Next Article in Journal
Design and Structure of a Non-Coaxial Multi-Focal Composite Fresnel Acoustic Lens for Synergistic Ultrasound Stimulation of Multiple Brain Regions
Previous Article in Journal
Measuring the Impact of Limb Asymmetry on Movement Irregularity and Complexity Changes During an Incremental Step Test in Para-Swimmers Using Inertial Measurement Units
Previous Article in Special Issue
Recent Advancement in Fluorescent Probes for Peroxynitrite (ONOO)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Endogenous γ-Secretase Is Linked to Phagocytic Activity in Microglial Cells

MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, 114, 16th Street, Charlestown, MA 02129, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Sensors 2025, 25(11), 3298; https://doi.org/10.3390/s25113298 (registering DOI)
Submission received: 6 March 2025 / Revised: 21 May 2025 / Accepted: 22 May 2025 / Published: 24 May 2025
(This article belongs to the Special Issue Fluorescence Sensors for Biological and Medical Applications)

Abstract

:
γ-Secretase has primarily been studied in neurons, whereas increasing evidence highlights its importance in microglia. Previous research has shown that the pharmacological inhibition of γ-secretase impairs microglial phagocytic activity. In this study, we used a genetically encoded Förster resonance energy transfer (FRET)-based biosensor to record γ-secretase activity, aiming to determine if naturally occurring cell-by-cell variations in endogenous γ-secretase activity are associated with phagocytic activity. Using the Notch1 N100 Y-T biosensor, we found that the regulation of endogenous γ-secretase activity varies among individual BV-2 microglial cells. Our multiplexed time-lapse imaging revealed that the phagocytosis of E. coli bioparticles was impaired in cells with lower γ-secretase activity compared to those with higher activity. Complementary biochemical analysis, utilizing Zymosan bioparticles and fluorescence-activated cell sorting (FACS), further demonstrated that cells with reduced phagocytic activity exhibited decreased endogenous γ-secretase activity. Collectively, our confirmatory study supports previous findings that microglial phagocytic activity is closely linked to γ-secretase and emphasizes the essential role of γ-secretase in microglia.

1. Introduction

Microglia are the resident macrophages of the central nervous system (CNS), and their normal function, activation, and dysfunction are tightly associated with development, aging, and brain diseases [1,2,3]. Microglia secrete cytokines and neurotropic factors that play pivotal roles in many aspects of immune responses in the CNS. Furthermore, microglia are responsible for phagocytosis, which is crucial for the removal of microbes, dead cells, and protein aggregates that could be harmful to neurons and their functions.
γ-Secretase is an enzyme complex responsible for the proteolytic processing of type I transmembrane proteins. The significance of γ-secretase is well established in neurons, particularly in the context of Notch signaling [4,5,6] and the processing of the amyloid precursor protein (APP) [7,8,9]. However, the role of γ-secretase in other cell types within the CNS remains unclear. Several studies have highlighted the involvement of γ-secretase in microglial migration and cytokine release (reviewed in [10]), while a more recent study has identified a pivotal role for γ-secretase in transcriptional regulation within microglia [11]. As described above, one of the essential functions of microglia is to phagocytose materials such as dead cells, debris, pathogens, and protein aggregates [1,2,3]. Notably, it has been reported that phagocytosis is impaired by the treatment of γ-secretase inhibitor [12], suggesting that γ-secretase is associated with phagocytic activity in microglia.
Several γ-secretase activity assays, such as the cell-free in vitro activity assay [13] and the cell-based reporter assay [14], have been previously developed; however, shortcomings in these assays do not permit an investigation of the dynamics of γ-secretase activity over time in individual cells. To better understand how endogenous γ-secretase activity is spatially and temporally regulated in live/intact cells, we recently developed Förster resonance energy transfer (FRET)-based biosensors for recording endogenous γ-secretase activity [15,16]. These biosensors have enabled us to detect that some cells exhibit higher while the others display lower γ-secretase activity, highlighting cell-by-cell heterogeneity in the regulation of endogenous γ-secretase activity. This unique capability of our biosensors has provided a new opportunity to ensure the correlation between γ-secretase and microglial phagocytic activity in more physiological settings. In this study, we aimed to address whether naturally occurring differences in endogenous γ-secretase activity define microglial phagocytic activities in individual cells.
Here, we record endogenous γ-secretase activity in BV-2 microglial cells using the Notch1 N100 YPet-mTurquoise-GL (N100 Y-T) biosensor [15]. Multiplexed time-lapse live-cell imaging revealed that phagocytic activity is impaired in the cells where endogenous γ-secretase activity is downregulated. Furthermore, we validated this finding using a complementary FACS and Western blot analysis. This new report confirms the previously identified link between γ-secretase and microglia phagocytosis and provides insights into how changes in endogenous γ-secretase activity regulate essential biological event(s) in various cell types.

2. Materials and Methods

2.1. Plasmid DNA, Antibodies, and Reagents

The plasmid encoding the Notch1 N100 Y-T biosensor was developed in a previous study [15]. An anti-APP C-terminus antibody was purchased from BioLegend (San Diego, CA, USA), anti-β-actin was from MilliporeSigma (Burlington, MA, USA), anti-cleaved Notch1 (Val1744) and anti-GAPDH antibodies were from Cell Signaling Technology, Inc (Dover, MA, USA), and anti-HA antibody was from Abcam (Cambridge, UK). γ-Secretase inhibitors DAPT and L-685,458 were purchased from Abcam and DMSO was from MilliporeSigma. pHrodo™ Red Escherichia coli (E. coli) BioParticles™, pHrodo™ Green Zymosan BioParticles™, and LysoTracker™ Deep Red were from Thermo Fisher Scientific (Waltham, MA, USA).

2.2. Cell Culture and Transfection

BV-2 cells were cultured in Opti-MEM Reduced Serum Medium (Thermo Fisher Scientific) with 5% FBS (Atlanta Biologicals Inc., Flowery Branch, GA, USA). The cells were authenticated using STR profiling and monitored for mycoplasma contamination. Lipofectamine 3000 (Thermo Fisher Scientific) was used for transient transfection according to the manufacturer’s instructions.

2.3. Confocal Microscopy and FRET

The Olympus FV3000RS Confocal Laser Scanning Microscope (Tokyo, Japan) was used for fluorescence imaging. The scope is equipped with a CO2/heating unit (Tokai-Hit, Fujinomiya, Japan) to maintain a suitable CO2 concentration and heating for live-cell imaging. Furthermore, the scope is equipped with the TruFocus Z drift compensation module to maintain focus during time-lapse imaging. A 10x/0.40NA objective was used for image acquisition.
For FRET detection, the cells expressing the N100 Y-T biosensor were excited by a laser at 405 nm, and the emission was simultaneously detected within 460–490 nm and 520–540 nm. A 520–540 nm over 460–490 nm emission ratio (i.e., Y/T ratio) was used as a readout of FRET. Pseudo-colored FRET images were generated using MATLAB version 8.4 (MathWorks, Natick, MA, USA). pHrodo™ Red E. coli BioParticles™ was excited by a laser at 561 nm, and emission was detected within 580–660 nm. A laser at 640 nm was used to excite LysoTracker™ DeepRed, and emission was detected within 670–770 nm. ImageJ version 1.54f was used to measure fluorescent intensity in regions of interest (ROIs).

2.4. Fluorescence-Activated Cell Sorting (FACS) and Western Blot

The Bio-Rad S3e (Hercules, CA, USA) or BD FACSMelody™ cell sorters (Franklin Lakes, NJ, USA) were used for cell sorting. The cells incubated with pHrodo™ Green Zymosan BioParticles™ were excited by a 488 nm laser, and the fluorescence emission was detected within 510–540 nm.
Proteins were extracted from cells using RIPA buffer (MilliporeSigma) with the Halt Protease Inhibitor Cocktail (Thermo Fisher Scientific). The protein concentration was determined using the BCA Protein Assay kit (Thermo Fisher Scientific). The samples were mixed with NuPAGETM LDS Sample Buffer and NuPAGETM Sample Reducing Agent (Thermo Fisher Scientific) and boiled for 3 min. Then, the samples were subjected to SDS-PAGE on NuPAGETM 4–12% Bis-Tris Protein gels (Thermo Fisher Scientific), followed by transfer to nitrocellulose membranes (Thermo Fisher Scientific) using the iBlot™ 2 Gel Transfer Device (Thermo Fisher Scientific). The membranes were incubated with primary and corresponding fluorophore-conjugated secondary antibodies and developed using the LI-COR Odyssey CLx scanner digital imaging system (LI-COR Biosciences, Lincoln, NE, USA).

2.5. Statistical Analysis

GraphPad Prism 9 (GraphPad Software, La Jolla, CA, USA) and Excel were used for the statistical analysis. The D’Agostino and Pearson omnibus normality test was used to examine the Gaussian distribution of the data and the variance equality. The one-sample t-test, unpaired t-test, and Mann–Whitney U test were used to compare the data. The Pearson correlation coefficient was measured to determine if γ-secretase activity correlates with phagocytic activity and/or lysosomal pH. In graphs, the mean and standard deviation (SD) were used as the center value and error bar, respectively. Three independent experiments were, at least, performed to ensure the reproducibility of the results.

3. Results

3.1. Recording Endogenous γ-Secretase Activity in Individual BV-2 Microglial Cells Using the N100 Y-T Biosensor

Pharmacological inhibition of γ-secretase is reported to impair phagocytic activity in microglial cells [12]. Here, we hypothesized that “naturally” altered endogenous γ-secretase activity is also associated with phagocytic activity in individual cells. To test the hypothesis, we employed the FRET-based Notch1 N100 Y-T biosensor, which enables recording endogenous γ-secretase activity on a cell-by-cell basis (Figure 1A) [15]. The N100 Y-T probe was transfected into BV-2 microglial cells, the cells were treated either with potent γ-secretase inhibitors DAPT (1 µM), L-685,458 (1 or 5 µM) or vehicle control for 16 hrs, and the cell lysates were subjected to Western blot. The Notch intracellular domain (NICD) Y-T fragment, which is produced from the N100 Y-T biosensor by γ-secretase cleavage, was significantly decreased by the treatment with γ-secretase inhibitors (Figure 1B), suggesting that endogenous γ-secretase successfully cleaves the N100 Y-T biosensor in BV-2 cells.
We then perform ratiometric spectral FRET analysis to determine if FRET between the donor and acceptor fluorophores within the N100 Y-T biosensor is changed due to γ-secretase cleavage. As such, BV-2 cells expressing the N100 Y-T biosensor were treated with 1 µM DAPT or vehicle control, the cells were excited by a laser at 405 nm, and emitted fluorescence from the donor (T: mTurquoise-GL) and acceptor (Y: YPet) was simultaneously detected using a confocal microscope. In image analysis, a region of interest (ROI) was generated over the entire cell, mTurquoise-GL and YPet fluorescence intensities were measured, and the average Y/T ratio was calculated for individual cells. We verified that the inhibition of the N100 Y-T biosensor cleavage by DAPT significantly increases FRET (Figure 1C), indicating that changes in FRET correlate with endogenous γ-secretase activity, and high FRET efficiency is associated with lower γ-secretase activity in BV-2 cells.

