Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2015

Open Access 01.12.2015 | Letter to the Editor

miRNAs and SAMHD1 regulation in vitro and in a model of HIV CNS disease

verfasst von: Kenneth W. Witwer, Erin L. Buchanan, Stephanie L. Myers, Melissa A. McAlexander

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2015

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Pilakka-Kanthikeel et al. recently reported higher levels of the retroviral restriction factor sterile alpha motif and histidine/aspartic acid domain-containing protein 1 (SAMHD1) in astrocytes than in microglia, suggesting that SAMHD1 levels might explain in part the relatively refractory nature of astrocytes to retroviral replication. These findings are consistent with our studies of simian and human immunodeficiency virus infection of astrocytes and macrophages. Similarly, a role for two host microRNAs in post-transcriptional regulation of SAMHD1 agrees with our in vitro results and those of others. However, data from an animal model of HIV neurologic disorders may not be consistent with robust miRNA-mediated regulation of SAMHD1 in vivo.
Hinweise

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

KWW conceived of, designed, and managed the study. ELB, MAM, SLM, and KWW performed experiments. ELB and KWW analyzed results. KWW wrote the manuscript. All authors read and approved the final manuscript.
Abkürzungen
3′ UTR
3′ untranslated region
CNS
central nervous system
HIV
human immunodeficiency virus
miRNA
microRNA
SAMHD1
SAM-domain and HD-domain containing protein 1

Letter to the editor

We were pleased to read the recent article of Pilakka-Kanthikeel and colleagues, reporting higher levels of the retroviral restriction factor sterile alpha motif and histidine/aspartic acid domain-containing protein 1 (SAMHD1) in astrocytes than in microglia [1]. The data suggest that SAMHD1 levels might explain in part the relatively refractory nature of astrocytes to retroviral replication. These findings are consistent with our studies of simian and human immunodeficiency virus (SIV and HIV) infection of astrocytes and macrophages ([2], "Regulation of the SAMHD1 transcript during retroviral infection of the central nervous system," submitted); see also the different results on SAMHD1 in elite suppressor populations [3-5]. Similarly, a role for two host microRNAs (miRNAs) in post-transcriptional regulation of SAMHD1 agrees with our results [2] and Swaminathan et al. [6].
At the same time, we are somewhat uncertain about the extent to which these miRNAs regulate SAMHD1 in vivo. Pilakka-Kanthikeel et al. transfected 50 nM miR-155 and miR-181 mimic or inhibitor and observed effects on SAMHD1 levels in cultured astrocytes [1]. In our hands, 10 nM miR-155 and miR-181—as well as several others including miR-34a and miR-150—reduced the expression of luciferase reporters fused to the SAMHD1 3′ untranslated region (3′ UTR), supporting direct regulation (Fig. 1a). However, the stoichiometries of these “all or nothing” assays are highly artificial, with exposures amounting to millions or tens of millions of oligonucleotides per cell. Additionally, and as reported previously by others [7], we observed significant cell death with miR-181a transfection, a potential explanation for the particularly large reduction in this condition. The authors did not report viability in their populations, but we wonder if this could explain some of the observed effects.
Seeking to determine whether there is a tissue-level, in vivo correlation between SAMHD1 and miRNAs found to regulate the transcript in vitro, we measured SAMHD1 and miRNA expression in a well-characterized simian model of HIV disease [8], including uninfected controls. In the model, thalamic SAMHD1 expression increased significantly after infection (p < 0.03), while miR-181a was significantly downregulated (p < 0.001). This regulation in opposite directions could be interpreted as consistent with a miR-181a-SAMHD1 interaction. However, further analysis revealed no correlation of miR-181a and SAMHD1 in any of the following groups: acute phase infection (Fig. 1b), progression to disease (Fig. 1c), or all samples, including uninfected controls (Fig. 1d). There was also no correlation of SAMHD1 with levels of miR-155 or miR-34a, which in fact tended to increase during infection (data not shown). These results recall a previous finding in which the RNA binding protein ZFP36 (tristetraprolin) could be suppressed by miRNAs in vitro but showed little evidence of negative correlation with the same miRNAs in vivo [9].
While our findings temper our initial enthusiasm about miRNA-mediated control of SAMHD1, we are not prepared to dismiss it entirely. Although the astrocyte is the most abundant cell type in the brain and appears to support higher levels of SAMHD1 expression than, e.g., macrophages ["Regulation of the SAMHD1 transcript during retroviral infection of the central nervous system," submitted] and microglia [1], the tissue-level profiling we performed may mask changes of miRNA or SAMHD1 expression in specific cells types or even the absence of certain miRNAs in disease-related cells [10]. Similarly, focusing on single miRNA-to-target interactions ignores the regulatory “whole picture,” since a given miRNA-to-target interaction is governed not only by the abundance of each partner but also by the abundance of other partners [11]. Finally, assuming adequate delivery methods, therapeutic antisense manipulation of anti-SAMHD1 miRNAs (for example, to help prevent cell-to-cell transmission during eradication therapy) could be performed at high concentrations not dissimilar to the in vitro manipulations that we and the authors performed. There may will be more to learn about miRNAs and SAMHD1 in the CNS.

Acknowledgements

This work was supported in part by NIH grant R21 AI102659 (KWW), NIMH pilot grant P30 MH075673 (to KWW), and by the National Center for Research Resources and the Office of Research Infrastructure Programs (ORIP) and the National Institutes of Health through grant number P40 OD013117.
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.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

KWW conceived of, designed, and managed the study. ELB, MAM, SLM, and KWW performed experiments. ELB and KWW analyzed results. KWW wrote the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Pilakka-Kanthikeel S, Raymond A, Atluri VS, Sagar V, Saxena SK, Diaz P, et al. Sterile alpha motif and histidine/aspartic acid domain-containing protein 1 (SAMHD1)-facilitated HIV restriction in astrocytes is regulated by miRNA-181a. J Neuroinflammation. 2015;12(1):66.PubMedCentralCrossRefPubMed Pilakka-Kanthikeel S, Raymond A, Atluri VS, Sagar V, Saxena SK, Diaz P, et al. Sterile alpha motif and histidine/aspartic acid domain-containing protein 1 (SAMHD1)-facilitated HIV restriction in astrocytes is regulated by miRNA-181a. J Neuroinflammation. 2015;12(1):66.PubMedCentralCrossRefPubMed
2.
Zurück zum Zitat Buchanan EL, Espinoza DA, Sisk JM, Moyer A, Clements JE, Witwer KW. Expression and regulation of host restriction factor SAMHD1 during lentiviral infection of astrocytes and macrophages. In: Conference on Retroviruses and Opportunistic Infections. Atlanta, GA, USA; CROI, LLC, Alexandria, 2013 Buchanan EL, Espinoza DA, Sisk JM, Moyer A, Clements JE, Witwer KW. Expression and regulation of host restriction factor SAMHD1 during lentiviral infection of astrocytes and macrophages. In: Conference on Retroviruses and Opportunistic Infections. Atlanta, GA, USA; CROI, LLC, Alexandria, 2013
3.
Zurück zum Zitat Riveira-Munoz E, Ruiz A, Pauls E, Permanyer M, Badia R, Mothe B, et al. Increased expression of SAMHD1 in a subset of HIV-1 elite controllers. J Antimicrob Chemother. 2014;69:3057–60.CrossRefPubMed Riveira-Munoz E, Ruiz A, Pauls E, Permanyer M, Badia R, Mothe B, et al. Increased expression of SAMHD1 in a subset of HIV-1 elite controllers. J Antimicrob Chemother. 2014;69:3057–60.CrossRefPubMed
4.
Zurück zum Zitat Abdel-Mohsen M, Raposo RA, Deng X, Li M, Liegler T, Sinclair E, et al. Expression profile of host restriction factors in HIV-1 elite controllers. Retrovirology. 2013;10:106.PubMedCentralCrossRefPubMed Abdel-Mohsen M, Raposo RA, Deng X, Li M, Liegler T, Sinclair E, et al. Expression profile of host restriction factors in HIV-1 elite controllers. Retrovirology. 2013;10:106.PubMedCentralCrossRefPubMed
5.
Zurück zum Zitat Buchanan EL, McAlexander MA, Witwer KW. SAMHD1 expression in blood cells of HIV-1 elite suppressors and viraemic progressors. J Antimicrob Chemother. 2014;70:954–6.CrossRefPubMed Buchanan EL, McAlexander MA, Witwer KW. SAMHD1 expression in blood cells of HIV-1 elite suppressors and viraemic progressors. J Antimicrob Chemother. 2014;70:954–6.CrossRefPubMed
6.
Zurück zum Zitat Swaminathan G, Navas-Martin S, Martin-Garcia J. MicroRNAs and HIV-1 infection: antiviral activities and beyond. J Mol Biol. 2014;426(6):1178–97.CrossRefPubMed Swaminathan G, Navas-Martin S, Martin-Garcia J. MicroRNAs and HIV-1 infection: antiviral activities and beyond. J Mol Biol. 2014;426(6):1178–97.CrossRefPubMed
7.
Zurück zum Zitat Ouyang YB, Lu Y, Yue S, Xu LJ, Xiong XX, White RE, et al. miR-181 regulates GRP78 and influences outcome from cerebral ischemia in vitro and in vivo. Neurobiol Dis. 2012;45(1):555–63.PubMedCentralCrossRefPubMed Ouyang YB, Lu Y, Yue S, Xu LJ, Xiong XX, White RE, et al. miR-181 regulates GRP78 and influences outcome from cerebral ischemia in vitro and in vivo. Neurobiol Dis. 2012;45(1):555–63.PubMedCentralCrossRefPubMed
8.
Zurück zum Zitat Beck SE, Queen SE, Witwer KW, Metcalf Pate KA, Mangus LM, Gama L, et al. Paving the path to HIV neurotherapy: predicting SIV CNS disease. Eur J Pharmacol. 2015;759:303–12.CrossRefPubMed Beck SE, Queen SE, Witwer KW, Metcalf Pate KA, Mangus LM, Gama L, et al. Paving the path to HIV neurotherapy: predicting SIV CNS disease. Eur J Pharmacol. 2015;759:303–12.CrossRefPubMed
9.
Zurück zum Zitat Liu J, Sisk JM, Gama L, Clements JE, Witwer KW. Tristetraprolin expression and microRNA-mediated regulation during simian immunodeficiency virus infection of the central nervous system. Mol Brain. 2013;6(1):40.PubMedCentralCrossRefPubMed Liu J, Sisk JM, Gama L, Clements JE, Witwer KW. Tristetraprolin expression and microRNA-mediated regulation during simian immunodeficiency virus infection of the central nervous system. Mol Brain. 2013;6(1):40.PubMedCentralCrossRefPubMed
10.
Zurück zum Zitat Kent OA, McCall MN, Cornish TC, Halushka MK. Lessons from miR-143/145: the importance of cell-type localization of miRNAs. Nucleic Acids Res. 2014;42(12):7528–38.PubMedCentralCrossRefPubMed Kent OA, McCall MN, Cornish TC, Halushka MK. Lessons from miR-143/145: the importance of cell-type localization of miRNAs. Nucleic Acids Res. 2014;42(12):7528–38.PubMedCentralCrossRefPubMed
Metadaten
Titel
miRNAs and SAMHD1 regulation in vitro and in a model of HIV CNS disease
verfasst von
Kenneth W. Witwer
Erin L. Buchanan
Stephanie L. Myers
Melissa A. McAlexander
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2015
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-015-0380-y

Weitere Artikel der Ausgabe 1/2015

Journal of Neuroinflammation 1/2015 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie