Skip to main content
PLOS ONE logoLink to PLOS ONE
. 2020 Aug 11;15(8):e0237066. doi: 10.1371/journal.pone.0237066

Lack of TLR4 modifies the miRNAs profile and attenuates inflammatory signaling pathways

Juan R Ureña-Peralta 1, Raúl Pérez-Moraga 2,3, Francisco García-García 2, Consuelo Guerri 1,*
Editor: Pratibha V Nerurkar4
PMCID: PMC7418977  PMID: 32780740

Abstract

TLR4 is a member of the toll-like receptors (TLR) immune family, which are activated by lipopolysaccharide, ethanol or damaged tissue, among others, by triggering proinflammatory cytokines release and inflammation. Lack of TLR4 protects against inflammatory processes and neuroinflammation linked with several neuropathologies. By considering that miRNAs are key post-transcriptional regulators of the proteins involved in distinct cellular processes, including inflammation, this study aimed to assess the impact of the miRNAs profile in mice cortices lacking the TLR4 response. Using mice cerebral cortices and next-generation sequencing (NGS), the findings showed that lack of TLR4 significantly reduced the quantity and diversity of the miRNAs expressed in WT mice cortices. The results also revealed a significant down-regulation of the miR-200 family, while cluster miR-99b/let-7e/miR-125a was up-regulated in TLR4-KO vs. WT. The bioinformatics and functional analyses demonstrated that TLR4-KO presented the systematic depletion of many pathways closely related to the immune system response, such as cytokine and interleukin signaling, MAPK and ion Channels routes, MyD88 pathways, NF-κβ and TLR7/8 pathways. Our results provide new insights into the molecular and biological processes associated with the protective effects of TLR-KO against inflammatory damage and neuroinflammation, and reveal the relevance of the TLR4 receptors response in many neuropathologies.

Introduction

Toll-like receptors (TLRs) are a family of pattern-recognition receptors (PRRs) that recognize conserved structural motifs in a wide array of pathogens or PAMPs (pathogen-associated molecular patterns), as well as damaged tissue or DAMPs (damage-associated molecular pattern) [1]. These receptors are expressed in immune cells and TLRs activation leads to the downstream stimulation of different signaling pathways that trigger the production of large amounts of inflammatory proteins and cytokines. These pathways include type I interferons (IFNs), antiviral proteins through the activation of interferon regulatory factor (IRF) 3, IRF7, activator protein-1 (AP-1) and nuclear factor-kappa B (NF-κβ) [2]. In the central nervous system, most TLRs are expressed in glial cells [3]. Recent evidence demonstrates the participation of the TLRs response in neurodegenerative disorders [4, 5].

TLR-signaling pathways are strictly regulated at many levels to prevent excessive inflammation and to achieve balanced output. Recently, several studies have demonstrated the regulatory role of miRNAs in the control of the immune response and TLRs to the pathogens and regulators involved in TLR-signaling [6, 7].

MicroRNAs (miRNAs, miRs) are short (17–24bp) RNA species expressed across cell types that are active against a high proportion of the transcriptome [8]. The sequence-complementary mechanism of miRNA activity exploits the combinatorial regulation of gene expression by repressing the translation of their complementary target genes [9]. These small RNAs play a crucial role in the regulation of diverse biological processes, such as tissue development and homeostasis [10], cell proliferation and differentiation, apoptosis, and immune system function [11]. However, dysregulated miRNAs contribute to the development of several diseases, such as cancer, and cardiovascular or neuroinflammatory and neurological diseases [12].

Ethanol is a neurotoxic compound and its abuse can cause neural damage. However, the mechanism of its neurotoxicity remains unclear. We have shown that by activating TLR4 receptors in glial cells [13, 14], ethanol triggers the release of cytokines and inflammatory mediators to cause neuroinflammation and brain damage in mice with chronic alcohol consumption [15]. The critical role of the TLR4 response in the neuroinflammatory effects of ethanol has been further supported by demonstrating that TLR4-deficient mice are protected against ethanol-induced neuroinflammation and neural damage [16]. To explore the regulatory action of miRNAs in the effects of ethanol on the brain and the influence of the TLR4 response, we used high-throughput sequencing methods along with a bioinformatics analysis in the cortices of the TLR4-WT and TLR4-KO mice treated with and without ethanol. The results demonstrate that ethanol treatment induces a differential expression of some miRNAs, such as the miR-183 cluster (miR-183C) (miR-96/-182/-183), miR-200a and miR-200b, which were down-regulated, while mirR-125b was up-regulated in the alcohol-treated WT versus (vs.) untreated mice. Some of these miRNAs modulate the target genes related to the genes associated with the innate immune TLR4 signaling response (Il1r1, Mapk14, Sirt1, Lrp6 and Bdnf). However, changes in the neuroinflammatory target genes associated with alcohol abuse were abolished mostly in the ethanol-treated TLR4-KO mice [17]. These results raise the question as to what the impact of the inflammatory or anti-inflammatory associated miRNAs profile is on mice lacking the TLR4 response.

Using high-throughput sequencing (NGS) data from the WT and TLR4 knockout mice cortices, along with a bioinformatics analysis, the aims of this study were to evaluate whether: 1) a differential or specific expression exists in certain miRNAs that modulate the inflammatory pathways associated with TLR4; 2) compensatory changes occur in certain anti-inflammatory miRNAs that could explain the reduction in neuroinflammation associated with alcohol intake.

Material & methods

Animals

Female wild-type (TLR4_WT, TLR4+/+, WT) (Harlan Ibérica S.L., Barcelona) and TLR4 knockout (TLR4_KO, TLR4-/-, KO) mice were used, which were kindly provided by Dr. S. Akira (Osaka University, Japan) with C57BL/6J genetic backgrounds. Animals were kept under controlled light/dark (12 h/12 h) conditions at 23°C and 60% humidity. The animal experiments were carried out in accordance with the guidelines set out in the European Communities Council Directive (86/609/ECC) and Spanish Royal Decree 1201/2005, and were approved by the Ethical Committee of Animal Experimentation of CIPF (Valencia, Spain).

Alcohol treatment

For the chronic alcohol treatment, 44 (11 animals/group) 7-week-old WT (C57BL/6J) and TLR4-KO female mice were housed (4 animals/cage) and maintained with either water (WT and TLR4-KO control) or water containing 10% (v/v) alcohol. They were placed on a solid diet ad libitum for 5 months. During this period, daily food and liquid intake were similar for both the WT and TLR4-KO mice and the alcohol-treated/untreated groups. Body weight gain at the end of the 5-month period was similar in both the WT (C57BL/6J) and TLR4-KO mice treated with or without alcohol, as previously described [15]. The peak blood alcohol levels (BALs) detected in mice after the chronic ethanol treatment were around ≈125 mg/dl (range of 87–140 mg/dl) in the ethanol-treated WT mice, and ≈ 122 mg/dl (range of 98–135 mg/dl) in the ethanol-treated-KO. The use of females instead of males was based on our previous studies showing that females were more vulnerable to the effects of ethanol than males [15].

Cerebral cortex dissection

Mice were sacrificed by cervical, brains were removed and cerebral cortices were dissected following the mouse brain atlas coordinates instructions [18]. Brain cortices were weighed and immediately snap-frozen in liquid nitrogen. Samples were stored at -80°C until processed.

Total RNA isolation

The frozen cortex samples (100–200 mg) were used for the total and small RNA (sRNA) extractions. Briefly, 100–200 mg of tissue were disrupted with 1 ml of QIAzol (Qiagen, Maryland, USA), followed by the phenol chloroform method [19]. Total RNA and sRNA were isolated using the miRNeasy columns from the Qiagen Kit to obtain a separate sample for each RNA type. sRNAs were used for the deep sequencing protocol. Total RNA was employed for RT-qPCR to evaluate miRNAs and genes.

DNA isolation and genotyping

The genomic DNA from the WT and TLR4-KO mice was isolated using the commercial Maxwell 16 mouse tail DNA purification kit and the Maxwell 16 Instrument (Promega, Barcelona, Spain). Following the manufacturer’s instructions, DNA was collected in 300 μl of elution buffer. DNA was amplified with specific primers designed to differentiate WT and TLR4-KO strains. For genotyping purposes, three primers were designed according to previous studies [20]: primer “b”, which was recognized by both genotypes (WT and TLR4-KO); primer “a”, which was specific for the WT mice; primer “c”, which was specific for the TLR4-KO mice. PCR was performed using 2 μl of DNA extract with master mix 2x PCR TaqNova-RED (Blirt, Gdańsk, Poland). The thermocycler (Eppendorf) program was 40 cycles: 30 seg 94 °C+ 90 seg 67 °C and 60 seg 74 °C. Amplicons were loaded in 1.5% agarose gel and visualized in BioRad. The employed primers are described in Table 1.

Table 1. The RT and RT-qPCR primers used.

PRIMER SEQUENCE
PRIMER A CGTGTAAACCAGCCAGGTTTTGAAGGC
PRIMER B TGTTGCCCTTCAGTCACAGAGACTCTG
PRIMER C TGTTGGGTCGTTTGTTCGGATCCGTCG
PPIA_F/PPIA_R GCGTCTGCTTCGAGCTGTTTGC / ACATGCTTGCCATCCAGC
TLR4_F/TLR4_R TGCCTCTCTTGCATCTGGCTGG/CTGTCAGTACCAAGGTTGAGAGCTGG
TRIL_F/TRIL_R CCAACGGCAACGAGATTG / CGGTTAGATTCCAGGTGTAGG
IL1Β_F/IL1B_R CTCATTGTGGCTGTGGAGAA / TCTAATGGGAACGTCACACA
IL6_F/IL6_R AAGCCAGAGTCCTTCAGAGAGA / TCTTGGTCCTTAGCCACTCCT
COX2_F/COX2_R CATTGACCAGAGCAGAGAGATG / GGCTTCCAGTATTGAGGAGAAC
IL1R_F/IL1R_R TGAAGAGCACAGAGGGGACT / CATTGATCCTGGGTCAGCTT
TRAF6_F/TRAF6_R AACGTCCTTTCCAGAAGTGC / GAATGTGCAAGGGATTGGAG
RT-MIR-592-5P GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATACGACACATCATCGA
PCR-MIR-592-5P ACACTCCAGCTGGGATTGTGTCAATATGCG
RT-MIR-377-3P GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATACGACACAAAAGTTG
PCR-MIR-377-3P ACACTCCAGCTGGGATCACACAAAGGCAAC
RT-MIR-382-3P GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATACGACAAAAAGTGTTG
PCR-MIR-382-3P ACACTCCAGCTGGGTCATTCACGGACAAC
RT-MIR-27B-5P GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATACGACGTTCACCAATC
PCR-MIR-27B-5P ACACTCCAGCTGGGAGAGCTTAGCTGATTG
RT-MIR-96-5P GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATACGACAGCAAAAATG
PCR-MIR-96-5P ACACTCCAGCTGGGTTTGGCACTAGCAC
RT-MIR-1982-3P GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATACGACCTGTGGGAGAAC
PCR-MIR-1982-3P ACACTCCAGCTGGGTCTCACCCTATGTTC
RT-MIR-5122 GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATACGACCACAGCCCCGG
PCR-MIR-5122 ACACTCCAGCTGGGCCGCGGGACCCGG
RT-MIR-182-5P GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATACGACCGGTGTGAGTTC
PCR-MIR-182-5P ACACTCCAGCTGGGTTTGGCAATGGTAGAAC
RT-MIR-183-5P GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATACGACAGTGAATTCTAC
PCR-MIR-183-5P ACACTCCAGCTGGGTATGGCACTGGTAG
RT-MIR-429-3P GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATACGACACGGCATTACC
PCR-MIR-429-3P ACACTCCAGCTGGGTAATACTGTCTGGTAAT
RT-MIR-141-3P GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATACGACCCATCTTTACCAG
PCR-MIR-141-3P ACACTCCAGCTGGGTAACACTGTCTGGTA
RT-MIR-9-5P GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATACGACTCATACAGCTAG
PCR-MIR-9-5P ACACTCCAGCTGGGTCTTTGGTTATCTAGC
RT-MIR-136-3P GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATACGACAAGACTCATTTG
PCR-MIR-136-3P ACACTCCAGCTGGGATCATCGTCTCAAATG
RT-MIR-99B-5P GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATACGACCGCAAGGTCGGT
PCR-MIR-99B-5P ACACTCCAGCTGGGCACCCGTAGAACCG
RT-MIR-125A-5P GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATACGACTCACAGGTTAAAG
PCR-MIR-125A-5P ACACTCCAGCTGGGTCCCTGAGACCCTTTAAC
RT-MIR-125B-5P GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATACGACTCACAAGTTAGG
PCR-MIR-125B-5P ACACTCCAGCTGGGTCCCTGAGACCCTAAC
REVERSE PRIMER CCAGTGCAGGGTCCGAGGT
TLR7_F / TLR7_R TGTGGACACGGAAGAGACAA / CCCTCAGGGATTTCTGTCAA
TLR8_F/ TLR8_R AACCATCGTCAACTGCATGA / CATTTGGGTGCTGTTGTTTG
IL12_F / IL12_R ACGGCCAGAGAAAAACTGAA / CTACCAAGGCACAGGGTCAT
INFΑ_F / INFA_R AGTGAGCTGACCCAGCAGAT / CAGGGGCTGTGTTTCTTCTC

RNA quantity and quality determinations

The quantities of each total RNA sample were determined using NanoDrop, and quantity and qualities were measured in an Agilent 2100 bioanalyzer. Total RNA integrity was analyzed by the RNA Nano6000 kit (Agilent Technologies, Santa Clara, CA, USA) and the sRNA kit was employed for sRNAs (Agilent Technologies, Santa Clara, CA, USA). The best nine samples for each condition were selected and combined to obtain three pooled samples, which gave 12 pooled sRNA samples. Briefly for each condition, nine animals were used and divided into three sample pools. With this approach, an attempt was made to minimize differences due to individuals and, in turn, to increase differences due to the studied variables. Then the sRNA profiles were measured again with the small-RNA kit (Agilent Technologies, Santa Clara, CA, USA) following the manufacturer’s instructions. Total RNA integrity was measured by the RNA Nano6000 kit (Agilent Technologies, Santa Clara, CA, USA).

Small RNA library preparation

First 100 ng of the sRNA fraction from the pooled cortex samples were used to prepare the sRNA libraries with the Truseq library prep Small RNA Sample Preparation kit (Illumina, San Diego, USA). These samples were employed for sequencing in HiSeq following the Illumina pooling manufacture’s guidelines. The cDNA from miRNAs was obtained by the Superscript II Reverse Transcriptase kit (Thermo Fisher Scientific, Carlsbad, CA, USA) and unique indices were introduced during PCR amplification for 15 cycles. The sRNA libraries were visualized and quantified in an Agilent 2100 bioanalyzer. A multiplexed pool was prepared that consisted of equimolar amounts of sRNA-derived libraries. Libraries were sequenced for 50 single read cycles in HiSeq2000 (Illumina).

Reverse transcription miRNA (miRNA-RT)

First of all, 500 ng of total RNA from cortical brain tissue were used. Samples were treated with DNase I (Invitrogen, Foster City, CA, USA) to avoid genomic DNA contamination. The retrotranscription reaction was run with specific RT-primers (1nM) (Integrated DNA Technologies, Inc.) for each analyzed miRNA using the High Capacity cDNA Reverse Transcription kit (Thermo Fisher Scientific, Foster City, CA, USA) following the manufacturer’s protocol; the specific primers are detailed in Table 1. The reaction was carried out in an Eppendorf 5341 Master Cycler (Eppendorf AG, Hamburg, Germany) at 25 °C for 10 min, then at 40 °C for 1 h, and finally at 85 °C for 5 min to inactivate the enzyme. Total RNA was also converted into cDNA. Briefly; 2 μg of total RNA from the cortical brain tissue were retrotranscribed with the High Capacity cDNA Reverse Transcription kit (Thermo Fisher Scientific, Foster City, CA, USA) following the manufacturer’s protocol.

Real-time quantitative PCR

RT-qPCR was performed in a Light Cycler® 480 System (Roche, Mannheim, Germany). The reactions contained Light Cycler 480 SYBR Green I Master (2X) (Roche Applied Science, Mannheim, Germany), 5 μM of the forward and reverse primers, and 1 μL of cDNA. The amplification efficiency (E) of the primers was calculated from the plot of the Cq values against the cDNA input according to the equation E = [10(-1/slope)]. The relative expression ratio of a target/reference gene was calculated by the Pfaffl method [21]. Housekeeping cyclophilin-A (Ppia) was used as an internal control for messengers and RNAU6 for small RNAs. The primer gene sequence is detailed in Table 1.

Bioinformatics /pipelines analysis

The preprocessing of reads was done with Cutadapt (version 2.0) [22]. After removing adapters, the trimmed sequences were aligned against the reference genome (GRCm38.pp6) by Bowtie2 (v. 2.3.5.1) [23]. As the genomic alignment of miRNAs is challenging given their size ranges (~21 nucleotides) [24], Bowtie2 was configured to increase sensitivity [25] (Fig 2A).

Fig 2.

Fig 2

Bioinformatics analysis and mature miRNA profile of samples: (A) Pipeline methodology used (see page 8). (B) The Top 10 abundant mature miRNA in samples, which were almost 50% of the total counts of the following mature miRNA: mmu-miR-181a-5p, mmu-miR-127-3p, mmu-miR-26a-5p, mmu-miR-434-3p, mmu-miR-22-3p and mmu-miR-128-3p.

A read count was performed by custom R scripts, and these scripts used the library RSubread [26]. In order to annotate the reads aligned against miRNAs, the genome coordinates from the miRBase/gff3 file [27] were used, which allowed the mature miRNA of interest to be detected and annotated. After generating the count matrix of the six samples, the gene expression data were evaluated by multidimensional scaling and clustering methods to detect any abnormal patterns in the samples. Finally, the Top 10 most abundant miRNAs by read counts in the samples were annotated, along with their corresponding GO terms (Biological Process) from the QuickGO database [28] for descriptive purposes.

The count matrix was normalized by the TMM method (Trimmed Mean of M values). The differential expressions were analyzed by the Bioconductor package of edgeR [29]. P-values were corrected from the False Discovery Rate (FDR) as proposed by Benjamini and Hochberg [30].

Gene Set Enrichment Analysis (GSEA)

The bioinformatics functional analysis was performed using the mdgsa (Multi-Dimensional Gene Set Analysis) package [31], while Cluster-Profiler [32] was utilized for the graphics/results representation. This functional profiling included several steps:

First, miRNA was linked with its targets using TargetScan Mouse (Release 7.2: August 2018). Two types of miRNA-to-Gene lists were used: one inferred by bioinformatic methods and the other validated by the experimental assays.

Second, the differential expression results were used in the mdgsa package to transform the miRNA expression level into a gene level, which allowed the gene inferred differential inhibition score or index to be obtained. This transferred index contained the effect of multiple miRNAs against the same gene. By this approach, it was possible to obtain a miRNA regulation model and the effects of many miRNAs against their target.

Third, the inferred gene index was used in a univariate gene set analysis [33].

The above methodology allowed us to correlate a large set of genes with different functional annotations (GO terms, KEGG and Reactome pathways, etc.). In fact as we herein obtained a ranking of genes with differential inhibition indices, it was possible to determine if a functional annotation was inhibited in either the WT or the TLR4-KO group. The employed functional annotations were Gene Ontology (Biological Process) terms [34], the Kyoto Encyclopedia of Genes and Genomes (KEGG) [35] and the Reactome pathways database [36]. Multiple testing corrections were made with the FDR developed by Benjamini and Hochberg. Data representation was carried out with ClusterProfiler, a Bioconductor package.

Statistical methods

The SPSS, version 17.0, and the R version 3.4.3 software 40 were used for the validation analysis and bioinformatics, respectively. The RT-qPCR data were analyzed by a Student’s t-test when comparing TLR4_WT and TLR4_KO. A two-way ANOVA was used in the ethanol treatment experiments when comparing more than two groups. Differences at a value of P < 0.05 were considered statistically significant.

Results

Lack of TLR4 attenuates inflammatory pathways

To assess whether lack of the TLR4 receptor would lower the expression of the inflammatory pathways, we firstly checked the genotypes of the WT and TLR4-KO mice. The agarose gel (see Fig 1A) illustrated that while the WT mice only showed a band that was derived by the amplification with primers “a” and “b”, the TLR4-KO mice only presented amplification when primers “c” and “b” were utilized, which confirmed the two herein used genotypes. We also proved the absence of mRNA TLR4 in the TLR4-KO mice (Fig 1B).

Fig 1. Lack of TLR4 alters the ethanol-induced immune response in mice cortices.

Fig 1

(A) The genotyping analysis of the wild-type (WT) and TLR4KO mice with specific primers for the locus of the TLR4 receptor were confirmed by agarose gel. (B) The RT-qPCR analysis in the WT and TLR4-KO mice cortices showed lack of TLR4 gene expression in the TLR4-KO mice. (C) The RT-qPCR analysis indicated the deregulation of some TLR4 pathway components, such as Il-6, Tril and IL1R, but not in others like the Traf6 gene. (D) The RT-qPCR analysis of IL1R1, IL1β IL6 and COX2 confirmed that ethanol treatment triggered the activation of specific TLR4 signaling pathway components in the WT mice cortices, but ethanol treatment effects were blocked or attenuated in the TLR4-KO mice. A statistical analysis was performed by the t-test for 1B and-1C and a two-way ANOVA for 1D. Asterisk indicates the significance with a p-value *<0.05, **<0.01 and **<0.001. n = 5–10.

We next measured the expression of the different genes and proteins involved in the TLR4 pathway. Fig 1B shows that while the gene expressions of Tril (TLR4 interactor with leucine-rich repeats) and interleukin Il-6 lowered in the TLR4-KO cortices, no change or up-regulation was observed in the gene expressions of Traf6 (tumor necrosis factor receptor-associated factor 6) and receptor Il-1r1, respectively, compared to the WT mice.

To further confirm our previous results, which demonstrated that lack of TLR4 attenuated the inflammatory response in TLR4-KO mice, we also evaluated the levels of some inflammatory-related genes in the mice cortices of the WT and TLR4-KO control with ethanol. Fig 1C shows that while ethanol treatment increased the gene expression levels of IL-1R, IL-1β, Il-6 and COX-2 in the WT mice cortices, the same ethanol treatment did not affect the levels of these genes in the TLR4-KO mice with the same treatment. These findings indicate that the inflammatory signaling response did not occur in the TLR4-KO mice cortices.

Analysis of the mature miRNA’s sequences from the WT and TLR4-KO mice cortices

With the deep sequencing analysis data (see Ureña et al., 2018, GEO Series accession number GSE120373), we next evaluated the mature miRNAs profile in the WT and TLR4-KO mice cortices. For this purpose, we followed the methodology scheme shown in Fig 2A, where sequences were mapped against the mouse reference genome (GRCm38.pp6) using Bowtie2 aligner (version 2.3.5.1). We also employed the miRNA annotation GFF file from miRbase (Release 22.1), which extracts the reads mapped to mature miRNAs sequences. The read count with R custom scripts and the Rsubread library were used to obtain a yield of 1.264.521 miRNA reads for the six samples.

Fig 2B shows the most abundant mature miRNA in our NGS data, where 50% of the total miRNA found belonged to only six miRNA: mmu-miR-181a-5p, mmu-miR-127-3p, mmu-miR-26a-5p, mmu-miR-434-3p, mmu-miR-22-3p and mmu-miR-128-3p. When focusing on samples separately, the MDS plot identified that both the WT and TLR4-KO samples clustered correctly. It is noteworthy that sample KO1 exhibited an abnormal pattern compared to samples KO2 and KO3. Therefore, we decided to eliminate this outliner sample to perform further bioinformatics analyses. In relation to this action, each individual sample was formed by a pool of three independent cortices.

Table 2 shows the functional annotation for the top miRNAs found. The more representative miRNA in the samples have GO terms that relate to the regulation of the biosynthesis of interleukins (mmu-miR-26a, mmu-miR-128-3p, mmu-miR-181a-5p) or neurodevelopment regulation (mmu-miR-22-3p, mmu-miR-26a, mmu-miR-125a-5p). We also found the positive regulation of microglial cell activation and neuroinflammatory response (mmu-miR-128-3p) or central nervous system myelin maintenance (mmu-miR-26a-5p), among others. These results indicate the major regulation of these routes in mice cortices. All the annotated miRNA has a GO term related to gene silencing by miRNA.

Table 2. The most represented mature miRNAs in mice cortices.

The mature miRNAs were annotated with Gene Ontology in relation to a biological process using the QuickGO database [27].

MIRNA NAME GO ID GO DESCRIPTION
MMU-MIR-22-3P GO:0035195 gene silencing by miRNA
GO:0051152 positive regulation of smooth muscle cell differentiation
GO:2001200 positive regulation of dendritic cell differentiation
MMU-MIR-26A-5P GO:0010459 negative regulation of heart rate
GO:0032286 central nervous system myelin maintenance
GO:0035195 gene silencing by miRNA
GO:0045379 negative regulation of the interleukin-17 biosynthetic process
GO:0045409 negative regulation of the interleukin-6 biosynthetic process
GO:0060371 regulation of atrial cardiac muscle cell membrane depolarization
GO:0090370 negative regulation of cholesterol efflux
GO:0150079 negative regulation of the neuroinflammatory response
GO:1903609 negative regulation of inward rectifier potassium channel activity
MMU-MIR-128-3P GO:0010985 negative regulation of lipoprotein particle clearance
GO:0035195 gene silencing by miRNA
GO:0042632 cholesterol homeostasis
GO:0055088 lipid homeostasis
GO:0090370 negative regulation of cholesterol efflux
GO:0150078 positive regulation of the neuroinflammatory response
GO:1903980 positive regulation of microglial cell activation
MMU-MIR-181A-5P GO:0032691 negative regulation of interleukin-1 beta production
GO:0032715 negative regulation of interleukin-6 production
GO:0032720 negative regulation of tumor necrosis factor production
GO:0035195 gene silencing by miRNA
GO:0050723 negative regulation of the interleukin-1 alpha biosynthetic process
GO:0050728 negative regulation of the inflammatory response
MMU-MIR-125A-5P GO:0010629 negative regulation of gene expression
GO:0035278 miRNA mediated inhibition of translation
GO:1903671 negative regulation of sprouting angiogenesis

Lack of TLR4 significantly reduces the quantity and diversity of the miRNAs expressed in the WT mice cortices

Using the information provided by the NGS technology, we analyzed the miRNAs that were either specific for each genotype or commonly expressed in the WT and TLR4-KO mice.

Fig 3A illustrates a Venn’s diagram which shows that while 365 miRNAs were commonly expressed in both genotypes, 52 miRNAs were exclusively expressed in the WT (Table 3; see also S1 Table), and only four miRNAs belonged exclusively to the TLR4-KO mice (Table 4).

Fig 3.

Fig 3

Unique miRNA profile for each genotype: (A) The Venn’s diagram illustrated that 365 miRNAs were commonly expressed in both genotypes; 52 miRNAs were exclusively expressed for the WT and only four miRNAs belonged to the TLR4-KO mice. (B) RT-qPCR confirmed the differential expression pattern of some miRNAs, e.g. miR-382-3p, miR-27b-5p, miR-592-5p and miR-377-3p, which were down-regulated in the TLR4-KO mice, while miR-96-5p and miR-5122 were not deregulated as predicted by NGS. A statistical analysis was performed by the t-test with n = 6–8. Statistical significance with a p-value of *<0.05 and **<0.01. (C) Bioinformatics enrichment analysis of specific miRNAs in the WT mice.

Table 3. Exclusive miRNAs in the TLR4-WT mice.

The table shows the total counts and a selected article about their function.

MIRNA ONLY IN TLR4-WT TLR4-WT1 TLR4-WT2 TLR4-WT3 TLR4-KO2 TLR4-KO3 LITERATURE PMID MATURE SEQUENCE
MMU-MIR-7A-1-3P 5 10 14 0 0 Differential stress induced c-Fos expression in the dorsal raphe and amygdala of high-responder/low-responder rats. [37] caacaaaucacagucugccaua
MMU-MIR-592-5P 7 10 9 0 0 Effects of MicroRNA-592-5p on hippocampal heuron injury following hypoxic-ischemic brain damage. [38] auugugucaauaugcgaugaugu
MMU-MIR-377-3P 7 9 3 0 0 miR-377-3p drives malignancy characteristics by up-regulating GSK-3β expression and activating the NF-κB pathway in hCRC cells. [39] aucacacaaaggcaacuuuugu
MMU-MIR-382-3P 6 2 11 0 0 miR-382-3p suppressed the IL-1β induced inflammatory response of chondrocytes byia the TLR4/MyD88/NF-κB signaling pathway. [40] ucauucacggacaacacuuuuu
MMU-MIR-27B-5P 3 10 5 0 0 Exosomal microRNA profiles from serum and cerebrospinal fluid in neurosyphilis. [41] agagcuuagcugauuggugaac
MMU-MIR-199A-3P 5 6 3 0 0 miR-199a-3p is involved in estrogen-mediated autophagy through the IGF-1/mTOR pathway in osteocyte-like MLO-Y4 cells. [42] acaguagucugcacauugguua
MMU-MIR-96-5P 3 4 7 0 0 miR-96-5p prevents hepatic stellate cell activation by inhibiting autophagy via ATG7. [43] uuuggcacuagcacauuuuugcu
MMU-MIR-345-3P 2 7 4 0 0 Mitochondria- associated microRNA expression profiling of heart failure. [44] ccugaacuaggggucuggagac

Table 4. Exclusive miRNAs in the TLR4-KO mice.

The table shows the total counts and a selected article about their function.

MIRNA ONLY IN TLR4-KO TLR4-WT1 TLR4-WT2 TLR4-WT3 TLR4-KO2 TLR4-KO3 LITERATURE PMID MATURE SEQUENCE
MMU-MIR-1982-3P 0 0 0 2 2 no items found ucucacccuauguucucccacag
MMU-MIR-5122 0 0 0 2 2 no items found ccgcgggacccggggcugug
MMU-MIR-5114 0 0 0 2 3 no items found acuggagacggaagcugcaaga
MMU-MIR-677-3P 0 0 0 3 2 no items found gaagccagaugccguuccugagaagg

When we looked at the specific miRNAs with higher counts (illustrated in Table 3). We noted that some, such as miR-382-3p, were related to the TLR4/MyD88/NF-κβ signaling pathway [40], and miR-592-5p was involved in brain injury [38]. Notably, these miRNAs were not detected in the TLR4-KO mice by our NGS data analysis. For the specific miRNA expressed in the TLR4-KO mice shown in Table 4, we were unable to find any relevant functions or pathway related to these miRNAs in the bibliography.

To confirm and validate the NGS data, we decided to evaluate the expression of some miRNAs related to inflammatory processes in the WT and TLR4-KO mice cortices by RT-qPCR (the selected miRNAs are depicted in red in Tables 3 and 4). Fig 3B shows the differential expression pattern of the selected miRNAs. Here we observed how miR-382-3p, miR-27b-5p, miR-592-5p and miR-377-3p, which were exclusively found miRNAs in the WT mice, showed a high expression level in the WT mice, but we noted a slight expression for the TLR4-KO mice. These findings demonstrate that RT-qPCR was a more sensitive technique than NGS.

We noted that miR-96-5p, miR-5122 and miR-1982 showed a very low expression in both genotypes, and no significant difference was found when comparing the WT vs. The TLR4-KO mice. It is noteworthy that when PCR cycles were used, miR-96-5p and miR-5122 were detected, but miR-1982 was not detected by real-time PCR.

After corroborating that the TLR4-KO genotype presented an altered miRNAs pattern in both quantity and diversity by RT-qPCR, we decided to perform a functional analysis with the 52 miRNAs found exclusively in the WT mice with the mdgsa package (Fig 3A). RT-qPCR provided us with information which indicated that the miRNAs not detected in TLR4-KO by the NGS data were detected by qPCR, but at a very low level compared to the WT mice. The bioinformatics results are offered in Fig 3C. Remarkably this figure depicts a strong regulation on routes like neuron death, apoptosis processes and synapsis organization, among others. These results suggest a lower regulation of TLR4-KO during these important routes/processes than the WT mice.

The analysis of the differential expression of miRNAs in the WT vs. TLR4-KO mice shows a deregulation of the miR-200 family and the miR-99b/let-7e/miR-125a cluster in TLR4-KO

We went on to evaluate the differential expression pattern of the miRNAs in the TLR4-WT vs. TLR4-KO mice. The dendrogram diagram illustrated in Fig 4A shows a marked difference in the expression levels of several miRNA in the WT and TLR4-KO mice cortices. Table 5 also provides complete information on the fold changes, statistics and p-values for the different miRNAs.

Fig 4.

Fig 4

Differential expression pattern between the TLR4 and TLR4-KO mice: (A) Heatmap diagram of the comparative TLR4 vs. TLR4-KO mice. (B) Sequence of the five miR-200 family members with the human genomic localization of two clusters containing miR-200 family members; http://atlasgeneticsoncology.org/Genes/GC_MIR141.html. (C) RT-qPCR shows the expression of some mature miRNAs selected in WT and TLR4-KO (miR-141-3p, miR429-5p, miR-182-5p, miR-136-3p, miR-9-5p, miR-125a-3p, miR-125b-5p, miR99a-5p) (n = 6–8). The statistical analysis was performed by the Test-t-. Statistical significance is denoted by a p-value of <0.05.

Table 5. Differential expression results for the miRNAs profiles in the comparative TLR4 vs. TLR4-KO mice.

miRNA logFC logCPM F PValue FDR
mmu-miR-182-5p -3.9719505 13.512520 404.36352 0e+00 0.00e+00
mmu-miR-183-5p -3.8092076 11.610893 244.90941 0e+00 0.00e+00
mmu-miR-429-3p -4.2064135 9.640591 143.75382 0e+00 0.00e+00
mmu-miR-200a-3p -5.0255762 8.843745 116.80459 0e+00 0.00e+00
mmu-miR-200b-3p -3.4471567 8.943224 87.78602 0e+00 0.00e+00
mmu-miR-141-3p -4.6305701 7.620256 58.62150 0e+00 0.00e+00
mmu-miR-9-5p -0.9452873 13.167504 30.57290 0e+00 6.00e-06
mmu-miR-451a -1.9476349 8.113536 28.13968 1e-07 1.92e-05
mmu-miR-200c-3p -2.5575429 7.183744 25.60193 4e-07 6.54e-05
mmu-miR-136-3p -1.2688583 9.777238 25.45641 5e-07 6.54e-05
mmu-miR-99b-5p 1.0160077 14.624300 50.12605 0e+00 0.00e+00
mmu-miR-125a-5p 0.9073845 15.386571 45.41301 0e+00 0.00e+00
mmu-miR-92b-3p 0.9775661 14.065904 42.16099 0e+00 0.00e+00
mmu-miR-125b-5p 0.8289091 14.400916 32.08300 0e+00 3.00e-06
mmu-miR-5121 0.9294424 11.644308 24.86325 6e-07 8.30e-05

When we considered the data in Fig 4A and Table 5, we saw the systematic down-regulation of the family miR-200 components in the TLR4-KO vs. WT mice. Fig 4B illustrates the two subfamilies: the first is composed of miR-141 and miR-200c located in mouse chromosome 6 (chromosome 12 in the human genome); the second subfamily includes miR-200a, miR-200b and miR-429, located in chromosome 4 in the mouse genome (chromosome 1 in the human genome).

In the first subfamily, miR-200a-3p and miR-141-3p were down-regulated 5.02- and 4.63-fold in TLR4-KO compared to WT. Other components like miR-429-3p, miR-200a and miR-200b showed a down-regulation of around 4.5-, 5- and 3.5–fold, respectively (Table 5 and Fig 4A). Notably, these miRNAs are associated with the response to LPS activity [45].

With the heatmap (Fig 4A), we also detected two miRNAs that formed part of the cluster formed by miR-99b/let-7e/miR-125a, which showed a higher expression in the TLR4-KO than in the WT mice. Interestingly, this cluster acts as a negative regulation on the TLR signaling pathway by controlling a set of target genes involved in the receptor pathway [46]. However, before assessing the functional analysis of the pathways modulated by these miRNAs, we validated them by RT-qPCR.

Fig 4C shows that all the miRNAs down-regulated by NGS were also down-regulated by RT-qPCR, which indicates that both techniques strongly correlate. The results showed that whereas the miR-200 family components were down-regulated in TLR4-KO, the miRNAs of cluster miR-99b/let-7e/miR-125a, such as miR-99b and mi-125a, tended to be up-regulated. These results indicate that specific miRNAs clusters are deregulated in the TLR4-KO mice cortices compared to the WT samples, which implies a differential miRNA profile related to the genotype.

Functional analysis of miRNAs shows the major depletion of the inflammatory pathways in the TLR4-KO mice

We next assessed the signaling pathways affected by the deregulation profile of the miRNAs in the cerebral cortices of those mice lacking the TLR4 receptor vs. the WT. For this purpose, a gene set analysis (GSA) was performed along with the Reactome, Biological Processes GO and KEGG pathways. These approaches allowed us to identify etiologic pathways and functional annotations and could, thus, provide novel biological insights [47]. Fig 5 illustrates the GSA results using the bioinformatics and experimental validated targets. The size of the round node was the number of genes in the function. Color was the adjusted p-value.

Fig 5. Enrichment analyses: Enrichment analyses results using miRNA to target the bioinformatics and experimental lists from the miR Database and TargetScan.

Fig 5

Analyses were performed with mdgsa [31] and clusterProfiler [32]. (A) Reactome pathways. (B) Biological Processes GO. (C) KEGG pathways. Abscissa axis indicates if the pathway or function is depleted in the WT or the knockout group. Ordinate axis displays the GO term or the pathway. The size of the round node is the number of genes in the function. Color is the adjusted p-value. Raw p-values were adjusted by FDR (Adjusted p-value < 0.05).

The Reactome pathways analysis (Fig 5A), run with an inferred bioinformatics targets list, revealed that TLR4-KO was depleted of numerous pathways linked with the immunological system and TLRs signaling. For instance, cytokines and interleukin signaling and immune system, “MyD88 dependent cascade initiated on endosome”, “Toll-like Receptor 7/8 (TLR 7/8) Cascade”, “TRAF-6 mediated induction of NF-kβ, MAP kinases upon TLR 7/8 or 9 activation”, etc.

Conversely in the WT group, the analysis showed depleted pathways in relation to the metabolism of inflammatory regulation molecules. The analysis run with the experimental inferred target lists also revealed depleted pathways related to the “Neuronal System” and “Potassium channels” in the knockout group, while the pathways associated with carbohydrate metabolism and olfactory signaling were depleted in the WT mice.

Functional profiling for Biological Processes indicated a significant reduction in several processes linked with the neurodevelopment and neuronal synapse processes in the TLR4-KO group, while the responses to the pheromones processes were depleted in the WT group (Fig 5B).

Finally, the KEGG pathways analysis demonstrated the activity depletion of diverse diseases routes, like diabetes and acute myeloid leukemia, as well as insulin and calcium signaling pathways, in the knockout group. However, the WT group revealed depleted pathways related to retinol and olfactory transduction routes (Fig 5C).

In short, by the NGS technology and bioinformatics approaches, the results revealed new insights into the molecular and biological processes associated with protective effects of TLR-KO against neuroinflammation, brain damage neurodegeneration and alcohol abuse.

Functional enrichment analysis of Toll-like Receptors 7/8

To further elucidate the hierarchical functions of miRNAs in gene regulatory networks in TLR4-KO vs. WT mice, we next performed a complementary and enrichment analysis of some routes obtained by the bioinformatics analysis. We selected Toll-like Receptor 7/8 (TLR 7/8) because these receptors have been related to TLR4 pathways and alcohol abuse [48]. We therefore performed a RT-qPCR analysis of TLR7 and TLR8, and of some genes associated with their signaling response, such as IL12 and INFα. Fig 6 shows that while ethanol, an inflammatory activator, increased the TLR8 mRNA levels in both genotypes, no changes in gene expression were noted in TLR7. With the downstream genes, while no changes were noted in Il12, INFα mRNA significantly increased with ethanol in the WT mice, but not in TR4-KO.

Fig 6. Lack of TLR4 dismisses INFα activation by TLR7/8 Receptors.

Fig 6

(A-D) The RT-qPCR analysis of the WT and TLR4-KO mice cortices with and without EtOH treatment, the relative quantification of receptors TLR7/8, Interleukin IL12 and INFα showed a differential expression of TLR8 by the ethanol inflammatory effect in both genotypes, but alcohol led to the activation of INFα only in the WT mice. A statistical analysis was performed by a two-way ANOVA with n = 6. Statistical significance with a p-value of *<0.05 and **<0.01.

Discussion

TLR4 receptors are highly conserved receptors that play a key role in the innate immune system by recognizing microbial subproducts, such as pathogen-associated molecular pattern and endogenous molecules, which results in inflammatory response activation [49, 50]. The activation of these receptors induces intracellular signaling by the myeloid differentiation factor 88 (MyD88) dependent and MyD88-independent pathways. The final event induces the activation of the downstream transcription factors, such as NF-κβ, among others, involved in cellular responses and the production of proinflammatory cytokines [51].

Recent studies have demonstrated the importance of TLR4 receptors in both metabolic functions and neuropathology because the absence of these receptors prevents metabolic damage related to fatty acids [52], ischemic injury [53] or neuroinflammatory, neurodegenerative and demyelination disorders [54, 55]. Our previous studies have shown the crucial participation of the TLR4 signaling response in ethanol-induced brain damage, neurodegeneration and behavioral dysfunction alteration [56, 57], effects which were not observed in alcohol-treated TLR4-KO mice. By NGS and bioinformatics tools, we recently identified miRNAs that were differentially expressed in chronic alcohol-treated vs. untreated WT or TLR4-KO mice cortices. In particular, we demonstrated the differential expression of the miR-183 cluster (miR-96/-182/-183), miR-200a and miR-200b, which were down-regulated, while mirR-125b was up-regulated in the alcohol-treated WT versus (vs.) untreated mice [17]. The functional enrichment of the miR-183C and miR-200a/b family target genes revealed that neuroinflammatory pathways networks are involved in TLR4 signaling associated with alcohol abuse. Notably, the neuroinflammatory target genes were abolished in TLR4-KO mice. These results raise the question about which miRNAs, genes or signaling proteins are down-regulated in TLR-KO to be able to exert their protective effects.

Here our results demonstrate that the TLR4-KO mice presented a significant lack or down-regulation in several mature miRNAs involved in the inflammatory response compared to the profile observed in the WT mice. Indeed the Venn’s diagram (Fig 2A) shows that 52 miRNAs were exclusively expressed in the WT mice, but these miRs had a very low expression in TLR4-KO, as demonstrated by RT-qPCR. Some of these down-regulated miRNAs, such as miR-382-3p, are involved in the regulation of IL-1β and TLR4 signaling inhibition [40], while miR-27b-5p participates in microbial infection defense [58]. The Venn’s diagram also illustrates that four miRNAs were exclusively expressed in TLR4-KO (miR-1982-3p, miR-5122, miR-677-3p, miR-5114). However, the information, functions and validated targets of this set of miRNAs are practically inexistent and only miR-5122 was detected by RT-qPCR with a very low expression. These results indicate that lack of TLR4 significantly alters the number and expression of several miRNAs. Indeed our previous studies demonstrated that ethanol treatment dysregulated miR-200s family and target genes (e.g. MAPK, IL1R1) in the WT, but not in the TLR4-KO mice treated with alcohol (17). Moreover, we showed that the miR-200 family was significant down-regulated in the TLR4-KO mice, which supports the absence of neuroinflammatory effects of ethanol on mice lacking this receptor.

The functional analysis of the miRNAs exclusively expressed in the WT mice cortices (Fig 3C) provided information on the role of these miRs in neuron death, apoptosis processes, synapsis organization, peptidyl serine signaling or synapsis structure and activity. Interestingly, the down-regulation of these miRNAs in the TLR4-KO mice offered some protection from neuronal death and brain injury in different pathologies. For instance, lack of TLR4 can protect mice from ischemic brain injury [59], brain damage/inflammation after experimental stroke [60], neuroinflammation, brain injury [15], Parkinson’s disease [61] and demyelination associated with alcohol abuse [16]. It is important to stress that the elimination of TLR4 not only abolished the activation of glial cells and the production of inflammatory cytokines in the brain, but could also block the recruitment of peripheral inflammatory cells and increase BBB permeability by participating in some neurodegenerative diseases [62].

From the Venn’s diagram, we also detected 362 miRNAs that were commonly expressed in both genotypes. Using the heatmap to assess the differential expression of the miRs in TLR4 vs. TLR4-KO (Fig 4A), we revealed the presence of different miRNAs belonging to the miR-200 family. All the components of this miRNAs family showed a down-regulation (miR-141 and miR-200a and miR-200b, miR-200c, miR-429) in TLR4-KO vs. WT. Interestingly, miR-200 components like miR-200b and miR-200c, alter the efficiency of TLR4 signaling through the MyD88-dependent pathway, and lack of the TLR4 receptor can modify miR-200 family levels by drastically lowering their expression [63]. Similarly, miR-141 and miR-200c regulate inflammation, while loss of miR-141/200c significantly reduces inflammation, hepatic steatosis and injury in non-alcoholic fatty livers in mice [64]. MiR-141-3p is also involved in chronic inflammatory pain [65], and miR-141 inhibition improves mortality, neurological deficits, and decreased infarct volumes during post-stroke social isolation in aged mice [66]. Likewise, miR-141 and other miR-200 family members also regulate oxidative stress by targeting the p38 (MAPK14) transcript [67, 68]. The heatmap data also demonstrated the up-regulation of the two components of the miR-99b/let-7e/miR-125a cluster (miR99b and miR-125a) in TLR4-KO vs. WT, whereas some studies have demonstrated the anti-inflammatory role of this cluster, which influences the production of pro-inflammatory cytokines in response to LPS [46].

The Functional Profiling for Biological Processes identified the pathways that were affected by the specific miRNA profile in each genotype. In general, inflammatory pathways are attenuated in the TLR-KO genotype [69], such as cytokine and interleukin signaling and inflammatory processes, as demonstrated after brain injury or diabetes in TLR4-KO [70, 71].

One interesting result was that lacks of TLR4 induced the depletion of other TLRs, like TLR7/8. These receptors are located in endosomal membranes and recognize single pathogenic stranded RNAs (ssRNA) and DNAs. TLR7/8 receptors have been associated with innate immunity to autoimmunity [72, 73]. Some studies also suggest that ethanol exposure increases TLR7 expression and the release of let-7b in microglia-derived microvesicles, which contributes to neuroimmune gene induction [74]. The functional analysis of these receptors demonstrated that while ethanol increased the TLR8 mRNA levels in both genotypes, no changes in gene expression were noted in TLR7 when comparing WT and TLR4-KO.

Other studies have demonstrated the role of miR-1906 as an important target of the TLR4 receptor in ischemic damage [75]. We detected miR-1906 in our samples, but with a very low expression. Other authors have suggested the role of either miR-182-5p in the neuroprotective effect of cerebral ischemia-reperfusion injury via regulation by TLR4 [76] or miR-124 and miR-146a in reversing neuropathic pain through the TLR4 receptor [77].

In short, the present study provides insights into the role of miRs associated with the TLR4 response. This receptor modulates and activates the inflammatory signaling response to trigger inflammation and tissue damage as lack of these receptors (TLR4-KO) significantly blocks or reduces the expression of the miRs associated with the inflammatory signaling response by contributing to immune disorders, including neurodegeneration and ischemic damage.

Supporting information

S1 Table

(PDF)

S1 Raw images

(TIF)

Acknowledgments

We are grateful to Dr. S. Akira, who provided us with the TLR4−/− mice, and to M. J. Morillo Bargues and R. Lopez Hidalgo for their excellent technical assistance.

Data Availability

All fataq files are available from the NCBI’s Gene Expression Omnibus database accessible through GEO Series accession number GSE120373.

Funding Statement

This work has been supported by grants from the Spanish Ministry of Science and Innovation (SAF2015-69187R)/ (URL: http://www.ciencia.gob.es/portal/site/MICINN/?lang_choosen=en). And the Institute Carlos III and FEDER funds (RTA-Network, RD16/0017/0004)/ (URL: https://www.isciii.es/Paginas/Inicio.aspx) granted to C.G. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1.Medzhitov R. Toll-like receptors and innate immunity. Nature Reviews Immunology. 2001;1(2):135 10.1038/35100529 [DOI] [PubMed] [Google Scholar]
  • 2.Akira S, Takeda K. Toll-like receptor signalling. Nature Reviews Immunology. 2004;4(7):499 10.1038/nri1391 [DOI] [PubMed] [Google Scholar]
  • 3.Mishra BB, Gundra UM, Teale JM. Expression and distribution of Toll-like receptors 11–13 in the brain during murine neurocysticercosis. Journal of Neuroinflammation. 2008;5(1):53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Arroyo DS, Soria JA, Gaviglio EA, Rodriguez-Galan MC, Iribarren P. Toll-like receptors are key players in neurodegeneration. International immunopharmacology. 2011;11(10):1415–21. 10.1016/j.intimp.2011.05.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Xiang W, Chao Z-Y, Feng D-Y. Role of Toll-like receptor/MYD88 signaling in neurodegenerative diseases. Reviews in the neurosciences. 2015;26(4):407–14. 10.1515/revneuro-2014-0067 [DOI] [PubMed] [Google Scholar]
  • 6.O’Connell RM, Rao DS, Baltimore D. microRNA regulation of inflammatory responses. Annual review of immunology. 2012;30:295–312. 10.1146/annurev-immunol-020711-075013 [DOI] [PubMed] [Google Scholar]
  • 7.O’neill LA, Sheedy FJ, McCoy CE. MicroRNAs: the fine-tuners of Toll-like receptor signalling. Nature reviews immunology. 2011;11(3):163 10.1038/nri2957 [DOI] [PubMed] [Google Scholar]
  • 8.Pasquinelli AE. MicroRNAs and their targets: recognition, regulation and an emerging reciprocal relationship. Nature Reviews Genetics. 2012;13(4):271 10.1038/nrg3162 [DOI] [PubMed] [Google Scholar]
  • 9.Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. cell. 2004;116(2):281–97. 10.1016/s0092-8674(04)00045-5 [DOI] [PubMed] [Google Scholar]
  • 10.Fichtlscherer S, De Rosa S, Fox H, Schwietz T, Fischer A, Liebetrau C, et al. Circulating microRNAs in patients with coronary artery disease. Circulation research. 2010;107(5):677–84. 10.1161/CIRCRESAHA.109.215566 [DOI] [PubMed] [Google Scholar]
  • 11.Schickel R, Boyerinas B, Park S, Peter M. MicroRNAs: key players in the immune system, differentiation, tumorigenesis and cell death. Oncogene. 2008;27(45):5959 10.1038/onc.2008.274 [DOI] [PubMed] [Google Scholar]
  • 12.Slota JA, Booth SA. MicroRNAs in neuroinflammation: implications in disease pathogenesis, biomarker discovery and therapeutic applications. Non-coding RNA. 2019;5(2):35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Blanco AM, Vallés SL, Pascual M, Guerri C. Involvement of TLR4/type I IL-1 receptor signaling in the induction of inflammatory mediators and cell death induced by ethanol in cultured astrocytes. The Journal of Immunology. 2005;175(10):6893–9. 10.4049/jimmunol.175.10.6893 [DOI] [PubMed] [Google Scholar]
  • 14.Fernandez-Lizarbe S, Pascual M, Guerri C. Critical role of TLR4 response in the activation of microglia induced by ethanol. The Journal of Immunology. 2009;183(7):4733–44. 10.4049/jimmunol.0803590 [DOI] [PubMed] [Google Scholar]
  • 15.Alfonso-Loeches S, Pascual-Lucas M, Blanco AM, Sanchez-Vera I, Guerri C. Pivotal role of TLR4 receptors in alcohol-induced neuroinflammation and brain damage. Journal of Neuroscience. 2010;30(24):8285–95. 10.1523/JNEUROSCI.0976-10.2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Alfonso-Loeches S, Pascual M, Gómez-Pinedo U, Pascual-Lucas M, Renau-Piqueras J, Guerri C. Toll-like receptor 4 participates in the myelin disruptions associated with chronic alcohol abuse. Glia. 2012;60(6):948–64. 10.1002/glia.22327 [DOI] [PubMed] [Google Scholar]
  • 17.Ureña-Peralta J, Alfonso-Loeches S, Cuesta-Diaz C, García-García F, Guerri C. Deep sequencing and miRNA profiles in alcohol-induced neuroinflammation and the TLR4 response in mice cerebral cortex. Scientific reports. 2018;8(1):15913 10.1038/s41598-018-34277-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Franklin KB, Paxinos G. The mouse brain in stereotaxic coordinates: Academic press; New York:; 2008. [Google Scholar]
  • 19.Chomczynski P, Sacchi N. Single-step method of RNA isolation by acid guanidinium thiocyanate-phenol-chloroform extraction. Analytical biochemistry. 1987;162(1):156–9. 10.1006/abio.1987.9999 [DOI] [PubMed] [Google Scholar]
  • 20.Hoshino K, Takeuchi O, Kawai T, Sanjo H, Ogawa T, Takeda Y, et al. Cutting edge: Toll-like receptor 4 (TLR4)-deficient mice are hyporesponsive to lipopolysaccharide: evidence for TLR4 as the Lps gene product. The Journal of Immunology. 1999;162(7):3749–52. [PubMed] [Google Scholar]
  • 21.Pfaffl MW. A new mathematical model for relative quantification in real-time RT–PCR. Nucleic acids research. 2001;29(9):e45–e. 10.1093/nar/29.9.e45 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Martin M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet journal. 2011;17(1):10–2. [Google Scholar]
  • 23.Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nature methods. 2012;9(4):357 10.1038/nmeth.1923 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Hammond SM. An overview of microRNAs. Advanced drug delivery reviews. 2015;87:3–14. 10.1016/j.addr.2015.05.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ziemann M, Kaspi A, El-Osta A. Evaluation of microRNA alignment techniques. Rna. 2016;22(8):1120–38. 10.1261/rna.055509.115 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Liao Y, Smyth GK, Shi W. The R package Rsubread is easier, faster, cheaper and better for alignment and quantification of RNA sequencing reads. Nucleic acids research. 2019;47(8):e47–e. 10.1093/nar/gkz114 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Kozomara A, Birgaoanu M, Griffiths-Jones S. miRBase: from microRNA sequences to function. Nucleic acids research. 2018;47(D1):D155–D62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Binns D, Dimmer E, Huntley R, Barrell D, O’donovan C, Apweiler R. QuickGO: a web-based tool for Gene Ontology searching. Bioinformatics. 2009;25(22):3045–6. 10.1093/bioinformatics/btp536 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26(1):139–40. 10.1093/bioinformatics/btp616 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. Journal of the Royal statistical society: series B (Methodological). 1995;57(1):289–300. [Google Scholar]
  • 31.Montaner D, Dopazo J. Multidimensional gene set analysis of genomic data. PloS one. 2010;5(4):e10348 10.1371/journal.pone.0010348 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Yu G, Wang L-G, Han Y, He Q-Y. clusterProfiler: an R package for comparing biological themes among gene clusters. Omics: a journal of integrative biology. 2012;16(5):284–7. 10.1089/omi.2011.0118 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Garcia-Garcia F, Panadero J, Dopazo J, Montaner D. Integrated gene set analysis for microRNA studies. Bioinformatics. 2016;32(18):2809–16. 10.1093/bioinformatics/btw334 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Consortium GO. The gene ontology resource: 20 years and still GOing strong. Nucleic acids research. 2018;47(D1):D330–D8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kanehisa M, Sato Y, Furumichi M, Morishima K, Tanabe M. New approach for understanding genome variations in KEGG. Nucleic acids research. 2018;47(D1):D590–D5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Croft D, Mundo AF, Haw R, Milacic M, Weiser J, Wu G, et al. The Reactome pathway knowledgebase. Nucleic acids research. 2013;42(D1):D472–D7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Cohen JL, Ata AE, Jackson NL, Rahn EJ, Ramaker RC, Cooper S, et al. Differential stress induced c-Fos expression and identification of region-specific miRNA-mRNA networks in the dorsal raphe and amygdala of high-responder/low-responder rats. Behavioural brain research. 2017;319:110–23. 10.1016/j.bbr.2016.11.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Sun L-Q, Guo G-L, Zhang S, Yang L-L. Effects of MicroRNA-592-5p on Hippocampal neuron injury following hypoxic-ischemic brain damage in neonatal mice-involvement of PGD2/DP and PTGDR. Cellular Physiology and Biochemistry. 2018;45(2):458–73. 10.1159/000486923 [DOI] [PubMed] [Google Scholar]
  • 39.Liu WY, Yang Z, Sun Q, Yang X, Hu Y, Xie H, et al. miR-377-3p drives malignancy characteristics via upregulating GSK-3β expression and activating NF-κB pathway in hCRC cells. Journal of cellular biochemistry. 2018;119(2):2124–34. 10.1002/jcb.26374 [DOI] [PubMed] [Google Scholar]
  • 40.Lei J, Fu Y, Zhuang Y, Zhang K, Lu D. miR-382-3p suppressed IL-1β induced inflammatory response of chondrocytes via the TLR4/MyD88/NF-κB signaling pathway by directly targeting CX43. Journal of cellular physiology. 2019. [DOI] [PubMed] [Google Scholar]
  • 41.Chen H, Zhou Y, Wang Z-Y, Yan B-X, Zhou W-F, Wang T-T, et al. Exosomal microRNA profiles from serum and cerebrospinal fluid in neurosyphilis. Sexually transmitted infections. 2019;95(4):246–50. 10.1136/sextrans-2018-053813 [DOI] [PubMed] [Google Scholar]
  • 42.Fu J, Hao L, Tian Y, Liu Y, Gu Y, Wu J. miR-199a-3p is involved in estrogen-mediated autophagy through the IGF-1/mTOR pathway in osteocyte-like MLO-Y4 cells. Journal of cellular physiology. 2018;233(3):2292–303. 10.1002/jcp.26101 [DOI] [PubMed] [Google Scholar]
  • 43.Yu K, Li N, Cheng Q, Zheng J, Zhu M, Bao S, et al. miR-96-5p prevents hepatic stellate cell activation by inhibiting autophagy via ATG7. Journal of Molecular Medicine. 2018;96(1):65–74. 10.1007/s00109-017-1593-6 [DOI] [PubMed] [Google Scholar]
  • 44.Wang X, Song C, Zhou X, Han X, Li J, Wang Z, et al. Mitochondria associated microRNA expression profiling of heart failure. BioMed research international. 2017;2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Xiao J, Tang J, Chen Q, Tang D, Liu M, Luo M, et al. miR-429 regulates alveolar macrophage inflammatory cytokine production and is involved in LPS-induced acute lung injury. Biochemical Journal. 2015;471(2):281–91. 10.1042/BJ20131510 [DOI] [PubMed] [Google Scholar]
  • 46.Curtale G, Renzi TA, Mirolo M, Drufuca L, Albanese M, De Luca M, et al. Multi-Step Regulation of the TLR4 Pathway by the miR-125a~ 99b~ let-7e Cluster. Frontiers in immunology. 2018;9:2037 10.3389/fimmu.2018.02037 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Pers TH. Gene set analysis for interpreting genetic studies. Human molecular genetics. 2016;25(R2):R133–R40. 10.1093/hmg/ddw249 [DOI] [PubMed] [Google Scholar]
  • 48.Massey VL, Qin L, Cabezas J, Caballeria J, Sancho-Bru P, Bataller R, et al. TLR 7-let-7 Signaling Contributes to Ethanol-Induced Hepatic Inflammatory Response in Mice and in Alcoholic Hepatitis. Alcoholism: Clinical and Experimental Research. 2018;42(11):2107–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140(6):805–20. 10.1016/j.cell.2010.01.022 [DOI] [PubMed] [Google Scholar]
  • 50.Yamamoto M, Takeda K. Current views of toll-like receptor signaling pathways. Gastroenterology research and practice. 2010;2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nature immunology. 2010;11(5):373 10.1038/ni.1863 [DOI] [PubMed] [Google Scholar]
  • 52.Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H, Flier JS. TLR4 links innate immunity and fatty acid–induced insulin resistance. The Journal of clinical investigation. 2006;116(11):3015–25. 10.1172/JCI28898 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Wang Y, Ge P, Zhu Y. TLR2 and TLR4 in the brain injury caused by cerebral ischemia and reperfusion. Mediators of inflammation. 2013;2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Rosenberger K, Dembny P, Derkow K, Engel O, Krüger C, Wolf SA, et al. Intrathecal heat shock protein 60 mediates neurodegeneration and demyelination in the CNS through a TLR4-and MyD88-dependent pathway. Molecular neurodegeneration. 2015;10(1):5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Perez-Pardo P, Dodiya HB, Engen PA, Forsyth CB, Huschens AM, Shaikh M, et al. Role of TLR4 in the gut-brain axis in Parkinson’s disease: a translational study from men to mice. Gut. 2019;68(5):829–43. 10.1136/gutjnl-2018-316844 [DOI] [PubMed] [Google Scholar]
  • 56.Pla A, Pascual M, Guerri C. Autophagy constitutes a protective mechanism against ethanol toxicity in mouse astrocytes and neurons. PloS one. 2016;11(4):e0153097 10.1371/journal.pone.0153097 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Pla A, Pascual M, Renau-Piqueras J, Guerri C. TLR4 mediates the impairment of ubiquitin-proteasome and autophagy-lysosome pathways induced by ethanol treatment in brain. Cell death & disease. 2014;5(2):e1066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Zhou R, Gong A-Y, Eischeid AN, Chen X-M. miR-27b targets KSRP to coordinate TLR4-mediated epithelial defense against Cryptosporidium parvum infection. PLoS pathogens. 2012;8(5):e1002702 10.1371/journal.ppat.1002702 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Hyakkoku K, Hamanaka J, Tsuruma K, Shimazawa M, Tanaka H, Uematsu S, et al. Toll-like receptor 4 (TLR4), but not TLR3 or TLR9, knock-out mice have neuroprotective effects against focal cerebral ischemia. Neuroscience. 2010;171(1):258–67. 10.1016/j.neuroscience.2010.08.054 [DOI] [PubMed] [Google Scholar]
  • 60.Caso JR, Pradillo JM, Hurtado O, Lorenzo P, Moro MA, Lizasoain I. Toll-like receptor 4 is involved in brain damage and inflammation after experimental stroke. Circulation. 2007;115(12):1599 10.1161/CIRCULATIONAHA.106.603431 [DOI] [PubMed] [Google Scholar]
  • 61.Campolo M, Paterniti I, Siracusa R, Filippone A, Esposito E, Cuzzocrea S. TLR4 absence reduces neuroinflammation and inflammasome activation in Parkinson’s diseases in vivo model. Brain, behavior, and immunity. 2019;76:236–47. 10.1016/j.bbi.2018.12.003 [DOI] [PubMed] [Google Scholar]
  • 62.Kempuraj D, Thangavel R, Selvakumar GP, Zaheer S, Ahmed ME, Raikwar SP, et al. Brain and peripheral atypical inflammatory mediators potentiate neuroinflammation and neurodegeneration. Frontiers in cellular neuroscience. 2017;11:216 10.3389/fncel.2017.00216 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Wendlandt EB, Graff JW, Gioannini TL, McCaffrey AP, Wilson ME. The role of microRNAs miR-200b and miR-200c in TLR4 signaling and NF-κB activation. Innate immunity. 2012;18(6):846–55. 10.1177/1753425912443903 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Tran M, Lee S-M, Shin D-J, Wang L. Loss of miR-141/200c ameliorates hepatic steatosis and inflammation by reprogramming multiple signaling pathways in NASH. JCI insight. 2017;2(21). 10.1172/jci.insight.96094 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Shen W, Xu X, Zhai N, Zhou Z, Shao J, Yu Y. Potential mechanisms of microRNA-141-3p to alleviate chronic inflammatory pain by downregulation of downstream target gene HMGB1: in vitro and in vivo studies. Gene therapy. 2017;24(6):353 10.1038/gt.2017.28 [DOI] [PubMed] [Google Scholar]
  • 66.Verma R, Ritzel RM, Harris NM, Lee J, Kim T, Pandi G, et al. Inhibition of miR-141-3p Ameliorates the Negative Effects of Poststroke Social Isolation in Aged Mice. Stroke. 2018;49(7):1701–7. 10.1161/STROKEAHA.118.020627 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Magenta A, Cencioni C, Fasanaro P, Zaccagnini G, Greco S, Sarra-Ferraris G, et al. miR-200c is upregulated by oxidative stress and induces endothelial cell apoptosis and senescence via ZEB1 inhibition. Cell death and differentiation. 2011;18(10):1628 10.1038/cdd.2011.42 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Mateescu B, Batista L, Cardon M, Gruosso T, De Feraudy Y, Mariani O, et al. miR-141 and miR-200a act on ovarian tumorigenesis by controlling oxidative stress response. Nature medicine. 2011;17(12):1627 10.1038/nm.2512 [DOI] [PubMed] [Google Scholar]
  • 69.Yao X, Liu S, Ding W, Yue P, Jiang Q, Zhao M, et al. TLR4 signal ablation attenuated neurological deficits by regulating microglial M1/M2 phenotype after traumatic brain injury in mice. Journal of neuroimmunology. 2017;310:38–45. 10.1016/j.jneuroim.2017.06.006 [DOI] [PubMed] [Google Scholar]
  • 70.Zhu H-t, Bian C, Yuan J-c, Chu W-h, Xiang X, Chen F, et al. Curcumin attenuates acute inflammatory injury by inhibiting the TLR4/MyD88/NF-κB signaling pathway in experimental traumatic brain injury. Journal of Neuroinflammation. 2014;11(1):59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Devaraj S, Tobias P, Jialal I. Knockout of toll-like receptor-4 attenuates the pro-inflammatory state of diabetes. Cytokine. 2011;55(3):441–5. 10.1016/j.cyto.2011.03.023 [DOI] [PubMed] [Google Scholar]
  • 72.Krieg AM, Vollmer J. Toll-like receptors 7, 8, and 9: linking innate immunity to autoimmunity. Immunological reviews. 2007;220(1):251–69. [DOI] [PubMed] [Google Scholar]
  • 73.Petes C, Odoardi N, Gee K. The toll for trafficking: toll-like receptor 7 delivery to the endosome. Frontiers in immunology. 2017;8:1075 10.3389/fimmu.2017.01075 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Coleman LG, Zou J, Crews FT. Microglial-derived miRNA let-7 and HMGB1 contribute to ethanol-induced neurotoxicity via TLR7. Journal of Neuroinflammation. 2017;14(1):22 10.1186/s12974-017-0799-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Xu X, Wen Z, Zhao N, Xu X, Wang F, Gao J, et al. MicroRNA-1906, a novel regulator of toll-like receptor 4, ameliorates ischemic injury after experimental stroke in mice. Journal of Neuroscience. 2017;37(43):10498–515. 10.1523/JNEUROSCI.1139-17.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Wang J, Xu Z, Chen X, Li Y, Chen C, Wang C, et al. MicroRNA-182-5p attenuates cerebral ischemia-reperfusion injury by targeting Toll-like receptor 4. Biochemical and biophysical research communications. 2018;505(3):677–84. 10.1016/j.bbrc.2018.09.165 [DOI] [PubMed] [Google Scholar]
  • 77.Grace PM, Strand KA, Galer EL, Maier SF, Watkins LR. MicroRNA-124 and microRNA-146a both attenuate persistent neuropathic pain induced by morphine in male rats. Brain research. 2018;1692:9–11. 10.1016/j.brainres.2018.04.038 [DOI] [PMC free article] [PubMed] [Google Scholar]

Decision Letter 0

Pratibha V Nerurkar

21 Apr 2020

PONE-D-19-31787

Lack of TLR4 modifies the miRNAs profile and attenuates inflammatory signaling pathways

PLOS ONE

Dear DR Guerri,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses all of the points raised during the review process.

We would appreciate receiving your revised manuscript by Jun 05 2020 11:59PM. When you are ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter.

To enhance the reproducibility of your results, we recommend that if applicable you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. For instructions see: http://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). This letter should be uploaded as separate file and labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. This file should be uploaded as separate file and labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. This file should be uploaded as separate file and labeled 'Manuscript'.

Please note while forming your response, if your article is accepted, you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out.

We look forward to receiving your revised manuscript.

Kind regards,

Pratibha V. Nerurkar, Ph.D

Academic Editor

PLOS ONE

Journal Requirements:

When submitting your revision, we need you to address these additional requirements.

1. Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at

http://www.journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and http://www.journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf

2. We note that you have included the phrase “data not shown” in your manuscript. Unfortunately, this does not meet our data sharing requirements. PLOS does not permit references to inaccessible data. We require that authors provide all relevant data within the paper, Supporting Information files, or in an acceptable, public repository. Please add a citation to support this phrase or upload the data that corresponds with these findings to a stable repository (such as Figshare or Dryad) and provide and URLs, DOIs, or accession numbers that may be used to access these data. Or, if the data are not a core part of the research being presented in your study, we ask that you remove the phrase that refers to these data.

3. Please include captions for your Supporting Information files at the end of your manuscript, and update any in-text citations to match accordingly. Please see our Supporting Information guidelines for more information: http://journals.plos.org/plosone/s/supporting-information.

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Yes

**********

2. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: I Don't Know

Reviewer #2: Yes

Reviewer #3: Yes

**********

3. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Yes

**********

4. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: No

Reviewer #2: Yes

Reviewer #3: Yes

**********

5. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: In the article entitled “Lack of TLR4 modifies the miRNAs profile and attenuates inflammatory signaling pathways,” by Urena-Peralta et al., the authors used the cerebral cortex from TLR4 KO and wildtype mice with next generation sequencing to determine how TLR4 regulates miRNA.

Major Concerns:

1) Why were female mice only used? Any role of the estrogen cycle?

2) Discussion is a bit long

Minor Concerns:

1) Please have a native English speaker edit for language

Reviewer #2: Comments to author

In the present study Ureña-Peralta and colleagues investigated the role of Toll-like 4 receptors (TLR4s) for the expression of microRNAs relevant to inflammatory pathways in mouse cerebral cortex. The authors utilized an experimental strategy, where cortices of transgenic TLR4 KO mice and WT controls, were analyzed with next generation sequencing targeting specific microRNAs, followed by validation of microRNA expression using qPCR. The authors first demonstrate the effectiveness of the global TLR4 KO approach on attenuating long-term EtOH-induced inflammatory markers (e.g. IL-1R, IL-1beta, IL-6 and COX2). They subsequently, found that absence of TLR4 significantly downregulated miR-382-3p, miR-27b-5p, miR-592-5p and miR-377-3p, as predicted by NGS; Some of these targets are relevant for TLR4/MyD88/NF-κB signaling and brain injury. Further, the authors investigated the differential expression pattern of the miRNAs and found that TLR4-KO mice exhibit downregulation of the miR-200 family and upregulation of the miR-99b/let-7e/miR-125a cluster. The authors further employed bioinformatics and functional analysis for exploring the role of the specific miRNA profile of the WTs vs TLR4-KOs on central molecular pathways involved in neuroinflammation, and found depletion of cytokine and interleukin signaling, MAPK and ion Channels routes, MyD88 pathways, NF-κB and TLR7/8 pathways. Though the comparative strategy of this study is elegant and the results are novel, additional experiments are needed to support the authors claims.

The sample-size used for the NGS analysis is very small (n=3 WT vs n=2 TLR4 KOs). This is a big concern as NGS technologies, as all other biotechnologies, are affected by both biological and technical variability. It would be beneficial to increase the n-number of the groups to gain statistical power and achieve comparability with your very clear qPCR data.

In your methods section, please provide details on brain extraction and dissection: Did you perfuse the brains? Did you use a brain matrix? What were your estimated coordinates and the thickness of the brain section?

It would be relevant to include in your discussion a comparison to the findings of your previous TLR4 study (Ureña-Peralta et al., 2018), as some of the miRNAs uncovered are similar.

Please discuss the limitations of the global TLR4 KO strategy: Are you expecting these effects to be due to CNS TLR4s or secondary effects due to peripheral immune-to-brain signaling, or blood-brain-barrier endothelial TLR4s?

General manuscript comments:

The image resolution of the figures is very poor! It in many cases not possible to read the figure labels. It makes it extremely difficult to evaluate consistency between what is written in the results-section and the actual data - Please revise.

Several of the figure-legends are incomplete, for instance some are missing n-numbers, while the figure legend for figure 1 is unstructured and missing (C) and (D). Please revise the figure-legends.

The language quality varies throughout the individual sections of the manuscript. The NGS-methods and discussion-sections are very well written, while the introduction has lacks (see minor comments below). Please make the language more coherent.

Language, typos and understanding corrections:

Please correct PAMPs from pattern associated molecular pattern to pathogen associated molecular pattern page 3 line 3.

Please revise the sentence: “The sequence-complementary mechanism of miRNA activity exploits combinatorial regulate the expression of genes by repressing the translation of their complementary target genes (9)”. Page 3, section 3, line 2. Do you mean: “….activity exploits combinatorial regulation of gene expression by….”?

Please revise aim 2 on page 4, it is unclear what you mean: “2) if there are compensatory changes in certain miRNAs that could explain the lack of effects with the neuroinflammation associated with alcohol intake”.

In the introduction you refer to your previous study (ref 17), which clearly is linked to this aim, however you don’t specify what the “lack of effects” were.

Furthermore, based on this study a question was raised “These results raise the question as to what the impact of miRNAs profile in mice is lacking TLR4 response?”. If this question is linked to aim 2, it needs to be further clarified, also it is unclear what you mean – please revise the question.

On page 5, under the Alcohol treatment section, line 10-11: You have written “obtained” several times, hence the sentence has become fragmented, also please state the actual BAL-values as this is useful information for the reader.

On page 10 under the statistical section, line 2, Western blotting results are mentioned. However, there is no description of such method or data elsewhere in the manuscript.

On page 19, line 1: “Figure 4B, obtained from, illustrate the two subfamilies…”, please specify what it was “obtained from” or simply delete the obtained from, it is unclear what you mean.

Reviewer #3: Summary

TLR4 is a member of toll-like receptors (TLRs). Lack of TLR4 has been shown to protect against inflammatory processes, neuroinflammation cause by chronic alcohol consumption. The authors aimed to understand whether the reduced inflammation in the cortex of TLR4 knock out mice was mediated by an alteration in the expression of miRNAs involved in inflammatory pathways. Using a next-generation sequencing (NGS) approach they screened the cerebral cortex of TLR4 knock out mice and wild type controls to identify miRNAs that were differentially regulated in TLR4 knock out mice and controls. Understanding how TLR4 mediates neuroinflammation in chronic alcohol consumption is very important because it could shed light on ways to modulate these processes and prevent brain damage.

Critique

1. The manuscript is technically sound and the statistical analysis has been performed appropriately and rigorously. The results obtained by sequencing are always validated by RT-PCR and the appropriate controls are included in each figure. Moreover the data support the conclusions.

2. One major concern is the observation that TLR4KO mice show depletion of TLR7/8. How can you be sure that the micro-RNA differential expression is not due to the lack of TLR7/8. Do TLR7/8 mice show micro-RNA-dysregulation?

3. The authors could focus on a few relevant micro-RNAs involved in inflammation that are differentially expressed and identify the effectors of the TLR4 signaling pathway that modulate these micro-RNAs.

4. The authors could then exogenously modulate the expression (overexpression or knockdown) or function of specific effectors in TLR4 KO mice to rescue the expression of specific micro-RNAs

5. While the statistical analysis has been performed appropriately, the description of the statistical analysis should be described for each panel (it is for example missing in Figure 3b).

6. Figure resolution is low and figure layout can be improved. In Figure 4, for example, some panels are extremely big (Figure 4a) while others are barely visible (Figure 4b).

7. Figure 1: Figure 1C-D description is missing in the figure legend.

8. Figure 4b: Specify that the chromosomal localization refers to the human genome both in the legend and the main text, where there is only reference to the mouse genome, which could create confusion.

**********

6. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

Reviewer #3: No

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files to be viewed.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email us at figures@plos.org. Please note that Supporting Information files do not need this step.

PLoS One. 2020 Aug 11;15(8):e0237066. doi: 10.1371/journal.pone.0237066.r002

Author response to Decision Letter 0


25 Jun 2020

Answer to reviewers

Reviewer #1:

Major Concerns:

1) Why were female mice only used? Any role of the estrogen cycle?

Thank you very much for you comment. We used female mice as previous results from our laboratory demonstrated that females are more sensitive to the neuroinflammatory effects of alcohol effects than males (Alfonso-Loeches et al., 2013; Pascual et al., 2017). A comment has been included on p. 5. Another reason was the price to perform next-generation sequencing (NGS). In fact we initially programmed to perform NGS in the cortices of males and females, but the prices to do NGS were twice as high as we had to add four additional groups (WT, WT + Ethanol, TLR4-KO, TLR4-KO +ethanol).

Regarding the effects of the estrogen cycle, although they are very important and could mediate some effects, we did not consider them in the present study.

2) Discussion is a bit long.

As suggested, we have attempt to reduce the Discussion but, at the same time, we have included some comments in response to the reviewers, which made the Discussion longer.

Minor Concerns:

1) Please have a native English speaker edit for language.

Thank you very much for your comment. The manuscript has been revised by a native English speaker.

Reviewer #2: Comments to author.

1) The sample-size used for the NGS analysis is very small (n=3 WT vs n=2 TLR4 KOs). This is a big concern as NGS technologies, as all other biotechnologies, are affected by both biological and technical variability. It would be beneficial to increase the n-number of the groups to gain statistical power and achieve comparability with your very clear qPCR data.

Thank you very much for your comment. As stated in the Material and Methods section (p. 7), each sample is a pool from three different mice cortices and, therefore, the three pools in the WT is the average of nine different cortices. However in TLR4 KO, we had problems with one pooled sample and we used only two pooled samples that were the average of six mice cortices. We have modified the section RNA Quantity and quality determinations paragraph to make it clearer (p. 7-8).

2) In your methods section, please provide details on brain extraction and dissection: Did you perfuse the brains? Did you use a brain matrix? What were your estimated coordinates and the thickness of the brain section?

Thank you very much for your question. On p. 5 we have described details of the procedure used for brain extraction and cortices dissection, and how cortices were processed. Indeed we have added a new section Cerebral cortex dissection (p. 5) in which we describe the specific procedure used. Cortices dissection was performed by the same person using atlas coordinates. Cortices were weighed and immediately snap-frozen in liquid nitrogen until used for further determination analyses. The weight of cortices was similar in all the groups.

3) It would be relevant to include in your discussion a comparison to the findings of your previous TLR4 study (Ureña-Peralta et al., 2018), as some of the miRNAs uncovered are similar.

As suggested, we have discussed and compared previous findings to the present results. In particular, we have discussed the relevance of some miRNAs that were dysregulated in the ethanol-treated WT mice, but not in TLR4-KO mice, as lack of TLR4 receptors down-regulates these miRNAs pathways (p. 24).

4) Please discuss the limitations of the global TLR4 KO strategy: Are you expecting these effects to be due to CNS TLR4s or secondary effects due to peripheral immune-to-brain signalling, or blood-brain-barrier endothelial TLR4s?

As suggested, we have discussed both the limitation of the TLR4 KO strategy to study neuroinflammation, and the potential involvement of peripheral immune-to-brain signaling in neuroinflammation (p. 24)

General manuscript comments:

5) The image resolution of the figures is very poor! It in many cases not possible to read the figure labels. It makes it extremely difficult to evaluate consistency between what is written in the results-section and the actual data. Please revise.

We appreciate your comment and have included the images in TIFF. We hope that they it is now easier to evaluate and observe the details and figure labels.

6) Several of the figure-legends are incomplete, for instance some are missing n-numbers, while the figure legend for figure 1 is unstructured and missing (C) and (D). Please revise the figure-legends.

Thank you very much for drawing our attention, and we apologize for these mistakes. We have revised all the figures legends and included the correct numbers. Figure 1 has been arranged.

7) The language quality varies throughout the individual sections of the manuscript. The NGS-methods and discussion-sections are very well written, while the introduction has lacks (see minor comments below). Please make the language more coherent.

We thank the reviewer for drawing our attention and have corrected the unclear text and sentences.

-Language, typos and understanding corrections:

-Please correct PAMPs from pattern associated molecular pattern to pathogen associated molecular pattern page 3 line 3.

Thank you very much. We have corrected all the typos.

-Please revise the sentence: “The sequence-complementary mechanism of miRNA activity exploits combinatorial regulate the expression of genes by repressing the translation of their complementary target genes (9). Page 3, section 3, line 2. Do you mean: (“…activity exploits combinatorial regulation of gene expression by…”?).

These sentences have been revised and appropriately changed.

-Please revise aim 2 on page 4, it is unclear what you mean: “2) if there are compensatory changes in certain miRNAs that could explain the lack of effects with the neuroinflammation associated with alcohol intake”.

Thank you, they have been corrected.

-In the introduction you refer to your previous study (ref 17), which clearly is linked to this aim, however you don’t specify what the “lack of effects” were.

We have clarified “lack of effects”.

-Furthermore, based on this study a question was raised “These results raise the question as to what the impact of miRNAs profile in mice is lacking TLR4 response?”. If this question is linked to aim 2, it needs to be further clarified, also it is unclear what you mean – please revise the question.

The question in aim 2 has been changed to a more specific aim.

-On page 5, under the Alcohol treatment section, line 10-11: You have written “obtained” several times; hence the sentence has become fragmented, also please state the actual BAL-values as this is useful information for the reader.

The sentence has been rewritten. Thank you for drawing our attention to this.

-On page 10 under the statistical section, line 2, Western blotting results are mentioned. However, there is no description of such method or data elsewhere in the manuscript.

On page 10, the sentence of the “Western blotting results” has been eliminated.

-On page 19, line 1: “Figure 4B, obtained from, illustrate the two subfamilies…”, please specify what it was “obtained from” or simply delete the obtained from, it is unclear what you mean.

As suggested, we have deleted “obtained from”.

We would like to thank the reviewer for his/her help and suggestions to clarify some parts of the text. All the comments have been taken into account and corrections made.

Reviewer #3: Summary

Critique

1) The manuscript is technically sound and the statistical analysis has been performed appropriately and rigorously. The results obtained by sequencing are always validated by RT-PCR and the appropriate controls are included in each figure. Moreover, the data support the conclusions.

2) One major concern is the observation that TLR4KO mice show depletion of TLR7/8. How can you be sure that the micro-RNA differential expression is not due to the lack of TLR7/8? Do TLR7/8 mice show micro-RNA-dysregulation?

Thank you very much for your appropriate comment. We have performed an additional experiment to evaluate the gene expression levels of receptors TLR7 and TLR8, as well as two genes involved in this pathway. In Fig. 6 we now show that while ethanol increased the TLR8 mRNA levels in both genotypes, no changes in gene expression were noted in TLR7 compared to WT and TLR4KO (see Fig. 6, the results on p. 22 and the Discussion on p. 25 ).

.3) The authors could focus on a few relevant micro-RNAs involved in inflammation that are differentially expressed and identify the effectors of the TLR4 signalling pathway that modulate these micro-RNAs.

Thank you for your comment; the genes analysed in figure 1 (Il1R1, Traf6 and IL6) in addition to the INFα, analyzed in the new figure (Fig. 6), are related with the miR200 family. We hope you have resolved your doubts.

4) The authors could then exogenously modulate the expression (overexpression or knockdown) or function of specific effectors in TLR4 KO mice to rescue the expression of specific micro-RNAs.

We appreciate your comment. In fact although we have attempted to exogenously modulate the expression of TLR4-KO in cortical glial cells in culture from the TLR4-KO mice, we still obtained no clear conclusions.

5) While the statistical analysis has been performed appropriately, the description of the statistical analysis should be described for each panel (it is for example missing in Figure 3b).

We appreciate your comment and have included the statistical analysis in all the figure legends.

6). Figure resolution is low and figure layout can be improved. In Figure 4, for example, some panels are extremely big (Figure 4a) while others are barely visible (Figure 4b).

Thank you very much for your comment. We have provided figures at higher resolutions. The panels for Fig. 4A and 4B have been modified.

7). Figure 1: Figure 1C-D description is missing in the figure legend.

We apologize for this error. We have modified the legend of Figure 1.

8). Figure 4b: Specify that the chromosomal localization refers to the human genome both in the legend and the main text, where there is only reference to the mouse genome, which could create confusion.

Thank you very much for your useful comment. We have now modified the text according to your suggestions.

Attachment

Submitted filename: Response to Reviewers.docx

Decision Letter 1

Pratibha V Nerurkar

21 Jul 2020

Lack of TLR4 modifies the miRNAs profile and attenuates inflammatory signaling pathways

PONE-D-19-31787R1

Dear Dr. Guerri,

We’re pleased to inform you that your manuscript has been judged scientifically suitable for publication and will be formally accepted for publication once it meets all outstanding technical requirements.

Within one week, you’ll receive an e-mail detailing the required amendments. When these have been addressed, you’ll receive a formal acceptance letter and your manuscript will be scheduled for publication.

An invoice for payment will follow shortly after the formal acceptance. To ensure an efficient process, please log into Editorial Manager at http://www.editorialmanager.com/pone/, click the 'Update My Information' link at the top of the page, and double check that your user information is up-to-date. If you have any billing related questions, please contact our Author Billing department directly at authorbilling@plos.org.

If your institution or institutions have a press office, please notify them about your upcoming paper to help maximize its impact. If they’ll be preparing press materials, please inform our press team as soon as possible -- no later than 48 hours after receiving the formal acceptance. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

Kind regards,

Pratibha V. Nerurkar, Ph.D

Academic Editor

PLOS ONE

Additional Editor Comments (optional):

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. If the authors have adequately addressed your comments raised in a previous round of review and you feel that this manuscript is now acceptable for publication, you may indicate that here to bypass the “Comments to the Author” section, enter your conflict of interest statement in the “Confidential to Editor” section, and submit your "Accept" recommendation.

Reviewer #1: All comments have been addressed

Reviewer #2: All comments have been addressed

Reviewer #3: All comments have been addressed

**********

2. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: (No Response)

Reviewer #3: (No Response)

**********

3. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

Reviewer #2: (No Response)

Reviewer #3: (No Response)

**********

4. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: (No Response)

Reviewer #3: (No Response)

**********

5. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: (No Response)

Reviewer #3: (No Response)

**********

6. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: the author should add sentence should be added that estrogen is a potential confounding variable in interpretation.

Reviewer #2: (No Response)

Reviewer #3: (No Response)

**********

7. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

Reviewer #3: No

Acceptance letter

Pratibha V Nerurkar

27 Jul 2020

PONE-D-19-31787R1

Lack of TLR4 modifies the miRNAs profile and attenuates inflammatory signaling pathways

Dear Dr. Guerri:

I'm pleased to inform you that your manuscript has been deemed suitable for publication in PLOS ONE. Congratulations! Your manuscript is now with our production department.

If your institution or institutions have a press office, please let them know about your upcoming paper now to help maximize its impact. If they'll be preparing press materials, please inform our press team within the next 48 hours. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information please contact onepress@plos.org.

If we can help with anything else, please email us at plosone@plos.org.

Thank you for submitting your work to PLOS ONE and supporting open access.

Kind regards,

PLOS ONE Editorial Office Staff

on behalf of

Dr. Pratibha V. Nerurkar

Academic Editor

PLOS ONE

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 Table

    (PDF)

    S1 Raw images

    (TIF)

    Attachment

    Submitted filename: Response to Reviewers.docx

    Data Availability Statement

    All fataq files are available from the NCBI’s Gene Expression Omnibus database accessible through GEO Series accession number GSE120373.


    Articles from PLoS ONE are provided here courtesy of PLOS

    RESOURCES