Skip to main content
  • Letter to the Editor
  • Open access
  • Published:

Technical considerations in detection of HERV-K in amyotrophic lateral sclerosis: selection of controls and the perils of qPCR

A Letter to the Editor Response to this article was published on 03 July 2019

The Original Article was published on 18 March 2019

Main text

The study by Garson et al. [12] failed to show a difference between the expression levels of Human endogenous retrovirus K (HERV-K) in amyotrophic lateral sclerosis (ALS) brain samples and controls by qPCR. However, several technical aspects need to be considered for the interpretation of their findings. Nearly half (11/23) the control samples had cancer. It is well known that HERV-K is activated in several types of cancer such as teratocarcinoma, germ cell tumors, melanoma, ovarian, and prostate cancer [4, 5, 14, 17, 23, 24, 34] and its expression is associated with various features of malignant cells [3, 25, 28, 30, 35]. Cancer cells can release viral-like particles containing viral products [3, 5, 23, 30]. These products would be expected to circulate within the brain vasculature and extracellular space hence can easily be detected in brain extracts. This is consistent with our observations where we were able to detect HERV-K transcripts in brain of patients with systemic cancer without any brain metastasis [6]. Garson et al., also found high levels of HERV-K transcripts in patients with cerebral infarcts. This is not surprising. Necrosis of the brain is likely to induce repair mechanisms that would include the presence of progenitor and stem cells. HERV-K and other endogenous retroviral elements are activated in these cells types and play an important role in cellular proliferation, similar to what is seen in cancer [11]. This raises two important questions: what the proper controls are to use in such studies and how does one reconcile the similarly opposite effects of HERV-K and other retroviral elements in cell proliferation and neurodegeneration. We have shown that HERV-K is specifically expressed in cortical neurons in a subpopulation of patients (~ 30%) with ALS [6, 21]. We did not see similar levels of activation in patients with other types of neurodegenerative disorders such as Alzheimer’s disease [21] and Parkinson’s disease [6]. Although other groups have shown the activation of other transposable elements in Alzheimer’s disease [13]. Similarly, increased circulating levels of antibodies directed against the HERV-K env have been found in serum and CSF of ALS patients [2].

Another important consideration in such studies is that ALS is a heterogeneous syndrome. Several genetic mutations have been associated with the disease, which suggest different pathophysiological pathways in these subtypes. Further, the clinical phenotypes may also vary. Some patients have predominant lower motor neuron involvement while others have predominant upper motor neuron involvement. A recent study classified ALS into three categories based on RNA seq analysis of the brain. One of these groups had activation of transposable elements including HERV-K that represented 20% of the samples [32]. Thus, a large enough sample size is necessary to capture these patients.

Garson et al. were meticulous in the design of their PCR assay and they followed the standard recommendations, checking for amplification specificity of the primers and calculating PCR efficiencies. However, if the goal of the study is to quantify RNA transcripts by qPCR, then the integrity of the samples measured by the RNA integrity number (RIN), is key [9]. RIN can have values ranging from 1 (totally degraded RNA) to 10 (intact RNA) [16]. While reference genes can control for small differences in RNA concentration and integrity [9], up to fourfold difference can be found in the relative expression of certain genes when comparing highly intact to degraded RNA [10] and negative and positive correlations between the RNA integrity and the relative expression have been seen depending on the target gene [27], or more accurately, depending on the differential resistance to degradation of the target and the reference gene. Studies have shown that the most important factor affecting expression levels of housekeeping genes, regardless of the diagnosis or brain tissue type, is the RIN number [22]. In human autopsy samples some degradation is to be expected. Factors such as post-mortem interval, pH of the brain, duration on the ventilator, duration of brain death etc., can affect the integrity of the RNA [31]. In the study by Garson et al., some RIN values were as low as 5, meaning 50% of the RNA was degraded. Most importantly, average RIN number was significantly different in ALS and controls. In contrast, in the study by Li et al. [21] only samples that had a RIN > 8 were used for the qPCR study and the experimental groups were matched for this critical variable.

As the authors did in this study, to control for small differences in RNA integrity and total amount of starting RNA, the use of a reference gene is strictly recommended. The selected reference gene should not be regulated or at least be minimally regulated and it must be sensitive to RNA degradation [10], logically in a similar manner to the target gene, because it is known that reference genes show different sensitivity to RNA degradation [10]. Garson et al. followed the international guidelines for qPCR, applying software (qBase+ and RefFinder) to determine the most stable reference genes among experimental groups. They selected one that was stable among ALS and control samples but, as they clearly state, the controls were more degraded (lower RIN number). Therefore, by choosing the housekeeping genes with the lowest variation among experimental groups, they selected the least sensitive to detect variations due to RNA integrity. So, the normalizations they performed and the attempts to show that the RIN number does not correlate with the normalized levels of HERV-K are not reliable, making it impossible to interpret the data.

Detection of transcripts that may be activated poses additional challenges in neurodegenerative diseases since the very cells of interest are the ones that are in various stages of degeneration or may be apoptotic. Analysis of expression by qPCR, due to its dependence on the appropriate selection of housekeeping genes, might not be ideal in these type of diseases. In a study by Rydbirk et al. [29], the authors tested a panel of reference genes to define the stability of the transcripts in neurodegenerative diseases and controls and then, validated them by analyzing the relative expression of a gene involved in neurodegeneration (GSK3B). GSK3B has been consistently implicated in neurodegeneration [19]. Its expression has been found altered in Alzheimer disease (AD) [15, 33] and Parkinson’s disease (PD) [1, 18]. Several functional studies have shown that the active enzyme encoded by GSK3B is elevated in these diseases [36] and is responsible for the phosphorylation of key proteins such as tau [8, 20] and alpha-synuclein [7], which leads to their pathological aggregation in the brain. GSK3B is a key player in the formation of beta-amyloid plaques [26]. Despite its clear association with neurodegeneration and the higher levels in AD and PD reported by several studies, Rydbirk et al. [29] could not find any difference in the relative expression of transcripts of GSK3B between AD or PD and controls when they used the most stable reference genes for normalization. They could only find higher expression of GSK3B in AD and PD when they used the most common reference genes (GAPDH and beta-actin) or the ones that they found as least stable.

A better method to use in these conditions might be RNAseq, where the quantification of the expression of a gene is “number of transcripts/million of total reads”. This method would be more adequate for neurodegenerative diseases since it controls for the possible decrease in global transcriptional activity in the disease. However, even this technique would not consider the differential susceptibility to degeneration of the various cell types in the cortex. Another method might be to perform laser capture microdissection of the neurons and then analyze the cells by RNA seq. The expression of the transcripts can also be visualized by in situ hybridization in tissue sections. Thus, due to the complexity of the problem and several technical challenges, no reliable conclusion can be made from the study by Garson et al., but further studies are needed using alternative methods and appropriate controls from brain samples with preserved integrity of the RNA to explore the expression of HERV-K in ALS.

Abbreviations

AD:

Alzheimer disease

ALS:

Amyotrophic lateral sclerosis

HERV-K:

Human endogenous retrovirus K

PD:

Parkinson disease

RIN:

RNA integrity number

References

  1. Armentero MT, Sinforiani E, Ghezzi C, Bazzini E, Levandis G, Ambrosi G et al (2011) Peripheral expression of key regulatory kinases in Alzheimer’s disease and Parkinson’s disease. Neurobiol Aging 32:2142–2151. https://doi.org/10.1016/j.neurobiolaging.2010.01.004

    Article  CAS  PubMed  Google Scholar 

  2. Arru G, Mameli G, Deiana GA, Rassu AL, Piredda R, Sechi E et al (2018) Humoral immunity response to human endogenous retroviruses K/W differentiates between amyotrophic lateral sclerosis and other neurological diseases. Eur J Neurol 25:1076–1e84. https://doi.org/10.1111/ene.13648

    Article  CAS  PubMed  Google Scholar 

  3. Bhardwaj N, Montesion M, Roy F, Coffin JM (2015) Differential expression of HERV-K (HML-2) proviruses in cells and virions of the teratocarcinoma cell line Tera-1. Viruses 7:939–968. https://doi.org/10.3390/v7030939

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Buscher K, Hahn S, Hofmann M, Trefzer U, Ozel M, Sterry W et al (2006) Expression of the human endogenous retrovirus-K transmembrane envelope, Rec and Np9 proteins in melanomas and melanoma cell lines. Melanoma Res 16:223–234

    Article  PubMed  Google Scholar 

  5. Buscher K, Trefzer U, Hofmann M, Sterry W, Kurth R, Denner J (2005) Expression of human endogenous retrovirus K in melanomas and melanoma cell lines. Cancer Res 65:4172–4180. https://doi.org/10.1158/0008-5472.CAN-04-2983

    Article  PubMed  Google Scholar 

  6. Douville R, Liu J, Rothstein J, Nath A (2011) Identification of active loci of a human endogenous retrovirus in neurons of patients with amyotrophic lateral sclerosis. Ann Neurol 69:141–151. https://doi.org/10.1002/ana.22149

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Duka T, Duka V, Joyce JN, Sidhu A (2009) Alpha-Synuclein contributes to GSK-3beta-catalyzed Tau phosphorylation in Parkinson’s disease models. FASEB J 23:2820–2830. https://doi.org/10.1096/fj.08-120410

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Ferrer I, Barrachina M, Puig B (2002) Glycogen synthase kinase-3 is associated with neuronal and glial hyperphosphorylated tau deposits in Alzheimer’s disease, Pick’s disease, progressive supranuclear palsy and corticobasal degeneration. Acta Neuropathol 104:583–591. https://doi.org/10.1007/s00401-002-002-0587-8

    Article  CAS  PubMed  Google Scholar 

  9. Fleige S, Pfaffl MW (2006) RNA integrity and the effect on the real-time qRT-PCR performance. Mol Asp Med 27:126–139. https://doi.org/10.1016/j.mam.2005.12.003

    Article  CAS  Google Scholar 

  10. Fleige S, Walf V, Huch S, Prgomet C, Sehm J, Pfaffl MW (2006) Comparison of relative mRNA quantification models and the impact of RNA integrity in quantitative real-time RT-PCR. Biotechnol Lett 28:1601–1613. https://doi.org/10.1007/s10529-006-9127-2

    Article  CAS  PubMed  Google Scholar 

  11. Fuchs NV, Loewer S, Daley GQ, Izsvak Z, Lower J, Lower R (2013) Human endogenous retrovirus K (HML-2) RNA and protein expression is a marker for human embryonic and induced pluripotent stem cells. Retrovirology 10:115. https://doi.org/10.1186/1742-4690-10-115

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Garson JA, Usher L, Al-Chalabi A, Huggett J, Day EF, McCormick AL (2019) Quantitative analysis of human endogenous retrovirus-K transcripts in postmortem premotor cortex fails to confirm elevated expression of HERV-K RNA in amyotrophic lateral sclerosis. Acta Neuropathol Commun 7:45. https://doi.org/10.1186/s40478-019-0698-2

    Article  PubMed  PubMed Central  Google Scholar 

  13. Guo C, Jeong HH, Hsieh YC, Klein HU, Bennett DA, De Jager PL et al (2018) Tau activates transposable elements in Alzheimer’s disease. Cell Rep 23:2874–2880. https://doi.org/10.1016/j.celrep.2018.05.004

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Herbst H, Sauter M, Mueller-Lantzsch N (1996) Expression of human endogenous retrovirus K elements in germ cell and trophoblastic tumors. Am J Pathol 149:1727–1735

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Hye A, Kerr F, Archer N, Foy C, Poppe M, Brown R et al (2005) Glycogen synthase kinase-3 is increased in white cells early in Alzheimer’s disease. Neurosci Lett 373:1–4. https://doi.org/10.1016/j.neulet.2004.10.031

    Article  CAS  PubMed  Google Scholar 

  16. Imbeaud S, Graudens E, Boulanger V, Barlet X, Zaborski P, Eveno E et al (2005) Towards standardization of RNA quality assessment using user-independent classifiers of microcapillary electrophoresis traces. Nucleic Acids Res 33:e56. https://doi.org/10.1093/nar/gni054

    Article  PubMed  PubMed Central  Google Scholar 

  17. Kurth R, Bannert N (2010) Beneficial and detrimental effects of human endogenous retroviruses. Int J Cancer 126:306–314. https://doi.org/10.1002/ijc.24902

    Article  CAS  PubMed  Google Scholar 

  18. Kwok JB, Hallupp M, Loy CT, Chan DK, Woo J, Mellick GD et al (2005) GSK3B polymorphisms alter transcription and splicing in Parkinson's disease. Ann Neurol 58:829–839. https://doi.org/10.1002/ana.20691

    Article  CAS  PubMed  Google Scholar 

  19. Lei P, Ayton S, Bush AI, Adlard PA (2011) GSK-3 in neurodegenerative diseases. Int J Alzheimers Dis 2011:189246. https://doi.org/10.4061/2011/189246

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Li T, Paudel HK (2006) Glycogen synthase kinase 3 beta phosphorylates Alzheimer’s disease-specific Ser (396) of microtubule-associated protein tau by a sequential mechanism. Biochemistry-US 45:3125–3133. https://doi.org/10.1021/bi051634r

    Article  CAS  Google Scholar 

  21. Li W, Lee MH, Henderson L, Tyagi R, Bachani M, Steiner J et al (2015) Human endogenous retrovirus-K contributes to motor neuron disease. Sci Transl Med 7:307ra153. https://doi.org/10.1126/scitranslmed.aac8201

    Article  PubMed  PubMed Central  Google Scholar 

  22. Lipska BK, Deep-Soboslay A, Weickert CS, Hyde TM, Martin CE, Herman MM et al (2006) Critical factors in gene expression in postmortem human brain: focus on studies in schizophrenia. Biol Psychiatry 60:650–658. https://doi.org/10.1016/j.biopsych.2006.06.019

    Article  CAS  PubMed  Google Scholar 

  23. Lower R, Lower J, Frank H, Harzmann R, Kurth R (1984) Human teratocarcinomas cultured in vitro produce unique retrovirus-like viruses. J Gen Virol 65(Pt 5):887–898. https://doi.org/10.1099/0022-1317-65-5-887

    Article  PubMed  Google Scholar 

  24. Muster T, Waltenberger A, Grassauer A, Hirschl S, Caucig P, Romirer I et al (2003) An endogenous retrovirus derived from human melanoma cells. Cancer Res 63:8735–8741

    CAS  PubMed  Google Scholar 

  25. Oricchio E, Sciamanna I, Beraldi R, Tolstonog GV, Schumann GG, Spadafora C (2007) Distinct roles for LINE-1 and HERV-K retroelements in cell proliferation, differentiation and tumor progression. Oncogene 26:4226–4233. https://doi.org/10.1038/sj.onc.1210214

    Article  CAS  PubMed  Google Scholar 

  26. Phiel CJ, Wilson CA, Lee VM, Klein PS (2003) GSK-3alpha regulates production of Alzheimer's disease amyloid-beta peptides. Nature 423:435–439. https://doi.org/10.1038/nature01640

    Article  CAS  PubMed  Google Scholar 

  27. Popova T, Mennerich D, Weith A, Quast K (2008) Effect of RNA quality on transcript intensity levels in microarray analysis of human post-mortem brain tissues. BMC Genomics 9:91. https://doi.org/10.1186/1471-2164-9-91

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Reis BS, Jungbluth AA, Frosina D, Holz M, Ritter E, Nakayama E et al (2013) Prostate cancer progression correlates with increased humoral immune response to a human endogenous retrovirus GAG protein. Clin Cancer Res 19:6112–6125. https://doi.org/10.1158/1078-0432.CCR-12-3580

    Article  CAS  PubMed  Google Scholar 

  29. Rydbirk R, Folke J, Winge K, Aznar S, Pakkenberg B, Brudek T (2016) Assessment of brain reference genes for RT-qPCR studies in neurodegenerative diseases. Sci Rep 6:37116. https://doi.org/10.1038/srep37116

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Schmitt K, Reichrath J, Roesch A, Meese E, Mayer J (2013) Transcriptional profiling of human endogenous retrovirus group HERV-K (HML-2) loci in melanoma. Genome Biol Evol 5:307–328. https://doi.org/10.1093/gbe/evt010

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Stan AD, Ghose S, Gao XM, Roberts RC, Lewis-Amezcua K, Hatanpaa KJ et al (2006) Human postmortem tissue: what quality markers matter? Brain Res 1123:1–11. https://doi.org/10.1016/j.brainres.2006.09.025

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Tam O, Rozhkov NV, Shaw R, Kim D, Hubbard I, Fennessey S, Propp N, The NYGC ALS Consortium, Fagegaltier D, Ostrow LW, Phatnani H, Ravits J, Dubnau J, Gale Hammell M (2019) Postmortem cortex samples identify distinct molecular subtypes of ALS: retrotransposon activation, oxidative stress, and activated glia. Biorxiv. https://doi.org/10.1101/574509

  33. Wang X, Chen Y, Wang X, Lu L (2010) Genetic regulatory network analysis for app based on genetical genomics approach. Exp Aging Res 36:79–93. https://doi.org/10.1080/03610730903418729

    Article  CAS  PubMed  Google Scholar 

  34. Wang-Johanning F, Liu J, Rycaj K, Huang M, Tsai K, Rosen DG et al (2007) Expression of multiple human endogenous retrovirus surface envelope proteins in ovarian cancer. Int J Cancer 120:81–90. https://doi.org/10.1002/ijc.22256

    Article  CAS  PubMed  Google Scholar 

  35. Wildschutte JH, Ram D, Subramanian R, Stevens VL, Coffin JM (2014) The distribution of insertionally polymorphic endogenous retroviruses in breast cancer patients and cancer-free controls. Retrovirology 11:62. https://doi.org/10.1186/s12977-014-0062-3 doi: 10.1186/PREACCEPT-1720768941312026

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Wills J, Jones J, Haggerty T, Duka V, Joyce JN, Sidhu A (2010) Elevated tauopathy and alpha-synuclein pathology in postmortem Parkinson’s disease brains with and without dementia. Exp Neurol 225:210–218. https://doi.org/10.1016/j.expneurol.2010.06.017

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Contributions

All authors read and approved the final manuscript.

Corresponding author

Correspondence to Avindra Nath.

Ethics declarations

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Garcia-Montojo, M., Li, W. & Nath, A. Technical considerations in detection of HERV-K in amyotrophic lateral sclerosis: selection of controls and the perils of qPCR. acta neuropathol commun 7, 101 (2019). https://doi.org/10.1186/s40478-019-0753-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40478-019-0753-z

Keywords