura, E.; Turkson, J.; Zhang, S.; Wang, T.; Sinibaldi, D.; Coppola, D.; et al. Constitutive Stat3 activity up-regulates VEGF expression and tumor angiogenesis. Oncogene 2002, 21, 2000–2008. [Google Scholar] [CrossRef] [Green Version]
- Howard, R.J.; Juszczak, E.; Ballard, C.G.; Bentham, P.; Brown, R.G.; Bullock, R.; Burns, A.S.; Holmes, C.; Jacoby, R.; Johnson, T.; et al. Donepezil for the treatment of agitation in Alzheimer’s disease. N. Engl. J. Med. 2007, 357, 1382–1392. [Google Scholar] [CrossRef] [Green Version]
- Tariot, P.N.; Farlow, M.R.; Grossberg, G.T.; Graham, S.M.; McDonald, S.; Gergel, I. Memantine treatment in patients with moderate to severe Alzheimer disease already receiving donepezil: A randomized controlled trial. JAMA 2004, 291, 317–324. [Google Scholar] [CrossRef]
- Porro, C.; Cianciulli, A.; Trotta, T.; Lofrumento, D.D.; Panaro, M.A. Curcumin regulates anti-inflammatory responses by JAK/STAT/SOCS signaling pathway in BV-2 microglial cells. Biology 2019, 8, 51. [Google Scholar] [CrossRef] [PubMed]
- Rusek, M.; Czuczwar, S.J. The role of curcumin in post-ischemic brain. In Cerebral Ischemia; Pluta, R., Ed.; Exon Publications: Brisbane, Australia, 2021; ISBN 978-0-6450017-9-2. [Google Scholar]
- Ma, C.; Wang, Y.; Dong, L.; Li, M.; Cai, W. Anti-inflammatory effect of resveratrol through the suppression of NF-κB and JAK/STAT signaling pathways. Acta Biochim. Biophys. Sin. 2015, 47, 207–213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Semwal, D.K.; Semwal, R.B.; Combrinck, S.; Viljoen, A. Myricetin: A dietary molecule with diverse biological activities. Nutrients 2016, 8, 90. [Google Scholar] [CrossRef] [Green Version]
- Taheri, Y.; Suleria, H.A.R.; Martins, N.; Sytar, O.; Beyatli, A.; Yeskaliyeva, B.; Seitimova, G.; Salehi, B.; Semwal, P.; Painuli, S.; et al. Myricetin bioactive effects: Moving from preclinical evidence to potential clinical applications. BMC Complement. Med. Ther. 2020, 20, 241. [Google Scholar] [CrossRef] [PubMed]
- Song, X.; Tan, L.; Wang, M.; Ren, C.; Guo, C.; Yang, B.; Ren, Y.; Cao, Z.; Li, Y.; Pei, J. Myricetin: A review of the most recent research. Biomed. Pharmacother. 2021, 134, 111017. [Google Scholar] [CrossRef] [PubMed]
- Clark, L.F.; Kodadek, T. The immune system and neuroinflammation as potential sources of blood-based biomarkers for Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease. ACS Chem. Neurosci. 2016, 7, 520–527. [Google Scholar] [CrossRef] [PubMed]
- Leung, R.; Proitsi, P.; Simmons, A.; Lunnon, K.; Güntert, A.; Kronenberg, D.; Pritchard, M.; Tsolaki, M.; Mecocci, P.; Kloszewska, I.; et al. Inflammatory proteins in plasma are associated with severity of Alzheimer’s disease. PLoS ONE 2013, 8, e64971. [Google Scholar] [CrossRef] [Green Version]
- Heppner, F.L.; Ransohoff, R.M.; Becher, B. Immune attack: The role of inflammation in Alzheimer disease. Nat. Rev. Neurosci. 2015, 16, 358–372. [Google Scholar] [CrossRef]
- Ferreira, S.T.; Clarke, J.R.; Bomfim, T.R.; De Felice, F.G. Inflammation, defective insulin signaling, and neuronal dysfunction in Alzheimer’s disease. Alzheimers Dement. 2014, 10, S76–S83. [Google Scholar] [CrossRef] [Green Version]
- McManus, R.M.; Heneka, M.T. Role of neuroinflammation in neurodegeneration: New insights. Alzheimers Res. Ther. 2017, 9, 14. [Google Scholar] [CrossRef] [Green Version]
- McGeer, P.L.; Schulzer, M.; McGeer, E.G. Arthritis and anti-inflammatory agents as possible protective factors for Alzheimer’s disease. Neurology 1996, 47, 425–432. [Google Scholar] [CrossRef] [PubMed]
- Gao, Q.; Liang, X.; Shaikh, A.S.; Zang, J.; Xu, W.; Zhang, Y. JAK/STAT signal transduction: Promising attractive targets for immune, inflammatory, and hematopoietic diseases. Curr. Drug Targets 2018, 19, 487–500. [Google Scholar] [CrossRef] [PubMed]
- Popiolek-Barczyk, K.; Mika, J. Targeting the microglial signaling pathways: New insights in the modulation of neuropathic pain. Curr. Med. Chem. 2016, 23, 2908–2928. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Rusek, M.; Smith, J.; El-Khatib, K.; Aikins, K.; Czuczwar, S.J.; Pluta, R. The Role of the JAK/STAT Signaling Pathway in the Pathogenesis of Alzheimer’s Disease: New Potential Treatment Target. Int. J. Mol. Sci. 2023, 24, 864. https://doi.org/10.3390/ijms24010864
Rusek M, Smith J, El-Khatib K, Aikins K, Czuczwar SJ, Pluta R. The Role of the JAK/STAT Signaling Pathway in the Pathogenesis of Alzheimer’s Disease: New Potential Treatment Target. International Journal of Molecular Sciences. 2023; 24(1):864. https://doi.org/10.3390/ijms24010864
Chicago/Turabian StyleRusek, Marta, Joanna Smith, Kamel El-Khatib, Kennedy Aikins, Stanisław J. Czuczwar, and Ryszard Pluta. 2023. "The Role of the JAK/STAT Signaling Pathway in the Pathogenesis of Alzheimer’s Disease: New Potential Treatment Target" International Journal of Molecular Sciences 24, no. 1: 864. https://doi.org/10.3390/ijms24010864