4.7 Article

Theranostic F-SLOH mitigates Alzheimer's disease pathology involving TFEB and ameliorates cognitive functions in Alzheimer's disease models

Journal

REDOX BIOLOGY
Volume 51, Issue -, Pages -

Publisher

ELSEVIER
DOI: 10.1016/j.redox.2022.102280

Keywords

Theranostic; Alzheimer's disease; A beta-targeting; A beta-aggregate inhibition; 3XTg-AD; 5XFAD

Funding

  1. Research Grants Council of Hong Kong Collaborative Research Fund [C2012-15G]
  2. Hong Kong Government [GRF/HKBU12101417, GRF/HKBU12100618, HMRF/17182541, HMRF/17182551]
  3. Hong Kong Baptist University [HKBU/RC-IRCs/17-18/03, IRCMS/19-20/H02]
  4. National Natural Science Foundation of China [NSFC 81703487, NSFC 81773926]
  5. Shenzhen Science and Technology Innovation Commission [JCYJ20180302174028790, JCYJ20180507184656626]

Ask authors/readers for more resources

The accumulation of phosphorylated Tau aggregates and A beta oligomers are crucial factors in the development of Alzheimer's disease. A study found that the fluorescent probe F-SLOH effectively inhibited A beta aggregation and reduced various neuropathological changes in AD mouse models by promoting autophagy lysosomal degradation pathway.
Accumulation of amyloid-beta (A beta) oligomers and phosphorylated Tau aggregates are crucial pathological events or factors that cause progressive neuronal loss, and cognitive impairments in Alzheimer's disease (AD). Current medications for AD have failed to halt, much less reverse this neurodegenerative disorder; therefore, there is an urgent need for the development of effective and safe drugs for AD therapy. In the present study, the in vivo therapeutic efficacy of an A beta-oligomer-targeted fluorescent probe, F-SLOH, was extensively investigated in 5XFAD and 3XTg-AD mouse models. We have shown that F-SLOH exhibits an efficient inhibitory activity against A beta aggregation in vivo, and acts as an effective theranostic agent for the treatment of multiple neuropathological changes in AD mouse models. F-SLOH has been found to significantly reduce not only the levels of A beta oligomers, Tau aggregates and plaques but also the levels of amyloid precursor protein (APP) and its metabolites via autophagy lysosomal degradation pathway (ALP) in the brains of 5XFAD and 3XTg-AD mice. It also reduces astrocyte activation and microgliosis ultimately alleviating neuro-inflammation. Furthermore, F-SLOH mitigates hyperphosphorylated Tau aggregates, synaptic deficits and ameliorates synaptic memory function, and cognitive impairment in AD mouse models. The mechanistic studies have shown that F-SLOH promotes the clearance of C terminal fragment 15 (CTF15) of APP and Paired helical filaments of Tau (PHF1) in stable cell models via the activation of transcription factor EB (TFEB). Moreover, F-SLOH promotes ALP and lysosomal biogenesis for the clearance of soluble, insoluble A beta, and phospho Tau. Our results unambiguously reveal effective etiological capabilities of theranostic F-SLOH to target and intervene multiple neuropathological changes in AD mouse models. Therefore, F-SLOH demonstrates tremendous therapeutic potential for treating AD in its early stage.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available