4.1 A bioinformatics workflow identified that TGFBR3 plays a crucial role in AD pathology
To systematically identify the molecular mechanisms underlying AD, a bioinformatic workflow was established for the discovery of crucial regulatory targets in AD (Fig. 1). After assessing the data quality, we analyzed the hallmarks for the RNA-seq cohort (ID: syn3157743) using GSEA (Supplementary Fig. 1). The results exhibited a significant enrichment of neuroinflammation-related hallmarks, including TNF-α signaling via NF-κB, INF-α response, TGF-β signaling, and IL6-JAK-STAT3 signaling (Fig. 2A).
To demonstrate the contribution of neuroinflammation to AD, we obtained 794 differentially expressed genes from the cohort. Then, several methods, including WGCNA, PPI, and biological process analysis, were combined to identify a group of crucial genes that connect neuroinflammation with typical lesions (CDR, Braak staging, and Aβ plaque). In WGCNA, the MEgray module was positively correlated with all three typical lesions of AD, and 379 genes associated with this module were collected (P < 0.01) using Pearson's correlation
analysis. (Fig. 2B and 2C). In PPI analysis, a total of 180 shared genes were collected from the top 200 differential genes calculated by CytoHubba and CentiScaPe 2.2 (Fig. 2D). In biological process analysis, neuroinflammation-related processes were identified, and 62 hit genes were integrated from these processes (Fig. 2E). Finally, we identified 18 shared genes from the three methods that connected neuroinflammation and typical lesions, which together accelerated the progression of AD (Fig. 2F).
We next ranked the 18 shared genes using the CFG method and identified 6 genes (TGFBR3, VIM, AGT, CD44, PLCE1, FN1) with a CFG score ≥ 3. (Table 2). The expression of 6 genes was then validated using data from multiple platforms, which revealed that only TGFBR3 was elevated across several brain regions (Supplementary Fig. 1). Notably, the KEGG enrichment analysis ranked TGF-β signaling as the top pathway (Fig. 2E), suggesting that TGFBR3 may be implicated in neuroinflammation-related AD lesions by mediating TGF-β signaling. Then, we conducted further investigation and discovered that TGFBR3 exhibited a positive correlation with CDR, plaque, and Braak staging (Fig. 2G-2I). Intriguingly, we also observed that TGFBR3 is highly expressed in microglia using single-cell sequencing analysis, indicating that microglia are implicated in the aforementioned process (Supplementary Fig. 2).
Table 2 The CFG rank of candidate genes in the AlzData online server
GENE
|
eQTLa
|
GWAS
|
PPIb
|
Early_DEGc
|
Pathology cor (Aβ)d
|
Pathology cor (tau) e
|
CFGf
|
TGFBR3
|
1
|
25
|
|
yes
|
0.527, ***
|
0.399, ns
|
4
|
VIM
|
3
|
0
|
APP, MAPT
|
yes
|
0.918, ***
|
0.700, **
|
4
|
AGT
|
1
|
0
|
APP, PSEN1, APOE
|
yes
|
-0.359, *
|
0.002, ns
|
4
|
CD44
|
2
|
0
|
APP, PSEN1
|
|
0.719, ***
|
0.793, ***
|
3
|
PLCE1
|
1
|
0
|
|
yes
|
0.683, ***
|
0.562, *
|
3
|
FN1
|
1
|
2
|
APP, MAPT, APOE
|
|
|
|
3
|
NPPA
|
4
|
0
|
APP, APOE
|
|
-0.006, ns
|
0.105, ns
|
2
|
HFE
|
1
|
0
|
|
|
0.786, ***
|
0.852, ***
|
2
|
ARHGEF6
|
0
|
NA
|
APOE
|
|
0.471, **
|
0.611, *
|
2
|
CD4
|
1
|
0
|
|
|
0.063, ns
|
-0.606, *
|
2
|
S100B
|
|
0
|
MAPT
|
|
0.406, **
|
0.313, ns
|
2
|
LTF
|
3
|
0
|
APP, APOE
|
|
-0.148, ns
|
0.459, ns
|
2
|
WLS
|
2
|
1
|
|
|
|
|
2
|
TFR2
|
1
|
0
|
|
|
|
|
1
|
PTPN3
|
1
|
0
|
|
|
-0.064, ns
|
-0.044, ns
|
1
|
IFIH1
|
|
0
|
|
|
|
|
0
|
TNFAIP3
|
0
|
0
|
|
|
0.025, ns
|
0.129, ns
|
0
|
aeQTL: expression of target gene is regulated by AD genetic variants
bPPI: target gene has significant physical interaction with APP, PSEN1, PSEN2, APOE or MAPT
cEarly_DEG: target gene is differentially expressed in AD mouse models before AD pathology emergence
dPathology cor (Aβ): correlation of target gene expression with AD pathology in Aβ line AD mouse models
ePathology cor (tau): correlation of target gene expression with AD pathology in tau line AD mouse models
fCFG: total CFG score of the target gene, 1 CFG point is added if any of the above evidence is significant, and the CFG point ranges from 0 to 5.
*P < 0.05, **P < 0.01, ***P< 0.001.
4.2 Increased sTGFBR3 levels are associated with AD
To further investigate the relationship between TGFBR3 expression and AD, we first stained brain tissue from wild-type mice of different ages using a monoclonal anti-mouse TGFBR3 antibody. This antibody was raised against amino acids 511-790 mapping near the C-terminus of the TGFBR3 extracellular domain. Staining with this TGFBR3 antibody was significantly increased with age (Fig. 3A and 3B). However, immunoblotting showed that only sTGFBR3 was increased with age (Fig. 3C and 3D). In addition, MT1-MMP and TGF-β1 were also increased with age, but P-Smad2/3 was decreased, indicating the abnormal accumulation of TGF-β1 and the blockade of TGF-β/Smad signaling (Fig. 3C, 3E-3F, Supplementary Fig. 4A-4B). We also found that sTGFBR3 and MT1-MMP had the same increasing trend in APP/PS1 mice expressing a chimeric mouse/human amyloid precursor protein (Mo/HuAPP695swe) and a mutant human presenilin 1 (PS1-dE9) by immunoblotting analysis (Fig. 3G-3I), indicating that the production of sTGFBR3 was sheared by MT1-MMP. Interestingly, the expression of full-length TGFBR3 (kDa: 180) was also increased in two samples (Fig. 3G). However, the biological function of sTGFBR3 still cannot be underestimated. Furthermore, we performed costaining for sTGFBR3 and Iba1 in 6-month-old APP/PS1 mice, suggesting that the elevated sTGFBR3 was almost entirely derived from microglia (Fig. 3M-3 N).
We then further examined postmortem temporal brain tissue from 3 patients with AD by RT‒qPCR, immunoblotting and immunostaining. First, RT‒qPCR and immunoblotting showed that TGFBR3 was significantly elevated in the temporal lobe compared with normal controls, but mainly sTGFBR3 was upregulated (Fig. 3J-3 L). Likewise, abnormal accumulation of TGF-β1 also existed in the brains of AD patients (Fig. 3J). Next, we performed costaining for human sTGFBR3 and Aβ, and the results suggested that sTGFBR3 was spatially correlated with Aβ plaques (Supplementary Fig. 3C). These data provide direct evidence that increased sTGFBR3 levels are associated with AD.
4.3 MT1-MMP inhibitor restored the impaired TGF-β/Smad signaling in LPS-induced BV2 cells
To verify whether sTGFBR3 was involved in blocking TGF-β/Smad signaling. We treated LPS-induced BV2 cells with marimastat, an inhibitor of MT1-MMP. Immunoblotting analysis showed that sTGFBR3 in the extracellular matrix was significantly increased, and P-Smad2/3 in the cytoplasm was decreased, suggesting the blockade of TGF-β/Smad signaling in LPS-induced BV2 cells (Fig. 4A-4G). However, these abnormalities were reversed in the coculture system of marimastat and LPS. The results showed that sTGFBR3 in the extracellular matrix was significantly decreased, and P-Smad2/3 in the cytoplasm was increased (Fig. 4A-4G). We also observed that the proinflammatory factors IL-6 and TNF-α were increased in LPS-induced BV2 cells, suggesting the formation of M1 proinflammatory microglia, which was also reversed by marimastat (Fig. 4A, 4H-4I), indicating that lowering sTGFBR3 could decrease inflammatory factors and promote microglial M2 polarization.