3.2. Phagocytosis Is Impaired in the BV-2 Cells with Lower γ-Secretase Activity

We next incubated BV-2 cells expressing the N100 Y-T biosensor with pHrodo™ Red E. coli BioParticles™ to perform multiplexed live-cell imaging analysis. These particles are pH-sensitive and thus non-fluorescent outside the cell but fluorescent bright red in phagosomes. In the imaging, we first conducted spectral FRET analysis to measure γ-secretase activity at the baseline on a cell-by-cell basis using the N100 Y-T biosensor, as described above. Then, we employed time-lapse live-cell imaging to track how long it took for individual cells to become positive for pHrodo™ Red E. coli BioParticles™ (Figure 2A). We discovered a statistically significant correlation between γ-secretase activity and the time to become E. coli BioParticles™-positive. Notably, cells exhibiting lower γ-secretase activity took longer to become E. coli BioParticles™-positive compared to those with higher activity (Figure 2B). We also divided the cells into two groups based on whether they were below or above the average mean of the Y/T FRET ratio at the baseline (t = 0), representing cell groups with higher or lower γ-secretase activity, respectively. We found that the cells with lower endogenous γ-secretase activity took a significantly longer time to become E. coli BioParticles™-positive compared to those with higher activity (Figure 2C).
To eliminate the possibility that lysosomal pH rather than phagocytic activity is tightly associated with γ-secretase activity in BV-2 microglial cells, we incubated the cells expressing the N100 Y-T biosensor with LysoTracker™ Deep Red that records lysosomal pH. Unlike the pHrodo™ Red E. coli BioParticles™ (Figure 2), we found no correlation between LysoTracker™ Deep Red fluorescence and the Y/T FRET ratio representing γ-secretase activity (Figure 3A). When we divided the cells into groups based on their Y/T FRET ratio, LysoTracker™ Deep Red fluorescence showed no significant difference between the two groups (Figure 3B). Furthermore, we confirmed that LysoTracker™ Deep Red fluorescence did not differ between BV-2 cells treated with γ-secretase inhibitor DAPT or those with the vehicle control (Figure 3C).
Lastly, to verify the result from the time-lapse multiplexed imaging that utilizes E. coli BioParticles™, BV-2 cells were first incubated with pHrodo™ Green Zymosan BioParticles™ for 16 hrs (Figure 4A), and then the cells were subjected to FACS to be sorted out based on the Zymosan fluorescence (Figure 4B). The numbers of Zymosan-positive cells decreased by the treatment of cytochalasin D, a potent phagocytosis inhibitor [17,18], evidencing that the fluorescence is dependent on the phagocytosis of Zymosan BioParticles™ (Figure 4C). Then, the sorted cells with distinct phagocytic activities were subjected to Western blot using an APP C-terminus antibody to assess γ-secretase activity. Strikingly, we found that APP C-terminal fragments, immediate endogenous substrates of γ-secretase, over the APP full-length ratio are significantly increased in the cells with lower phagocytic activity compared to those with higher phagocytic activity (Figure 4D). This suggests that BV-2 cells with reduced phagocytic activity exhibit lower endogenous γ-secretase activity and further supports the link between γ-secretase and microglial phagocytosis.

4. Discussion

The great importance of γ-secretase and its pivotal roles in biology and diseases have been well established; still, how γ-secretase activity is spatial and temporally regulated remains unclear. Our recent development of genetically encoded FRET-based biosensors has allowed for endogenous γ-secretase activity to be recorded over time [15], on a cell-by-cell basis [15,16,19], with subcellular resolution [20,21], not only in vitro but also in vivo [22]. Moreover, the unique capabilities of the biosensors have enabled us to detect the cell-by-cell heterogeneity in endogenous γ-secretase activity and further determine the consequences of naturally altered γ-secretase activity in individual neurons (Yokomizo et al., 2024, in revision). In the present study, we employed the Notch1 N100 Y-T biosensor [15] to record γ-secretase activity in BV-2 microglial cells (Figure 1). Our findings indicate that phagocytic activity (Figure 2 and Figure 4), but not lysosomal pH (Figure 3), is decreased in cells exhibiting lower endogenous γ-secretase activity.
The membrane-bound enzymatic complex γ-secretase is responsible for the proteolytic cleavage of various transmembrane substrates [23]. The enzyme functions at a critical intersection between various cellular pathways such as Notch [4,5,6,24] and most notably APP proteolytic processing [7,8,9], where it facilitates the development of one of the pathological hallmarks of AD. γ-Secretase does not solely function in the APP pathway; thus, γ-secretase inhibitors alone are insufficient to slow down AD progression [25,26], despite demonstrating an ability to reduce the β-amyloid concentration in the CNS [27]. Several studies have highlighted the essential roles played by γ-secretase in microglia, including the modulation of pro-inflammatory cytokine expression and release [28,29] and the regulation of microglial migration [30].
The phagocytic clearance of cell debris, pathogens, and protein aggregates is one of the core functions of microglia [1,2,3,31]. The previous literature has connected γ-secretase to phagocytic activity using a γ-secretase inhibitor [12], establishing a clear cause-and-effect relationship. However, there is limited knowledge about whether microglial phagocytosis in individual cells relies on endogenous γ-secretase activity. Hence, we utilized our FRET-based biosensor and pHrodo™ E. coli BioParticles™ in time-lapse live-cell imaging and realized a heterogeneity of γ-secretase activity on a cell-by-cell basis, which correlated to a similar heterogeneity of microglial phagocytic activity (Figure 2). This correlation was further verified by pHrodo™ Zymosan BioParticles™ adapted to flow cytometry and cell sorting. We identified that the cells demonstrating reduced γ-secretase processing of APP-CTFs were also the cells that had a lower phagocytic efficiency of Zymosan BioParticles™ (Figure 4). Of note, we visualized a decrease in fluorescence in response to a phagocytic inhibitor cytochalasin D treatment, demonstrating the reliability of Zymosan fluorescence in capturing phagocytic activity (Figure 4). We also ensured that our measurements using pHrodo™ E. coli BioParticles™ truly represented phagocytosis rather than changes in lysosomal pH (Figure 3). Altogether, this study employed two different assays (i.e., time-lapse imaging, FACS cell sorting and Western blot) and two distinct bioparticles (i.e., pHrodo™ E. coli and Zymosan BioParticles™) to validate the correlation between phagocytosis and endogenous γ-secretase activity.
Microglial phagocytic activity is regulated by γ-secretase; however, the underlying molecular mechanisms are not fully understood. The triggering receptor expressed on myeloid cells 2 (TREM2) is a type I membrane protein exclusively expressed in microglia [32] and is tightly associated with microglial phagocytic activity [33,34,35]. TREM2 undergoes the regulated sequential intramembrane proteolysis first by α-secretase(s) and then by γ-secretase [36,37,38,39]. To initiate downstream signaling upon ligand activation, TREM2 requires a complex formation with adapter proteins (e.g., DAP12) [32]. It has been reported that the inhibition of γ-secretase activity decreases TREM2-dependent phagocytosis in BV-2 cells [40]. Mechanistically, γ-secretase inhibition results in the accumulation of TREM2 C-term fragments (TREM2 CTFs), limiting the interaction of DAP12 with the functional full-length receptor and thus decreasing DAP12 phosphorylation [36]. TREM2 may, therefore, link between diminished endogenous γ-secretase activity and microglial phagocytosis. Detailed mechanistic research is necessary to establish the exact molecular link between γ-secretase and microglia phagocytosis. Furthermore, phagocytic behavior and regulating factors in BV-2 cells may differ from those of primary microglia and/or microglia in the brain in vivo; thus, validation of the findings in more physiologically relevant models is also necessary.
Lastly, the Y/T ratio from the N100 Y-T biosensor is correlated with the time until pHrodo™ E. coli BioParticles™ positivity in a statistically significant manner (Figure 2C), and its Pearson correlation coefficient (r = 0.3276) is larger than the cut-off used in biological science [41,42]. However, the correlation is mild; therefore, the possible reasons behind the non-robust correlation would be worthwhile to be discussed. First, we think it could be because microglia phagocytic activity is regulated not only by γ-secretase but also by many other factors. Second, the non-robustness could be also related to the design of our biosensor. Whereas we are confident that our FRET biosensor can sensitively record endogenous γ-secretase activity over time on a cell-by-cell basis in live cells, which are fully validated by the series of our previous publications [15,16,19,20,21,22], the exact “purity” of the correlation between the Y/T ratio and γ-secretase activity remains unclear due to the logistics behind the design of the biosensor. The FRET phenomenon relies on orientation and proximity between the donor and acceptor fluorophores. To make our biosensor more proximity-dependent, excluding the contribution of orientation and aiming to increase sensitivity, we optimized the linker length between the donor and acceptor fluorophores from 20 amino acids to 80 amino acids [15]. Yet, our recent findings using multiple different cell types in vitro and in vivo suggest that orientation still plays a role [22]. These findings are particularly important since our FRET biosensor, the N100 Y-T biosensor in this study, uses the SAGG repeat flexible linker, which randomizes the orientation between the donor and acceptor of the sensor as well as that of the biosensor cleavage product. Therefore, we expect that this randomness may create the “dirtiness” in the Y/T ratio, contributing to the appearance of a less striking correlation between the Y/T ratio and the time until pHrodo™ E. coli BioParticles™ positivity.

5. Conclusions

This study verifies a previously discovered link between γ-secretase and phagocytosis in BV-2 microglial cells. Using the N100 Y-T biosensor, we demonstrate that 1) endogenous γ-secretase activity is differently regulated in individual BV-2 cells and, while the correlation is not strong, 2) phagocytic activity is reduced in the cells with diminished endogenous γ-secretase activity. This finding is consistent with earlier research that reported impaired phagocytosis following γ-secretase inhibitor treatment [12]. This study also sheds light on the capability of genetically encoded biosensors to record endogenous γ-secretase activity with single-cell resolution, facilitating a deeper understanding of the spatiotemporal regulation of γ-secretase and its consequences in various cell types beyond neurons.

Author Contributions

Conceptualization, M.M.; methodology, M.M.; validation, E.W., M.C.Q.H., and S.T.; formal analysis, E.W., M.C.Q.H., and S.T.; investigation, E.W., M.C.Q.H., and S.T.; resources, M.S., N.W., and M.Y.; data curation, E.W., M.C.Q.H., and S.T.; writing—original draft preparation, E.W.; writing—review and editing, M.M.; visualization, E.W., M.C.Q.H., and M.M.; supervision, M.M.; project administration, M.M.; funding acquisition, M.M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the National Institute of Health grants AG079838 (M.M.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data are visualized and included in the manuscript. The raw data used to create the corresponding graphs can be shared by the corresponding author upon reasonable requests.

Acknowledgments

We are grateful to Florian Perrin, Priyanka Sinha, and Oksana Berezovska (MGH Neurology) for constructive discussions. We also appreciate Lori Chibnik (MGH Neurology and Harvard T.H. Chan School of Public Health) for her advice on the statistical analysis.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
APP amyloid precursor protein
FACS fluorescence-activated cell sorting
FRET Förster resonance energy transfer
Y-T YPet-mTurquoise-GL

References

  1. Li, Q.; Barres, B.A. Microglia and macrophages in brain homeostasis and disease. Nat. Rev. Immunol. 2018, 18, 225–242. [Google Scholar] [CrossRef] [PubMed]
  2. Prinz, M.; Jung, S.; Priller, J. Microglia Biology: One Century of Evolving Concepts. Cell 2019, 179, 292–311. [Google Scholar] [CrossRef] [PubMed]
  3. Pallarés-Moratalla, C.; Bergers, G. The ins and outs of microglial cells in brain health and disease. Front. Immunol. 2024, 15, 1305087. [Google Scholar] [CrossRef]
  4. De Strooper, B.; Annaert, W.; Cupers, P.; Saftig, P.; Craessaerts, K.; Mumm, J.S.; Schroeter, E.H.; Schrijvers, V.; Wolfe, M.S.; Ray, W.J.; et al. A presenilin-1-dependent gamma-secretase-like protease mediates release of Notch intracellular domain. Nature 1999, 398, 518–522. [Google Scholar] [CrossRef]
  5. Costa, R.M.; Honjo, T.; Silva, A.J. Learning and memory deficits in Notch mutant mice. Curr. Biol. 2003, 13, 1348–1354. [Google Scholar] [CrossRef]
  6. Alberi, L.; Liu, S.; Wang, Y.; Badie, R.; Smith-Hicks, C.; Wu, J.; Pierfelice, T.J.; Abazyan, B.; Mattson, M.P.; Kuhl, D.; et al. Activity-induced Notch signaling in neurons requires Arc/Arg3.1 and is essential for synaptic plasticity in hippocampal networks. Neuron 2011, 69, 437–444. [Google Scholar] [CrossRef]
  7. De Strooper, B.; Saftig, P.; Craessaerts, K.; Vanderstichele, H.; Guhde, G.; Annaert, W.; Von Figura, K.; Van Leuven, F. Deficiency of presenilin-1 inhibits the normal cleavage of amyloid precursor protein. Nature 1998, 391, 387–390. [Google Scholar] [CrossRef]
  8. Wolfe, M.S.; Xia, W.; Ostaszewski, B.L.; Diehl, T.S.; Kimberly, W.T.; Selkoe, D.J. Two transmembrane aspartates in presenilin-1 required for presenilin endoproteolysis and gamma-secretase activity. Nature 1999, 398, 513–517. [Google Scholar] [CrossRef]
  9. Saura, C.A.; Chen, G.; Malkani, S.; Choi, S.Y.; Takahashi, R.H.; Zhang, D.; Gouras, G.K.; Kirkwood, A.; Morris, R.G.; Shen, J. 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. 2005, 25, 6755–6764. [Google Scholar] [CrossRef]
  10. Walter, J.; Kemmerling, N.; Wunderlich, P.; Glebov, K. γ-Secretase in microglia–implications for neurodegeneration and neuroinflammation. J. Neurochem. 2017, 143, 445–454. [Google Scholar] [CrossRef]
  11. Hou, P.; Zielonka, M.; Serneels, L.; Martinez-Muriana, A.; Fattorelli, N.; Wolfs, L.; Poovathingal, S.; T’Syen, D.; Balusu, S.; Theys, T.; et al. The γ-secretase substrate proteome and its role in cell signaling regulation. Mol. Cell 2023, 83, 4106–4122.e10. [Google Scholar] [CrossRef] [PubMed]
  12. Farfara, D.; Trudler, D.; Segev-Amzaleg, N.; Galron, R.; Stein, R.; Frenkel, D. γ-Secretase component presenilin is important for microglia β-amyloid clearance. Ann. Neurol. 2011, 69, 170–180. [Google Scholar] [CrossRef]
  13. Kakuda, N.; Funamoto, S.; Yagishita, S.; Takami, M.; Osawa, S.; Dohmae, N.; Ihara, Y. Equimolar production of amyloid beta-protein and amyloid precursor protein intracellular domain from beta-carboxyl-terminal fragment by gamma-secretase. J. Biol. Chem. 2006, 281, 14776–14786. [Google Scholar] [CrossRef]
  14. Cao, X.; Südhof, T.C. A transcriptionally [correction of transcriptively] active complex of APP with Fe65 and histone acetyltransferase Tip60. Science 2001, 293, 115–120. [Google Scholar] [CrossRef]
  15. Maesako, M.; Sekula, N.M.; Aristarkhova, A.; Feschenko, P.; Anderson, L.C.; Berezovska, O. Visualization of PS/γ-Secretase Activity in Living Cells. iScience 2020, 23, 101139. [Google Scholar] [CrossRef]
  16. Houser, M.C.Q.; Hou, S.S.; Perrin, F.; Turchyna, Y.; Bacskai, B.J.; Berezovska, O.; Maesako, M. A Novel NIR-FRET Biosensor for Reporting PS/γ-Secretase Activity in Live Cells. Sensors 2020, 20, 5980. [Google Scholar] [CrossRef]
  17. Axline, S.G.; Reaven, E.P. Inhibition of phagocytosis and plasma membrane mobility of the cultivated macrophage by cytochalasin B. Role of subplasmalemmal microfilaments. J. Cell Biol. 1974, 62, 647–659. [Google Scholar] [CrossRef]
  18. Goddette, D.W.; Frieden, C. Actin polymerization. The mechanism of action of cytochalasin D. J. Biol. Chem. 1986, 261, 15974–15980. [Google Scholar] [CrossRef]
  19. Houser, M.C.Q.; Turchyna, Y.; Perrin, F.; Chibnik, L.; Berezovska, O.; Maesako, M. Limited Substrate Specificity of PS/γ-Secretase Is Supported by Novel Multiplexed FRET Analysis in Live Cells. Biosensors 2021, 11, 169. [Google Scholar] [CrossRef]
  20. Maesako, M.; Houser, M.C.Q.; Turchyna, Y.; Wolfe, M.S.; Berezovska, O. Presenilin/γ-Secretase Activity Is Located in Acidic Compartments of Live Neurons. J. Neurosci. 2022, 42, 145–154. [Google Scholar] [CrossRef]
  21. McKendell, A.K.; Houser, M.C.Q.; Mitchell, S.P.C.; Wolfe, M.S.; Berezovska, O.; Maesako, M. In-Depth Characterization of Endo-Lysosomal Aβ in Intact Neurons. Biosensors 2022, 12, 663. [Google Scholar] [CrossRef] [PubMed]
  22. Hou, S.S.; Ikegawa, Y.; Kwon, Y.; Wieckiewicz, N.; Houser, M.C.Q.; Lundin, B.; Bacskai, B.J.; Berezovska, O.; Maesako, M. Recording γ-secretase activity in living mouse brains. Elife 2024, 13, RP96848. [Google Scholar] [CrossRef]
  23. Güner, G.; Lichtenthaler, S.F. The substrate repertoire of γ-secretase/presenilin. Semin. Cell Dev. Biol. 2020, 105, 27–42. [Google Scholar] [CrossRef]
  24. Wong, P.C.; Zheng, H.; Chen, H.; Becher, M.W.; Sirinathsinghji, D.J.; Trumbauer, M.E.; Chen, H.Y.; Price, D.L.; Van der Ploeg, L.H.; Sisodia, S.S. Presenilin 1 is required for Notch1 and DII1 expression in the paraxial mesoderm. Nature 1997, 387, 288–292. [Google Scholar] [CrossRef]
