Acessibilidade / Reportar erro

Enterovirus infection and its relationship with neurodegenerative diseases

Abstract

Neurodegenerative diseases (NDs) are increasingly common, especially in populations with higher life expectancies. They are associated mainly with protein metabolism and structure changes, leading to neuronal cell death. Viral infections affect these cellular processes and may be involved in the etiology of several neurological illnesses, particularly NDs. Enteroviruses (EVs) frequently infect the central nervous system (CNS), causing neurological disease. Inflammation, disruption of the host autophagy machinery, and deregulation and accumulation/misfolding of proteins are the main alterations observed after infection by an EV. In this perspective, we discuss the most recent findings on the subject, examining the possible role of EVs in the development of NDs, and shedding light on the putative role played by these viruses in developing NDs.

Key words:
enterovirus; neurodegenerative diseases; amyotrophic lateral sclerosis; Alzheimer’s disease; Parkinson’s disease


Neurodegenerative diseases (NDs) are highly debilitating illnesses and the most common causes of severe and fatal dementia. Tauopathies, synucleinopathies, and TDP-43 proteinopathies are the most prevalent NDs and can be categorised primarily by clinical traits and the anatomic distribution of neurodegeneration (Table).11. Dugger BN, Dickson DW. Pathology of neurodegenerative diseases. Cold Spring Harb Perspect Biol. 2017; 9(7): a028035. These disorders have pathological hallmarks, including the loss of specific subset neurons in certain brain regions and damage to network synaptic connections that lead to a decline in brain function. Furthermore, recent studies indicate that neuroinflammation is critical for ND pathogenesis and progression.22. Guzman-Martinez L, Maccioni RB, Andrade V, Navarrete LP, Pastor MG, Ramos-Escobar N. Neuroinflammation as a common feature of neurodegenerative disorders. Front Pharmacol. 2019; 12(10): 1008.,33. Chen WW, Zhang X, Huang WJ. Role of neuroinflammation in neurodegenerative diseases (Review). Mol Med Rep. 2016; 13(4): 3391-6. Another one of the hallmarks of NDs is the increase in intra- and extracellular amyloid deposits. Amyloid deposits are well-ordered aggregates of misfolded proteins that spontaneously co-assemble into oligomers and reorganise into beta-sheet-rich fibrils.44. Soto C, Pritzkow S. Protein misfolding, aggregation, and conformational strains in neurodegenerative diseases. Nat Neurosci. 2018; 21(10): 1332-40. Additionally, oxidative stress, programmed cell death, and proteotoxic stress associated with changes in the ubiquitin-proteasomal and autophagosomal/lysosomal systems significantly affect the progression of NDs.

Most NDs occur sporadically and are considered multifactorial diseases associated with aging and a complex interaction of epigenetic factors, such as genetics, biological networks (relationships between humans and other organisms), development, and environmental issues.55. Migliore L, Coppedè F. Genetics, environmental factors and the emerging role of epigenetics in neurodegenerative diseases. Mutat Res. 2009; 667(1-2): 82-97.,66. Onisiforou A, Spyrou GM. Identification of viral-mediated pathogenic mechanisms in neurodegenerative diseases using network-based approaches. Brief Bioinform. 2021; 22(6): bbab141. Our understanding of how these factors contribute to the development of NDs needs to improve. Numerous studies on NDs and pathogenic organisms have been conducted in recent years, and viruses have mostly been reported as risk factors for the development of neurodegeneration and dementia.77. Abbott A. Are infections seeding some cases of Alzheimer's disease? Nature. 2020; 587(7832): 22-5.,88. Lotz SK, Blackhurst BM, Reagin KL, Funk KE. Microbial infections are a risk factor for neurodegenerative diseases. Front Cell Neurosci. 2021; 7(15): 691136.,99. Wouk J, Rechenchoski DZ, Rodrigues BCD, Ribelato EV, Faccin-Galhardi LC. Viral infections and their relationship to neurological disorders. Arch Virol. 2021; 166(3): 733-53.,1010. Leblanc P, Vorberg IM. Viruses in neurodegenerative diseases: more than just suspects in crimes. PLoS Pathog. 2022; 4; 18(8): e1010670. Additionally, emerging evidence suggests that alterations induced in the gut microbiome may play a significant role in the development of NDs. For instance, many viral infections can cause a microbiome-gut-brain axis imbalance (dysbiosis) and are associated with neurodegeneration either directly (through viral infection on neural cells) or indirectly [as the viral infection may trigger a systemic inflammation by increasing gut permeability, which may increase levels of circulating lipopolysaccharides causing the release of inflammatory cytokines in the central nervous system (CNS)].1111. Follmer C. Viral infection-induced gut dysbiosis, neuroinflammation, and a-synuclein aggregation: updates and perspectives on COVID-19 and neurodegenerative disorders. ACS Chem Neurosci. 2020; 11(24): 4012-6.,1212. Fang P, Kazmi SA, Jameson KG, Hsiao EY. The microbiome as a modifier of neurodegenerative disease risk. Cell Host Microbe. 2020; 28(2): 201-22.

TABLE
Overview of some neurodegenerative conditions

Neurodegenerative mechanisms associated with viral infection

The three main mechanisms through which viral infection of the CNS may lead to NDs are protein metabolism alterations, direct conversion and aggregation of cellular proteins by viral factors, and deleterious post-translational modifications (Fig. 1).

Fig. 1:
mechanisms leading to neurodegenerative diseases after virus infection. Virus infection can collectively trigger different alterations in cell processes through three main mechanisms: 1. It alters protein metabolism, perturbing protein synthesis and degradation (protein ubiquitination, degradation at the proteasome, and autophagy/aggrephagy). It also affects chaperon prevention/correction of protein misfolding. 2. Viruses can directly interact with proteins, leading to structural conversion, starting aggregation, and forming small toxic aggregates and amyloid fibrils. These fibrils can seed monomer aggregation, representing a self-replicating process. Different virus molecules, such as proteins, lipids, and nucleic acids, are supposed to show this property. 3. Viruses perturb post-translational protein modifications, modulating protein function and oligomerisation. For example, Tau hyperphosphorylation, associated with Alzheimer’s disease, can be triggered by a virus infection, leading to the formation of neurofibrillary tangles and cell dysfunction. This figure was designed at Biorender.com.

Cellular proteostasis results from the action of coordinated networks that maintain a dynamic balance between protein translation, folding, and degradation. Typically, cells avoid forming aggregates, or if they already exist, the cells destroy them in order to prevent any potentially harmful effects.1313. Chopra G, Shabir S, Yousuf S, Kauts S, Bhat SA, Mir AH, et al. Proteinopathies: deciphering physiology and mechanisms to develop effective therapies for neurodegenerative diseases. Mol Neurobiol. 2022; 59: 7513-40. However, preservation of proteome stability is challenging once cells are exposed to certain environmental stressors. Viral infection is one such stressor whereby the viruses hijack the cellular machinery, alter proteostasis to allow replication and then leave the cell to infect other nearby cells (Fig. 1).

Some viruses can utilise aggregate clearance to increase their replication or evade the immune response.1414. Muscolino E, Luoto LM, Brune W. Viral induced protein aggregation: a mechanism of immune evasion. Int J Mol Sci. 2021; 22(17): 9624. Heat shock proteins (HSP) such as HSP 40, HSP 70, and HSP 90 have been extensively studied in this area, and they are involved in various stages of viral infection.1515. Zhang X, Yu W. Heat shock proteins and viral infection. Front Immunol. 2022; 13: 947789. HSPs play a vital role in the lifecycle of proteins, including protein folding and refolding, transport, degradation, assembly, activity regulation, and translocation, as well as the depolymerisation of protein aggregates.1515. Zhang X, Yu W. Heat shock proteins and viral infection. Front Immunol. 2022; 13: 947789. The interaction of viral proteins with HSPs can change the apoptosis pathways in which they are involved, making it easier to activate caspase cascades and induce apoptosis to release viruses while evading the immune system.

Additionally, viruses alter the proteasome system to prevent the ubiquitination of viral proteins, leading to their accumulation in the cell.1616. Valerdi KM, Hage A, van Tol S, Rajsbaum R, Giraldo MI. The role of the host ubiquitin system in promoting replication of emergent viruses. Viruses. 2021; 13(3): 369. This scenario, therefore, leads to the accumulation of cellular proteins that also induce their degradation, deposition, and toxicity, rendering cells more vulnerable to protein misfolding. Additionally, antiviral cell responses involving the release of pro-inflammatory cytokines and chemokines can lead to protein misfolding and aggregation, potentially accelerating the onset of genetic forms of NDs and exacerbating preexisting abnormalities.1717. Shelkovnikova TA, An H, Skelt L, Tregoning JS, Humphreys IR, Buchman VL. Antiviral immune response as a trigger of FUS proteinopathy in amyotrophic lateral sclerosis. Cell Rep. 2019; 29(13): 4496-508e4.

Viral proteins can interact with various biomolecules inside the cell, directly altering the activity of some proteins and affecting their solubility and stability. In the context of NDs, for example, HIV-Tat (HIV trans-activator of transcription protein) interacts with Aβ peptide in vitro, shaping its fibril structure and increasing its toxicity to hippocampal neuron cultures.1818. Hategan A, Bianchet MA, Steiner J, Karnaukhova E, Masliah E, Fields A, et al. HIV Tat protein and amyloid-ß peptide form multifibrillar structures that cause neurotoxicity. Nat Struct Mol Biol. 2017; 24(4): 379-86. Glycoprotein B of the herpes simplex virus (HSV-1) can seed Aβ aggregation, accelerating fibril formation in vitro, showing toxicity to neuronal cultures,1919. Cribbs DH, Azizeh BY, Cotman CW, LaFerla FM. Fibril formation and neurotoxicity by a herpes simplex virus glycoprotein B fragment with homology to the Alzheimer's A beta peptide. Biochemistry. 2000; 39(20): 5988-94. and could exacerbate neurodegeneration in vivo.

Neurotropic influenza A virus induces the conversion of the cellular prion protein (PrPC) into the infectious scrapie prion (PrPSc), and subsequent aggregation, when infecting neuroblastoma cells.2020. Hara H, Chida J, Uchiyama K, Pasiana AD, Takahashi E, Kido H, et al. Neurotropic influenza A virus infection causes prion protein misfolding into infectious prions in neuroblastoma cells. Sci Rep. 2021; 11(1): 10109. Both nucleic acids (NA) and lipids are also known to induce protein aggregation.2121. Angelli JN, Passos YM, Brito JMA, Silva JL, Cordeiro Y, Vieira TCRG. Rabbit PrP is partially resistant to in vitro aggregation induced by different biological cofactors. Front Neurosci. 2021; 15: 689315.,2222. Silva JL, Cordeiro Y. The "Jekyll and Hyde" actions of nucleic acids on the prion-like aggregation of proteins. J Biol Chem. 2016; 291(30): 15482-90.,2323. Tetz G, Pinho M, Pritzkow S, Mendez N, Soto C, Tetz V. Bacterial DNA promotes Tau aggregation. Sci Rep. 2020; 10(1): 2369.,2424. Tetz G, Tetz V. Bacterial extracellular DNA promotes ß-amyloid aggregation. microorganisms. 2021; 9(6): 1301. In this context, exogenous NAs are strong candidate inducers that should be considered. Thus, cells may become more prone to protein misfolding due to the interaction of viral factors with host amyloidogenic proteins (Fig. 1). Even if only transient, this interaction may play an important triggering role since these proteins self-perpetuate when aggregated.2525. Riek R, Eisenberg DS. The activities of amyloids from a structural perspective. Nature. 2016; 539(7628): 227-35.

Post-translational modification (PTM) is a well-characterised cell strategy to modulate protein function. Nonnative PTM as tau hyperphosphorylation is known to affect its conformation leading to neurofibrillary tangle formation and cell damage (Fig. 1).2626. De-Paula VJ, Radanovic M, Diniz B, Forlenza O. Alzheimer's disease. In: Harris J, editor. Protein aggregation and fibrillogenesis in cerebral and systemic amyloid disease. Subcellular Biochemistry. Vol 65. Dordrecht: Springer; 2012. HSV-1 infection up-regulates the kinases responsible for Tau phosphorylation (GSK3β-glycogen synthase kinase-3β and PKA-protein kinase A) in human neuroblastoma-infected cells,2727. Wozniak MA, Frost AL, Itzhaki RF. Alzheimer's disease-specific tau phosphorylation is induced by herpes simplex virus type 1. J Alzheimers Dis. 2009; 16: 341-50. leading to hyperphosphorylation and neuronal loss in primary cell culture.2828. Zambrano A, Solis L, Salvadores N, Cortés M, Lerchundi R, Otth C. Neuronal cytoskeletal dynamic modification and neurodegeneration induced by infection with herpes simplex virus type 1. J Alzheimers Dis. 2008; 14(3): 259-69. Treatment with acyclovir, which inhibits HSV-1 replication, decreased Tau phosphorylation in cells.2929. Wozniak MA, Frost AL, Preston CM, Itzhaki RF. Antivirals reduce the formation of key Alzheimer's disease molecules in cell cultures acutely infected with herpes simplex virus type 1. PLoS One. 2011; 6(10): e25152. HIV-tat also increases Tau phosphorylation in mice.3030. Giunta B, Hou H, Zhu Y, Rrapo E, Tian J, Takashi M, et al. HIV-1 Tat contributes to Alzheimer's disease-like pathology in PSAPP mice. Int J Clin Exp Pathol. 2009; 2(5): 433-43. Therefore, viral infections can modulate PTMs, thus enhancing neurodegeneration.

It is worth noting that the accumulation and abnormal location of toxic aggregated proteins is one of the clinical hallmarks of neurodegenerative illnesses like Alzheimer’s disease (AD), amyotrophic lateral sclerosis (ALS), and Parkinson’s disease (PD). In this perspective, we present a critical view of the relationship between enterovirus infection and neurodegenerative disorders, shedding light on the putative role played by these viruses in developing NDs.

Enteroviruses

Globally, enteroviruses (EVs) are one of the major causes of human viral infections. EVs are small, icosahedral-shaped, non-enveloped, positive-sense, single-stranded RNA viruses with a capsid composed of four structural proteins (VP1-VP4) belonging to the genus Enterovirus and Picornaviridae family. EVs have been classified into fifteen species: EV A-L and human rhinoviruses A-C. However, only EV A to D species and rhinoviruses are known to cause human infection. Due to recurrent mutations and/or recombination, these viruses exhibit a high level of genomic diversity.3131. Muslin C, Mac Kain A, Bessaud M, Blondel B, Delpeyroux F. Recombination in enteroviruses, a multi-step modular evolutionary process. Viruses. 2019; 11(9): 859. While most EVs are transmitted primarily by the faecal-oral route and can advance from the primary site of infection (gastrointestinal tract) to other tissues, some EV-types (e.g., rhinoviruses and EV-D68) can cause respiratory infection and spread via respiratory secretion. Although EV infections in most patients are often mild and self-limiting, these viruses can cause severe CNS illnesses, particularly aseptic meningitis (AM), acute flaccid paralysis (AFP), encephalitis, and acute flaccid myelitis (AFM), which can be fatal. Enteroviruses are the most common cause of aseptic meningitis worldwide. Most of the existing CNS infections associated with EVs have been caused by EV-A71, CVA2, and CVA4 (EV-A species), E6, E11, E30, CVB5 (EV-B species), EV-C99 and CVA24 (EV-C species) and EV-D68 (EV-D species).3232. Sousa Jr IP, Dos Santos FB, de Paula VS, Vieira TCRG, Dias HG, Barros CA, et al. Viral and prion infections associated with central nervous system syndromes in Brazil. Viruses. 2021; 13(7): 1370.,3333. Suresh S, Rawlinson WD, Andrews PI, Stelzer-Braid S. Global epidemiology of nonpolio enteroviruses causing severe neurological complications: a systematic review and meta-analysis. Rev Med Virol. 2020; 30(1): e2082.

EVs are able to enter into the CNS through a mechanism known as “Trojan Horse” or via neuromuscular junctions using retrograde axonal transport.3232. Sousa Jr IP, Dos Santos FB, de Paula VS, Vieira TCRG, Dias HG, Barros CA, et al. Viral and prion infections associated with central nervous system syndromes in Brazil. Viruses. 2021; 13(7): 1370.,3434. Majer A, McGreevy A, Booth TF. Molecular pathogenicity of enteroviruses causing neurological disease. Front Microbiol. 2020; 11: 540. The pathways used by EVs to bypass the blood-brain barrier (BBB) and enter the CNS, the mechanisms of neuronal cell death, and the neuropathogenesis and immunology triggered by EVs during CNS infection have been recently reviewed.3434. Majer A, McGreevy A, Booth TF. Molecular pathogenicity of enteroviruses causing neurological disease. Front Microbiol. 2020; 11: 540.,3535. Chen BS, Lee HC, Lee KM, Gong YN, Shih SR. Enterovirus and encephalitis. Front Microbiol. 2020; 11: 261.

The role of enterovirus infection in neurodegenerative diseases

Over the past decades, infections caused by numerous infectious agents have been associated with the pathogenesis of NDs, although this idea is still controversial.3636. Piekut T, Hurla M, Banaszek N, Szejn P, Dorszewska J, Kozubski W, et al. Infectious agents and Alzheimer's disease. J Integr Neurosci. 2022; 21(2): 73. To date, little is known about the potential contribution of enteroviruses in the development of NDs through neuroinflammatory pathways or aggravating the pathogenic alterations that were not necessarily caused by a viral infection, favoring disease progression. EV infection leads to an alteration of the metabolism of the proteins associated with these pathways.

Some studies have highlighted the potential relationship between neurodegeneration induction and AD through Toll-like receptors (TLRs) activation signaling in enterovirus infections. TLRs are a group of receptors that are extensively expressed in several cell types, including immunological, intestinal, lung, and neural cells. They have been linked to inflammatory responses and pathological conditions, including neurodegeneration.3737. Luo Z, Su R, Wang W, Liang Y, Zeng X, Shereen MA, et al. EV71 infection induces neurodegeneration via activating TLR7 signaling and IL-6 production. PLoS Pathog. 2019; 15(11): e1008142.,3838. Fiebich BL, Batista CRA, Saliba SW, Yousif NM, de Oliveira ACP. Role of microglia TLRs in neurodegeneration. Front Cell Neurosci. 2018; 12: 329. Additionally, unfolded protein response (UPR) and oxidative stress are two factors that are linked to TLRs and present in different NDs.3838. Fiebich BL, Batista CRA, Saliba SW, Yousif NM, de Oliveira ACP. Role of microglia TLRs in neurodegeneration. Front Cell Neurosci. 2018; 12: 329.,3939. García-González P, Cabral-Miranda F, Hetz C, Osorio F. Interplay between the unfolded protein response and immune function in the development of neurodegenerative diseases. Front Immunol. 2018; 9: 2541. Recently, Luo and coworkers suggested that upon EV-A71 infection, neural cells can trigger the pathogenesis in the brain (mice and human) through the TLR7 pathway inducing neurodegeneration.3737. Luo Z, Su R, Wang W, Liang Y, Zeng X, Shereen MA, et al. EV71 infection induces neurodegeneration via activating TLR7 signaling and IL-6 production. PLoS Pathog. 2019; 15(11): e1008142. Another study found that EV-A71 infection can cause PRSS3 retention in human neuroblastoma SH-SY5Y cells. This essential serine protease acts as a signaling agent and is secreted and transported via the cellular secretory pathway, playing a critical role in the CNS.4040. Rattanakomol P, Srimanote P, Tongtawe P, Khantisitthiporn O, Supasorn O, Thanongsaksrikul J. Host neuronal PRSS3 interacts with enterovirus A71 3A protein and its role in viral replication. Sci Rep. 2022; 12(1): 12846. PRSS3 accumulation may involve the gain of cytotoxic function. Indeed, enteroviruses infection disrupts the host autophagy machinery at different steps and remodels the secretory pathways inside the cells.3434. Majer A, McGreevy A, Booth TF. Molecular pathogenicity of enteroviruses causing neurological disease. Front Microbiol. 2020; 11: 540.,4141. Mohamud Y, Luo H. The intertwined life cycles of enterovirus and autophagy. Virulence. 2019; 10(1): 470-80. EV-induced autophagy dysregulation may directly harm cells due to the loss of the protection conferred by autophagy. It is important to highlight the pathogenic roles associated with autophagy impairment in several NDs.4242. Nixon RA. The role of autophagy in neurodegenerative disease. Nat Med. 2013; 19(8): 983-97.

Notably, when infected by EV-A71, neural cells from the mouse brain suffered a depolarisation of aquaporin-4 (AQP4).4343. Wang H, Yuan M, Yang E, Chen D, Su A, Wu Z. Enterovirus 71 infection induced Aquaporin-4 depolarization by increasing matrix metalloproteinase-9 activity. Neurosci Lett. 2021; 759: 136049. In addition to controlling body water balance and water flow in and out of the brain parenchyma, APQ4 may affect several pathways, and decreasing its expression has been closely associated with the development of AD.4444. Tamtaji OR, Behnam M, Pourattar MA, Jafarpour H, Asemi Z. Aquaporin 4: a key player in Parkinson's disease. J Cell Physiol. 2019; 234(12): 21471-8. The changes brought about by EV-A71 infection on APQ4 may therefore disrupt its ability to clear out beta-amyloid and tau proteins, the accumulation of which is a hallmark of AD and can start years before the disease’s onset.44. Soto C, Pritzkow S. Protein misfolding, aggregation, and conformational strains in neurodegenerative diseases. Nat Neurosci. 2018; 21(10): 1332-40.,4343. Wang H, Yuan M, Yang E, Chen D, Su A, Wu Z. Enterovirus 71 infection induced Aquaporin-4 depolarization by increasing matrix metalloproteinase-9 activity. Neurosci Lett. 2021; 759: 136049.

Interestingly, recent findings have revealed Aβ to be an antimicrobial peptide, and it has been hypothesised that Aβ deposition may represent an effective innate immune reaction to infection.4545. Fulop T, Witkowski JM, Bourgade K, Khalil A, Zerif E, Larbi A, et al. Can an infection hypothesis explain the beta amyloid hypothesis of Alzheimer's disease? Front Aging Neurosci. 2018; 10: 224. Thus, Aβ production may initially be beneficial for defending against microorganisms, but it will become progressively more harmful as the infection becomes chronic or reactivated over time. Regarding the activity of Aβ on EV infection, a recent study has demonstrated that Aβ1-42 effectively inhibited EV-A71 in the early stages (attachment and uncoating) of the virus cycle in different cell lines, including neural cells.4646. Zhong M, Wang H, Yan H, Wu S, Wang K, Yang L, et al. Effects and mechanism of Aß1-42 on EV-A71 replication. Virol J. 2022; 19(1): 151. Additionally, EV-A71 infection also induced Aβ production and accumulation in SH-SY5Y cells.4646. Zhong M, Wang H, Yan H, Wu S, Wang K, Yang L, et al. Effects and mechanism of Aß1-42 on EV-A71 replication. Virol J. 2022; 19(1): 151.

Several viruses (including enteroviruses) are well-known to be associated with both acute and chronic viral parkinsonism, even though none have been found in Lewy body deposition on examination of post-mortem brain tissues; instead, they mimic the signs and symptoms of the disease.4747. Jang H, Boltz DA, Webster RG, Smeyne RJ. Viral parkinsonism. Biochim Biophys Acta. 2009; 1792(7): 714-21. Nevertheless, a few studies have demonstrated that people with a history of poliovirus infection may show an increased risk of developing PD over time.4848. Nielsen NM, Rostgaard K, Hjalgrim H, Aaby P, Askgaard D. Poliomyelitis and Parkinson disease. JAMA. 2002; 287(13): 1650-1.,4949. Kang JH, Lin HC. Comorbidity profile of poliomyelitis survivors in a Chinese population: a population-based study. J Neurol. 2011; 258(6): 1026-33. Also, in the brainstem neurons of PD patients, virus-like particles and enteroviruses (poliovirus and coxsackievirus) antigens have been identified.5050. Dourmashkin RR, McCall SA, Dourmashkin N, Hannah MJ. Virus-like particles and enterovirus antigen found in the brainstem neurons of Parkinson's disease. F1000Res. 2018; 7: 302. In addition to the direct neuronal cytopathogenic effects of viral infection, these findings suggest that enterovirus infection in PD could directly or indirectly alter the metabolism of α-synuclein. Indeed, a recent study found that α-synuclein is up-regulated and associated with α-synuclein inclusion body formation and dysfunctional autophagy machinery in neurons during coxsackievirus B3 (CVB3) infection.5151. Park SJ, Jin U, Park SM. Interaction between coxsackievirus B3 infection and a-synuclein in models of Parkinson's disease. PLoS Pathog. 2021; 17(10): e1010018. Additionally, the authors observed α-synuclein aggregates in the cell body of midbrain neurons of mice infected with CVB3.5151. Park SJ, Jin U, Park SM. Interaction between coxsackievirus B3 infection and a-synuclein in models of Parkinson's disease. PLoS Pathog. 2021; 17(10): e1010018. These modifications might act as triggers for PD. Fig. 2 shows the potential effect of enterovirus infections in neural cells and its impact on developing NDs.

Fig. 2:
the potential effect of enterovirus (EV) infections in neural cells and its impact on developing neurodegenerative diseases (NDs). (A) After gaining access to the central nervous system (CNS), EV infects neural cells, leading to different alterations in cell processes and decreasing the number of neurons. (B) EV infection could stimulate neural cells to produce amyloid proteins as defense mechanisms, wrapping and neutralising the virus. These amyloid proteins constitute the amyloid plaques, which activate microglia releasing pro-inflammatory cytokines. (C) The blood-brain barrier (BBB) can be disturbed from outside the brain via peripheral inflammatory mediators and dysbiosis, as well as from within the brain by the mediators generated by neural cells that have been stimulated by EV infections.

ALS is a neurodegenerative disorder for which the greatest evidence exists linking it to EV infection. Numerous studies have reported EV genome detection in the brain/spinal cord and cerebrospinal fluid of ALS patients, even though the available data are controversial.5252. Xue YC, Feuer R, Cashman N, Luo H. Enteroviral infection: the forgotten link to amyotrophic lateral sclerosis? Front Mol Neurosci. 2018; 11: 63.) and references therein Additionally, EV seroprevalence is higher in ALS patients than in controls, regardless of disease progression,5353. Cermelli C, Vinceti M, Beretti F, Pietrini V, Nacci G, Pietrosemoli P, et al. Risk of sporadic amyotrophic lateral sclerosis associated with seropositivity for herpesviruses and echovirus-7. Eur J Epidemiol. 2003; 18(2): 123-7. and a high EV-detection rate is observed in spinal cord neurons and CSF of these patients.5454. Berger MM, Kopp N, Vital C, Redl B, Aymard M, Lina B. Detection and cellular localization of enterovirus RNA sequences in spinal cord of patients with ALS. Neurology. 2000; 54(1): 20-5.,5555. Vandenberghe N, Leveque N, Corcia P, Brunaud-Danel V, Salort-Campana E, Besson G, et al. Cerebrospinal fluid detection of enterovirus genome in ALS: a study of 242 patients and 354 controls. Amyotroph Lateral Scler. 2010; 11(3): 277-82. Unfortunately, most of these studies present different EV-detection rates resulting in controversial results, which were not always reproducible,5656. Walker MP, Schlaberg R, Hays AP, Bowser R, Lipkin WI. Absence of echovirus sequences in brain and spinal cord of amyotrophic lateral sclerosis patients. Ann Neurol. 2001; 49(2): 249-53.,5757. Nix WA, Berger MM, Oberste MS, Brooks BR, McKenna-Yasek DM, Brown Jr RH, et al. Failure to detect enterovirus in the spinal cord of ALS patients using a sensitive RT-PCR method. Neurology. 2004; 62(8): 1372-7. likely due to the use of fresh versus archived tissues, the nature of the sample, and methodological differences. However, a recent study found that sublethal infection (mimicking a persistent infection) with CVB3 in ALS genetically susceptible mice resulted in early onset, increased motor impairment, and decreased lifespan.5858. Xue YC, Liu H, Mohamud Y, Bahreyni A, Zhang J, Cashman NR, et al. Sublethal enteroviral infection exacerbates disease progression in an ALS mouse model. J Neuroinflammation. 2022; 19(1): 16. In addition, pro-inflammatory cytokine/chemokine gene expression increased significantly, independent of immune cell infiltration in the CNS of mice infected with CVB3.5858. Xue YC, Liu H, Mohamud Y, Bahreyni A, Zhang J, Cashman NR, et al. Sublethal enteroviral infection exacerbates disease progression in an ALS mouse model. J Neuroinflammation. 2022; 19(1): 16. The authors reported that all these modifications induced ALS-related pathologies in the CNS of mice. Interestingly, upon CVB3 infection, TDP-43, a major pathological hallmark in sporadic ALS, is also translocated from the nucleus to the cytoplasm (it is predominantly located in the nucleus), in addition to its decreased solubility and increased cytoplasmic accumulation.5959. Fung G, Shi J, Deng H, Hou J, Wang C, Hong A, et al. Cytoplasmic translocation, aggregation, and cleavage of TDP-43 by enteroviral proteases modulate viral pathogenesis. Cell Death Differ. 2015; 22(12): 2087-97. This translocation is observed in patients who have ALS.6060. Lee EB, Lee VM, Trojanowski JQ. Gains or losses: molecular mechanisms of TDP43-mediated neurodegeneration. Nat Rev Neurosci. 2011; 13(1): 38-50.

In conclusion

Although EVs are thought to be highly lytic viruses that typically cause acute infections, investigations have shown that EVs can establish a persistent infection in many tissues over time, including neural cells, and can be reactivated either spontaneously or in response to external stimulations.6161. Zhang X, Zheng Z, Shu B, Liu X, Zhang Z, Liu Y, et al. Human astrocytic cells support persistent coxsackievirus B3 infection. J Virol. 2013; 87(22): 12407-21.,6262. Feuer R, Ruller CM, An N, Tabor-Godwin JM, Rhoades RE, Maciejewski S, et al. Viral persistence and chronic immunopathology in the adult central nervous system following Coxsackievirus infection during the neonatal period. J Virol. 2009; 83(18): 9356-69. Furthermore, chronic EV infection can produce pro-inflammatory cytokines and activate inflammatory reactions in the CNS. These findings imply that a risk factor or cause of NDs may be chronic EV infection.

Overall, although it is widely recognised that EV infections may cause CNS diseases, their involvement in the development of neurodegeneration and dementia, including the mechanisms involved, is not yet fully understood. The recent findings significantly impact most aspects and may suggest an association between the development of NDs and enterovirus infection, although this must be interpreted cautiously. The changes observed during EV infections can lead to a decrease in the number of neurons. The virus-induced damage may exacerbate the effects of typical aging-related neuronal degeneration and thus precipitate the symptoms of ND. Further clinical studies must be conducted to detect EVs in patients with NDs to establish the connection between EV infection and NDs. Causality is difficult to verify since the infection occurs long before clinical signs appear. It will also be interesting to understand the potential impact of EV infection in neural cells, including its role in protein accumulation in secretory vesicles, the disruption of the autophagy machinery, and neuroinflammation, as well as whether persistent EV infection is one of the factors involved in the development of NDs. Finally, EV infection can result in many other health-related disorders, such as diabetes and dysbiosis, which are risk factors for NDs. Therefore, it seems reasonable to investigate the potential relationship between these conditions and viral-induced neurodegeneration. Although significant progress has been made in recent years, more compelling data is still required to link NDs to EV infection.

REFERENCES

  • 1
    Dugger BN, Dickson DW. Pathology of neurodegenerative diseases. Cold Spring Harb Perspect Biol. 2017; 9(7): a028035.
  • 2
    Guzman-Martinez L, Maccioni RB, Andrade V, Navarrete LP, Pastor MG, Ramos-Escobar N. Neuroinflammation as a common feature of neurodegenerative disorders. Front Pharmacol. 2019; 12(10): 1008.
  • 3
    Chen WW, Zhang X, Huang WJ. Role of neuroinflammation in neurodegenerative diseases (Review). Mol Med Rep. 2016; 13(4): 3391-6.
  • 4
    Soto C, Pritzkow S. Protein misfolding, aggregation, and conformational strains in neurodegenerative diseases. Nat Neurosci. 2018; 21(10): 1332-40.
  • 5
    Migliore L, Coppedè F. Genetics, environmental factors and the emerging role of epigenetics in neurodegenerative diseases. Mutat Res. 2009; 667(1-2): 82-97.
  • 6
    Onisiforou A, Spyrou GM. Identification of viral-mediated pathogenic mechanisms in neurodegenerative diseases using network-based approaches. Brief Bioinform. 2021; 22(6): bbab141.
  • 7
    Abbott A. Are infections seeding some cases of Alzheimer's disease? Nature. 2020; 587(7832): 22-5.
  • 8
    Lotz SK, Blackhurst BM, Reagin KL, Funk KE. Microbial infections are a risk factor for neurodegenerative diseases. Front Cell Neurosci. 2021; 7(15): 691136.
  • 9
    Wouk J, Rechenchoski DZ, Rodrigues BCD, Ribelato EV, Faccin-Galhardi LC. Viral infections and their relationship to neurological disorders. Arch Virol. 2021; 166(3): 733-53.
  • 10
    Leblanc P, Vorberg IM. Viruses in neurodegenerative diseases: more than just suspects in crimes. PLoS Pathog. 2022; 4; 18(8): e1010670.
  • 11
    Follmer C. Viral infection-induced gut dysbiosis, neuroinflammation, and a-synuclein aggregation: updates and perspectives on COVID-19 and neurodegenerative disorders. ACS Chem Neurosci. 2020; 11(24): 4012-6.
  • 12
    Fang P, Kazmi SA, Jameson KG, Hsiao EY. The microbiome as a modifier of neurodegenerative disease risk. Cell Host Microbe. 2020; 28(2): 201-22.
  • 13
    Chopra G, Shabir S, Yousuf S, Kauts S, Bhat SA, Mir AH, et al. Proteinopathies: deciphering physiology and mechanisms to develop effective therapies for neurodegenerative diseases. Mol Neurobiol. 2022; 59: 7513-40.
  • 14
    Muscolino E, Luoto LM, Brune W. Viral induced protein aggregation: a mechanism of immune evasion. Int J Mol Sci. 2021; 22(17): 9624.
  • 15
    Zhang X, Yu W. Heat shock proteins and viral infection. Front Immunol. 2022; 13: 947789.
  • 16
    Valerdi KM, Hage A, van Tol S, Rajsbaum R, Giraldo MI. The role of the host ubiquitin system in promoting replication of emergent viruses. Viruses. 2021; 13(3): 369.
  • 17
    Shelkovnikova TA, An H, Skelt L, Tregoning JS, Humphreys IR, Buchman VL. Antiviral immune response as a trigger of FUS proteinopathy in amyotrophic lateral sclerosis. Cell Rep. 2019; 29(13): 4496-508e4.
  • 18
    Hategan A, Bianchet MA, Steiner J, Karnaukhova E, Masliah E, Fields A, et al. HIV Tat protein and amyloid-ß peptide form multifibrillar structures that cause neurotoxicity. Nat Struct Mol Biol. 2017; 24(4): 379-86.
  • 19
    Cribbs DH, Azizeh BY, Cotman CW, LaFerla FM. Fibril formation and neurotoxicity by a herpes simplex virus glycoprotein B fragment with homology to the Alzheimer's A beta peptide. Biochemistry. 2000; 39(20): 5988-94.
  • 20
    Hara H, Chida J, Uchiyama K, Pasiana AD, Takahashi E, Kido H, et al. Neurotropic influenza A virus infection causes prion protein misfolding into infectious prions in neuroblastoma cells. Sci Rep. 2021; 11(1): 10109.
  • 21
    Angelli JN, Passos YM, Brito JMA, Silva JL, Cordeiro Y, Vieira TCRG. Rabbit PrP is partially resistant to in vitro aggregation induced by different biological cofactors. Front Neurosci. 2021; 15: 689315.
  • 22
    Silva JL, Cordeiro Y. The "Jekyll and Hyde" actions of nucleic acids on the prion-like aggregation of proteins. J Biol Chem. 2016; 291(30): 15482-90.
  • 23
    Tetz G, Pinho M, Pritzkow S, Mendez N, Soto C, Tetz V. Bacterial DNA promotes Tau aggregation. Sci Rep. 2020; 10(1): 2369.
  • 24
    Tetz G, Tetz V. Bacterial extracellular DNA promotes ß-amyloid aggregation. microorganisms. 2021; 9(6): 1301.
  • 25
    Riek R, Eisenberg DS. The activities of amyloids from a structural perspective. Nature. 2016; 539(7628): 227-35.
  • 26
    De-Paula VJ, Radanovic M, Diniz B, Forlenza O. Alzheimer's disease. In: Harris J, editor. Protein aggregation and fibrillogenesis in cerebral and systemic amyloid disease. Subcellular Biochemistry. Vol 65. Dordrecht: Springer; 2012.
  • 27
    Wozniak MA, Frost AL, Itzhaki RF. Alzheimer's disease-specific tau phosphorylation is induced by herpes simplex virus type 1. J Alzheimers Dis. 2009; 16: 341-50.
  • 28
    Zambrano A, Solis L, Salvadores N, Cortés M, Lerchundi R, Otth C. Neuronal cytoskeletal dynamic modification and neurodegeneration induced by infection with herpes simplex virus type 1. J Alzheimers Dis. 2008; 14(3): 259-69.
  • 29
    Wozniak MA, Frost AL, Preston CM, Itzhaki RF. Antivirals reduce the formation of key Alzheimer's disease molecules in cell cultures acutely infected with herpes simplex virus type 1. PLoS One. 2011; 6(10): e25152.
  • 30
    Giunta B, Hou H, Zhu Y, Rrapo E, Tian J, Takashi M, et al. HIV-1 Tat contributes to Alzheimer's disease-like pathology in PSAPP mice. Int J Clin Exp Pathol. 2009; 2(5): 433-43.
  • 31
    Muslin C, Mac Kain A, Bessaud M, Blondel B, Delpeyroux F. Recombination in enteroviruses, a multi-step modular evolutionary process. Viruses. 2019; 11(9): 859.
  • 32
    Sousa Jr IP, Dos Santos FB, de Paula VS, Vieira TCRG, Dias HG, Barros CA, et al. Viral and prion infections associated with central nervous system syndromes in Brazil. Viruses. 2021; 13(7): 1370.
  • 33
    Suresh S, Rawlinson WD, Andrews PI, Stelzer-Braid S. Global epidemiology of nonpolio enteroviruses causing severe neurological complications: a systematic review and meta-analysis. Rev Med Virol. 2020; 30(1): e2082.
  • 34
    Majer A, McGreevy A, Booth TF. Molecular pathogenicity of enteroviruses causing neurological disease. Front Microbiol. 2020; 11: 540.
  • 35
    Chen BS, Lee HC, Lee KM, Gong YN, Shih SR. Enterovirus and encephalitis. Front Microbiol. 2020; 11: 261.
  • 36
    Piekut T, Hurla M, Banaszek N, Szejn P, Dorszewska J, Kozubski W, et al. Infectious agents and Alzheimer's disease. J Integr Neurosci. 2022; 21(2): 73.
  • 37
    Luo Z, Su R, Wang W, Liang Y, Zeng X, Shereen MA, et al. EV71 infection induces neurodegeneration via activating TLR7 signaling and IL-6 production. PLoS Pathog. 2019; 15(11): e1008142.
  • 38
    Fiebich BL, Batista CRA, Saliba SW, Yousif NM, de Oliveira ACP. Role of microglia TLRs in neurodegeneration. Front Cell Neurosci. 2018; 12: 329.
  • 39
    García-González P, Cabral-Miranda F, Hetz C, Osorio F. Interplay between the unfolded protein response and immune function in the development of neurodegenerative diseases. Front Immunol. 2018; 9: 2541.
  • 40
    Rattanakomol P, Srimanote P, Tongtawe P, Khantisitthiporn O, Supasorn O, Thanongsaksrikul J. Host neuronal PRSS3 interacts with enterovirus A71 3A protein and its role in viral replication. Sci Rep. 2022; 12(1): 12846.
  • 41
    Mohamud Y, Luo H. The intertwined life cycles of enterovirus and autophagy. Virulence. 2019; 10(1): 470-80.
  • 42
    Nixon RA. The role of autophagy in neurodegenerative disease. Nat Med. 2013; 19(8): 983-97.
  • 43
    Wang H, Yuan M, Yang E, Chen D, Su A, Wu Z. Enterovirus 71 infection induced Aquaporin-4 depolarization by increasing matrix metalloproteinase-9 activity. Neurosci Lett. 2021; 759: 136049.
  • 44
    Tamtaji OR, Behnam M, Pourattar MA, Jafarpour H, Asemi Z. Aquaporin 4: a key player in Parkinson's disease. J Cell Physiol. 2019; 234(12): 21471-8.
  • 45
    Fulop T, Witkowski JM, Bourgade K, Khalil A, Zerif E, Larbi A, et al. Can an infection hypothesis explain the beta amyloid hypothesis of Alzheimer's disease? Front Aging Neurosci. 2018; 10: 224.
  • 46
    Zhong M, Wang H, Yan H, Wu S, Wang K, Yang L, et al. Effects and mechanism of Aß1-42 on EV-A71 replication. Virol J. 2022; 19(1): 151.
  • 47
    Jang H, Boltz DA, Webster RG, Smeyne RJ. Viral parkinsonism. Biochim Biophys Acta. 2009; 1792(7): 714-21.
  • 48
    Nielsen NM, Rostgaard K, Hjalgrim H, Aaby P, Askgaard D. Poliomyelitis and Parkinson disease. JAMA. 2002; 287(13): 1650-1.
  • 49
    Kang JH, Lin HC. Comorbidity profile of poliomyelitis survivors in a Chinese population: a population-based study. J Neurol. 2011; 258(6): 1026-33.
  • 50
    Dourmashkin RR, McCall SA, Dourmashkin N, Hannah MJ. Virus-like particles and enterovirus antigen found in the brainstem neurons of Parkinson's disease. F1000Res. 2018; 7: 302.
  • 51
    Park SJ, Jin U, Park SM. Interaction between coxsackievirus B3 infection and a-synuclein in models of Parkinson's disease. PLoS Pathog. 2021; 17(10): e1010018.
  • 52
    Xue YC, Feuer R, Cashman N, Luo H. Enteroviral infection: the forgotten link to amyotrophic lateral sclerosis? Front Mol Neurosci. 2018; 11: 63.
  • 53
    Cermelli C, Vinceti M, Beretti F, Pietrini V, Nacci G, Pietrosemoli P, et al. Risk of sporadic amyotrophic lateral sclerosis associated with seropositivity for herpesviruses and echovirus-7. Eur J Epidemiol. 2003; 18(2): 123-7.
  • 54
    Berger MM, Kopp N, Vital C, Redl B, Aymard M, Lina B. Detection and cellular localization of enterovirus RNA sequences in spinal cord of patients with ALS. Neurology. 2000; 54(1): 20-5.
  • 55
    Vandenberghe N, Leveque N, Corcia P, Brunaud-Danel V, Salort-Campana E, Besson G, et al. Cerebrospinal fluid detection of enterovirus genome in ALS: a study of 242 patients and 354 controls. Amyotroph Lateral Scler. 2010; 11(3): 277-82.
  • 56
    Walker MP, Schlaberg R, Hays AP, Bowser R, Lipkin WI. Absence of echovirus sequences in brain and spinal cord of amyotrophic lateral sclerosis patients. Ann Neurol. 2001; 49(2): 249-53.
  • 57
    Nix WA, Berger MM, Oberste MS, Brooks BR, McKenna-Yasek DM, Brown Jr RH, et al. Failure to detect enterovirus in the spinal cord of ALS patients using a sensitive RT-PCR method. Neurology. 2004; 62(8): 1372-7.
  • 58
    Xue YC, Liu H, Mohamud Y, Bahreyni A, Zhang J, Cashman NR, et al. Sublethal enteroviral infection exacerbates disease progression in an ALS mouse model. J Neuroinflammation. 2022; 19(1): 16.
  • 59
    Fung G, Shi J, Deng H, Hou J, Wang C, Hong A, et al. Cytoplasmic translocation, aggregation, and cleavage of TDP-43 by enteroviral proteases modulate viral pathogenesis. Cell Death Differ. 2015; 22(12): 2087-97.
  • 60
    Lee EB, Lee VM, Trojanowski JQ. Gains or losses: molecular mechanisms of TDP43-mediated neurodegeneration. Nat Rev Neurosci. 2011; 13(1): 38-50.
  • 61
    Zhang X, Zheng Z, Shu B, Liu X, Zhang Z, Liu Y, et al. Human astrocytic cells support persistent coxsackievirus B3 infection. J Virol. 2013; 87(22): 12407-21.
  • 62
    Feuer R, Ruller CM, An N, Tabor-Godwin JM, Rhoades RE, Maciejewski S, et al. Viral persistence and chronic immunopathology in the adult central nervous system following Coxsackievirus infection during the neonatal period. J Virol. 2009; 83(18): 9356-69.
  • Financial support: CAPES, FAPERJ, FIOCRUZ, CNPq.

Publication Dates

  • Publication in this collection
    20 Mar 2023
  • Date of issue
    2023

History

  • Received
    31 Oct 2022
  • Accepted
    24 Jan 2023
Instituto Oswaldo Cruz, Ministério da Saúde Av. Brasil, 4365 - Pavilhão Mourisco, Manguinhos, 21040-900 Rio de Janeiro RJ Brazil, Tel.: (55 21) 2562-1222, Fax: (55 21) 2562 1220 - Rio de Janeiro - RJ - Brazil
E-mail: memorias@fiocruz.br