4.4 sTGFBR3 is associated with cognitive decline in APP/PS1 transgenic mice
Having established that sTGFBR3 levels are strongly associated with AD, we then assessed the relevance of our findings for dementia. To this end, the sTGFBR3 knockdown model was established using a lentivirus to interfere with the expression of sTGFBR3 in the brains of APP/PS1 mice. Notably, sTGFBR3 expression was significantly increased in APP/PS1 mice compared with wild-type controls (Fig. 5A-5B). In contrast, sTGFBR3 expression was decreased in APP/PS1 mice with lentivirus intervention, demonstrating efficient viral transduction and sTGFBR3 knockdown (Fig. 5A-5B). Next, we found a significant association of higher sTGFBR3 expression with increased measures of cognitive decline by the MWM task. APP/PS1 mice spent more time reaching the target platform during the training period and displayed severe loss of spatial learning and memory compared with wild-type controls. In contrast, sTGFBR3 knockdown significantly improved task learning (Fig. 5C). Furthermore, during the test period, sTGFBR3 knockdown resulted in a significantly decreased time to reach the target platform and an increased number of platform crossings but no significant alteration in swimming speed, which suggested alleviation of cognitive impairment (Fig. 5D-5G).
We next determined whether sTGFBR3 knockdown affected Aβ deposition in APP/PS1 mice. As shown in Fig. 5H-5I, the number of Aβ plaques was markedly decreased in lentivirus-treated APP/PS1 mice compared to shRNA ctrl-treated APP/PS1 mice, indicating that sTGFBR3 knockdown could alleviate the Aβ plaque burden in the cortex and hippocampus of APP/PS1 transgenic mice. Consistently, ELISA analysis revealed a significant reduction in both soluble Aβ40 and Aβ42 in the temporal and hippocampal extracts (Fig. 5J-5 M), suggesting that sTGFBR3 plays a crucial role in promoting Aβ deposition in APP/PS1 mice. Furthermore, to clarify whether the decline in Aβ content was due to the decrease in Aβ production or the increase in Aβ clearance, we quantified the protein expression of full-length APP and BACE1. Immunoblotting analysis showed that the critical regulators APP and BACE1 were significantly decreased in lentivirus-treated APP/PS1 mice (Fig. 5N-5P), suggesting that sTGFBR3 knockdown influenced APP metabolism. Here, it is unclear whether sTGFBR3 knockdown affects the clearance of Aβ.
4.5 The effect sTGFBR3 knockdown on microglial function in APP/PS1 mice
Abnormal phagocytosis of microglia mediates the propagation of Aβ within the brain in AD [39]. We therefore examined the effect of sTGFBR3 knockdown on microglial chemotaxis and phagocytosis. As shown in Fig. 6A-6B and S4A-S4B, triple staining for Aβ, Iba1 and TREM2 was used to characterize the chemotaxis and phagocytosis of microglia for Aβ plaques in APP/PS1 mice. The results showed that sTGFBR3 knockdown significantly decreased the number of microglia around plaques. Meanwhile, immunoblotting analysis showed that Iba1, CCR2 and MCP1 were significantly decreased in lentivirus-treated APP/PS1 mice (Fig. S4C, S4E, Supplementary Fig. 4F-4H), indicating that the chemotaxis of microglia to Aβ plaques was weakened.
Moreover, TREM2 mediates abnormal Aβ phagocytosis by microglia [40]. TREM2 staining indicated that phagocytosis from microglia to Aβ plaques was enhanced compared to a lower number of microglia (Fig. 6A-6B). Immunoblotting analysis showed similar results (Fig. S4C-S4D). In addition, we found that sTGFBR3 knockdown significantly decreased the levels of the proinflammatory factors IL-6 and TNF-α but increased the levels of the anti-inflammatory factors IL-4 and IL-10 (Supplementary Fig. 5A-5G). Moreover, sTGFBR3 knockdown significantly decreased NLRP3 and NF-κB p65 (Supplementary Fig. 5H-5J), indicating that microglia polarized into the M2 phenotype from the M1 phenotype. These results suggested that the polarization of the microglia M2 phenotype restores phagocytosis but reduces diffusion of the Aβ protein.
TGF-β/Smad signaling is involved in the pathological process of AD, including Aβ uptake, neuroinflammation and chemotaxis of microglia.[41,42] Immunoblotting analysis showed that P-Smad2/3 was significantly increased in lentivirus-treated APP/PS1 mice (Fig. 6D-6E), suggesting that TGF-β1/Smad3 signaling was enhanced and Aβ uptake in the AD brain was increased. Finally, Griess Reagent analysis showed that NO release was significantly decreased in lentivirus-treated APP/PS1 mice compared with an increased level in APP/PS1 mice (Fig. 6F).
Overall, the knockdown of sTGFBR3 improved TGF-β1/Smad3 signaling and weakened non-Smad-dependent NF-κB signaling. These changes polarized microglia into the M2 phenotype and enhanced the phagocytosis of microglia while limiting their chemotaxis.
4.6 sTGFBR3 knockdown inhibits LPS-induced neuroinflammation in vivo
The above studies have implicated sTGFBR3 in microglial neuroinflammation. To further investigate the role of sTGFBR3 in neuroinflammation, we established an acute LPS injection mouse model as previously described [36]. lentivirus was used to interfere with the expression of TGFBR3 in C57BL/6 mice. Four weeks later, an acute neuroinflammation model was induced by intraperitoneal injection of LPS. We then tested sTGFBR3 protein expression in the brain. sTGFBR3 expression was significantly increased in LPS-induced mice compared with wild-type controls. In contrast, sTGFBR3 expression was decreased in lentivirus-treated LPS-induced mice, demonstrating efficient viral transduction and sTGFBR3 knockdown (Fig. 6G). Next, we found that intraperitoneal injection of LPS significantly increased the levels of certain proinflammatory cytokines, including TNF-α and IL-6. Notably, sTGFBR3 knockdown markedly inhibited the levels of these cytokines (Fig. 6H-6I), suggesting a critical role for sTGFBR3 in mediating neuroinflammation.
Moreover, the changes in microglial morphology were analyzed by Iba1 staining. LPS injections induced significant changes in microglial morphology, characterized by decreased branch numbers, end-point voxels, max branch length, and a significant increase in total branch length. sTGFBR3 knockdown significantly rescued LPS-induced morphological changes (Fig. 6K-6 N).
4.7 sTGFBR3 is associated with cognitive decline in the STZ-induced tau hyperphosphorylation mouse model
To investigate the role of sTGFBR3 in tau hyperphosphorylation, an STZ-induced tau hyperphosphorylation mouse model was established as described earlier [38]. The expression of sTGFBR3 in the C57BL/6 mouse brain was interfered with by the above method. Four weeks later, a tau hyperphosphorylation mouse model was induced by intraperitoneal injection of STZ. Except for the effective sTGFBR3 knockdown in lentivirus-treated STZ-induced mice, we also observed that TGF-β1/Smad3 signaling was significantly enhanced compared with the decreased density of P-Smad2/3 in STZ-induced mice (Fig. 7A-7B). We then checked whether sTGFBR3 knockdown affected learning and memory impairment using the MWM task. STZ-induced mice spent more time reaching the platform in the training period, suggesting the presence of a cognitive deficit. Notably, sTGFBR3 knockdown significantly improved task learning (Fig. 7C). During the test period, sTGFBR3 knockdown resulted in a significantly decreased time to reach the platform, increased preference for the target quadrant, and increased number of platform crossings. However, no significant alteration in swimming speed suggested alleviation of cognitive impairment (Fig. 7D-7F).
Mechanistically, immunoblotting analysis showed that P-GSK3β (Ser9) was significantly increased and P-tau (Thr181, Ser202+Thr205, and Ser404) was significantly decreased in lentivirus-treated STZ-induced mice compared with the reverse changes in STZ-induced mice. This result suggests that tau hyperphosphorylation was reduced (Fig. 7G-7I). The structural integrity of neurons is the basis of learning and memory, and the destruction of neuronal structures often presupposes damage to cognitive function [43]. We found that the neurons in the cortex and hippocampus suffered from impairment in STZ-induced mice by Nissl staining, characterized by atrophy, deep staining, and a decreased number of neurons. However, sTGFBR3 knockdown rescued the impairment to neurons and restored the number of neurons (Supplementary Fig. 6A-6B). Immunofluorescence analysis revealed that the function of neurons was obviously improved by staining NeuN and MAP2 in lentivirus-treated STZ-induced mouse brains (Fig. 7J-7 L). Meanwhile, immunoblotting analysis showed that the density of synapse-related proteins, including PSD95, SYP, and SYT, was significantly increased in lentivirus-treated STZ-induced mouse brains (Supplementary Fig. 6C-6F), suggesting that sTGFBR3 knockdown restored synaptic plasticity.
Overall, sTGFBR3 knockdown reduced tau hyperphosphorylation, restored neuronal function, and maintained synaptic plasticity by restoring TGF-β1/Smad3 signaling in STZ-induced tau hyperphosphorylation.