Научная статья на тему 'INTERRELATION BETWEEN MIRNA AND MRNA EXPRESSION IN HT-29 LINE CELLS UNDER HYPOXIA'

INTERRELATION BETWEEN MIRNA AND MRNA EXPRESSION IN HT-29 LINE CELLS UNDER HYPOXIA Текст научной статьи по специальности «Биологические науки»

CC BY
48
11
i Надоели баннеры? Вы всегда можете отключить рекламу.
i Надоели баннеры? Вы всегда можете отключить рекламу.
iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.
i Надоели баннеры? Вы всегда можете отключить рекламу.

Текст научной работы на тему «INTERRELATION BETWEEN MIRNA AND MRNA EXPRESSION IN HT-29 LINE CELLS UNDER HYPOXIA»

INTERRELATION BETWEEN MIRNA AND MRNA EXPRESSION IN HT-29 LINE CELLS UNDER HYPOXIA

Nersisyan SA1, Galatenko AV2-3, Maltseva DV1-4 Ushkaryov YuA1, AG Tonevitskiy AG1'4

1 Faculty of Biology and Biotechnology, HSE University, Moscow, Russia

2 Faculty of Mechanics and Mathematics, Lomonosov Moscow State University, Moscow, Russia

3 Moscow Center for Fundamental and Applied Mathematics, Moscow, Russia

4 Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia

Hypoxia accompanies various pathophysiological processes, including progression of tumors and metastasis. One of the mechanisms of molecular response of cells to hypoxia implies recruitment of specific miRNAs that regulate the expression of their target genes. This study aimed to evaluate the hypoxia-induced change in expression of miRNAs and their target genes in the HT-29 human colorectal adenocarcinoma cell line with the help of integrated miRNA and mRNA sequencing. To simulate hypoxia, the cells were treated with cobalt (II) chloride. We registered a significant change in expression of sixteen human miRNAs. Six of them (hsa-miR-18a-5p, hsa-miR-22-3p, hsa-miR-27a-5p, hsa-miR-182-5p, hsa-miR-215 -5p, hsa-miR-425-5p) had a significant proportion of target genes that had the expression changing in the opposite direction. Based on the bioinformatic analysis of interactions between differentially expressed transcription factors and miRNAs, we built a possible regulatory network with its main hubs being HIF-1a, p65, c-Myc, and Egr1 (encoded by the HIF1A, RELA, MYC and EGR1 genes).

Keywords: hypoxia, miRNA, mRNA, transcriptome, sequencing, intestinal epithelium, HIF-1a, HT-29

Funding: the work was supported financially by the Russian Science Foundation (agreement #17-14-01338).

Author contribution: SA Nersisyan and AV Galatenko — sequencing data processing, bioinformatic analysis, functional gene analysis, building the regulatory network of interactions, manuscript preparation; DV Maltseva — cell culturing work, preparation of samples for sequencing, manuscript preparation for publication; YA Ushkaryov — discussion of the results, article text review; AG Tonevitsky — research organization, analysis of the results, article manuscript preparation.

Compliance with ethical standards: the study was approved by the Ethics Committee of Amur State Medical Academy (Protocol № 10 dated November 20, 2019); the study conformed with the guidelines for the medical research involving human subjects. Voluntary informed consent was obtained from all the participants.

1X1 Correspondence should be addressed: Diana V. Maltseva Vavilova, 7, Moscow, 117321; dmaltseva@gmail.com

Received: 26.10.2020 Accepted: 22.11.2020 Published online: 07.12.2020

DOI: 10.24075/brsmu.2020.074

ВЗАИМОСВЯЗЬ ИЗМЕНЕНИЯ ЭКСПРЕССИИ микроРНК И МРНК В КЛЕТКАХ ЛИНИИ НТ-29 В УСЛОВИЯХ ГИПОКСИИ

С. А. Нерсисян1, А. В. Галатенко23 Д. В. Мальцева1,4 Ю. А. Ушкарев1, А. Г. Тоневицкий1,4

1 Национальный исследовательский университет «Высшая школа экономики», Москва, Россия

2 Московский государственный университет имени М. В. Ломоносова, Москва, Россия

3 Московский центр фундаментальной и прикладной математики, Москва, Россия

4 Институт биоорганической химии имени М. М. Шемякина и Ю. А. Овчинникова, Москва, Россия

Гипоксия возникает в различных патофизиологических процессах, включая прогрессирование опухолевых заболеваний и метастазирование. Один из механизмов молекулярного ответа клеток на гипоксию состоит в рекрутировании специфичных микроРНК, регулирующих экспрессию своих генов-мишеней. Целью работы было оценить изменения экспрессии микроРНК и их генов-мишеней в клеточной линии колоректальной аденокарциномы человека HT-29 в ответ на гипоксию с помощью интегрированного секвенирования микроРНК и мРНК. Для моделирования условий гипоксии клетки обрабатывали хлоридом кобальта (II). Было обнаружено достоверное изменение экспрессии 16 человеческих микроРНК, шесть из которых (hsa-miR-18a-5p, hsa-miR-22-3p, hsa-miR-27a-5p, hsa-miR-182-5p, hsa-miR- 215-5p, hsa-miR-425-5p) имели статистически значимую долю генов-мишеней с противоположным направлением изменения экспрессии. На основании биоинформатического анализа взаимодействий дифференциально экспрессированных факторов транскрипции и микроРНК была построена возможная регуляторная сеть, основыми узлами которой оказались HIF-1 а, p65, с-Myc и Egr1 (кодируемые генами HIF1A, RELA, MYC и EGR1).

Ключевые слова: гипоксия, микроРНК, мРНК, транкриптом, секвенирование, эпителий кишечника, HIF-1 а, НТ-29 Финансирование: работа выполнена при финансовой поддержке Российского научного фонда (соглашение № 17-14-01338).

Вклад авторов: С. А. Нерсисян и А. В. Галатенко — обработка данных секвенирования, биоинформатический анализ, функциональный анализ генов, построение регуляторной сети взаимодействий, подготовка рукописи статьи; Д. В. Мальцева — работа с клетками, подготовка образцов для секвенирования, подготовка рукописи к публикации; Ю. А. Ушкарев — обсуждение результатов, рецензирование текста статьи; А. Г. Тоневицкий — организация исследования, анализ полученных результатов, подготовка рукописи статьи.

Соблюдение этических стандартов: исследование проведено с соблюдением этических принципов Хельсинкской декларации Всемирной медицинской ассоциации.

[><1 Для корреспонденции: Диана Васильевна Мальцева

ул. Вавилова, д. 7, г Москва, 117321; dmaltseva@gmail.com

Статья получена: 26.10.2020 Статья принята к печати: 22.11.2020 Опубликована онлайн: 07.12.2020 DOI: 10.24075/vrgmu.2020.074

Hypoxia plays a role in the development and course of a number of pathological conditions, such as cardiovascular disorders [1] and tumor developments [2]. Hypoxia models staged in vitro allow studying the response to hypoxia at the molecular and cellular levels. One of the traditional models involves chemical agents that activate the hypoxia signaling pathways. CoCl2 is one of the most common chemical hypoxia induction agents, since this substance causes direct and long-term stabilization of the hypoxia-induced factors 1 and 2 (HIF-1, HIF-2) [3].

MiRNAs are short non-coding RNAs; their main function is to suppress genes post-transcriptionally [4]. Usually, one miRNA has dozens of target genes, while 3'-UTR of a gene may have binding sites for hundreds of miRNAs [5]. It has been shown that interactions between miRNAs and their target genes play an important role in intercellular communication [6] and pathogenesis of many diseases, including various types of tumors [7, 8].

A number of studies aimed to investigate the role and functional activity of cellular miRNAs under the hypoxic stress conditions. It was found that some miRNAs, such as miR-210 or miR-27, are altered by hypoxia in many cells, and the differential expression of miRNAs and their targeting usually depend on the mechanism of hypoxia induction and cell type [9]. A relationship between the patterns of hypoxia-driven changes in miRNA expression and tumors has also been established: most of the miRNAs associated with the tumors can be affected by hypoxia [10].

This study aimed to investigate the effect of hypoxia on the miRNA profile and transcriptome in the HT-29 human colorectal adenocarcinoma line cells and to identify the potential key molecules involved in the response to hypoxia.

METHODS

Kit Illumina (Illumina; USA). Each sample was sequenced on an Illumina NextSeq 550 to obtain 75 nucleotide reading frames at one end.

The libraries for miRNA sequencing were prepared from the total RNA samples using the NEBNext Multiplex Small RNA Library Prep Kit for Illumina. Each sample was sequenced in the Illumina NextSeq 550 to obtain 50 unidirectional nucleotide reading frames.

We sequenced mRNA and miRNA for three biological iterations, with each of them having four technical iterations set up.

Processing of the sequencing results

FASTQ file quality was assessed with the help of FastQC v0.11.9 (Babraham Bioinformatics; UK). One of the CoCl2-treated replicates did not pass quality control at the microRNA sequencing stage. The adapters were cut using Cutadapt v2.10 [11]. The resulting mRNA fragment sequences were mapped to the human genome (GENCODE GRCh38.p13) using STAR v2.7.5b [12]. MiRDeep2 v2.0.1.2 package [13] enabled compilation of the miRNA expression matrix.

The sequencing library depths were normalized with the Trimmed Mean of M-values (TMM) algorithm available in the edgeR v3.30.3 package [14] with default background noise filtering. The same package was used to generate normalized mRNA and miRNA expression matrices in Reads Per Kilobase of transcript per Million mapped reads (RPKM) and Reads Per Million mapped reads (RPM) units, respectively. We took the logarithms of the values obtained with 2 as the base. For further processing, we only used the highly expressed transcripts and cut off the bottom 5% of genes and 50% of miRNAs based on their mean RPKM/RPM values.

Cell cultivation and processing

Human colorectal adenocarcinoma cells HT-29 (ATCC; USA) were cultured in McCoy's 5A medium (Thermo Fisher Scientific; USA) containing 10% fetal bovine serum (Thermo Fisher Scientific; USA). Penicillin (100 U/ml) and streptomycin (100 mg/ml) were added to the nutrient medium. The cells were seeded into 6-well plates in the amount of 4 x 105 cells per well, then cultured in a humidified atmosphere at +37 °C and 5% CO2 for 48 h. To induce hypoxia, a water solution of cobalt chloride (CoCl2) was prepared, added to the medium to the concentration of 300 pM, and incubated for 24 h. Three biological iterations were performed for both control and treatment group cells.

RNA isolation

The cells were lysed in the Qiazol Lysis Reagent (Qiagen; Germany) for subsequent total RNA extraction with the help of the Qiagen miRNeasy Mini Kit (Qiagen, Hilden; Germany). The amount of isolated RNA was determined using a Nanodrop device (Thermo Fisher Scientific; USA). Agilent High Sensitivity DNA Kit (Agilent Technologies; USA) and a Bioanalyzer 2100 capillary electrophoresis instrument (Agilent Technologies; USA) enabled analysis of quality of the isolated RNA samples. The RIN (RNA integrity number) parameter value for all samples was above 9.0.

Preparation of libraries and sequencing

The libraries for mRNA sequencing were obtained from the total RNA samples using the Illumina Stranded mRNA Library Prep

Evaluation of the differential expression and overrepresented signaling pathways

We used DESeq2 v1.28.1 [15] to analyze the differential expression and applied the Benjamini-Hochberg procedure to determine the false detection rate (FDR). The differences with FDR below the threshold value of 0.05 were considered significant. DAVID v6.8 online service [16] enabled the analysis of overrepresented signal pathways.

Prediction of the miRNA targets

At the first stage of miRNA target prediction we exported a list of miRNA-gene interactions from TargetScan v7.2 [17]. Then, we selected the negative expression correlation miRNA-gene pairs from The Cancer Genome Atlas Colon Adenocarcinoma (TCGA-COAD) cohort [18]. The initial miRNA/mRNA expression matrices for tumor samples were obtained from the GDC Data Portal (https://portal.gdc.cancer.gov/) and converted into the RPKM/RPM tables using the procedure described above. Next, we calculated the Spearman correlation for each miRNA and the predicted target gene. The thresholds of 0.05 and -0.1 were applied to the FDR and correlation values, respectively.

Building the regulatory network of interactions between transcription factors and miRNA

We took the information on the regulatory interactions of transcription factors and miRNAs from the curated TransmiR v2.0 database [19]. The resulting interaction network was built and visualized in the yED Graph Editor (yWorks GmbH; Germany).

А

B

ALDOC PFKP

MKNK2 VEGFA HKDC1 CDKN1A MAP2K1

Cobalt, repl Cobalt, rep2 Cobalt, rep3 Control, repl Control, rep2 Control, rep3

Cobalt, repl Cobalt, rep2 Cobalt, rep3 Control, repl Control, rep2 Control, rep3

Fig. 1. Differential expression of genes associated with hypoxia. A. Glycolysls/gluconeogenesls. B. HIF-1 signaling pathway

RESULTS

Effect of Cobalt (II) Chloride on gene expression in HT-29 cells

To chemically induce hypoxia, HT-29 cells were treated with cobalt (II) chloride for 24 h. Analysis of sequencing of the RNA isolated from HT-29 control cells and treated with cobalt chloride showed a statistically significant change in the expression of 2511 genes that encode proteins, this change being 2-fold and greater in response to hypoxia. The search for overrepresented metabolic pathways identified 25 genes associated with the switch of aerobic metabolism to anaerobic glycolysis (KEGG pathway hsa00010 Glycolysis/Gluconeogenesis, FDR = 2.04 x 10-4; Fig. 1A), and also demonstrated activation of the HIF-1 signaling pathway (KEGG pathway hsa04066 HIF-1 signaling pathway, FDR = 4.45 x 10-3) that allows cobalt chloride to simulate

Table 1. Differentially expressed miRNAs

hypoxia (Fig. 1B). In addition to the HIF-1 pathway, several other signaling cascades involved in the response to hypoxia were activated, including NF-kB [20] and AMPK [21] (attachment 1).

The most overrepresented category corresponded to the genes encoding proteins of proteasome complexes (KEGG pathway hsa03050 Proteasome, FDR = 2,02 x 10-16). In particular, 33 genes were significantly activated in response to hypoxia, including 6 out of 6 ATPases, 11 out of 12 subunits of the 26S proteasome lacking ATPase activity, 7 out of 8 a-subunits and 7 out of 11 p-subunits of the 20S proteasome, as well as the proteasome maturation protein POMP and proteasome activator subunit PSME4 (attachment 2). We have registered an increase in the expression of UBB, UBC, UBA52 and RPS27A genes, the increase being 3.1, 8.7, 2.0 and 1.6-fold, respectively. These genes encode ubiquitin, which is needed for proteasome-dependent protein degradation.

MiRNA Mean expression level in control (RPM) Expression change, times* FDR

hsa-miR-210-3p 372.38 2.40 4.01 x 10-20

hsa-miR-4521 452.54 -2.48 2.31 x 10-18

hsa-miR-615-3p 739.12 -1.90 5.30 x 10-10

hsa-miR-22-3p 1032.99 1.65 5.21 x 10-6

hsa-miR-425-5p 751.22 -1.44 8.52 x 10-4

hsa-let-7a-3p 631.34 -1.44 2.87 x 10-3

hsa-miR-32-5p 594.48 -1.43 3.18 x 10-3

hsa-miR-215-5p 2604.64 1.49 6.61 x 10-3

hsa-miR-224-5p 4385.37 1.41 0.0123

hsa-miR-182-5p 3935.22 1.49 0.0144

hsa-miR-1260b 550.76 -1.35 0.0187

hsa-miR-1260a 531.53 -1.34 0.0241

hsa-miR-27a-5p 158.10 1.51 0.0317

hsa-miR-30b-5p 1509.37 1.31 0.0317

hsa-miR-10a-3p 2102.20 -1.29 0.0417

hsa-miR-18a-5p 225.40 -1.39 0.0444

Note: * — positive and negative values indicate the cellular miRNA level drop or growth in response to hypoxia.

JUN

CDKN1A

HSF2

\ /

i к

CDX2

^^^-210-3^^ (^^-224-5^^

Fig. 2. Regulatory network of interactions between transcription factors (TF) and miRNA, induced by hypoxia. Rectangles are TFs, ellipses — miRNAs. Expression growths and drops are shown in green and red, respectively. Arrows indicate activation of the expression, T-shaped lines — its suppression

We have also detected a change in the expression of genes that encode proteins involved in focal adhesion (integrins and laminins). Accordingly, the expression levels of laminin subunits a3, p3, Y1, and Y2 were increased 5.5, 4.6, 3.1 and 4.5-fold, respectively. Three of them (a3, p3, Y2) can form a heterotrimer and thus generate laminin 332, also known as laminin-5 [22]. The expression of integrin subunits changed in a different direction: subunits aE, aV and p1 showed the growth of 1.8, 2.2, 1.8 times, while for subunits a1, a2, p3 and p8 the registered expression level decrease was 2.3, 2.1, 1.7 and 2.7 times.

Effect of hypoxia on the expression of miRNAs and their target genes

We have registered 16 miRNAs (Table 1) showing a significant change in the expression as a response to the treatment of cells with cobalt chloride. One of them was hsa-miR-210-3p, the only miRNA that is cited in all papers published as increasing expression in response to hypoxia [23]. The level of several confirmed hsa-miR-210-3p mRNA targets involved in mitochondrial metabolism and apoptosis induction decreased in response to hypoxia: GPD1L by 2.3 times, CASP8AP2 by 1.7 times, and AIFM3 by 8 times.

