Skip to main content
  • Short report
  • Open access
  • Published:

Deficiency of terminal complement pathway inhibitor promotes neuronal tau pathology and degeneration in mice

Abstract

Background

The neuronal microtubule-associated protein tau becomes hyperphosphorylated and forms aggregates in tauopathies but the processes leading to this pathological hallmark are not understood. Because tauopathies are accompanied by neuroinflammation and the complement cascade forms a key innate immune pathway, we asked whether the complement system has a role in the development of tau pathology.

Findings

We tested this hypothesis in two mouse models, which expressed either a central inhibitor of complement or lacked an inhibitor of the terminal complement pathway. Complement receptor-related gene/protein y is the natural inhibitor of the central complement component C3 in rodents. Expressing a soluble variant (sCrry) reduced the number of phospho-tau (AT8 epitope) positive neurons in the brain stem, cerebellum, cortex, and hippocampus of aged P301L mutant tau/sCrry double-transgenic mice compared with tau single-transgenic littermates (JNPL3 line). CD59a is the major inhibitor of formation of the membrane attack complex in mice. Intrahippocampal injection of adeno-associated virus encoding mutant human P301L tau into Cd59a−/− mice resulted in increased numbers of AT8-positive cells compared with wild-type controls. This was accompanied by neuronal and synaptic loss and reduced dendritic integrity.

Conclusions

Our data in two independent mouse models with genetic changes in key regulators of the complement system support the hypothesis that the terminal pathway has an active role in the development of tau pathology. We propose that inhibition of the terminal pathway may be beneficial in tauopathies.

Findings

Intraneuronal insoluble deposits of the microtubule-associated protein tau are found in neurodegenerative diseases commonly known as tauopathies [1]. One of the causes leading to these deposits in sporadic tauopathies may be aberrant phosphorylation of tau. A common feature in Alzheimer’s disease (AD), the most prevalent tauopathy, and other tauopathies is activation of immune pathways in the brain. The complement system is a key innate immune pathway, which is fully expressed in the brain, independent of peripheral contribution, and exerts critical homeostatic cerebral functions in development and aging (for extensive discussions and citations of relevant original articles about the role of complement system in the brain see [2, 3]). Brains of patients with Pick’s disease (a pure tauopathy), AD, or individuals with Down’s syndrome with AD-pathology are found to have tangle-bearing neurons that are decorated with complement proteins, including the membrane attack complex (MAC). The presence of MAC in the brain even at early stages of AD or the deposition of products of complement activation in aged normal brains [4] suggests a lack of proper inhibitory control of the cascade with age and disease. Indeed, in affected brain regions of AD patients, levels of the main inhibitor of the MAC, CD59, are reduced [5, 6]. These histological and biochemical findings in human beings open the question whether complement activation and formation of the MAC, in particular, are involved in the development of tau pathology.

To test this hypothesis we crossed human mutant P301L tau transgenic mice (line JNLP3, herein called ‘tau transgenic’, a model for a genetic form of a pure tauopathy [7]; all described mouse experiments have been approved by the Palo Alto Veterans Hospital Institutional Review Board for Animal Experiments) with transgenic mice overexpressing a soluble form of the murine complement receptor 1-related gene/protein y (sCrry) [8] (Figure 1A). Tau transgenic (n = 13) and double-transgenic tau/sCrry mice (n = 17) were aged until the first mice started to present with the described lack of hind limb splay or weight loss in this line [7]. The symptoms developed, however, only in very few mice and only after 15 to 20 months, which was much slower and later in life than reported for the original line on a mixed genetic background. Mice were sacrificed and 30 μm floating cryotome brain sections were stained according to the mouse-on-mouse protocol (Vector Labs) with the phospho-tau specific antibody AT8 (0.5 μg/ml, mouse monoclonal antibody; Pierce Thermo Scientific). After developing with immunoperoxidase and diaminobenzidine, the number of AT8-positive cell bodies was counted blindly by two independent observers using light microscopy. AT8 immunoreactivity was observed in the neocortex, deep cerebellar nuclei, and, most prominently, brainstem but only in about one-third of all mice (Figure 1B). Despite this lack of consistent penetration of tau pathology, the proportion of mice that developed tau pathology in brainstem was significantly higher in tau transgenic mice (6/13) compared with the complement-inhibited tau/sCrry transgenic mice (2/17) (Figure 1C; two-tailed Fisher’s exact test P = 0.049). An increased inter-mouse variability of tau pathology has been described previously for the JNLP3 line after crossing it for several generations onto the C57BL/6 background [9]. Here, tau and tau/sCrry transgenic mice had a mixed background with at least 70% calculated contribution of the C57BL/6 J background. Neuroinflammation was quantified by relative staining intensity of microglial marker CD68 (diluted 1:50, FA-11; Serotec) in the same brain region. CD68 staining intensity correlated highly with the abundance of AT8-positive cells (Figure 1D,E). Together, this points to an involvement of innate immune pathways and in particular the complement system in tau pathology.

Figure 1
figure 1

Abnormal tau phosphorylation is reduced in genetically complement-inhibited P301L tau transgenic mice. A, Simplified illustration of activation of the complement cascade in mice and its hypothetical effect on tau phosphorylation in the brain. Spontaneous (alternative pathway) or antibody-mediated (classical pathway) activation can initiate the complement cascade in the aging brain and if insufficiently controlled by inhibitory proteins (for example, Crry on C3 convertases or CD59a in the terminal pathway in mice) this can lead to the formation of a self-integrating membrane pore formed by C5b-9, also known as the membrane attack complex (MAC). The MAC disrupts cellular homeostasis by sublytic or lytic mechanisms potentially leading to activation of kinases, which could contribute to phosphorylation of tau protein. B, Examples of high and low tau pathology in various brain regions of 15 to 20 months aged P301L tau or P301L tau/sCrry transgenic mice, respectively, as detected by immunoreactivity of phospho-tau specific antibody AT8. C, Number of AT8-positive cells in the brainstem of 15 to 20 months aged tau (n = 13) and tau/sCrry (n = 17) transgenic mice. Each symbol indicates the mean from 5 to 6 sections per mouse (solid line is the group mean). The proportion of mice that developed tau pathology above the overall mean (dashed line) was significantly higher in tau mice than in the complement-inhibited tau/sCrry transgenic mice (2×2 contingency table, two-tailed Fisher’s exact test). D, Immunoreactivity for microglia marker CD68 in the brainstem of the two mice shown in B. E, Average number of AT8-positive cells in the brainstem plotted against % area of CD68 immunoreactivity in the same brain area. Linear regression with trend line (solid line) and 95% confidence intervals (C.I., dashed lines) are indicated; r2 as goodness-of-fit is significant (95% C.I. for the slope and P = 0.0001, two-tailed F test). Scale bar, 100 μm.

Insertion of the MAC into the cell membrane in mice is tightly controlled by CD59a in mice and in human beings by CD59 [10] (Figure 1A). A lack thereof leaves cells more susceptible to spontaneous and induced attack by the MAC [10, 11]. To test the effect of CD59a-deficiency on tau phosphorylation in the brain, Cd59a−/− mice (n = 5) [11] and wild-type littermates (n = 3) aged 4.5 months were injected stereotaxically with an adeno-associated virus (AAV2 serotype) encoding human P301L mutant tau in the right hippocampus and with an AAV2 encoding green fluorescent protein (GFP, internal control) in the left hippocampus. Two microliters of each AAV2 (tau or GFP; 8 × 1012 vg/ml [12]) were injected with a 10 μl Hamilton syringe for 2 min at a rate of 1 μl/min in a nanoinjector system. The needle was allowed to remain in the brain for an additional 2 min. The coordinates were anterior/posterior from bregma −2.0 mm, lateral +/− 1.5 mm, and dorsal/ventral 1.4 mm. After 5 months, mice were sacrificed and prepared for histological analysis as described previously. We observed a strong cytoplasmic and neuritic GFP signal in the granular layer of AAV2-GFP injected hippocampi of wild-type and Cd59−/− mice, consistent with robust expression of GFP and efficient transduction by AAV2. Occasionally, GFP-positive neurites were also present in the contralateral hemisphere (Figure 2A). The hippocampus of Cd59a−/− mice expressing human P301L mutant tau had significantly higher levels of AT8-positive cells than the respective hippocampus of their wild-type littermates (Figure 2A,B; P = 0.036, Mann–Whitney U test). AT8 immunoreactivity was detected in neuronal cell bodies, neurites, and axons in the CA2 and CA3 regions of the tau-injected side (Figure 2A). Relative staining intensity of microglial marker CD68 did not correlate with immunoreactivity for tau phosphorylation (Figure 3A,B). Tau phosphorylation was, however, accompanied by significant reduction of hippocampal immunoreactivity for markers of neuronal (NeuN and microtubule-associated protein 2, MAP-2; both antibodies at 1:500, Millipore) and synaptic integrity (synaptophysin, 1:500, Millipore) (Figure 3C-H; ANOVA and post-hoc Tukey-Kramer test). In conclusion, lack of CD59a promotes abnormal tau phosphorylation and loss of neuronal and synaptic integrity in mouse brains producing human P301L mutant tau pointing to an active role of the terminal complement pathway in tau pathology.

Figure 2
figure 2

Lack of CD59a promotes abnormal tau phosphorylation, which is accompanied by neuronal and synaptic degeneration. A, Immunoreactivity of AT8 phosphorylated tau in the granular cell layer of the CA2/3 region of Cd59a−/− mice (n = 5) and wild-type (wt) littermate controls (n = 3) 5 months after intrahippocampal injection of adeno-associated virus (AAV2) encoding human P301L mutant tau (arrowheads indicates needle track). Contralateral hippocampus was injected with AAV2 encoding green fluorescent protein (AAV2-GFP), resulting in cytoplasmic and neuritic distribution of GFP. Occasionally, GFP-positive neuritis can also be observed in the AAV2-P301L tau-injected hemisphere. B, Number of AT8-positive cell bodies in the entire AAV2-P301L tau-injected hippocampus in Cd59a−/− mice and wild-type littermate controls. Each symbol indicates the mean number of 3 to 4 sections per mouse. Significance was calculated by Mann–Whitney U test; *P ≤ 0.05. Scale bar, 100 μm.

Figure 3
figure 3

Neuronal and synaptic degeneration in AAV2-P301L tau-injected hemisphere. Microglial marker (A,B, CD68), neuronal (C,D, NeuN), dendritic (E,F, MAP-2), and synaptic (G,H, synaptophysin) integrity was quantified based on mean % area of immunoreactivity (IR, for example, neuropil occupied for the respective markers) (B,F,H) or IR alone (D). MAP-2 was quantified lateral to injection site to avoid overlap with the GFP signal. Each symbol indicates the mean number of 3 to 4 sections per mouse. Significance was calculated by ANOVA and post-hoc Tukey-Kramer test; *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001. Scale bar, 100 μm (A), 200 μm (B), and 50 μm (D,E).

Earlier studies in mouse models of tauopathies reported a strong link between microglia activation and development of tau pathology [1317] or vice versa [18]. Whereas some identified this link after administration of an exogenous trigger of inflammation [1416], we observed a significant correlation between microglial activation and the number of AT8-positive cells in tau transgenic mice in the absence of exogenous stimuli (Figure 1D,E), which is consistent with findings in the brains of AD patients [19].

A growing number of studies show that the complement system probably has multiple functions in normal and injured brain and this may be relevant for AD [2, 3]. For instance, overexpression of sCrry or ablating C3 in APP transgenic mouse models of AD accelerated formation of amyloid-β plaques and neurodegeneration [20, 21]. This apparently protective effector function of the central component of the complement cascade may involve the opsonization of plaques followed by clearance of amyloid (Figure 1A). In contrast, full activation of the complement system and the terminal or lytic pathway can lead to formation of the MAC with possibly detrimental consequences. If not properly controlled by CD59, the MAC can generate differently sized (lytic and sublytic) pores in the cell membrane. Such pores then lead to increased Ca2+ influx, which may trigger depolarization of the membrane and activation of kinases in the cell [22]. Thus, full complement activation involving the terminal pathway and MAC formation may be upstream in the activation of kinases, such as MAPK, PKC, JNK, or PI3K/AKT, which have been implicated in the regulation of tau phosphorylation. Cellular and in vivo experiments also demonstrate that MAC formation in neurons can induce seizures and excitotoxicity, which promote neurodegeneration [23]. Excitotoxicity and seizures have also been proposed to contribute to cognitive decline in AD mouse models and patients. Interestingly, the absence of tau protects APP transgenic and wild-type mice from excitotoxic insults and prevents behavioral deficits [24]. Whether MAC formation and excitotoxicity are linked through tau would need to be studied in more detail.

Neuropathological analyses in human beings and our in vivo data do indeed point to an active role of the terminal complement pathway in the development of tau pathology, a neuropathological hallmark of AD and other tauopathies. Intriguingly, recent genome-wide association studies identified independently genetic variants of complement receptor 1 (CR1/CD35) and clusterin to be associated with AD ([25], replicated by others: http://www.alzgene.org). CR1/CD35 is one of the human functional analogs of the murine Crry and clusterin, which is also known as apolipoprotein J or complement lysis inhibitor, acts just one step upstream of CD59. It is interesting that, out of all immune-function-related proteins, two key regulators of the central component and the terminal cascade seem to be genetically linked with AD. It remains to be shown whether variants of genes coding for complement components or regulators are associated with other tauopathies as well.

In summary, complement proteins are key components of the innate immune system and have been implicated in homeostasis as well as degeneration of the human brain. Our data in two independent mouse models with genetic alterations in complement regulatory proteins support the hypothesis that the terminal pathway has an active role in the development of tau pathology. We propose that inhibition of the terminal pathway and, more specifically, inhibition of the MAC may be beneficial in tauopathies.

Abbreviations

AAV2:

Adeno-associated virus serotype 2

AD:

Alzheimer’s disease

CR1:

Complement receptor 1

GFP:

Green fluorescent protein

IR:

Immunoreactivity

MAC:

Membrane attack complex

MAP-2:

Microtubule-associated protein 2

sCrry:

Soluble complement receptor 1-related gene/protein y

wt:

Wild-type.

References

  1. Lee VM, Goedert M, Trojanowski JQ: Neurodegenerative tauopathies. Annu Rev Neurosci 2001, 24:1121–1159.

    Article  CAS  PubMed  Google Scholar 

  2. Stephan AH, Barres BA, Stevens B: The complement system: an unexpected role in synaptic pruning during development and disease. Annu Rev Neurosci 2012, 35:369–389.

    Article  CAS  PubMed  Google Scholar 

  3. Veerhuis R, Nielsen HM, Tenner AJ: Complement in the brain. Mol Immunol 2011,48(14):1592–1603.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Loeffler DA, Camp DM, Schonberger MB, Singer DJ, LeWitt PA: Early complement activation increases in the brain in some aged normal subjects. Neurobiol Aging 2004,25(8):1001–1007.

    Article  CAS  PubMed  Google Scholar 

  5. Yasojima K, McGeer EG, McGeer PL: Complement regulators C1 inhibitor and CD59 do not significantly inhibit complement activation in Alzheimer disease. Brain Res 1999,833(2):297–301.

    Article  CAS  PubMed  Google Scholar 

  6. Yang LB, Li R, Meri S, Rogers J, Shen Y: Deficiency of complement defense protein CD59 may contribute to neurodegeneration in Alzheimer’s disease. J Neurosci 2000,20(20):7505–7509.

    CAS  PubMed  Google Scholar 

  7. Lewis J, McGowan E, Rockwood J, Melrose H, Nacharaju P, Van Slegtenhorst M, Gwinn-Hardy K, Murphy MP, Baker M, Yu X, Duff K, Hardy J, Corral A, Lin W-L, Yen S-H, Dickson D, Davies P, Hutton M: Neurofibrillary tangles, amyotrophy and progressive motor disturbance in mice expressing mutant (P301L) tau protein. Nat Genet 2000,25(4):402–405.

    Article  CAS  PubMed  Google Scholar 

  8. Quigg RJ, He C, Lim A, Berthiaume D, Alexander JJ, Kraus D, Holers VM: Transgenic mice overexpressing the complement inhibitor Crry as a soluble protein are protected from antibody-induced glomerular injury. J Exp Med 1998, 188:1321–1331.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Bolmont T, Clavaguera F, Meyer-Luehmann M, Herzig MC, Radde R, Staufenbiel M, Lewis J, Hutton M, Tolnay M, Jucker M: Induction of tau pathology by intracerebral infusion of amyloid-β-containing brain extract and by amyloid-β deposition in APP x Tau transgenic mice. Am J Pathol 2007,171(6):2012–2020.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Baalasubramanian S, Harris CL, Donev RM, Mizuno M, Omidvar N, Song WC, Morgan BP: CD59a is the primary regulator of membrane attack complex assembly in the mouse. J Immunol 2004,173(6):3684–3692.

    Article  CAS  PubMed  Google Scholar 

  11. Holt DS, Botto M, Bygrave AE, Hanna SM, Walport MJ, Morgan BP: Targeted deletion of the CD59 gene causes spontaneous intravascular hemolysis and hemoglobinuria. Blood 2001,98(2):442–449.

    Article  CAS  PubMed  Google Scholar 

  12. Ubhi K, Rockenstein E, Doppler E, Mante M, Adame A, Patrick C, Trejo M, Crews L, Paulino A, Moessler H, Masliah E: Neurofibrillary and neurodegenerative pathology in APP-transgenic mice injected with AAV2-mutant TAU: neuroprotective effects of Cerebrolysin. Acta Neuropathol (Berl) 2009,117(6):699–712.

    Article  CAS  Google Scholar 

  13. Jaworski T, Lechat B, Demedts D, Gielis L, Devijver H, Borghgraef P, Duimel H, Verheyen F, Kugler S, Van Leuven F: Dendritic degeneration, neurovascular defects, and inflammation precede neuronal loss in a mouse model for tau-mediated neurodegeneration. Am J Pathol 2011,179(4):2001–2015.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Bhaskar K, Konerth M, Kokiko-Cochran ON, Cardona A, Ransohoff RM, Lamb BT: Regulation of tau pathology by the microglial fractalkine receptor. Neuron 2010,68(1):19–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Kitazawa M, Oddo S, Yamasaki TR, Green KN, LaFerla FM: Lipopolysaccharide-induced inflammation exacerbates tau pathology by a cyclin-dependent kinase 5-mediated pathway in a transgenic model of Alzheimer’s disease. J Neurosci 2005,25(39):8843–8853.

    Article  CAS  PubMed  Google Scholar 

  16. Li Y, Liu L, Barger SW, Griffin WS: Interleukin-1 mediates pathological effects of microglia on tau phosphorylation and on synaptophysin synthesis in cortical neurons through a p38-MAPK pathway. J Neurosci 2003,23(5):1605–1611.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Yoshiyama Y, Higuchi M, Zhang B, Huang SM, Iwata N, Saido TC, Maeda J, Suhara T, Trojanowski JQ, Lee VM: Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron 2007,53(3):337–351.

    Article  CAS  PubMed  Google Scholar 

  18. Zilka N, Stozicka Z, Kovac A, Pilipcinec E, Bugos O, Novak M: Human misfolded truncated tau protein promotes activation of microglia and leukocyte infiltration in the transgenic rat model of tauopathy. J Neuroimmunol 2009,209(1–2):16–25.

    Article  CAS  PubMed  Google Scholar 

  19. DiPatre PL, Gelman BB: Microglial cell activation in aging and Alzheimer disease: partial linkage with neurofibrillary tangle burden in the hippocampus. J Neuropathol Exp Neurol 1997,56(2):143–149.

    Article  CAS  PubMed  Google Scholar 

  20. Wyss-Coray T, Yan F, Lin AH, Lambris JD, Alexander JJ, Quigg RJ, Masliah E: Prominent neurodegeneration and increased plaque formation in complement-inhibited Alzheimer’s mice. Proc Natl Acad Sci USA 2002,99(16):10837–10842.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Maier M, Peng Y, Jiang L, Seabrook TJ, Carroll MC, Lemere CA: Complement C3 deficiency leads to accelerated amyloid beta plaque deposition and neurodegeneration and modulation of the microglia/macrophage phenotype in amyloid precursor protein transgenic mice. J Neurosci 2008,28(25):6333–6341.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Tegla CA, Cudrici C, Trippe R 3rd, Rus V, Niculescu F, Rus H: Membrane attack by complement: the assembly and biology of terminal complement complexes. Immunol Res 2011,51(1):45–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Xiong ZQ, Qian W, Suzuki K, McNamara JO: Formation of complement membrane attack complex in mammalian cerebral cortex evokes seizures and neurodegeneration. J Neurosci 2003,23(3):955–960.

    CAS  PubMed  Google Scholar 

  24. Roberson ED, Scearce-Levie K, Palop JJ, Yan F, Cheng IH, Wu T, Gerstein H, Yu GQ, Mucke L: Reducing endogenous tau ameliorates amyloid β-induced deficits in an Alzheimer’s disease mouse model. Science 2007,316(5825):750–754.

    Article  CAS  PubMed  Google Scholar 

  25. Lambert JC, Heath S, Even G, Campion D, Sleegers K, Hiltunen M, Combarros O, Zelenika D, Bullido MJ, Tavernier B, Letenneur L, Bettens K, Berr C, Pasquier F, Fiévet N, Barberger-Gateau P, Engelborghs S, De Deyn P, Mateo I, Franck A, Helisalmi S, Porcellini E, Hanon O, de Pancorbo MM, Lendon C, Dufouil C, Jaillard C, Leveillard T, Alvarez V, The European Alzheimer’s Disease Initiative Investigators: Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer’s disease. Nat Genet 2009,41(10):1094–1099.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Drs. S. Tomlinson (Medical University of South Carolina, USA) and BP Morgan (Cardiff University School of Medicine, UK) for providing the Cd59a−/− mice, and J Warner and LH Mondshein for technical assistance. This work was supported by the Swiss National Science Foundation (postdoctoral fellowship PBBEB-102096 to MB), NIH (AG18440, AG02074, AG10435 to EM), National Institute on Aging (AG20603 and AG27505 to TW-C), and the Department of Veterans Affairs (TW-C).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Markus Britschgi or Tony Wyss-Coray.

Additional information

Competing interests

MB is currently a full-time employee of Roche/F. Hoffmann-La Roche Ltd., Basel, Switzerland. The data related to this manuscript were generated when MB was still at Stanford University and the manuscript was written when MB was already at Roche. The authors declare that they have no competing interests.

Authors’ contributions

MB and TW-C designed research and wrote the paper; MB, YT-U, HJ, ER, and EM performed research; MB, EM, and TW-C analyzed data. All authors read and approved the final manuscript.

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Britschgi, M., Takeda-Uchimura, Y., Rockenstein, E. et al. Deficiency of terminal complement pathway inhibitor promotes neuronal tau pathology and degeneration in mice. J Neuroinflammation 9, 220 (2012). https://doi.org/10.1186/1742-2094-9-220

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1742-2094-9-220

Keywords