  25. Coric, V.; van Dyck, C.H.; Salloway, S.; Andreasen, N.; Brody, M.; Richter, R.W.; Soininen, H.; Thein, S.; Shiovitz, T.; Pilcher, G.; et al. Safety and tolerability of the γ-secretase inhibitor avagacestat in a phase 2 study of mild to moderate Alzheimer disease. Arch. Neurol. 2012, 69, 1430–1440. [Google Scholar] [CrossRef]
  26. Doody, R.S.; Raman, R.; Farlow, M.; Iwatsubo, T.; Vellas, B.; Joffe, S.; Kieburtz, K.; He, F.; Sun, X.; Thomas, R.G.; et al. A phase 3 trial of semagacestat for treatment of Alzheimer’s disease. N. Engl. J. Med. 2013, 369, 341–350. [Google Scholar] [CrossRef]
  27. Bateman, R.J.; Siemers, E.R.; Mawuenyega, K.G.; Wen, G.; Browning, K.R.; Sigurdson, W.C.; Yarasheski, K.E.; Friedrich, S.W.; Demattos, R.B.; May, P.C.; et al. A gamma-secretase inhibitor decreases amyloid-beta production in the central nervous system. Ann. Neurol. 2009, 66, 48–54. [Google Scholar] [CrossRef]
  28. Cao, Q.; Li, P.; Lu, J.; Dheen, S.T.; Kaur, C.; Ling, E.A. Nuclear factor-κB/p65 responds to changes in the Notch signaling pathway in murine BV-2 cells and in amoeboid microglia in postnatal rats treated with the γ-secretase complex blocker DAPT. J. Neurosci. Res. 2010, 88, 2701–2714. [Google Scholar] [CrossRef]
  29. Jayadev, S.; Case, A.; Eastman, A.J.; Nguyen, H.; Pollak, J.; Wiley, J.C.; Möller, T.; Morrison, R.S.; Garden, G.A. Presenilin 2 is the predominant γ-secretase in microglia and modulates cytokine release. PLoS ONE 2010, 5, e15743. [Google Scholar] [CrossRef]
  30. Kemmerling, N.; Wunderlich, P.; Theil, S.; Linnartz-Gerlach, B.; Hersch, N.; Hoffmann, B.; Heneka, M.T.; de Strooper, B.; Neumann, H.; Walter, J. Intramembranous processing by γ-secretase regulates reverse signaling of ephrin-B2 in migration of microglia. Glia 2017, 65, 1103–1118. [Google Scholar] [CrossRef]
  31. Sierra, A.; Abiega, O.; Shahraz, A.; Neumann, H. Janus-faced microglia: Beneficial and detrimental consequences of microglial phagocytosis. Front. Cell. Neurosci. 2013, 7, 6. [Google Scholar] [CrossRef] [PubMed]
  32. Colonna, M. TREMs in the immune system and beyond. Nat. Rev. Immunol. 2003, 3, 445–453. [Google Scholar] [CrossRef]
  33. Kawabori, M.; Kacimi, R.; Kauppinen, T.; Calosing, C.; Kim, J.Y.; Hsieh, C.L.; Nakamura, M.C.; Yenari, M.A. Triggering receptor expressed on myeloid cells 2 (TREM2) deficiency attenuates phagocytic activities of microglia and exacerbates ischemic damage in experimental stroke. J. Neurosci. 2015, 35, 3384–3396. [Google Scholar] [CrossRef]
  34. Xiang, X.; Werner, G.; Bohrmann, B.; Liesz, A.; Mazaheri, F.; Capell, A.; Feederle, R.; Knuesel, I.; Kleinberger, G.; Haass, C. TREM2 deficiency reduces the efficacy of immunotherapeutic amyloid clearance. EMBO Mol. Med. 2016, 8, 992–1004. [Google Scholar] [CrossRef]
  35. Jay, T.R.; Miller, C.M.; Cheng, P.J.; Graham, L.C.; Bemiller, S.; Broihier, M.L.; Xu, G.; Margevicius, D.; Karlo, J.C.; Sousa, G.L.; et al. TREM2 deficiency eliminates TREM2+ inflammatory macrophages and ameliorates pathology in Alzheimer’s disease mouse models. J. Exp. Med. 2015, 212, 287–295. [Google Scholar] [CrossRef]
  36. Wunderlich, P.; Glebov, K.; Kemmerling, N.; Tien, N.T.; Neumann, H.; Walter, J. Sequential proteolytic processing of the triggering receptor expressed on myeloid cells-2 (TREM2) protein by ectodomain shedding and γ-secretase-dependent intramembranous cleavage. J. Biol. Chem. 2013, 288, 33027–33036. [Google Scholar] [CrossRef]
  37. Schlepckow, K.; Kleinberger, G.; Fukumori, A.; Feederle, R.; Lichtenthaler, S.F.; Steiner, H.; Haass, C. An Alzheimer-associated TREM2 variant occurs at the ADAM cleavage site and affects shedding and phagocytic function. EMBO Mol. Med. 2017, 9, 1356–1365. [Google Scholar] [CrossRef]
  38. Thornton, P.; Sevalle, J.; Deery, M.J.; Fraser, G.; Zhou, Y.; Ståhl, S.; Franssen, E.H.; Dodd, R.B.; Qamar, S.; Gomez Perez-Nievas, B.; et al. TREM2 shedding by cleavage at the H157-S158 bond is accelerated for the Alzheimer’s disease-associated H157Y variant. EMBO Mol. Med. 2017, 9, 1366–1378. [Google Scholar] [CrossRef]
  39. Steiner, A.; Schlepckow, K.; Brunner, B.; Steiner, H.; Haass, C.; Hagn, F. γ-Secretase cleavage of the Alzheimer risk factor TREM2 is determined by its intrinsic structural dynamics. EMBO J. 2020, 39, e104247. [Google Scholar] [CrossRef]
  40. Glebov, K.; Wunderlich, P.; Karaca, I.; Walter, J. Functional involvement of γ-secretase in signaling of the triggering receptor expressed on myeloid cells-2 (TREM2). J. Neuroinflamm. 2016, 13, 17. [Google Scholar] [CrossRef]
  41. Miot, H.A. Correlation analysis in clinical and experimental studies. J. Vasc. Bras. 2018, 17, 275–279. [Google Scholar] [CrossRef]
  42. Rusakov, D.A. A misadventure of the correlation coefficient. Trends Neurosci. 2023, 46, 94–96. [Google Scholar] [CrossRef]
Figure 1. The N100 Y-T biosensor reports γ-secretase activity in BV-2 microglial cells: (A) schematic presentation of the N100 Y-T biosensor and its cleavage by γ-secretase. (B) Western blotting using HA and cleaved Notch1 antibodies. HA antibody captures both full-length N100 Y-T and γ-secretase cleavage product: Notch intracellular domain (NICD) Y-T. In contrast, the cleaved Notch1 antibody only recognizes NICD Y-T, showing its reduction by γ-secretase inhibitors: DAPT (1 µM), L-685,458 (1 or 5 µM) for 16 h of treatment. (C) Spectral FRET analysis shows that DAPT (1 µM) treatment significantly increases YPet/mTurquoise-GL emission ratio (Y/T ratio). Scale bar 20 µm, N = 60 cells, Mann–Whitney U test, **** p < 0.0001.
Figure 1. The N100 Y-T biosensor reports γ-secretase activity in BV-2 microglial cells: (A) schematic presentation of the N100 Y-T biosensor and its cleavage by γ-secretase. (B) Western blotting using HA and cleaved Notch1 antibodies. HA antibody captures both full-length N100 Y-T and γ-secretase cleavage product: Notch intracellular domain (NICD) Y-T. In contrast, the cleaved Notch1 antibody only recognizes NICD Y-T, showing its reduction by γ-secretase inhibitors: DAPT (1 µM), L-685,458 (1 or 5 µM) for 16 h of treatment. (C) Spectral FRET analysis shows that DAPT (1 µM) treatment significantly increases YPet/mTurquoise-GL emission ratio (Y/T ratio). Scale bar 20 µm, N = 60 cells, Mann–Whitney U test, **** p < 0.0001.