The following analysis was performed to assess the overall functional effects of the cellular miRNA content fluctuations induced by hypoxia. The TargetScan resource enabled compilation of the list of potential miRNA targets. Since some interactions between miRNA and target mRNA inhibit translation without affecting the level of mRNA expression, analyzing the samples from 441 colon adenocarcinoma patients (taken from the TCGA-COAD database, Table S3) we searched for miRNA-target mRNA pairs showing a significant negative correlation. Next, we crossed the resulting list with the list of genes that had the expression changed at least two-fold in the direction opposite that peculiar to the corresponding miRNA. The result were six miRNAs with a statistically significant number of deregulated target genes (hypergeometric test; p < 0.05): hsa-miR-18a-5p, hsa-miR-22-3p, hsa-miR-27a-5p, hsa-miR -182-5p, hsa-miR-215-5p, hsa-miR-425-5p (attachment 4).

Building the network of regulatory interactions between transcription factors and miRNA

To better understand the mechanisms underlying the aberrant expression of microRNAs, we analyzed the possibility of regulation of microRNAs with transcription factors (TF). In particular, we

Table 2. MiRNAs showing differential expression patterns specific to the particular miRNA arms

MIRNA Mean expressIon level In control (RPM) ExpressIon change, tImes* FDR

hsa-let-7a-5p 21537.87 1.07 0.899

hsa-let-7a-3p 631.34 -1.44 2.87 x 10-3

hsa-mIR-10a-5p 100119.91 1.09 0.872

hsa-mIR-10a-3p 2102.20 -1.29 0.0417

iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.

hsa-mIR-27a-5p 158.10 1.51 0.0317

hsa-mIR-27a-3p 6321.04 1.04 0.929

Note: * — positive and negative values indicate the cellular miRNA level drop or growth in response to hypoxia.

considered TFs that had their mRNA representation significantly changing (two-fold or greater) in response to hypoxia and searched for the miRNAs they regulate in the TransmiR database of regulatory TF-microRNA interactions. As a result, we identified 30 TF-miRNA interactions between 15 TFs and 11 miRNAs. We have also considered the reciprocal miRNA-induced TF silence to build a complete regulatory network at these nodes (Fig. 2). As is shown, the four TFs encoded by the EGR1, HIF1A, MYC, and RELA genes simultaneously regulate several miRNAs, while most miRNAs are regulated by the TF ensembles.

DISCUSSION

This study investigated changes in the transcriptome landscape of HT-29 cells in response to hypoxia induced by cobalt (II) chloride. The investigation relied on the mRNA and miRNA integrative sequencing technique. In addition to the activation of HIF-1, the canonical signaling pathway, we have shown that the expression of integrins and laminins changes, too. These proteins play a critical role in cell adhesion and interactions with the extracellular matrix. The latter is of particular importance, since recent data indicate a close relationship between the microenvironment formed by hypoxia and the metastatic progression of tumors, including colon adenocarcinoma [24]. One of the possible mechanisms for the metastatic spread of tumors is associated with laminin 332. By interacting with various receptors on the cell surface (including integrins a6^4 and a3p1, epidermal growth factor receptor and syndecan 1), as well as some other components of the basal membrane, laminin 332 regulates the process of oncogenesis, promotes invasion and survival of tumor cells [25]. The increased expression of the Y1-arm of laminins (encoded by the LAMC1 gene) may also play a role in the progression of tumor diseases, as has been shown in uterine carcinoma [26].

The analysis of profile of the small noncoding RNAs revealed several miRNAs expressed differentially in response to hypoxia. Some of these miRNAs were already reported as having their expression changed under hypoxia, including hsa-miR-210-3p

[23], hsa-miR-27a-5p [27], hsa-miR-182-5p [28]. Four miRNAs (hsa-miR-30b-5p, hsa-miR-32-5p, hsa-miR-425-5p, hsa-miR-1260a, and hsa-miR-1260b) were not previously covered in the published papers in connection with the cellular response to hypoxia. This can be explained both by the cellular specificity of the response and by the cobalt chloride's stream effects.

Particular attention should be paid to the miRNAs that show differential patterns of expression specific to a particular miRNA arm (arm-specific differential expression patterns). Namely, this applies to the miRNAs that had only the passenger arms hsa-let-7a, hsa-mir-10a and hsa-mir-27a regulated, while the expression of their guide arms did not change (Table 2). We have recently reported a similar observation for miR-21-3p (passenger arm) in mouse lungs; it showed an eight-fold increase in expression in response to the SARS-CoV infection, while the guide arm of the same miRNA increased only threefold [29]. One of the most promising theories that could explain this phenomenon is the regulation of miRNA arms by RNA-binding proteins [30].

The analysis of regulatory interactions between TF and miRNA showed that HIF-1, p65, c-Myc, and EGR1 (encoded by the HIF1A, RELA, MYC and EGR1 genes) are the key factors regulating transcription of differentially expressed miRNAs (see Fig. 2). Three of the considered miRNAs demonstrated reciprocal capabilities and suppressed some of the TFs. In particular, HIF1A is a confirmed target for hsa-miR-18a-5p that is suppressed by multiple TFs.

CONCLUSION

The performed integrative sequencing of miRNA/mRNA allowed revealing significant changes in the transcriptome and miRNA profile in HT-29 cells in response to the CoCl2-induced hypoxia. We have shown that differential expression of several of the miRNAs can cause significant changes in the expression of their target mRNAs. The analysis of the regulatory interactions between transcription factors and miRNAs revealed possible mechanisms underlying the observed response to hypoxia.

References

1. Semenza GL. Hypoxia-inducible factor 1 and cardiovascular disease. Annu Rev Physiol. 2014; 76: 39-56.

2. Muz B, de la Puente P, Azab F, Azab AK. The role of hypoxia in cancer progression, angiogenesis, metastasis, and resistance to therapy. Hypoxia. 2015; 83.

3. Muñoz-Sánchez J, Chánez-Cárdenas ME. The use of cobalt chloride as a chemical hypoxia model. J Appl Toxicol. 2019; 39 (4): 556-70.

4. Cai Y, Yu X, Hu S, Yu J. A Brief Review on the Mechanisms of miRNA Regulation. Genomics Proteomics Bioinformatics. Elsevier; 2009; 7 (4): 147-54.

5. Hendrickson DG, Hogan DJ, McCullough HL, Myers JW, Herschlag D, Ferrell JE, et al. Concordant regulation of translation and mRNA abundance for hundreds of targets of a human microRNA. PLoS Biol. 2009; 7 (11).

6. Turchinovich A, Tonevitsky AG, Cho WC, Burwinkel B. Check and mate to exosomal extracellular miRNA: new lesson from a new approach. Front Mol Biosci. Frontiers; 2015; 2: 11.

7. Visone R, Croce CM. MiRNAs and cancer. Am J Pathol. 2009; 174 (4): 1131-8.

8. Shkurnikov M, Nikulin S, Nersisyan S, Poloznikov A, Zaidi S, Baranova A, et al. LAMA4-Regulating miR-4274 and Its Host

Gene SORCS2 Play a Role in IGFBP6-Dependent Effects on Phenotype of Basal-Like Breast Cancer. Front Mol Biosci. 2019; 6. 20.

9. Lacedonia D, Scioscia G, Palladino GP, Gallo C, Carpagnano GE, Sabato R, et al. MicroRNA expression profile during different conditions of hypoxia. Oncotarget. 2018; 9 (80): 35114-22. 21.

10. Shen G, Li X, Jia YF, Piazza GA, Xi Y. Hypoxia-regulated microRNAs in human cancer. Acta Pharmacol Sin. 2013; 34 (3): 336-41.

11. Martin M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet.journal. 2011; 17 (1): 10. 22.

12. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S,

et al. STAR: Ultrafast universal RNA-seq aligner. Bioinformatics. 23. 2013; 29 (1): 15-21.

13. Friedländer MR, Mackowiak SD, Li N, Chen W, Rajewsky N. miRDeep2 accurately identifies known and hundreds of novel 24. microRNA genes in seven animal clades. Nucleic Acids Res. Oxford Academic. 2012; 40 (1): 37-52. 25.

14. Robinson MD, McCarthy DJ, Smyth GK. edgeR: A Bioconductor package for differential expression analysis of digital gene 26. expression data. Bioinformatics. 2009; 26 (1): 139-40.

15. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. BioMed Central. 2014; 15 (12): 550. 27.

16. Huang DW, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources.

Nat Protoc. Nature Publishing Group. 2009; 4 (1): 44-57. 28.

17. Agarwal V, Bell GW, Nam JW, Bartel DP. Predicting effective microRNA target sites in mammalian mRNAs. Elife. 2015; 4 (AUGUST2015). 29.

18. Muzny DM, Bainbridge MN, Chang K, Dinh HH, Drummond JA, Fowler G, et al. Comprehensive molecular characterization of human colon and rectal cancer. Nature. 2012; 487 (7407): 330-7. 30.

19. Tong Z, Cui Q, Wang J, Zhou Y. TransmiR v2.0: An updated transcription factor-microRNA regulation database. Nucleic Acids

Res. 2019; 47 (D1): D253--D258.

Culver C, Sundqvist A, Mudie S, Melvin A, Xirodimas D, Rocha S. Mechanism of hypoxia-induced NF-kB. Mol Cell Biol. 2010; 30 (20): 4901-21.

Mungai PT, Waypa GB, Jairaman A, Prakriya M, Dokic D, Ball MK, et al. Hypoxia Triggers AMPK Activation through Reactive Oxygen Species-Mediated Activation of Calcium Release-Activated Calcium Channels. Mol Cell Biol. 2011; 31 (17): 3531-45. Domogatskaya A, Rodin S, Tryggvason K. Functional Diversity of Laminins. Annu Rev Cell Dev Biol. 2012; 28 (1): 523-53. Bavelloni A, Ramazzotti G, Poli A, Piazzi M, Focaccia E, Blalock W, et al. Mirna-210: A current overview. Anticancer Res. 2017; 37 (12): 6511-21.

Rankin EB, Giaccia AJ. Hypoxic control of metastasis. Sci (80- ). 2016; 352 (6282): 175-80.

Maltseva D V., Rodin SA. Laminins in metastatic cancer. Mol Biol. Springer; 2018; 52 (3): 350-71.

Kashima H, Wu RC, Wang Y, Sinno AK, Miyamoto T, Shiozawa T, et al. Laminin C1 expression by uterine carcinoma cells is associated with tumor progression. Gynecol Oncol. Elsevier Inc.; 2015; 139 (2): 338-44.

Zhang J, Qiu W, Ma J, Wang Y, Hu Z, Long K, et al. miR-27a-5p Attenuates Hypoxia-induced Rat Cardiomyocyte Injury by Inhibiting Atg7. Int J Mol Sci. 2019; 20 (10). Zhang Y, Fang J, Ma H. Inhibition of miR-182-5p protects cardiomyocytes from hypoxia-induced apoptosis by targeting CIAPIN1. Biochem Cell Biol. 2018; 96 (5): 646-54. Nersisyan S, Engibaryan N, Gorbonos A, Kirdey K, Makhonin A, Tonevitsky A. Potential role of cellular miRNAs in coronavirus-host interplay. PeerJ. 2020; 8: e9994.

Van Kouwenhove M, Kedde M, Agami R. MicroRNA regulation by RNA-binding proteins and its implications for cancer. Nat Rev Cancer. 2011; 11 (9): 644-56.

Литература

1. Semenza GL. Hypoxia-inducible factor 1 and cardiovascular disease. Annu Rev Physiol. 2014; 76: 39-56.

2. Muz B, de la Puente P, Azab F, Azab AK. The role of hypoxia in cancer progression, angiogenesis, metastasis, and resistance to 14. therapy. Hypoxia. 2015; B3.

3. Muñoz-Sánchez J, Chánez-Cárdenas ME. The use of cobalt chloride as a chemical hypoxia model. J Appl Toxicol. 2019; 39 15. (4): 556-70.

4. Cai Y, Yu X, Hu S, Yu J. A Brief Review on the Mechanisms

of miRNA Regulation. Genomics Proteomics Bioinformatics. 16. Elsevier; 2009; 7 (4): 147-54.

5. Hendrickson DG, Hogan DJ, McCullough HL, Myers JW, Herschlag D, Ferrell JE, et al. Concordant regulation of translation 17. and mRNA abundance for hundreds of targets of a human microRNA. PLoS Biol. 2009; 7 (11).

6. Turchinovich A, Tonevitsky AG, Cho WC, Burwinkel B. Check and 18. mate to exosomal extracellular miRNA: new lesson from a new approach. Front Mol Biosci. Frontiers; 2015; 2: 11.

7. Visone R, Croce CM. MiRNAs and cancer. Am J Pathol. 2009; 19. 174 (4): 1131-B.

8. Shkurnikov M, Nikulin S, Nersisyan S, Poloznikov A, Zaidi S, Baranova A, et al. LAMA4-Regulating miR-4274 and Its Host 20. Gene SORCS2 Play a Role in IGFBP6-Dependent Effects on Phenotype of Basal-Like Breast Cancer. Front Mol Biosci. 2019; 6.

9. Lacedonia D, Scioscia G, Palladino GP, Gallo C, Carpagnano GE, 21. Sabato R, et al. MicroRNA expression profile during different conditions of hypoxia. Oncotarget. 201B; 9 (B0): 35114-22.

10. Shen G, Li X, Jia YF, Piazza GA, Xi Y. Hypoxia-regulated microRNAs in human cancer. Acta Pharmacol Sin. 2013; 34 (3): 336-41. 22.

11. Martin M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet.journal. 2011; 17 (1): 10. 23.

12. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: Ultrafast universal RNA-seq aligner. Bioinformatics. 2013; 29 (1): 15-21. 24.

13. Friedländer MR, Mackowiak SD, Li N, Chen W, Rajewsky N.

miRDeep2 accurately identifies known and hundreds of novel microRNA genes in seven animal clades. Nucleic Acids Res. Oxford Academic. 2012; 40 (1): 37-52.

Robinson MD, McCarthy DJ, Smyth GK. edgeR: A Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2009; 26 (1): 139-40. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. BioMed Central. 2014; 15 (12): 550.

Huang DW, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. Nature Publishing Group. 2009; 4 (1): 44-57. Agarwal V, Bell GW, Nam JW, Bartel DP. Predicting effective microRNA target sites in mammalian mRNAs. Elife. 2015; 4 (AUGUST2015).

Muzny DM, Bainbridge MN, Chang K, Dinh HH, Drummond JA, Fowler G, et al. Comprehensive molecular characterization of human colon and rectal cancer. Nature. 2012; 487 (7407): 330-7. Tong Z, Cui Q, Wang J, Zhou Y. TransmiR v2.0: An updated transcription factor-microRNA regulation database. Nucleic Acids Res. 2019; 47 (D1): D253--D258.

Culver C, Sundqvist A, Mudie S, Melvin A, Xirodimas D, Rocha S. Mechanism of hypoxia-induced NF-kB. Mol Cell Biol. 2010; 30 (20): 4901-21.

Mungai PT, Waypa GB, Jairaman A, Prakriya M, Dokic D, Ball MK, et al. Hypoxia Triggers AMPK Activation through Reactive Oxygen Species-Mediated Activation of Calcium Release-Activated Calcium Channels. Mol Cell Biol. 2011; 31 (17): 3531-45. Domogatskaya A, Rodin S, Tryggvason K. Functional Diversity of Laminins. Annu Rev Cell Dev Biol. 2012; 28 (1): 523-53. Bavelloni A, Ramazzotti G, Poli A, Piazzi M, Focaccia E, Blalock W, et al. Mirna-210: A current overview. Anticancer Res. 2017; 37 (12): 6511-21.

Rankin EB, Giaccia AJ. Hypoxic control of metastasis. Sci (80- ). 2016; 352 (6282): 175-80.

25. Мальцева Д. В., Родин С. A. Ламинины и метастазирование опухолей. Мол. Биол. 2018; 52 (3): 350-71.

26. Kashima H, Wu RC, Wang Y, Sinno AK, Miyamoto T, Shiozawa T, et al. Laminin C1 expression by uterine carcinoma cells is associated with tumor progression. Gynecol Oncol. Elsevier Inc.; 2015; 139 (2): 338-44.

27. Zhang J, Qiu W, Ma J, Wang Y, Hu Z, Long K, et al. miR-27a-5p Attenuates Hypoxia-induced Rat Cardiomyocyte Injury by Inhibiting Atg7. Int J Mol Sci. 2019; 20 (10).

28. Zhang Y, Fang J, Ma H. Inhibition of miR-182-5p protects cardiomyocytes from hypoxia-induced apoptosis by targeting CIAPIN1. Biochem Cell Biol. 2018; 96 (5): 646-54.

29. Nersisyan S, Engibaryan N, Gorbonos A, Kirdey K, Makhonin A, Tonevitsky A. Potential role of cellular miRNAs in coronavirus-host interplay. PeerJ. 2020; 8: e9994.

30. Van Kouwenhove M, Kedde M, Agami R. MicroRNA regulation by RNA-binding proteins and its implications for cancer. Nat Rev Cancer. 2011; 11 (9): 644-56.

i Надоели баннеры? Вы всегда можете отключить рекламу.