Skip to main content

MINI REVIEW article

Front. Neurosci., 14 October 2021
Sec. Neurodegeneration

Porphyromonas gingivalis-Induced Neuroinflammation in Alzheimer’s Disease

  • Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway

“Chronic” periodontitis and its keystone pathogen Porphyromonas gingivalis have repeatedly been associated with Alzheimer’s disease (AD). Pathological hallmarks in AD are brain accumulations of amyloid-beta and neurofibrillary tangles consisting of aggregated and hyperphosphorylated tau. In addition, neuroinflammation induced by P. gingivalis has increasingly been recognized as a factor in the pathogenesis of AD. The present mini-review discusses possible mechanisms for the induction of neuroinflammation by P. gingivalis in AD, involving factors such as pro-inflammatory mediators, amyloid-beta, tau, microglia, cathepsin B, and protein kinase R. Inflammagens of P. gingivalis such as lipopolysaccharide and gingipains are also discussed.

Introduction

“Chronic” periodontitis is a disease affecting the supporting tissues of the teeth. Untreated, it may end with tooth loss. It is a widely prevalent disease in adults all over the world (Eke et al., 2016) and has in several reports been associated with Alzheimer’s disease (AD) (for a review, see Olsen, 2021). Porphyromonas gingivalis, which is considered a keystone bacterium in “chronic” periodontitis (Socransky et al., 1998; Darveau et al., 2012; Hajishengallis et al., 2012), has been detected in the brains of subjects with AD together with its toxic proteases—gingipains (Dominy et al., 2019). Also, P. gingivalis DNA was found in AD brains and cerebrospinal fluid of clinical AD patients. In other studies, P. gingivalis lipopolysaccharide (LPS) was detected in human AD brains and in the brains from transgenic mice serving as AD models (Poole et al., 2013; Ishida et al., 2017).

Several animal studies have indicated that P. gingivalis can induce neuroinflammation in the brain of AD patients (see later), and neuroinflammation has increasingly been suggested to have a substantial role in the progression of the neuropathological changes taking place in AD (Ilievski et al., 2018). This mini-review will deal with neuroinflammation in AD induced by P. gingivalis and possible mechanisms for this induction.

Neuroinflammation and Porphyromonas gingivalis

Alzheimer’s disease is our commonest neurological disease characterized by cognitive decline and accumulation of amyloid-beta (Aβ) plaques and neurofibrillary tangles (NTFs). Neuroinflammation has increasingly been considered as another hallmark of AD. P. gingivalis-LPS-induced neuroinflammation was proposed to play an important role in the cognitive impairment of C57BL/6 mice (Zhang et al., 2018). Hu et al. (2020) found that periodontitis induced by P. gingivalis-LPS promoted neuroinflammation by activating the Toll-like receptor 4/nuclear factor-kappa B signaling pathway and was associated with learning and memory impairment in Sprague-Dawley rats. In another study, P. gingivalis periodontal infection was proposed to cause cognitive impairment by releasing pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), interleukin (IL)-6, and IL-1β in the brain tissues of middle-aged mice (Ding et al., 2018). Similarly, Memedovski et al. (2020) reported that P. gingivalis LPS induced classical and alternative activation of rat brain microglia with concomitant release of cytokines and chemokines.

Amyloid-Beta and Porphyromonas gingivalis

Amyloid-beta is known to be an activator of microglia. On the one hand, microglia can release inflammatory mediators such as inflammatory cytokines, complement components, chemokines, and free radicals that all contribute to Aβ production and accumulation. On the other hand, microglia can play a beneficial role in generating anti-Aβ antibodies and stimulating the clearance of Aβ plaques (Cai et al., 2014). According to these authors, a vicious cycle of inflammation occurs between Aβ accumulation, activated microglia, and microglia inflammatory mediators, which promotes Aβ deposition and neuroinflammation. This idea diverges from the general notion that Aβ production and neuroinflammation are independent processes.

Nie et al. (2019) reported that chronic exposure to P. gingivalis LPS led to the accumulation of Aβ in the brain of middle-aged mice. Such exposure also induced peripheral Aβ accumulation in inflammatory monocytes/macrophages. This suggested that monocytes/macrophages can serve as a circulating pool of Aβ in patients with periodontitis. Similarly, Leira et al. (2019) reported that P. gingivalis-induced LPS in periodontitis produced increased serum levels of Aβ peptides. In mice, oral P. gingivalis infection caused brain colonization and increased production of the amyloid plaque component Aβ142 (Dominy et al., 2019). Importantly, the neuroinflammation established by P. gingivalis in the mice could be reduced by gingipain inhibition.

Tau Protein and Porphyromonas gingivalis

As mentioned, NFTs are created from hyperphosphorylated tau—a protein that stabilizes microtubules (for a review, see Kinney et al., 2018). In AD, hyperphosphorylated tau is removed from microtubules resulting in a collapse of the microtubule structure and thereby disrupted cellular functions for protein trafficking and cellular morphology, formation of tau aggregates, loss of neuronal function, and apoptosis.

There is a clear relationship between P. gingivalis and tau. Gingipains may cleave procaspase-3 to activate caspase-3 (Urnowey et al., 2006). The latter has been associated with tau phosphorylation (Chu et al., 2017) and tau cleavage (Sandhu et al., 2017). Dominy et al. (2019) found tau to be a target of gingipain proteolysis and suggested that tau pathology in AD brains may be caused by transneural spread of P. gingivalis, tau damage by gingipain proteolysis, and activation of human proteases. They also hypothesized that gingipains might be a driver of a compensatory increase in tau production of AD patients.

Tang et al. (2021) confirmed that peripheral P. gingivalis infection caused tau hyperphosphorylation, preventing tau from fulfilling its role as a microtubule-stabilizing protein, leaving it to self-assembly. In P. gingivalis-injected rats, the severity of phosphorylated tau at the AD-related sites Thr181 and Thr231 and the number of activated astrocytes were greater than in the hippocampus. Also, the levels of IL-1β, IL-6, and TNF-α in the rat serum and hippocampus were increased. Furthermore, the activity of protein phosphatase 2A (PP2A) was significantly inhibited in the hippocampus of these rats. Inhibition of PP2A and application of a PP2A promoter efficiently decreased IL-1β-induced tau hyperphosphorylation in HT-22 cells. Although systemic inflammation was identified as the driver of tau phosphorylation, the specificity of P. gingivalis producing this effect was not assessed. Laurent et al. (2018) and Didonna (2020) emphasized tauopathies and neuroinflammatory processes as a vicious circle that works together in the pathogenesis of AD. A link between pro-inflammatory cytokine signaling and hyperphosphorylation of tau has also been reported (Domingues et al., 2017). Of note, usnic acid derivatives were found to inhibit tau aggregation and neuroinflammation (Shi et al., 2020).

A novel mechanism of tau-seed-affected microglia was demonstrated by activation of the NLRP3–ASC inflammasome (Stancu et al., 2019). This inflammasome is an important sensor of innate immunity. Olsen and Singhrao (2016) and Olsen and Yilmaz (2016) reviewed the plausible contribution of specific bacteria playing a role in influencing the activity of the NLRP3 inflammasome in AD progression. P. gingivalis was found to have several mechanisms for modulating innate immunity by limiting the activation of the NLRP3 inflammasome. Among them, ATP-/P2X7-signaling is associated not only with periodontitis but also with the development of several systemic diseases, including AD.

Microglia and Porphyromonas gingivalis

Hu et al. (2020) observed that P. gingivalis LPS-induced periodontitis caused learning and memory impairment in rats through neuroinflammation induced by significant activation of microglia and astrocytes in the brain cortex. Microglia activation may precede tau pathology (Yoshiyama et al., 2007). Memedovski et al. (2020) reported that 18-h in vitro stimulation with ultrapure P. gingivalis LPS caused classical and alternative activation of rat brain microglia with the release of cytokines and chemokines.

The gingipains Rgp and Kgp have important effects on brain-residing microglia, being responsible for P. gingivalis-induced cell migration of microglia and expression of pro-inflammatory mediators by activating the protease-activated receptor 2 (Liu et al., 2017; Nonaka and Nakanishi, 2020). The subsequent activation of phosphoinositide 3-kinase/Akt and mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) kinase/ERK pathways resulted in cell migration and inflammatory response in microglia. Here, the gingipains of P. gingivalis cooperatively contributed to cell migration of microglia toward the infected site (brain) and induction of neuroinflammation after reaching it.