Sensors 25 03298 g001
Figure 2. Time-lapse live-cell imaging reveals that BV-2 cells with lower γ-secretase activity display decreased phagocytic activity: (A) BV-2 cells expressing the N100 Y-T biosensor were incubated with pHrodo™ Red E. coli BioParticles™. γ-Secretase activity at the baseline was calculated, and the time until becoming positive with E. coli BioParticles™ was measured on a cell-by-cell basis. Scale bar 100 µm. (B) Positive correlation between baseline Y/T ratio (a higher ratio means lower γ-secretase activity) and the time until becoming E. coli BioParticles™-positive. N = 58 cells, Pearson correlation coefficient, * p < 0.05. R2 = 0.1073, RSME = 0.8619. MAE = 0.0002. (C) Significantly longer time until becoming pHrodo™ Red E. coli BioParticles™ in the BV-2 cells with lower γ-secretase (i.e., Y/T ratio is higher than the average mean) compared to those with higher activity. N = 28–30 cells, Mann–Whitney U test, * p < 0.05.
Figure 2. Time-lapse live-cell imaging reveals that BV-2 cells with lower γ-secretase activity display decreased phagocytic activity: (A) BV-2 cells expressing the N100 Y-T biosensor were incubated with pHrodo™ Red E. coli BioParticles™. γ-Secretase activity at the baseline was calculated, and the time until becoming positive with E. coli BioParticles™ was measured on a cell-by-cell basis. Scale bar 100 µm. (B) Positive correlation between baseline Y/T ratio (a higher ratio means lower γ-secretase activity) and the time until becoming E. coli BioParticles™-positive. N = 58 cells, Pearson correlation coefficient, * p < 0.05. R2 = 0.1073, RSME = 0.8619. MAE = 0.0002. (C) Significantly longer time until becoming pHrodo™ Red E. coli BioParticles™ in the BV-2 cells with lower γ-secretase (i.e., Y/T ratio is higher than the average mean) compared to those with higher activity. N = 28–30 cells, Mann–Whitney U test, * p < 0.05.
Sensors 25 03298 g002
Figure 3. Not distinct lysosomal pH between the BV-2 cells with lower vs. higher γ-secretase activity: (A) BV-2 cells expressing the N100 Y-T biosensor were incubated with LysoTrackerTM DeepRed. γ-Secretase activity at the baseline was calculated and LysoTrackerTM DeepRed fluorescence was measured on a cell-by-cell basis, showing that there is no significant correlation. N = 289 cells. p = 0.146. Pearson correlation coefficient. (B) No significant difference in LysoTrackerTM DeepRed intensity between the BV-2 cells with lower γ-secretase (i.e., Y/T ratio is higher than the average mean) and those with higher activity (p = 0.07). N = 130–159 cells, Mann–Whitney U test. n.s. means not significant. (C) LysoTrackerTM DeepRed intensity is not significantly different between BV-2 cells treated with DAPT (1 µM) or vehicle control for 16 hrs (p = 0.91). N = 143–289 cells, Mann–Whitney U test. n.s.: not significant.
Figure 3. Not distinct lysosomal pH between the BV-2 cells with lower vs. higher γ-secretase activity: (A) BV-2 cells expressing the N100 Y-T biosensor were incubated with LysoTrackerTM DeepRed. γ-Secretase activity at the baseline was calculated and LysoTrackerTM DeepRed fluorescence was measured on a cell-by-cell basis, showing that there is no significant correlation. N = 289 cells. p = 0.146. Pearson correlation coefficient. (B) No significant difference in LysoTrackerTM DeepRed intensity between the BV-2 cells with lower γ-secretase (i.e., Y/T ratio is higher than the average mean) and those with higher activity (p = 0.07). N = 130–159 cells, Mann–Whitney U test. n.s. means not significant. (C) LysoTrackerTM DeepRed intensity is not significantly different between BV-2 cells treated with DAPT (1 µM) or vehicle control for 16 hrs (p = 0.91). N = 143–289 cells, Mann–Whitney U test. n.s.: not significant.
Sensors 25 03298 g003
Figure 4. FACS and Western blot verify the link between γ-secretase and phagocytosis: (A) BV-2 cells phagocytosed pHrodo™ Green Zymosan BioParticles™, (B) which were subjected to FACS to isolate and extract the cells with higher and lower Zymosan BioParticles™ fluorescence. (C) The number of cells exhibiting Zymosan BioParticles™ positivity decreased by 100 nM cytochalasin D, a phagocytosis inhibitor, treatment. (D) The cell lysates from BV-2 cells with higher or lower Zymosan BioParticles™ fluorescence were subjected to Western blot using an APP C-terminus antibody. Increased APP-C-terminal fragments (CTFs), endogenous substrates of γ-secretase, over APP full-length ratio in the cells with lower phagocytic activity (1.497 ± 0.52 vs. high phagocytic activity set as 1) suggest decreased γ-secretase activity in the cell populations. N = 3 independent experiments. * p < 0.05, one-sample t-test.
Figure 4. FACS and Western blot verify the link between γ-secretase and phagocytosis: (A) BV-2 cells phagocytosed pHrodo™ Green Zymosan BioParticles™, (B) which were subjected to FACS to isolate and extract the cells with higher and lower Zymosan BioParticles™ fluorescence. (C) The number of cells exhibiting Zymosan BioParticles™ positivity decreased by 100 nM cytochalasin D, a phagocytosis inhibitor, treatment. (D) The cell lysates from BV-2 cells with higher or lower Zymosan BioParticles™ fluorescence were subjected to Western blot using an APP C-terminus antibody. Increased APP-C-terminal fragments (CTFs), endogenous substrates of γ-secretase, over APP full-length ratio in the cells with lower phagocytic activity (1.497 ± 0.52 vs. high phagocytic activity set as 1) suggest decreased γ-secretase activity in the cell populations. N = 3 independent experiments. * p < 0.05, one-sample t-test.
Sensors 25 03298 g004
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.

