Skip to main content

Advertisement

Log in

Endogenous Retrovirus-K and Nervous System Diseases

  • Infection (ML Solbrig, Section Editor)
  • Published:
Current Neurology and Neuroscience Reports Aims and scope Submit manuscript

Abstract

A new appreciation of the microbiome is changing the way we perceive human health and disease. The holobiontic nature of humans is even etched into our DNA in the form of viral symbionts. Empirical evidence for the presence of endogenous retroviruses (ERVs) in the human genome and their activity in homeostatic and pathologic states has accumulated; however, no causal relationship with human disease has been established to date. In this review, we will focus on the role of endogenous retrovirus-K in neurologic disease. Specifically, we will attempt to reconcile the pathologic contribution of ERVK in disparate neurologic diseases by providing evidence as to inter-individual differences in ERVK genotypes, addressing the molecular regulation of ERVK, and provide detailed examples of ERVK-mediated processes in nervous system diseases.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. Weiss RA. The discovery of endogenous retroviruses. Retrovirology. 2006;3:67.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Marchi E, Kanapin A, Magiorkinis G, Belshaw R. Unfixed endogenous retroviral insertions in the human population. J Virol. 2014;88(17):9529–37. This study highlights the inter-individual variability of ERVK polymorphisms, and reveals novel ERVK insertions, which are not annotated in the human reference genome. Marchi et al also predict that ERVK has been active in the human germline as recently as 250,000 years ago.

  3. Shin W, Lee J, Son SY, et al. Human-specific HERV-K insertion causes genomic variations in the human genome. PLoS One. 2013;8:e60605.

  4. van der Kuyl AC. HIV infection and HERV expression: a review. Retrovirology. 2012;9:6.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Hanke K, Hohn O, Liedgens L, et al. Staufen-1 interacts with the human endogenous retrovirus family HERV-K(HML-2) rec and gag proteins and increases virion production. J Virol. 2013;87:11019–30.

  6. Contreras-Galindo R, Kaplan MH, Contreras-Galindo AC, et al. Characterization of human endogenous retroviral elements in the blood of HIV-1-infected individuals. J Virol. 2012;86:262–76.

  7. Contreras-Galindo R, Kaplan MH, Leissner P, et al. Human endogenous retrovirus K (HML-2) elements in the plasma of people with lymphoma and breast cancer. J Virol. 2008;82:9329–36.

  8. Dube D, Contreras-Galindo R, He S, et al. Genomic flexibility of human endogenous retrovirus type K. J Virol. 2014;88(17):9673–82.

  9. Subramanian RP, Wildschutte JH, Russo C, Coffin JM. Identification, characterization, and comparative genomic distribution of the HERV-K (HML-2) group of human endogenous retroviruses. Retrovirology. 2011;8:90.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  10. Belshaw R, Dawson AL, Woolven-Allen J, et al. Genomewide screening reveals high levels of insertional polymorphism in the human endogenous retrovirus family HERV-K(HML2): implications for present-day activity. J Virol. 2005;79:12507–14.

  11. Contreras-Galindo R, Kaplan MH, He S, et al. HIV infection reveals widespread expansion of novel centromeric human endogenous retroviruses. Genome Res. 2013;23:1505–13. Here, expression of a novel ERVK (HML-2) provirus termed K11 is identified in HIV-1 infection. Multiple K11 copies are found in centromeric regions of human chromosomes and not yet annotated in the human genome assembly.

  12. Moyes DL, Martin A, Sawcer S, et al. The distribution of the endogenous retroviruses HERV-K113 and HERV-K115 in health and disease. Genomics. 2005;86:337–41.

  13. Turner G, Barbulescu M, Su M, et al. Insertional polymorphisms of full-length endogenous retroviruses in humans. Curr Biol. 2001;11:1531–5.

  14. Hughes JF, Coffin JM. Human endogenous retrovirus K solo-LTR formation and insertional polymorphisms: implications for human and viral evolution. Proc Natl Acad Sci U S A. 2004;101:1668–72.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  15. Stauffer Y, Marguerat S, Meylan F, et al. Interferon-alpha-induced endogenous superantigen. a model linking environment and autoimmunity. Immunity. 2001;15:591–601.

  16. Tai AK, O’Reilly EJ, Alroy KA, et al. Human endogenous retrovirus-K18 Env as a risk factor in multiple sclerosis. Mult Scler. 2008;14:1175–80.

  17. de la Hera B, Varade J, Garcia-Montojo M, et al. Role of the human endogenous retrovirus HERV-K18 in autoimmune disease susceptibility: study in the Spanish population and meta-analysis. PLoS One. 2013;8:e62090.

  18. Dickerson F, Rubalcaba E, Viscidi R, et al. Polymorphisms in human endogenous retrovirus K-18 and risk of type 2 diabetes in individuals with schizophrenia. Schizophr Res. 2008;104:121–6.

  19. Nyegaard M, Demontis D, Thestrup BB, et al. No association of polymorphisms in human endogenous retrovirus K18 and CD48 with schizophrenia. Psychiatr Genet. 2012;22:146–8.

  20. Balada E, Ordi-Ros J, Vilardell-Tarres M. Molecular mechanisms mediated by human endogenous retroviruses (HERVs) in autoimmunity. Rev Med Virol. 2009;19:273–86.

    Article  PubMed  CAS  Google Scholar 

  21. Perron H, Lang A. The human endogenous retrovirus link between genes and environment in multiple sclerosis and in multifactorial diseases associating neuroinflammation. Clin Rev Allergy Immunol. 2010;39:51–61.

    Article  PubMed  CAS  Google Scholar 

  22. Frank O, Giehl M, Zheng C, et al. Human endogenous retrovirus expression profiles in samples from brains of patients with schizophrenia and bipolar disorders. J Virol. 2005;79:10890–901.

  23. Douville R, Liu J, Rothstein J, Nath A. Identification of active loci of a human endogenous retrovirus in neurons of patients with amyotrophic lateral sclerosis. Ann Neurol. 2011;69:141–51. This article describes the expression of ERVK RNA from specific loci in the cortical brain tissue of patients with ALS. Immunohistologic staining revealed that ERVK reverse transcriptase protein expression was localized in prefrontal and motor cortical neurons in ALS-affected individuals.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  24. Jeong BH, Lee YJ, Carp RI, Kim YS. The prevalence of human endogenous retroviruses in cerebrospinal fluids from patients with sporadic Creutzfeldt-Jakob disease. J Clin Virol. 2010;47:136–42.

    Article  PubMed  CAS  Google Scholar 

  25. Maksakova IA, Mager DL, Reiss D. Keeping active endogenous retroviral-like elements in check: the epigenetic perspective. Cell Mol Life Sci. 2008;65:3329–47.

    Article  PubMed  CAS  Google Scholar 

  26. Fuchs NV, Kraft M, Tondera C, et al. Expression of the human endogenous retrovirus (HERV) group HML-2/HERV-K does not depend on canonical promoter elements but is regulated by transcription factors Sp1 and Sp3. J Virol. 2011;85:3436–8.

  27. Knossl M, Lower R, Lower J. Expression of the human endogenous retrovirus HTDV/HERV-K is enhanced by cellular transcription factor YY1. J Virol. 1999;73:1254–61.

    PubMed  CAS  PubMed Central  Google Scholar 

  28. Katoh I, Mirova A, Kurata S, et al. Activation of the long terminal repeat of human endogenous retrovirus K by melanoma-specific transcription factor MITF-M. Neoplasia. 2011;13:1081–92.

  29. Ono M, Kawakami M, Ushikubo H. Stimulation of expression of the human endogenous retrovirus genome by female steroid hormones in human breast cancer cell line T47D. J Virol. 1987;61:2059–62.

    PubMed  CAS  PubMed Central  Google Scholar 

  30. Goering W, Ribarska T, Schulz WA. Selective changes of retro element expression in human prostate cancer. Carcinogenesis. 2011;32:1484–92.

    Article  PubMed  CAS  Google Scholar 

  31. Manghera M, Douville RN. Endogenous retrovirus-K promoter: a landing strip for inflammatory transcription factors? Retrovirology. 2013;10:16. Using a bioinformatics approach, this paper predicts that prototypical ERVK promoters contain multiple conserved binding sites for proinflammatory transcription factors, particularly Nuclear Factor-kappa B (NF-κB) and Interferon Response Factors (IRFs). An interesting feature of the ERVK promoter is the presence of 2 conserved Interferon Stimulated Response Elements (ISREs), which are known to bind IRFs. These findings suggest that augmented levels of proinflammatory transcription factors, such as NF-κB and IRF1, during neuroinflammation may be responsible for enhanced ERVK transcription in a variety of neurodegenerative conditions.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  32. Tateishi T, Yamasaki R, Tanaka M, et al. CSF chemokine alterations related to the clinical course of amyotrophic lateral sclerosis. J Neuroimmunol. 2010;222:76–81.

  33. Aebischer J, Moumen A, Sazdovitch V, et al. Elevated levels of IFNgamma and LIGHT in the spinal cord of patients with sporadic amyotrophic lateral sclerosis. Eur J Neurol. 2012;19(752–9):e745–56.

  34. Monji A, Kato T, Kanba S. Cytokines and schizophrenia: microglia hypothesis of schizophrenia. Psychiatry Clin Neurosci. 2009;63:257–65.

    Article  PubMed  CAS  Google Scholar 

  35. Mameli G, Astone V, Arru G, et al. Brains and peripheral blood mononuclear cells of multiple sclerosis (MS) patients hyperexpress MS-associated retrovirus/HERV-W endogenous retrovirus, but not Human herpes virus 6. J Gen Virol. 2007;88:264–74.

  36. Shi Q, Xie WL, Zhang B, et al. Brain microglia were activated in sporadic CJD but almost unchanged in fatal familial insomnia and G114V genetic CJD. Virol J. 2013;10:216.

  37. Freimanis G, Hooley P, Ejtehadi HD, et al. A role for human endogenous retrovirus-K (HML-2) in rheumatoid arthritis: investigating mechanisms of pathogenesis. Clin Exp Immunol. 2010;160:340–7.

  38. Morozov VA, Dao Thi VL, Denner J. The transmembrane protein of the human endogenous retrovirus—K (HERV-K) modulates cytokine release and gene expression. PLoS One. 2013;8:e70399. This paper demonstrates that ERVK virions and recombinant ERVK TM protein can inhibit the proliferation of human immune cells. The recombinant TM protein as well as ERVK virions also stimulated the expression and secretion of several cytokines, including the soluble TNF receptor II (sTNFRII) and Interleukin 10 (IL-10). An immunosuppressive state induced by the antiproliferative and anti-inflammatory effects of ERVK TM protein may allow tumor cells to escape immune detection. Thus, enhanced expression of ERVK env in multiple cancers may be responsible for promoting tumor proliferation.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  39. Ariza ME, Williams MV. A human endogenous retrovirus K dUTPase triggers a TH1, TH17 cytokine response: does it have a role in psoriasis? J Invest Dermatol. 2011;131:2419–27.

    Article  PubMed  CAS  Google Scholar 

  40. Rachita HR, Nagarajaram HA. Viral proteins that bridge unconnected proteins and components in the human PPI network. Mol Biosyst. 2014;10(9):2448–58.

  41. Matsuzaki T, Nakagawa M, Nagai M, et al. HTLV-I-associated myelopathy (HAM)/tropical spastic paraparesis (TSP) with amyotrophic lateral sclerosis-like manifestations. J Neurovirol. 2000;6:544–8.

  42. Verma A, Berger JR. ALS syndrome in patients with HIV-1 infection. J Neurol Sci. 2006;240:59–64.

    Article  PubMed  CAS  Google Scholar 

  43. MacGowan DJ, Scelsa SN, Imperato TE, et al. A controlled study of reverse transcriptase in serum and CSF of HIV-negative patients with ALS. Neurology. 2007;68:1944–6.

  44. McCormick AL, Brown Jr RH, Cudkowicz ME, Al-Chalabi A, Garson JA. Quantification of reverse transcriptase in ALS and elimination of a novel retroviral candidate. Neurology. 2008;70:278–83.

    Article  PubMed  CAS  Google Scholar 

  45. Steele AJ, Al-Chalabi A, Ferrante K, et al. Detection of serum reverse transcriptase activity in patients with ALS and unaffected blood relatives. Neurology. 2005;64:454–8.

  46. Hadlock KG, Miller RG, Jin X, et al. Elevated rates of antibody reactivity to HML-2/HERV-K but not other endogenous retroviruses in ALS. Amyotroph Lateral Scler Motor Neuron Disord. 2004;5:63.

    Google Scholar 

  47. Alfahad T, Nath A. Retroviruses and amyotrophic lateral sclerosis. Antivir Res. 2013;99:180–7.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  48. Weis S, Llenos IC, Sabunciyan S, et al. Reduced expression of human endogenous retrovirus (HERV)-W GAG protein in the cingulate gyrus and hippocampus in schizophrenia, bipolar disorder, and depression. J Neural Transm. 2007;114:645–55.

  49. Huang W, Li S, Hu Y, et al. Implication of the env gene of the human endogenous retrovirus W family in the expression of BDNF and DRD3 and development of recent-onset schizophrenia. Schizophr Bull. 2011;37:988–1000.

  50. Yao Y, Schroder J, Nellaker C, et al. Elevated levels of human endogenous retrovirus-W transcripts in blood cells from patients with first episode schizophrenia. Genes Brain Behav. 2008;7:103–12.

  51. Perron H, Mekaoui L, Bernard C, et al. Endogenous retrovirus type W GAG and envelope protein antigenemia in serum of schizophrenic patients. Biol Psychiatry. 2008;64:1019–23.

  52. Karlsson H, Bachmann S, Schroder J, et al. Retroviral RNA identified in the cerebrospinal fluids and brains of individuals with schizophrenia. Proc Natl Acad Sci U S A. 2001;98:4634–9.

  53. Yolken RH, Karlsson H, Yee F, Johnston-Wilson NL, Torrey EF. Endogenous retroviruses and schizophrenia. Brain Res Rev. 2000;31:193–9.

    Article  PubMed  CAS  Google Scholar 

  54. Leboyer M, Tamouza R, Charron D, Faucard R, Perron H. Human endogenous retrovirus type W (HERV-W) in schizophrenia: a new avenue of research at the gene-environment interface. World J Biol Psychiatry. 2013;14:80–90.

    Article  PubMed  Google Scholar 

  55. Diem O, Schaffner M, Seifarth W, Leib-Mosch C. Influence of antipsychotic drugs on human endogenous retrovirus (HERV) transcription in brain cells. PLoS One. 2012;7:e30054. Schizophrenia patients analyzed in previous studies (which often showed elevated levels of ERVs), were almost all taking medications such as antipsychotics. Since some neuroleptics and antidepressants are known to influence gene expression, in this study they attempted to determine if medications commonly prescribed to schizophrenics influence the expression of ERVs. Overall, they found that some cell types and postmortem brain tissue show upregulation of several types of HERVs with valproic acid treatment, but these did not include ERVK (HML2). Their results suggest that antipsychotic medication may contribute to increased expression of select ERV groups in patients with neuropsychiatric diseases.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  56. Hegyi H. GABBR1 has a HERV-W LTR in its regulatory region—a possible implication for schizophrenia. Biol Direct. 2013;8:5.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  57. Cohen CJ, Lock WM, Mager DL. Endogenous retroviral LTRs as promoters for human genes: a critical assessment. Gene. 2009;448:105–14.

    Article  PubMed  CAS  Google Scholar 

  58. Buzdin A, Kovalskaya-Alexandrova E, Gogvadze E, Sverdlov E. At least 50 % of human-specific HERV-K (HML-2) long terminal repeats serve in vivo as active promoters for host nonrepetitive DNA transcription. J Virol. 2006;80:10752–62.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  59. Suntsova M, Gogvadze EV, Salozhin S, et al. Human-specific endogenous retroviral insert serves as an enhancer for the schizophrenia-linked gene PRODH. Proc Natl Acad Sci U S A. 2013;110:19472–7. A human specific (hs) ERV belonging to the ERVK (HML-2) group is involved in the transcriptional regulation of a schizophrenia-related gene, PRODH. PRODH regulates proline catabolism, and is integral in normal functioning of the CNS; several mutations in this gene are associated with neuropsychiatric disorders, including schizophrenia. In cells expressing PRODH, hsERV PRODH is hypomethylated. Using bioinformatics they predicted that the hsERV PRODH LTR contains transcription factor binding sites for SOX2 and NF-κB1, when these genes were overexpressed in vitro, only over-expression of SOX2 resulted in a strong enhancer effect of hsERV PRODH .

  60. Kempf L, Nicodemus KK, Kolachana B, et al. Functional polymorphisms in PRODH are associated with risk and protection for schizophrenia and fronto-striatal structure and function. PLoS Genet. 2008;4:e1000252.

  61. Grace AA. Dopamine system dysregulation by the hippocampus: implications for the pathophysiology and treatment of schizophrenia. Neuropharmacology. 2012;62:1342–8.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  62. Shi J, Levinson DF, Duan J, et al. Common variants on chromosome 6p22.1 are associated with schizophrenia. Nature. 2009;460:753–7.

  63. Fatemi SH, Folsom TD, Rooney J, Thuras PD. Expression of GABAA alpha2-, beta1- and epsilon-receptors are altered significantly in the lateral cerebellum of subjects with schizophrenia, major depression and bipolar disorder. Translation. Psychiatry. 2013;3:e303.

    CAS  Google Scholar 

  64. Fatemi SH, Folsom TD, Thuras PD. Deficits in GABA(B) receptor system in schizophrenia and mood disorders: a postmortem study. Schizophr Res. 2011;128:37–43.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Karlsson H, Schroder J, Bachmann S, Bottmer C, Yolken RH. HERV-W-related RNA detected in plasma from individuals with recent-onset schizophrenia or schizoaffective disorder. Mol Psychiatry. 2004;9:12–3.

    Article  PubMed  CAS  Google Scholar 

  66. Perron H, Germi R, Bernard C, et al. Human endogenous retrovirus type W envelope expression in blood and brain cells provides new insights into multiple sclerosis disease. Mult Scler. 2012;18:1721–36.

  67. Brudek T, Christensen T, Aagaard L, et al. B cells and monocytes from patients with active multiple sclerosis exhibit increased surface expression of both HERV-H Env and HERV-W Env, accompanied by increased seroreactivity. Retrovirology. 2009;6:104.

  68. Garcia-Montojo M, Dominguez-Mozo M, Arias-Leal A, et al. The DNA copy number of human endogenous retrovirus-W (MSRV-type) is increased in multiple sclerosis patients and is influenced by gender and disease severity. PLoS One. 2013;8:e53623.

  69. Schmitt K, Richter C, Backes C, et al. Comprehensive analysis of human endogenous retrovirus group HERV-W locus transcription in multiple sclerosis brain lesions by high-throughput amplicon sequencing. J Virol. 2013;87:13837–52.

  70. Alvarez-Lafuente R, Garcia-Montojo M, De Las Heras V, et al. Herpes viruses and human endogenous retroviral sequences in the cerebrospinal fluid of multiple sclerosis patients. Mult Scler. 2008;14:595–601.

  71. Laufer G, Mayer J, Mueller BF, Mueller-Lantzsch N, Ruprecht K. Analysis of transcribed human endogenous retrovirus W env loci clarifies the origin of multiple sclerosis-associated retrovirus env sequences. Retrovirology. 2009;6:37.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Johnston JB, Silva C, Holden J, et al. Monocyte activation and differentiation augment human endogenous retrovirus expression: implications for inflammatory brain diseases. Ann Neurol. 2001;50:434–42.

  73. Hsiao FC, Tai AK, Deglon A, et al. EBV LMP-2A employs a novel mechanism to transactivate the HERV-K18 superantigen through its ITAM. Virology. 2009;385:261–6.

  74. Sutkowski N, Chen G, Calderon G, Huber BT. Epstein-Barr virus latent membrane protein LMP-2A is sufficient for transactivation of the human endogenous retrovirus HERV-K18 superantigen. J Virol. 2004;78:7852–60.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  75. Serafini B, Rosicarelli B, Franciotta D, et al. Dysregulated Epstein-Barr virus infection in the multiple sclerosis brain. J Exp Med. 2007;204:2899–912.

  76. Mameli G, Poddighe L, Mei A, et al. Expression and activation by Epstein Barr virus of human endogenous retroviruses-W in blood cells and astrocytes: inference for multiple sclerosis. PLoS One. 2012;7:e44991.

  77. Mameli G, Madeddu G, Mei A, et al. Activation of MSRV-type endogenous retroviruses during infectious mononucleosis and Epstein-Barr virus latency: the missing link with multiple sclerosis? PLoS One. 2013;8:e78474.

  78. Pertel T, Hausmann S, Morger D, et al. TRIM5 is an innate immune sensor for the retrovirus capsid lattice. Nature. 2011;472:361–5. This paper demonstrates that TRIM5 acts as a typical pattern recognition receptor, capable of detecting the retroviral capsid lattice. The engagement of TRIM5 with the retroviral capsid proteins stimulates inflammatory innate immune signaling mediated by AP-1 and NF-κB transcription factors, which is crucial for restricting retroviral replication.

  79. Rolland A, Jouvin-Marche E, Viret C, et al. The envelope protein of a human endogenous retrovirus-W family activates innate Immunity. through CD14/TLR4 and promotes Th1-like responses. J Immunol. 2006;176:7636–44.

  80. Nexo BA, Hansen B, Nissen KK, et al. Restriction genes for retroviruses influence the risk of multiple sclerosis. PLoS One. 2013;8:e74063.

  81. do Olival GS, Faria TS, Nali LH, et al. Genomic analysis of ERVWE2 locus in patients with multiple sclerosis: absence of genetic association but potential role of human endogenous retrovirus type W elements in molecular mimicry with myelin antigen. Front Microbiol. 2013;4:172.

  82. Gonzalez-Hernandez MJ, Cavalcoli JD, Sartor MA, et al. Regulation of the HERV-K (HML-2) transcriptome by the HIV-1 Tat protein. J Virol. 2014;88(16):8924–35.

  83. Bhat RK, Rudnick W, Antony JM, et al. Human endogenous retrovirus-K(II) envelope induction protects neurons during HIV/AIDS. PLoS One. 2014;9:e97984. Bhat et al describe the neuronal expression of ERVK (HML-2) envelope protein in brain tissue from HIV-infected and uninfected individuals. In vitro and murine models suggest that the ERVK (HML-2) transmembrane protein is protective against HIV-1 Vpr-mediated toxicity. Thus, exaptation of ERVK env may be a neuroprotective mechanism under pathologic conditions.

  84. Garrison KE, Jones RB, Meiklejohn DA, et al. T cell responses to human endogenous retroviruses in HIV-1 infection. PLoS Pathog. 2007;3:e165.

  85. Michaud HA, SenGupta D, de Mulder M, et al. Cutting edge: an antibody recognizing ancestral endogenous virus glycoproteins mediates antibody-dependent cellular cytotoxicity on HIV-1-infected cells. J Immunol. 2014;193(4):1544–8. This study examines how an antibody targeting the ERVK transmembrane protein facilitates NK killing of HIV-1 infected cells. The humoral response against ERVK in HIV-infected individuals may play a role in antibody-dependent cellular cytotoxicity, and could be used in novel immunomodulatory or neuroprotective strategies.

  86. Michaud HA, de Mulder M, SenGupta D, et al. Trans-activation, post-transcriptional maturation, and induction of antibodies to HERV-K (HML-2) envelope transmembrane protein in HIV-1 infection. Retrovirology. 2014;11:10.

  87. Lebeau G, DesGroseillers L, Sossin W, Lacaille JC. mRNA binding protein staufen 1-dependent regulation of pyramidal cell spine morphology via NMDA receptor-mediated synaptic plasticity. Mol Brain. 2011;4:22.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  88. Chatel-Chaix L, Boulay K, Mouland AJ, Desgroseillers L. The host protein Staufen1 interacts with the Pr55Gag zinc fingers and regulates HIV-1 assembly via its N-terminus. Retrovirology. 2008;5:41.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Brinzevich D, Young GR, Sebra R, et al. HIV-1 interacts with human endogenous retrovirus K (HML-2) envelopes derived from human primary lymphocytes. J Virol. 2014;88:6213–23.

  90. Lee Y-J, Jeong B-H, Choi E-K, Kim Y-S. Involvement of endogenous retroviruses in prion diseases. Pathogens. 2013;2:533–43.

    Article  Google Scholar 

  91. Adler V, Zeiler B, Kryukov V, et al. Small, highly structured RNAs participate in the conversion of human recombinant PrP(Sen) to PrP(Res) in vitro. J Mol Biol. 2003;332:47–57.

  92. Leblanc P, Baas D, Darlix JL. Analysis of the interactions between HIV-1 and the cellular prion protein in a human cell line. J Mol Biol. 2004;337:1035–51.

    Article  PubMed  CAS  Google Scholar 

  93. Leblanc P, Alais S, Porto-Carreiro I, et al. Retrovirus infection strongly enhances scrapie infectivity release in cell culture. EMBO J. 2006;25:2674–85.

  94. Veerhuis R, Hoozemans JJ, Janssen I, et al. Adult human microglia secrete cytokines when exposed to neurotoxic prion protein peptide: no intermediary role for prostaglandin E2. Brain Res. 2002;925:195–203.

  95. Kim JI, Ju WK, Choi JH, et al. Expression of cytokine genes and increased nuclear factor-kappa B activity in the brains of scrapie-infected mice. Brain Res Mol Brain Res. 1999;73:17–27.

  96. Lu Y, Liu A, Zhou X, et al. Prion peptide PrP106-126 induces inducible nitric oxide synthase and proinflammatory cytokine gene expression through the activation of NF-kappaB in macrophage cells. DNA Cell Biol. 2012;31:833–8.

  97. Letendre SL, Woods SP, Ellis RJ, et al. Lithium improves HIV-associated neurocognitive impairment. AID. 2006;20:1885–8.

  98. Jones-Brando LV, Buthod JL, Holland LE, Yolken RH, Torrey EF. Metabolites of the antipsychotic agent clozapine inhibit the replication of human immunodeficiency virus type 1. Schizophr Res. 1997;25:63–70.

    Article  PubMed  CAS  Google Scholar 

  99. Stommel EW, Graber D, Montanye J, Cohen JA, Harris BT. Does treating schizophrenia reduce the chances of developing amyotrophic lateral sclerosis? Med Hypotheses. 2007;69:1021–8.

    Article  PubMed  CAS  Google Scholar 

  100. Moots RJ, Al-Saffar Z, Hutchinson D, et al. Old drug, new tricks: haloperidol inhibits secretion of proinflammatory cytokines. Ann Rheum Dis. 1999;58:585–7.

Download references

Compliance with Ethics Guidelines

Conflict of Interest

Mamneet Manghera and Jennifer Ferguson declare that they have no conflict of interest. Renée Douville has received establishment and operating grants from the Manitoba Health Research Council, an operating grant from the Manitoba Medical Services Foundation, a starter grant from the ALS Association, and major and discretionary awards from the University of Winnipeg.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Renée Douville.

Additional information

This article is part of the Topical Collection on Infection

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Manghera, M., Ferguson, J. & Douville, R. Endogenous Retrovirus-K and Nervous System Diseases. Curr Neurol Neurosci Rep 14, 488 (2014). https://doi.org/10.1007/s11910-014-0488-y

Download citation

  • Published:

  • DOI: https://doi.org/10.1007/s11910-014-0488-y

Keywords

Navigation