The interaction between genetic factors, microglia, and P. gingivalis was reviewed by Olsen and Singhrao (2020). It was suggested that genes for apolipoprotein, clusterin, CD33, triggering receptor expressed on myeloid cells-2, tyrosine kinase binding protein (TYR-OBP), and complement receptors could affect microglia. Most of these genes can also be affected by P. gingivalis via its mastering of immune suppression.

Cathepsin B and Porphyromonas gingivalis

Cathepsin B (CatB), a lysosomal cysteine protease, was suggested to have an important role in the initiation of neuroinflammation and neural dysfunction after chronic systemic exposure to LPS from P. gingivalis in mice. Thus, Wu et al. (2017) found that such exposure to P. gingivalis LPS induced AD-like phenotypes, including microglia-mediated neuroinflammation, intracellular Aβ accumulation in neurons, and reduced learning and memory functions in middle-aged mice in a CatB-dependent manner. As already mentioned, chronic systemic P. gingivalis infection induced Aβ accumulation in inflammatory monocytes/macrophages. This occurred via activation of CatB/nuclear factor kappa B signaling (Nie et al., 2019). CatB has been suggested as a potential therapeutic target for preventing the initiation and progression of periodontitis-related AD (Nakanishi et al., 2020).

Protein Kinase R and Porphyromonas gingivalis

Protein kinase R (PKR) is a 551 amino acid protein responsible for a key part of the defense against bacterial and viral infections in neurons (Dabo and Meurs, 2012; Marchal et al., 2014). This inflammation-associated kinase protein directly phosphorylates several abnormal and disease-modifying residues within tau, such as Thr181, Ser199/202, Thr231, Ser396, Ser404, and Ser409 (Reimer et al., 2021). The PKR-mediated phosphorylations actively dislocate tau from microtubules in cells. Also, PKR overexpression and knockdown increased and decreased, respectively, tau protein and mRNA levels in cells. It was noteworthy that acute encephalopathy in wild-type mice, induced by intracranial Langat virus infection, resulted in robust inflammation and PKR upregulation, which was followed by abnormally phosphorylated full-length and truncated tau. PKR can be capable of triggering pathological modification of tau independent of other kinases after brain inflammation. This might be the initial pathological seed in tauopathies such as AD and in chronic encephalopathy with severe inflammation. PKR inhibition reduced phosphorylation of soluble tau in the brain of transgenic rTg4510 tau mice (Reimer et al., 2021). Inhibition of PKR also prevented long-term potentiation and memory impairment in AD mouse models (Hwang et al., 2017). Furthermore, PKR inhibition reduced neuronal loss, motor deficits, and memory deficits in mice models of AD (Mouton-Liger et al., 2015; Segev et al., 2015; Reimer et al., 2021).

A direct relationship between P. gingivalis and PKR has not yet been demonstrated. However, PKR, a ubiquitously expressed serine–threonine kinase, is activated by indirect binding to bacterial LPS or pro-inflammatory cytokines such as TNF-α, IL-1, and interferon-gamma (for a review, see Reimer et al., 2021). PKR directly regulates tau, and activation of PKR has been associated with different tauopathies such as AD, Parkinson’s disease, and Huntington’ disease (Peel et al., 2001; Chang et al., 2002; Bando et al., 2005; Paquet et al., 2012; Lourenco et al., 2013; Ma et al., 2013). Because PKR is activated indirectly by LPS and specific cytokines, this could contribute to the correlation of “chronic” periodontitis and P. gingivalis brain levels with AD (Reimer et al., 2021). Bacterial infections and inflammation could also make neurons vulnerable to degeneration and thus initiate the onset of neurodegenerative diseases such as AD (Deleidi and Isacson, 2012). In this situation, activated PKR could initiate abnormal tau phosphorylation.

Concluding Remarks

Neuroinflammation seems to have a substantial role in the pathogenesis of AD. This supports neuroinflammation as a third disease hallmark of the disease. P. gingivalis with its inflammagens, gingipains, and LPS, both detected in the brains of AD subjects, could be major factors inducing neuroinflammation. P. gingivalis particularly affects Aβ, tau, microglia, CatB, and possibly PKR. A vicious cycle of inflammation probably occurs between several of these players where the interaction is complex and not yet fully understood.

Although not specifically related to P. gingivalis, PKR stands out as an inflammation-associated kinase of particular interest because it is ubiquitously expressed and an important part of the defense against bacterial infections in neurons. It is activated indirectly by LPS and specific pro-inflammatory cytokines and has been linked to AD. PKR is co-localized with abnormally phosphorylated tau in AD brains and directly regulates tau expression. Thus, PKR activated by P. gingivalis-induced brain infection/inflammation/pro-inflammatory cytokines may precede tau phosphorylation and thus participate in the etiology of AD. This should be studied.

Author Contributions

The author confirms being the sole contributor of this work and has approved it for publication.

Conflict of Interest

The author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Bando, Y., Onuki, R., Katayama, T., Manabe, T., Kudo, T., Taira, K., et al. (2005). Double-strand RNA dependent protein kinase (PKR) is involved in the extrastriatal degeneration in Parkinson’s disease and Huntington’s disease. Neurochem. Int. 46, 11–18. doi: 10.1016/j.neuint.2004.07.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Cai, Z., Hussain, M. D., and Yan, L.-J. (2014). Microglia, neuroinflammation, and beta-amyloid protein in Alzheimer’s disease. Int. J. Neurosci. 124, 307–321. doi: 10.3109/00207454.2013.833510

PubMed Abstract | CrossRef Full Text | Google Scholar

Chang, R. C., Wong, A. K., Ng, H. K., and Hugon, J. (2002). Phosphorylation of eukaryotic initiation factor-2-alpha (eIF2alpha) is associated with neuronal degeneration in Alzheimer’s disease. NeuroReport 13, 2429–2432. doi: 10.1097/00001756-200212200-00011

PubMed Abstract | CrossRef Full Text | Google Scholar

Chu, J., Lauretti, E., and Praticò, D. (2017). Caspase-3-dependent cleavage of Akt modulates tau phosphorylation via GSK3β kinase: implications for Alzheimer’s disease. Mol. Psychiatry 22, 1002–1008. doi: 10.1038/mp.2016.214

PubMed Abstract | CrossRef Full Text | Google Scholar

Dabo, S., and Meurs, E. F. (2012). dsRNA-dependent protein kinase PKR and its role in stress, signaling and HCV infection. Viruses 4, 2598–2635. doi: 10.3390/v4112598

PubMed Abstract | CrossRef Full Text | Google Scholar

Darveau, R. P., Hajishengallis, G., and Curtis, M. A. (2012). Porphyromonas gingivalis as a potential community activist. J. Dent. Res. 91, 816–820.

Google Scholar

Deleidi, M., and Isacson, O. (2012). Viral and inflammatory triggers of neurodegenerative diseases. Sci. Transl. Med. 4:121s3. doi: 10.1126/scitranslmed.3003492

PubMed Abstract | CrossRef Full Text | Google Scholar

Didonna, A. (2020). Tau at the interface between neurodegeneration and neuroinflammation. Genes Immun. 21, 288–300. doi: 10.1038/s41435-020-00113-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Ding, Y., Ren, J., Yu, H., Yu, W., and Zhou, Y. (2018). Porphyromonas gingivalis, a periodontitis causing bacterium, induces memory impairment and age-dependent neuroinflammation in mice. Immun. Ageing 15:6. doi: 10.1186/s12979-017-0110-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Domingues, C., da Cruz, E., Silva, O. A. B., and Henriques, A. G. (2017). Impact of cytokines and chemokines on Alzheimer’s disease neuropathological hallmarks. Curr. Alzheimer Res. 14, 870–882. doi: 10.2174/1567205014666170317113606

PubMed Abstract | CrossRef Full Text | Google Scholar

Dominy, S. S., Lynch, C., Ermini, F., Benedyk, M., Marczyk, A., Konradi, A., et al. (2019). Porphyromonas gingivalis in Alzheimer’s disease brains: evidence for disease causation and treatment with small-molecule inhibitors. Sci. Adv. 5:eaau3333. doi: 10.1126/sciadv.aau3333

PubMed Abstract | CrossRef Full Text | Google Scholar

Eke, P. I., Wei, L., Borgnakke, W. S., Thornton-Evans, G., Zhang, X., Lu, H., et al. (2016). Periodontitis prevalence in adults ≥ 65 years of age, in the USA. Periodontol. 2000 72, 76–95. doi: 10.1111/prd.12145

PubMed Abstract | CrossRef Full Text | Google Scholar

Hajishengallis, G., Darveau, R. P., and Curtis, M. A. (2012). The keystone-pathogen hypothesis. Nat. Rev. Microbiol. 10, 717–725. doi: 10.1038/nrmicro2873

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, Y., Li, H., Zhang, J., Zhang, X., Xia, X., Qiu, C., et al. (2020). Periodontitis induced by P. gingivalis-LPS is associated with neuroinflammation and learning and memory impairment in Sprague-Dawley rats. Front. Neurosc. 14:658. doi: 10.3389/fnins.2020.00658

PubMed Abstract | CrossRef Full Text | Google Scholar

Hwang, K. D., Bak, M. S., Kim, S. J., Rhee, S., and Lee, Y. S. (2017). Restoring synaptic plasticity and memory in mouse models of Alzheimer’s disease by PKR inhibition. Mol. Brain 10:57. doi: 10.1186/s13041-017-0338-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Ilievski, V., Zuchowska, P. K., Green, S. J., Toth, P. T., Ragozzino, M. E., Le, K., et al. (2018). Chronic oral application of a periodontal pathogen results in brain inflammation, neurodegeneration and amyloid beta production in wild type mice. PLoS One 13:e0204941. doi: 10.1371/journal.pone.0204941

PubMed Abstract | CrossRef Full Text | Google Scholar

Ishida, N., Ishihara, Y., Ishida, K., Tada, H., Funaki-Kato, Y., Hagiwara, M., et al. (2017). Periodontitis induced by bacterial infection exacerbates features of Alzheimer’s disease in transgenic mice. NPJ Aging Mech. Dis. 3:15. doi: 10.1038/s41514-017-0015-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Kinney, J. W., Bemiller, S. M., Murtishaw, A. S., Leisgang, A. M., Salazar, A. M., and Lamb, B. T. (2018). Inflammation as a central mechanism in Alzheimer’s disease. Alzheimers Dement. 4, 575–590. doi: 10.1016/j.trci.2018.06.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Laurent, C., Buée, L., and Blum, D. (2018). Tau and neuroinflammation: what impact for Alzheimer’s disease and tauopathies? Biomed. J. 41, 21–33. doi: 10.1016/j.bj.2018.01.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Leira, Y., Iglesias-Rey, R., Gómez-Lado, N., Aguiar, P., Campos, F., D’Aiuto, F., et al. (2019). Porphyromonas gingivalis lipopolysaccharide-induced periodontitis and serum amyloid-beta peptides. Arch. Oral Biol. 99, 120–125. doi: 10.1016/j.archoralbio.2019.01.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, Y., Wu, Z., Nakanishi, Y., Ni, J., Hayashi, Y., Takayama, F., et al. (2017). Infection of microglia with Porphyromonas gingivalis promotes cell migration and an inflammatory response through the gingipain-mediated activation of protease-activated receptor-2 in mice. Sci. Rep. 7:11759. doi: 10.1038/s41598-017-12173-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Lourenco, M. V., Clarke Lourenco, M. V., Clarke, J. R., Frozza, R. L., Bomfim, T. R., Forny-Germano, L., et al. (2013). TNF-α mediates PKR-dependent memory impairment and brain IRS-1 inhibition induced by Alzheimer’s β-amyloid oligomers in mice and monkeys. Cell Metab. 18, 831–843. doi: 10.1016/j.cmet.2013.11.00

CrossRef Full Text | Google Scholar

Ma, T., Trinh, M. A., Wexler, A. J., Bourbon, C., Gatti, E., Pierre, P., et al. (2013). Suppression of eIF2α kinases alleviates Alzheimer’s disease-related plasticity and memory deficits. Nat. Neurosci. 16, 1299–1305. doi: 10.1038/nn.3486

PubMed Abstract | CrossRef Full Text | Google Scholar

Marchal, J. A., Lopez, G. J., Peran, M., Comino, A., Delgado, J. R., Garcia-Garcia, J. A., et al. (2014). The impact of PKR activation: from neurodegeneration to cancer. FASEB J. 28, 1965–1974. doi: 10.1096/fj.13-248294

PubMed Abstract | CrossRef Full Text | Google Scholar

Memedovski, Z., Czerwonka, E., Han, J., Mayer, J., Luce, M., Klemm, L. C., et al. (2020). Classical and alternative activation of rat microglia treated with ultrapure Porphyromonas gingivalis lipopolysaccharide in vitro. Toxins 12:333. doi: 10.3390/toxins12050333

PubMed Abstract | CrossRef Full Text | Google Scholar

Mouton-Liger, F., Rebillat, A.-S., Gourmaud, S., Paquet, C., Leguen, A., and Dumurgier, J. (2015). PKR downregulation prevents neurodegeneration and β-amyloid production in a thiamine-deficient model. Cell Death Dis. 6:e1594. doi: 10.1038/cddis.2014.552

PubMed Abstract | CrossRef Full Text | Google Scholar

Nakanishi, H., Nonaka, S., and Wu, Z. (2020). Microglial cathepsin B and Porphyromonas gingivalis gingipains as potential therapeutic targets for sporadic Alzheimer’s disease. CNS Neurol. Disord. Drug Targets 19, 495–502. doi: 10.2174/1871527319666200708125130

PubMed Abstract | CrossRef Full Text | Google Scholar

Nie, R., Wu, Z., Ni, J., Zeng, F., Yu, W., Zhang, Y., et al. (2019). Porphyromonas gingivalis infection induces amyloid-β accumulation in monocytes/macrophages. J. Alzheimers Dis. 72, 479–494. doi: 10.3233/JAD-190298

PubMed Abstract | CrossRef Full Text | Google Scholar

Nonaka, S., and Nakanishi, H. (2020). Secreted gingipains from Porphyromonas gingivalis induce microglia migration through endosomal signaling by protease-activated receptor 2. Neurochem. Int. 140:104840. doi: 10.1016/j.neuint.2020.104840

PubMed Abstract | CrossRef Full Text | Google Scholar

Olsen, I. (2021). Porphyromonas gingivalis may seek the Alzheimer’s disease brain to acquire iron from its surplus. J. Alzheimer’s Dis. Rep. 5, 79–86. doi: 10.3233/ADR-200272

PubMed Abstract | CrossRef Full Text | Google Scholar

Olsen, I., and Singhrao, S. K. (2016). Inflammasome involvement in Alzheimer’s disease. J. Alzheimers Dis. 54, 45–53. doi: 10.3233/JAD-160197

PubMed Abstract | CrossRef Full Text | Google Scholar

Olsen, I., and Singhrao, S. K. (2020). Interaction between genetic factors, Porphyromonas gingivalis and microglia to promote Alzheimer’s disease. J. Oral Microbiol. 12:1820834. doi: 10.1080/20002297.2020.1820834

PubMed Abstract | CrossRef Full Text | Google Scholar

Olsen, I., and Yilmaz, Ö (2016). Modulation of inflammasome activity by Porphyromonas gingivalis in periodontitis and associated systemic diseases. J. Oral Microbiol. 8:30385. doi: 10.3402/jom.v8.30385

PubMed Abstract | CrossRef Full Text | Google Scholar

Paquet, C., Mouton-Liger, F., Meurs, E. F., Mazot, P., Bouras, C., Pradier, L., et al. (2012). The PKR activator PACT is induced by Aβ: involvement in Alzheimer’s disease. Brain Pathol. 22, 219–229. doi: 10.1111/j.1750-3639.2011.00520.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Peel, A. L., Rao, R. V., Cottrell, B. A., Hayden, M. R., Ellerby, L. M., and Bredesen, D. E. (2001). Double-stranded RNA-dependent protein kinase, PKR, binds preferentially to Huntington’s disease (HD) transcripts and is activated in HD tissue. Hum. Mol. Genet. 10, 1531–1538. doi: 10.1093/hmg/10.15.1531

PubMed Abstract | CrossRef Full Text | Google Scholar

Poole, S., Singhrao, S. K., Kesavalu, L., Curtis, M. A., and Crean, S. (2013). Determining the presence of periodontopathic virulence factors in short-term postmortem Alzheimer’s disease brain tissue. J. Alzheimers Dis. 36, 665–677. doi: 10.3233/JAD-121918

PubMed Abstract | CrossRef Full Text | Google Scholar

Reimer, L., Betzer, C., Kofoed, R. H., Volbracht, C., Fog, K., Kurhade, C., et al. (2021). PKR kinase directly regulates tau expression and Alzheimer’s disease-related tau phosphorylation. Brain Pathol. 31, 103–119. doi: 10.1111/bpa.12883

PubMed Abstract | CrossRef Full Text | Google Scholar

Sandhu, P., Naeem, M. M., Lu, C., Kumarathasan, P., Gomes, J., and Basak, A. (2017). Ser422 phosphorylation blocks human Tau cleavage by caspase-3: Biochemical implications to Alzheimer’s disease. Bioorg. Med. Chem. Lett. 27, 642–652. doi: 10.1016/j.bmcl.2016.11.08

CrossRef Full Text | Google Scholar

Segev, Y., Barrera, I., Ounallah-Saad, H., Wibrand, K., Sporild, I., and Livne, A. (2015). PKR inhibition rescues memory deficit and ATF4 overexpression in ApoE ε4 human replacement mice. J. Neurosci. 35, 12986–12993. doi: 10.1523/JNEUROSCI.5241-14.2015

PubMed Abstract | CrossRef Full Text | Google Scholar

Shi, C.-J., Peng, W., Zhao, J.-H., Yang, H.-L., Qu, L.-L., and Wang, C. (2020). Usnic acid derivatives as tau-aggregation and neuroinflammation inhibitors. Eur. J. Med. Chem. 187:111961. doi: 10.1016/j.ejmech.2019.111961

PubMed Abstract | CrossRef Full Text | Google Scholar

Socransky, S. S., Haffajee, A. D., Cugini, M. A., Smith, C., and Kent, R. L. Jr. (1998). Microbial complexes in subgingival plaque. J. Clin. Periodontol. 25, 134–144. doi: 10.1111/j.1600-051x.1998.tb02419.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Stancu, I.-C., Cremers, N., Vanrusselt, H., Couturier, J., Vanoosthuyse, A., Kessels, S., et al. (2019). Aggregated tau activates NLRP3–ASC inflammasome exacerbating exogenously seeded and non-exogenously seeded tau pathology in vivo. Acta Neuropathol. 137, 599–617. doi: 10.1007/s00401-018-01957-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Tang, Z., Liang, D., Cheng, M., Su, X., Liu, R., Zhang, Y., et al. (2021). Effects of Porphyromonas gingivalis and its underlying mechanisms on Alzheimer-like tau hyperphosphorylation in Sprague-Dawley rats. J. Mol. Neurosci. 71, 89–100. doi: 10.1007/s12031-020-01629-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Urnowey, S., Ansai, T., Bitko, V., Nakayam, K., Takehara, T., and Barik, S. (2006). Temporal activation of anti- and pro-apoptotic factors in human gingival fibroblasts infected with the periodontal pathogen, Porphyromonas gingivalis: potential role of bacterial proteases in host signalling. BMC Microbiol. 6:26. doi: 10.1186/1471-2180-6-26

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, Z., Ni, J., Liu, Y., Teeling, J. L., Takayama, F., Collcutt, A., et al. (2017). Cathepsin B plays a critical role in inducing Alzheimer’s disease-like phenotypes following chronic systemic exposure to lipopolysaccharide from Porphyromonas gingivalis in mice. Brain Behav. Immun. 65, 350–361. doi: 10.1016/j.bbi.2017.06.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Yoshiyama, Y., Higuchi, M., Zhang, B., Huang, S.-M., Iwata, N., Saido, T. C., et al. (2007). Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron 53, 337–351. doi: 10.1016/j.neuron.2007.01.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, J., Yu, C., Zhang, X., Chen, H., Dong, J., Lu, W., et al. (2018). Porphyromonas gingivalis lipopolysaccharide induces cognitive dysfunction, mediated by neuronal inflammation via activation of the TLR4 signaling pathway in C57BL/6 mice. J Neuroinflammation 15:37. doi: 10.1186/s12974-017-1052-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: amyloid-beta, tau, microglia, cathepsin B, protein kinase R, lipopolysaccharide, gingipains

Citation: Olsen I (2021) Porphyromonas gingivalis-Induced Neuroinflammation in Alzheimer’s Disease. Front. Neurosci. 15:691016. doi: 10.3389/fnins.2021.691016

Received: 14 April 2021; Accepted: 27 August 2021;
Published: 14 October 2021.

Edited by:

Tibor Hortobágyi, University of Szeged, Hungary

Reviewed by:

Erin Grace Reed-Geaghan, Northeast Ohio Medical University, United States
Istvan Varga, University of Debrecen, Hungary

Copyright © 2021 Olsen. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Ingar Olsen, ingar.olsen@odont.uio.no

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.