Share and Cite

MDPI and ACS Style

Williams, E.; Houser, M.C.Q.; Torres, S.; Wieckiewicz, N.; Sadek, M.; Yokomizo, M.; Maesako, M. Endogenous γ-Secretase Is Linked to Phagocytic Activity in Microglial Cells. Sensors 2025, 25, 3298. https://doi.org/10.3390/s25113298

AMA Style

Williams E, Houser MCQ, Torres S, Wieckiewicz N, Sadek M, Yokomizo M, Maesako M. Endogenous γ-Secretase Is Linked to Phagocytic Activity in Microglial Cells. Sensors. 2025; 25(11):3298. https://doi.org/10.3390/s25113298

Chicago/Turabian Style

Williams, Emily, Mei C. Q. Houser, Sebastian Torres, Natalia Wieckiewicz, Michael Sadek, Midori Yokomizo, and Masato Maesako. 2025. "Endogenous γ-Secretase Is Linked to Phagocytic Activity in Microglial Cells" Sensors 25, no. 11: 3298. https://doi.org/10.3390/s25113298

APA Style

Williams, E., Houser, M. C. Q., Torres, S., Wieckiewicz, N., Sadek, M., Yokomizo, M., & Maesako, M. (2025). Endogenous γ-Secretase Is Linked to Phagocytic Activity in Microglial Cells. Sensors, 25(11), 3298. https://doi.org/10.3390/s25113298

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop