Open Access

Advances in microRNA regulation of deep vein thrombosis through venous vascular endothelial cells (Review)

  • Authors:
    • Chucun Fang
    • Feng Huang
    • Mengting Yao
    • Zilong Wang
    • Jiacheng Ma
    • Dongwen Wu
    • Tianting Guo
    • Fei Zhang
    • Jianwen Mo
  • View Affiliations

  • Published online on: April 9, 2024     https://doi.org/10.3892/mmr.2024.13220
  • Article Number: 96
  • Copyright: © Fang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Deep vein thrombosis (DVT) is a prevalent clinical venous thrombotic condition that often manifests independently or in conjunction with other ailments. Thrombi have the propensity to dislodge into the circulatory system, giving rise to complications such as pulmonary embolism, thereby posing a significant risk to the patient. Virchow proposed that blood stagnation, alterations in the vessel wall and hypercoagulation are primary factors contributing to the development of venous thrombosis. Vascular endothelial cells (VECs) constitute the initial barrier to the vascular wall and are a focal point of ongoing research. These cells exert diverse stimulatory effects on the bloodstream and secrete various regulatory factors that uphold the dynamic equilibrium between the coagulation and anticoagulation processes. MicroRNAs (miRNAs) represent a class of non‑coding RNAs present in eukaryotes, characterized by significant genetic and evolutionary conservation and displaying high spatiotemporal expression specificity. Typically ranging from 20 to 25 bases in length, miRNAs can influence downstream gene transcription through RNA interference or by binding to specific mRNA sites. Consequently, advancements in understanding the molecular mechanisms of miRNAs, including their functionalities, involve modulation of vascular‑associated processes such as cell proliferation, differentiation, secretion of inflammatory factors, migration, apoptosis and vascular remodeling regeneration. miRNAs play a substantial role in DVT formation via venous VECs. In the present review, the distinct functions of various miRNAs in endothelial cells are outlined and recent progress in comprehending their role in the pathogenesis and clinical application of DVT is elucidated.

Introduction

MicroRNAs (miRNAs) are a class of non-coding RNAs characterized by very short sequences, typically containing approximately 20–25 bases (13). Traditionally, miRNAs exert their biological functions by binding to specific sites on mRNA molecules, thereby influencing the transcription of downstream genes. This binding occurs through essentially complementary pairing with gene sequences on the target mRNAs, organizing the translation process of the bound mRNA and thus regulating the expression levels of corresponding proteins. In addition to the classical mechanism of gene expression downregulation, miRNAs exhibit other non-classical regulatory mechanisms that warrant further investigation (47). Research into miRNAs has seen a significant increase, revealing their involvement in the development of various systemic diseases such as cardiovascular (8), digestive (9) and respiratory (10) diseases. Moreover, they play a unique and biologically significant role in numerous biological processes.

Previous research suggests that the canonical pathways for miRNA production play critical roles in multiple biological processes. Genes containing miRNAs undergo transcription, translation and initial processing into pre-miRNAs within the nucleus. These pre-miRNAs then traverse the nuclear membrane via transmembrane proteins into the cytoplasm, where they undergo further processing and modifications to become mature miRNAs. Initially, genes harboring miRNAs are transcribed by RNA polymerase II into primary miRNAs (pri-miRNAs), characterized by one or more hairpin structures with 5′ caps and 3′ polyadenylate (A) tails (1113). Within the nucleus, a microprocessor complex, consisting of a DROSHA endonuclease molecule and two molecular chaperones, including DGCR8 with an RNase III structural domain, cleaves and modifies pri-miRNAs into 50–70 nucleotide pre-miRNAs (14). Subsequently, pre-miRNAs are transported across the nuclear membrane to the cytoplasm by the transmembrane protein exportin 5. In the cytoplasm, Dicer, containing two RNase III endonuclease structures, cleaves pre-miRNAs into mature miRNA duplexes ~22 nucleotides in length (15). Finally, with the assistance of molecular chaperones such as HSC70/HSP90 (16,17), duplex miRNAs form a silencing complex known as the RNA-induced silencing complex (RISC), which includes the Argonaute (AGO) protein possessing shearing functionality (18,19). This complex catalyzes the separation of duplex miRNAs into two single-stranded miRNAs by disrupting the hydrogen bonds of the base complementary sequences. The strand with a stronger binding affinity to the 5′ end of the AGO protein within its inner pocket is designated as the guide strand (20,21), while the other strand, known as the passenger strand (22), undergoes degradation by cytoplasmic RNA enzymes. Once a stable RISC structure forms, miRNAs bind to specific sites on mRNA molecules, thereby silencing mRNA transcription and exerting biological effects. This process delineates the classical formation pathway of miRNAs.

It is generally accepted that miRNAs interact with target genes by inhibiting translation. However, some miRNAs interact with target genes with less complementarity, overlooking the fact that certain miRNAs can lead to target gene degradation (23). Numerous studies suggest that, contrary to the common misconception, miRNAs typically do not fully silence target gene mRNAs but rather decrease their expression (24,25). However, over time, miRNAs exhibit both temporal and tissue expression specificity, which explains why mere inhibition or promotion does not fully reflect the biological functions of miRNAs (26). A number of studies have reported that miRNAs have seven non-classical regulatory molecular mechanisms (27); however, this does not fully outline the non-classical regulatory functions of miRNAs. Despite the current limited understanding of the biological functions of miRNAs, further investigation into cellular function regulation by miRNAs is warranted (2,28,29).

Cardiovascular endothelial cells serve dual roles in blood coagulation, with both anticoagulant and procoagulant functions, thereby preserving the equilibrium necessary for proper blood flow (30). Initially, the intact endothelium acts as a physical barrier, segregating platelets, clotting factors and the highly procoagulant endothelial matrix from the bloodstream (31). Furthermore, endothelial cells contribute to anti-platelet aggregation mechanisms. They produce prostacyclin (3234) and nitric oxide (NO) (3537), which inhibit platelet aggregation, along with adenosine diphosphatase (3840), which degrades ADP and prevents platelet agglutination. Additionally, the vascular endothelium synthesizes antithrombin and coagulation factors. It generates thrombomodulin, which binds to circulating thrombin, triggering the activation of the anticoagulant factor protein C. This synergistic action with protein S, also synthesized by the endothelium, leads to the inactivation of coagulation factors V and VIII (4144). They synthesize membrane-associated heparin-like molecules that bind to antithrombin III and inactivate thrombin, as well as coagulation factors X and IX (45,46). Additionally, VECs promote fibrinolysis. They internalize tissue-type plasminogen activator, leading to the degradation of fibrin deposited on the endothelial cell surface into fibrin degradation products. The latter exhibits anticoagulant effects (4749). Damage to the cardiovascular endothelium constitutes one of the three major pathological processes of thrombosis. Following endothelial cell injury, subendothelial collagen becomes exposed, activating platelets and coagulation factor VII, thereby initiating the endogenous coagulation pathway (50,51). Simultaneously, injured endothelial cells release tissue factor (TF), which activates coagulation factor VII and triggers the exogenous coagulation pathway (52,53). Consequently, VECs play a multidimensional role in thrombus formation and regulation, serving as key cells in maintaining the dynamic balance between the coagulation and anticoagulation systems. Recently, miRNAs have been recognized as common risk factors for deep vein thrombosis (DVT) (54). Therefore, in the present review, the diverse functions of different miRNAs in endothelial cells are summarized and recent advances in their pathogenesis and clinical application in DVT are discussed.

miRNAs in endothelial cells regulate DVT

miRNAs reverse DVT

Several studies on DVT have highlighted the role of miRNAs in directly regulating or indirectly influencing the function of venous VECs, thus impacting thrombosis (30,54). In recent years, researchers have employed genetic techniques to initially screen for aberrantly expressed miRNAs in the blood of patients with DVT. Subsequently, the target genes these miRNAs act upon, their effects on the function of VECs and their involvement in DVT formation have been investigated. These miRNAs play a crucial role in regulating the function of VECs, inhibiting apoptosis and ultimately preventing thrombus formation. For instance, a previous study revealed that miR-9-5p (55) reduces TRPM7 expression by activating the PI3K/Akt/autophagy pathway, thereby enhancing endothelial progenitor cell migration, invasion and angiogenesis. Conversely, the upregulation of histone deacetylase 3 can elevate miR-19b levels, which in turn mediates peroxisome proliferator-activated receptor γ to deactivate nuclear factor-κB (NF-κB), thus mitigating inflammation (56). Decreased levels of miR-125a-5p (57) have been shown to lead to increased expression of myeloid cell leukemia sequence 1, promoting VEC migration and angiogenesis, consequently inhibiting DVT. miRNAs with similar biological functions include miR-9 (55,58), miR-19b (56,59), miR-29c-3p (60), miR-125a-5p (57), miR-126 (61), miR-143-3p (62), miR-150 (6365), miR-205 (66), miR-411 (67) and miR-3120 (68), which primarily regulate the autophagic pathway in PI3K/Akt cells, thus exerting influence on this pathway. Li and Ni (69) found that miR-26a regulates the NF-κB signaling pathway by binding to protein kinase C-δ mRNA, thereby inhibiting the expression levels of inflammatory factor mRNAs and reducing the risk of DVT. Sun et al (70) revealed that miR-103a-3p can inhibit the expression of inflammatory factors such as TF, plasminogen activator inhibitors, interleukin (IL)-6 and IL-8, ultimately disrupting the inflammatory response. miR-5189-3p can inhibit apoptosis of VECs through the Notch signaling pathway (71); miR-195/582 (72) maintains the homeostasis of the intracellular environment by targeting and inhibiting the 3′-UTR of post-transcriptional nitric oxide synthase 3 in VECs, which in turn inhibits NO release. The miR-125a-3p/IL-1 receptor type 1 axis and miR-136-5p inhibit the secretion of inflammatory mediators in the blood of rat femoral veins, resulting in morphological changes in thrombus length and weight, and inhibition of thrombus formation (73,74). A number of miRNAs inhibit thrombosis by promoting the proliferation of VECs, increasing their viability and attenuating cellular damage. These miRNAs are summarized as miR-21 (75,76), miR-195 (77), miR-204-5p (78), miR-296-5p (79), miR-342-3p (80) and miR-361-5p (81). The aforementioned studies demonstrated that among various miRNAs, some could inhibit VEC injury, improve endothelial cell inflammation and autophagy and inhibit thrombus formation. Therefore, miRNA mimics or inhibitors may serve as potential anticoagulants (Fig. 1; Table I).

Table I.

miRNAs and target proteins that reverse DVT formation.

Table I.

miRNAs and target proteins that reverse DVT formation.

miRNAChange in expressionTarget gene(s)(Refs.)
miR-9UpregulationFGF5/TRPM3(55,58)
miR-19bUpregulationPPARγ/NF-κB(56)
miR-19bUpregulationTGFβ2(59)
miR-29c-3pDownregulationMDM2(60)
miR-125a-5pDownregulationMCL-1(57)
miR-126UpregulationPIK3R2(61)
miR-143-3pUpregulationATG2B(62)
miR-150DownregulationSRCIN1(63)
miR-150UpregulationAkt/FOXO1(64)
miR-150Upregulationc-Myb(65)
miR-205UpregulationPTEN(66)
miR-411DownregulationHIF-2α(67)
miR-3120DownregulationMMP1(68)
miR-26aDownregulationPRKCD(69)
miR-103a-3pUpregulationCXCL12(70)
miR-125a-3pDownregulationIL1R1(73)
miR-136-5pUpregulationIL-6 and CRP(74)
miR-5189-3pDownregulationJAG1(71)
miR-195/582-NOS3(72)
miR-21UpregulationFASLG(75)
miR-21UpregulationIL6R(76)
miR-195DownregulationGABARAPL1(77)
miR-204-5pUpregulationSPRED1(78)
miR-296-5pUpregulationS100A4(79)
miR-342-3pUpregulationEDNRA(80)
miR-361-5pDownregulationFGF1(81)

[i] miRNA, microRNA; DVT, deep vein thrombosis.

miRNAs promote DVT

Functionally, miRNAs can enhance the secretion of inflammatory factors, proliferation, migration and VEC angiogenesis. They accelerate injury and apoptosis of VECs and cause shape elongation, aggregation and cytoskeletal rearrangement of VECs, thereby promoting DVT formation. Previous studies have reported that miR-122 (82), miR-181a-5p (83), miR-195-5p (84), miR-206 (85), miR-338-5p (86), miR-383-5p (87), miR-448 (88), miR-483-3p (89), miR-525p-5p (90) and let-7e-5p (91) can act on their respective target proteins to promote DVT formation. These aforementioned studies suggest that miRNAs can promote the release of inflammatory factors and accelerate oxidative stress-mediated cellular injury, leading to the loss of barrier and material transport functions and exacerbation of ischemic and hypoxic damage to the vascular microenvironment. The various damaging effects aforementioned disrupt the blood system balance and the procoagulant state accelerates DVT formation.

Additionally, miRNAs have been found to promote thrombus formation in patients with cancer. Venous thromboembolism (VTE) poses a significant risk to individuals with cancer, leading to increased morbidity and mortality, with miRNAs playing a role in this process (9294). For instance, miRNA-135a directly affects forkhead box M1 and metastasis suppressor 1, thereby promoting metastasis of hepatocellular carcinoma cells and facilitating portal vein thrombosis (95). Oto et al (96) identified seven miRNAs, including miR-423-5p, as biological markers capable of predicting venous thrombosis in pancreatic ductal adenocarcinoma and distal extrahepatic cholangiocarcinoma. Similarly, four miRNAs, including miR-3652, were found to be significantly downregulated in patients with colorectal cancer with VTE, suggesting their potential as novel predictive biomarkers (97). Morelli et al (98) observed a positive correlation between plasma levels of miR-145 and the absence of VTE, proposing miR-145 as a potential target for VTE prevention (98). These studies collectively demonstrate the dysregulated expression of miRNAs in the plasma of patients with cancer, their involvement in cancer cell metastasis and their role in promoting cancer-associated thrombosis formation. Consequently, numerous investigations have centered on utilizing miRNAs as biological markers for cancer-associated thrombosis (99) (Table II).

Table II.

miRNA and target proteins that promote DVT formation.

Table II.

miRNA and target proteins that promote DVT formation.

miRNAChange in expressionTarget gene(s)(Refs.)
miRNA-122Upregulationp53(82)
miR-181a-5pDownregulationPcyox1l(83)
miR-195-5pUpregulationBcl-2(84)
miR-206UpregulationGJA1(85)
miR-338-5pDownregulationIL-6(86)
miR-383-5pDownregulationBCL2L11(87)
miR-448DownregulationSIRT1(88)
miR-483-3pDownregulationSRF(89)
miR-525p-5pDownregulationBax(90)
let-7e-5pDownregulationFASLG(91)
miRNA-135aUpregulationFOXM1(93)

[i] miRNA, microRNA; DVT, deep vein thrombosis.

miRNAs can promote thrombus dissolution

Fibrinolytic enzyme activation within the newly formed thrombus and the release of lysozyme from cell disintegration allow for gradual thrombus dissolution. The lysis of thrombi depends on numerous factors and previous studies suggest that miRNAs are among them. miR-21 (75), miR-92a-3p (100), miR-126 (61,101), miRNA-136-5p (102) and miR-361-5p (81) increase the expression level of fibrinolytic enzymes, inhibiting the production and activity of fibrinogen and thrombin while promoting thrombus lysis. Additionally, seven additional miRNAs have been reported to be aberrantly expressed in patients with DVT after anticoagulation therapy and stable thrombosis (103). These studies could provide new targets and insights for thrombolysis. While miRNAs have the function of promoting DVT, this effect did not result in complete thrombus lysis and miRNAs were not identified as the most direct or critical factors for thrombus dissolution. The aforementioned studies have only demonstrated reduced thrombus size in animal models and have not been applied in a clinical setting (Table III).

Table III.

miRNAs and target proteins that promote thrombus dissolution.

Table III.

miRNAs and target proteins that promote thrombus dissolution.

miRNAChange in expressionTarget gene(s)(Refs.)
miR-21UpregulationFASLG(75)
miR-92a-3pDownregulationHmgcr(100)
miR-126UpregulationPIK3R2(61,101)
miR-136-5pUpregulationTXNIP(102)
miR-361-5pDownregulationFGF1(81)

[i] miRNA, microRNA.

miRNAs play a crucial role as transcriptional regulators within the human blood system, with considerable research centered on human umbilical vein endothelial cells or endothelial progenitor cells. Consequently, the present review emphasized the role of miRNAs in the regulation of VEC injury, which in turn affects DVT formation by modulating cellular injury. Previous studies have highlighted the involvement of miRNAs in various facets of thrombosis. miRNAs not only impact the function of VECs but also influence platelet activation (104), coagulation factor secretion (105,106) and fibrinolytic enzyme function (107). However, of note, while miRNAs are not primary determinants in thrombus formation and dissolution, they play a significant regulatory role in these processes. These investigations reveal that individual miRNAs can modulate different proteins in VECs and conversely, the same proteins may be subject to regulation by multiple miRNAs. This complexity suggests that the mechanisms through which miRNAs regulate thrombosis are highly intricate.

Clinical application of miRNAs in DVT

Extraction and detection techniques for miRNAs in the clinic

With the increasing focus on research regarding DVT, a growing number of researchers have turned their attention to utilizing miRNAs as biomarkers for DVT (108). Currently, miRNAs are primarily extracted and detected from plasma (109). In patients with DVT, various factors influence changes in miRNA expression in plasma (110), including: i) Active release from VECs, ii) platelet and red blood cell lysis, iii) cellular exosome secretion and iv) the influence of other unknown factors. miRNAs that enter the circulatory system are primarily in the form of nucleic acid-protein complexes (111). Conversely, miRNAs present as monomers are small in molecular weight, convey limited genetic information and are highly susceptible to degradation. Therefore, the majority of miRNAs detected in the circulatory system are in the form of nucleic acid-protein complexes rather than miRNA monomers (110). Detecting circulating miRNAs relies on two main methods: i) Microarrays/chips and ii) reverse transcription-quantitative PCR (RT-qPCR) (112). While microarrays excel at screening hundreds of miRNAs per sample and offer quantitative analysis, their complexity, high cost and limitation to small sample sizes hinder their clinical use, relegating them mainly to research purposes (113). RT-qPCR stands out due to its affordability, ease of use and ability to perform both relative and absolute quantification, rendering it the preferred choice for clinical applications (114). Therefore, the current clinical approach for miRNA detection involves initially using miRNA microarrays to screen out miRNAs with aberrant expression profiles, followed by validation through RT-qPCR to identify the target miRNAs (112).

miRNAs as markers for diagnosing DVT

As miRNAs are highly conserved genetically and stable within the circulatory system, investigations have been initiated into the utilization of aberrantly expressed miRNAs in plasma of patients, as markers for early DVT diagnosis (108). miRNAs present in the circulatory system offer diagnostic advantages over traditional biomarkers for several reasons: i) Circulating miRNAs demonstrate greater stability and reproducibility; ii) they impose less damage to samples, are more convenient to detect and are easier to analyze and iii) alterations in circulating miRNA expression occur earlier, facilitating early detection and diagnosis (115). The high early diagnostic value of miR-582, miR-195, miR-532 (116) and miR-488 (88) in serum for the development of DVT has been well-documented. Similarly, upregulation of miR-424-5p and miR-125a-5p expression levels, alongside downregulation of miR-136-5p and miR-223-3p expression levels, has been observed in patients with DVT compared with healthy participants (108,117). These alterations are associated with a hypercoagulable state of the blood, indicating potential diagnostic value for these miRNAs. By contrast, the diagnostic potential of miRNAs has been investigated by examining their relationship with D-dimers. For instance, the simultaneous detection of D-dimer and miR-96 (118) or miRNA-320a/b (119) has been shown to enhance the diagnostic accuracy of DVT. However, this does not imply that miRNAs surpass D-dimers in diagnostic value; the latter remain indispensable. In summary, the fluctuation in miRNA levels in blood during DVT is influenced by various factors and their specificity is not high. Consequently, studies utilizing miRNAs as diagnostic markers for VTE are still in the experimental stage and have not been extensively implemented in clinical settings. Nevertheless, monitoring changes in miRNA expression levels may offer a more precise assessment of risk levels in patients with DVT (54). Further research into miRNAs is likely to uncover their diagnostic potential to a greater extent.

Advances in miRNA therapy for DVT

Recent studies on miRNA therapy for patients with DVT have revealed significant progress (120). Research into miRNA therapy for various diseases has indicated that miRNA overexpression effectively regulates the transcriptional process of various genes, inhibits tissue inflammatory responses and reduces adverse effects in patients with DVT (121,122). A previous study suggested that when miRNAs enter the pulmonary veins following a DVT episode, they regulate and control cardiomyocyte activity, thus protecting the heart from DVT-induced damage (103). There is potential for treating vascular endothelial cell injury through miRNA upregulation or downregulation, which could pave the way for new therapeutic approaches to DVT intervention. However, challenges persist for miRNA-based therapies, including the need for specific miRNA delivery within tissues, targeted receptor binding and dose optimization (61). To date, the direct miRNA use for DVT therapy has not been reported. Nonetheless, in the future, integrating miRNAs into novel therapeutic strategies may revolutionize DVT treatment.

Conclusions and future perspectives

miRNAs play a crucial role in regulating DVT formation via VECs, rendering them a valuable addition to clinical studies on DVT. These small RNA molecules exert biological effects on DVT formation by modulating VECs. However, there have been no reports of direct antagonistic effects between functionally distinct miRNAs regulating DVT formation. Consequently, the complexity of the miRNA regulatory network in DVT surpasses previous understanding. The vascular endothelium, being in direct contact with the vein wall, maintains a dynamic balance between procoagulation and anticoagulation. miRNAs influence the function of VECs through various pathways, including autophagy and inflammation, disturbing this delicate balance between pro- and anti-coagulation systems. Ultimately, this disruption indirectly regulates thrombus formation. It is unclear whether miRNAs mainly act on venous VECs, thereby regulating the biological function of these cells and potentially exacerbating the coagulation process following vascular endothelium damage. Thus, miRNAs do not directly participate in the endogenous coagulation pathway but rather modulate the process of thrombus formation. Furthermore, the expression levels of miRNAs in plasma are influenced by numerous factors, diminishing their sensitivity and specificity. There is no evidence indicating that the diagnostic efficacy of miRNAs surpasses that of traditional coagulation function tests. Similarly, while the thrombolytic potential of miRNAs may equal or exceed that of conventional thrombolytic drugs, no targeted thrombolytic medications or therapeutic approaches utilizing miRNAs have been developed. Consequently, there has been no significant clinical breakthrough or application of miRNAs in the diagnosis and treatment of DVT. Nevertheless, studying the biological interactions between miRNAs and VECs is crucial for advancing the comprehension of the intricate process of thrombosis. Continuous research and development of extraction, detection and analytical techniques for miRNAs will provide robust support and technical assurances for DVT research. As exploration in this field advances, the future holds promise for expanded applications of miRNA-based disease prevention and treatment. Investigating the regulatory mechanisms of miRNAs in thrombosis holds significant value, offering new insights for early DVT diagnosis and identifying fresh targets for thrombolysis and personalized therapy.

Acknowledgements

Not applicable.

Funding

The present study was supported by The National Natural Science Foundation of China (grant no. 82160375), The Natural Science Foundation of Jiangxi Province (grant no. 20202BABL206035) and The Jiangxi Ganzhou Science and Technology Program Project (grant no. 2023LNS26841).

Availability of data and materials

Not applicable.

Authors' contributions

JMo conceived the theme and reviewed the manuscript. CF and FH wrote the first draft of the manuscript. ZW, MY and JMa revised parts of the manuscript. DW, TG and FZ helped revise the manuscript and reviewed the literature. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Bartel DP: Metazoan MicroRNAs. Cell. 173:20–51. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Matsuyama H and Suzuki HI: Systems and synthetic microRNA biology: From biogenesis to disease pathogenesis. Int J Mol Sci. 21:1322019. View Article : Google Scholar : PubMed/NCBI

3 

Mohr AM and Mott JL: Overview of microRNA biology. Semin Liver Dis. 35:3–11. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Thamotharan S, Chu A, Kempf K, Janzen C, Grogan T, Elashoff DA and Devaskar SU: Differential microRNA expression in human placentas of term intra-uterine growth restriction that regulates target genes mediating angiogenesis and amino acid transport. PLoS One. 12:e01764932017. View Article : Google Scholar : PubMed/NCBI

5 

Lucas T, Schäfer F, Müller P, Eming SA, Heckel A and Dimmeler S: Light-inducible antimiR-92a as a therapeutic strategy to promote skin repair in healing-impaired diabetic mice. Nat Commun. 8:151622017. View Article : Google Scholar : PubMed/NCBI

6 

Siomi H and Siomi MC: Posttranscriptional regulation of microRNA biogenesis in animals. Mol Cell. 38:323–332. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Karreth FA, Tay Y, Perna D, Ala U, Tan SM, Rust AG, DeNicola G, Webster KA, Weiss D, Perez-Mancera PA, et al: In vivo identification of tumor-suppressive PTEN ceRNAs in an oncogenic BRAF-induced mouse model of melanoma. Cell. 147:382–395. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Vegter EL, van der Meer P, de Windt LJ, Pinto YM and Voors AA: MicroRNAs in heart failure: From biomarker to target for therapy. Eur J Heart Fail. 18:457–468. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Jung H, Kim JS, Lee KH, Tizaoui K, Terrazzino S, Cargnin S, Smith L, Koyanagi A, Jacob L, Li H, et al: Roles of microRNAs in inflammatory bowel disease. Int J Biol Sci. 17:2112–2123. 2021. View Article : Google Scholar : PubMed/NCBI

10 

Weidner J, Bartel S, Kılıç A, Zissler UM, Renz H, Schwarze J, Schmidt-Weber CB, Maes T, Rebane A, Krauss-Etschmann S and Rådinger M: Spotlight on microRNAs in allergy and asthma. Allergy. 76:1661–1678. 2021. View Article : Google Scholar : PubMed/NCBI

11 

Rodriguez A, Griffiths-Jones S, Ashurst JL and Bradley A: Identification of mammalian microRNA host genes and transcription units. Genome Res. 14:1902–1910. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Lee Y, Jeon K, Lee JT, Kim S and Kim VN: MicroRNA maturation: Stepwise processing and subcellular localization. EMBO J. 21:4663–4670. 2002. View Article : Google Scholar : PubMed/NCBI

13 

Lee Y, Kim M, Han J, Yeom KH, Lee S, Baek SH and Kim VN: MicroRNA genes are transcribed by RNA polymerase II. EMBO J. 23:4051–4060. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Nguyen TA, Jo MH, Choi YG, Park J, Kwon SC, Hohng S, Kim VN and Woo JS: Functional anatomy of the human microprocessor. Cell. 161:1374–1387. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Lee Y, Ahn C, Han J, Choi H, Kim J, Yim J, Lee J, Provost P, Rådmark O, Kim S and Kim VN: The nuclear RNase III Drosha initiates microRNA processing. Nature. 425:415–419. 2003. View Article : Google Scholar : PubMed/NCBI

16 

Ha M and Kim VN: Regulation of microRNA biogenesis. Nature reviews. Nat Rev Mol Cell Biol. 15:509–524. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Iwasaki S, Kobayashi M, Yoda M, Sakaguchi Y, Katsuma S, Suzuki T and Tomari Y: Hsc70/Hsp90 chaperone machinery mediates ATP-dependent RISC loading of small RNA duplexes. Mol Cell. 39:292–299. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Frank F, Sonenberg N and Nagar B: Structural basis for 5′-nucleotide base-specific recognition of guide RNA by human AGO2. Nature. 465:818–822. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Suzuki HI, Katsura A, Yasuda T, Ueno T, Mano H, Sugimoto K and Miyazono K: Small-RNA asymmetry is directly driven by mammalian Argonautes. Nat Struct Mol. 22:512–521. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Khvorova A, Reynolds A and Jayasena SD: Functional siRNAs and miRNAs exhibit strand bias. Cell. 115:209–216. 2003. View Article : Google Scholar : PubMed/NCBI

21 

Schwarz DS, Hutvágner G, Du T, Xu Z, Aronin N and Zamore PD: Asymmetry in the assembly of the RNAi enzyme complex. Cell. 115:199–208. 2003. View Article : Google Scholar : PubMed/NCBI

22 

Chiang HR, Schoenfeld LW, Ruby JG, Auyeung VC, Spies N, Baek D, Johnston WK, Russ C, Luo S, Babiarz JE, et al: Mammalian microRNAs: Experimental evaluation of novel and previously annotated genes. Genes Dev. 24:992–1009. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Giraldez AJ, Mishima Y, Rihel J, Grocock RJ, Van Dongen S, Inoue K, Enright AJ and Schier AF: Zebrafish MiR-430 promotes deadenylation and clearance of maternal mRNAs. Science. 312:75–79. 2006. View Article : Google Scholar : PubMed/NCBI

24 

Baek D, Villen J, Shin C, Camargo FD, Gygi SP and Bartel DP: The impact of microRNAs on protein output. Nature. 455:64–71. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Selbach M, Schwanhäusser B, Thierfelder N, Fang Z, Khanin R and Rajewsky N: Widespread changes in protein synthesis induced by microRNAs. Nature. 455:58–63. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Landgraf P, Rusu M, Sheridan R, Sewer A, Iovino N, Aravin A, Pfeffer S, Rice A, Kamphorst AO, Landthaler M, et al: A mammalian microRNA expression atlas based on small RNA library sequencing. Cell. 129:1401–1414. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Dragomir MP, Knutsen E and Calin GA: SnapShot: Unconventional miRNA functions. Cell. 174:1038–1038.e1. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Neubauer K and Zieger B: Endothelial cells and coagulation. Cell Tissue Res. 387:391–398. 2022. View Article : Google Scholar : PubMed/NCBI

29 

Krüger-Genge A, Blocki A, Franke RP and Jung F: Vascular endothelial cell biology: An update. Int J Mol Sci. 20:44112019. View Article : Google Scholar : PubMed/NCBI

30 

Di Nisio M, van Es N and Büller HR: Deep vein thrombosis and pulmonary embolism. Lancet. 388:3060–3073. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Juchem G, Weiss DR, Knott M, Senftl A, Förch S, Fischlein T, Kreuzer E, Reichart B, Laufer S and Nees S: Regulation of coronary venular barrier function by blood borne inflammatory mediators and pharmacological tools: Insights from novel microvascular wall models. Am J Physiol Heart Circ Physiol. 302:H567–H581. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Moncada S, Gryglewski R, Bunting S and Vane JR: An enzyme isolated from arteries transforms prostaglandin endoperoxides to an unstable substance that inhibits platelet aggregation. Nature. 263:663–665. 1976. View Article : Google Scholar : PubMed/NCBI

33 

Moncada S, Higgs EA and Vane JR: Human arterial and venous tissues generate prostacyclin (prostaglandin x), a potent inhibitor of platelet aggregation. Lancet. 1:18–20. 1977. View Article : Google Scholar : PubMed/NCBI

34 

Cines DB, Pollak ES, Buck CA, Loscalzo J, Zimmerman GA, McEver RP, Pober JS, Wick TM, Konkle BA, Schwartz BS, et al: Endothelial cells in physiology and in the pathophysiology of vascular disorders. Blood. 91:3527–3561. 1998.PubMed/NCBI

35 

Panza JA, Quyyumi AA, Brush JE Jr and Epstein SE: Abnormal endothelium-dependent vascular relaxation in patients with essential hypertension. N Engl J Med. 323:22–27. 1990. View Article : Google Scholar : PubMed/NCBI

36 

Marti CN, Gheorghiade M, Kalogeropoulos AP, Georgiopoulou VV, Quyyumi AA and Butler J: Endothelial dysfunction, arterial stiffness, and heart failure. J Am Coll Cardiol. 60:1455–1469. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Radomski MW, Palmer RM and Moncada S: Endogenous nitric oxide inhibits human platelet adhesion to vascular endothelium. Lancet. 2:1057–1058. 1987. View Article : Google Scholar : PubMed/NCBI

38 

Deaglio S and Robson SC: Ectonucleotidases as regulators of purinergic signaling in thrombosis, inflammation, and immunity. Adv Pharmacol. 61:301–332. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Marcus AJ, Safier LB, Hajjar KA, Ullman HL, Islam N, Broekman MJ and Eiroa AM: Inhibition of platelet function by an aspirin-insensitive endothelial cell ADPase. Thromboregulation by endothelial cells. J Clin Invest. 88:1690–1696. 1991. View Article : Google Scholar : PubMed/NCBI

40 

Fuentes E and Palomo I: Extracellular ATP metabolism on vascular endothelial cells: A pathway with pro-thrombotic and anti-thrombotic molecules. Vascul Pharmacol. 75:1–6. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Esmon CT: Structure and functions of the endothelial cell protein C receptor. Crit Care Med. 32 (Suppl 5):S298–S301. 2004. View Article : Google Scholar : PubMed/NCBI

42 

Giri H, Panicker SR, Cai X, Biswas I, Weiler H and Rezaie AR: Thrombomodulin is essential for maintaining quiescence in vascular endothelial cells. Proc Natl Acad Sci USA. 118:e20222481182021. View Article : Google Scholar : PubMed/NCBI

43 

Iba T and Levy JH: Inflammation and thrombosis: Roles of neutrophils, platelets and endothelial cells and their interactions in thrombus formation during sepsis. J Thromb Haemost. 16:231–241. 2018. View Article : Google Scholar : PubMed/NCBI

44 

Griffin JH, Evatt B, Zimmerman TS, Kleiss AJ and Wideman C: Deficiency of protein C in congenital thrombotic disease. J Clin Invest. 68:1370–1373. 1981. View Article : Google Scholar : PubMed/NCBI

45 

Ofosu FA, Modi GJ, Smith LM, Cerskus AL, Hirsh J and Blajchman MA: Heparan sulfate and dermatan sulfate inhibit the generation of thrombin activity in plasma by complementary pathways. Blood. 64:742–747. 1984. View Article : Google Scholar : PubMed/NCBI

46 

Mann KG, Butenas S and Brummel K: The dynamics of thrombin formation. Arterioscler Thromb Vasc Biol. 23:17–25. 2003. View Article : Google Scholar : PubMed/NCBI

47 

Loskutoff DJ and Edgington TE: Synthesis of a fibrinolytic activator and inhibitor by endothelial cells. Proc Natl Acad Sci USA. 74:3903–3907. 1977. View Article : Google Scholar : PubMed/NCBI

48 

Huber D, Cramer EM, Kaufmann JE, Meda P, Massé JM, Kruithof EK and Vischer UM: Tissue-type plasminogen activator (t-PA) is stored in Weibel-Palade bodies in human endothelial cells both in vitro and in vivo. Blood. 99:3637–3645. 2002. View Article : Google Scholar : PubMed/NCBI

49 

Henderson SJ, Weitz JI and Kim PY: Fibrinolysis: Strategies to enhance the treatment of acute ischemic stroke. J Thromb Haemost. 16:1932–1940. 2018. View Article : Google Scholar : PubMed/NCBI

50 

Levi M and van der Poll T: Coagulation and sepsis. Thromb Res. 149:38–44. 2017. View Article : Google Scholar : PubMed/NCBI

51 

Levi M and van der Poll T: Inflammation and coagulation. Crit Care Med. 38 (Suppl 2):S26–S34. 2010. View Article : Google Scholar : PubMed/NCBI

52 

Mackman N, Tilley RE and Key NS: Role of the extrinsic pathway of blood coagulation in hemostasis and thrombosis. Arterioscler Thromb Vasc Biol. 27:1687–1693. 2007. View Article : Google Scholar : PubMed/NCBI

53 

Zelaya H, Rothmeier AS and Ruf W: Tissue factor at the crossroad of coagulation and cell signaling. J Thromb Haemost. 16:1941–1952. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Sun LL, Li WD, Lei FR and Li XQ: The regulatory role of microRNAs in angiogenesis-related diseases. J Cell Mol Med. 22:4568–4587. 2018. View Article : Google Scholar : PubMed/NCBI

55 

Zhou DM, Sun LL, Zhu J, Chen B, Li XQ and Li WD: MiR-9 promotes angiogenesis of endothelial progenitor cell to facilitate thrombi recanalization via targeting TRPM7 through PI3K/Akt/autophagy pathway. J Cell Mol Med. 24:4624–4632. 2020. View Article : Google Scholar : PubMed/NCBI

56 

Wang J, Xu X, Li P, Zhang B and Zhang J: HDAC3 protects against atherosclerosis through inhibition of inflammation via the microRNA-19b/PPARγ/NF-κB axis. Atherosclerosis. 323:1–12. 2021. View Article : Google Scholar : PubMed/NCBI

57 

Yu J, Jin Y, Xu C, Fang C, Zhang Z, Chen L and Xu G: Downregulation of miR-125a-5p promotes endothelial progenitor cell migration and angiogenesis and alleviates deep vein thrombosis in mice via upregulation of MCL-1. Mol Biotechnol. 65:1664–1678. 2023. View Article : Google Scholar : PubMed/NCBI

58 

Zhu J, Sun LL, Li WD and Li XQ: Clarification of the role of miR-9 in the angiogenesis, migration, and autophagy of endothelial progenitor cells through RNA sequence analysis. Cell Transplant. 29:9636897209639362020. View Article : Google Scholar : PubMed/NCBI

59 

Liang HZ, Li SF, Zhang F, Wu MY, Li CL, Song JX, Lee C and Chen H: Effect of endothelial microparticles induced by hypoxia on migration and angiogenesis of human umbilical vein endothelial cells by delivering MicroRNA-19b. Chin Med J (Engl). 131:2726–2733. 2018. View Article : Google Scholar : PubMed/NCBI

60 

Lou Z, Ma H, Li X, Zhang F, Du K and Wang B: Hsa_circ_0001020 accelerates the lower extremity deep vein thrombosis via sponging miR-29c-3p to promote MDM2 expression. Thromb Res. 211:38–48. 2022. View Article : Google Scholar : PubMed/NCBI

61 

Meng Q, Wang W, Yu X, Li W, Kong L, Qian A, Li C and Li X: Upregulation of MicroRNA-126 contributes to endothelial progenitor cell function in deep vein thrombosis via its target PIK3R2. J Cell Biochem. 116:1613–1623. 2015. View Article : Google Scholar : PubMed/NCBI

62 

Zhang W, Chen P, Zong H, Ding Y and Yan R: MiR-143-3p targets ATG2B to inhibit autophagy and promote endothelial progenitor cells tube formation in deep vein thrombosis. Tissue Cell. 67:1014532020. View Article : Google Scholar : PubMed/NCBI

63 

Wang W, Zhu X, Du X, Xu A, Yuan X, Zhan Y, Liu M and Wang S: MiR-150 promotes angiogensis and proliferation of endothelial progenitor cells in deep venous thrombosis by targeting SRCIN1. Microvasc Res. 123:35–41. 2019. View Article : Google Scholar : PubMed/NCBI

64 

Du X, Hu N, Yu H, Hong L, Ran F, Huang D, Zhou M, Li C and Li X: miR-150 regulates endothelial progenitor cell differentiation via Akt and promotes thrombus resolution. Stem Cell Res Ther. 11:3542020. View Article : Google Scholar : PubMed/NCBI

65 

Wang W, Li C, Li W, Kong L, Qian A, Hu N, Meng Q and Li X: MiR-150 enhances the motility of EPCs in vitro and promotes EPCs homing and thrombus resolving in vivo. Thromb Res. 133:590–598. 2014. View Article : Google Scholar : PubMed/NCBI

66 

Sun LL, Xiao L, Du XL, Hong L, Li CL, Jiao J, Li WD and Li XQ: MiR-205 promotes endothelial progenitor cell angiogenesis and deep vein thrombosis recanalization and resolution by targeting PTEN to regulate Akt/autophagy pathway and MMP2 expression. J Cell Mol Med. 23:8493–8504. 2019. View Article : Google Scholar : PubMed/NCBI

67 

Ai P, Shen B, Pan H, Chen K, Zheng J and Liu F: MiR-411 suppressed vein wall fibrosis by downregulating MMP-2 via targeting HIF-1α. J Thromb Thrombolysis. 45:264–273. 2018. View Article : Google Scholar : PubMed/NCBI

68 

Li WD, Zhou DM, Sun LL, Xiao L, Liu Z, Zhou M, Wang WB and Li XQ: LncRNA WTAPP1 promotes migration and angiogenesis of endothelial progenitor cells via MMP1 through MicroRNA 3120 and Akt/PI3K/autophagy pathways. Stem Cells. 36:1863–1874. 2018. View Article : Google Scholar : PubMed/NCBI

69 

Li Z and Ni J: Role of microRNA-26a in the diagnosis of lower extremity deep vein thrombosis in patients with bone trauma. Exp Ther Med. 14:5069–5074. 2017.PubMed/NCBI

70 

Sun S, Chai S, Zhang F and Lu L: Overexpressed microRNA-103a-3p inhibits acute lower-extremity deep venous thrombosis via inhibition of CXCL12. IUBMB Life. 72:492–504. 2020. View Article : Google Scholar : PubMed/NCBI

71 

Lu J, Fang Q and Ge X: Role and mechanism of mir-5189-3p in deep vein thrombosis of lower extremities. Ann Vasc Surg. 77:288–295. 2021. View Article : Google Scholar : PubMed/NCBI

72 

Qin JZ, Wang SJ and Xia C: microRNAs regulate nitric oxide release from endothelial cells by targeting NOS3. J Thromb Thrombolysis. 46:275–282. 2018. View Article : Google Scholar : PubMed/NCBI

73 

Yang B and Zhang Z: Suppression of long intergenic non-protein coding RNA 1123 constrains lower extremity deep vein thrombosis via microRNA-125a-3p to target interleukin 1 receptor type 1. Bioengineered. 13:13452–13461. 2022. View Article : Google Scholar : PubMed/NCBI

74 

Ou M, Hao S, Chen J, Zhao S, Cui S and Tu J: Downregulation of interleukin-6 and C-reactive protein underlies a novel inhibitory role of microRNA-136-5p in acute lower extremity deep vein thrombosis. Aging (Albany NY). 12:21076–21090. 2020. View Article : Google Scholar : PubMed/NCBI

75 

Du X, Hong L, Sun L, Sang H, Qian A, Li W, Zhuang H, Liang H, Song D, Li C, et al: miR-21 induces endothelial progenitor cells proliferation and angiogenesis via targeting FASLG and is a potential prognostic marker in deep venous thrombosis. J Transl Med. 17:2702019. View Article : Google Scholar : PubMed/NCBI

76 

Wang W, Yuan X, Xu A, Zhu X, Zhan Y, Wang S and Liu M: Human cancer cells suppress behaviors of endothelial progenitor cells through miR-21 targeting IL6R. Microvasc Res. 120:21–28. 2018. View Article : Google Scholar : PubMed/NCBI

77 

Mo J, Zhang D and Yang R: MicroRNA-195 regulates proliferation, migration, angiogenesis and autophagy of endothelial progenitor cells by targeting GABARAPL1. Biosci Rep. 36:e003962016. View Article : Google Scholar : PubMed/NCBI

78 

Ding M, Chi G, Li F, Wang B, Shao C and Song W: Up-regulated miR-204-5p promoted the migration, invasion, and angiogenesis of endothelial progenitor cells to enhance the thrombolysis of rats with deep venous thrombosis by targeting SPRED1. Exp Cell Res. 411:1129852022. View Article : Google Scholar : PubMed/NCBI

79 

Pan Z, Zhang Y, Li C, Yin Y, Liu R, Zheng G, Fan W, Zhang Q, Song Z, Guo Z, et al: MiR-296-5p ameliorates deep venous thrombosis by inactivating S100A4. Exp Biol Med (Maywood). 246:2259–2268. 2021. View Article : Google Scholar : PubMed/NCBI

80 

Pan Z, Chen Q, Ding H and Li H: MicroRNA-342-3p loaded by human umbilical cord mesenchymal stem cells-derived exosomes attenuates deep vein thrombosis by downregulating EDNRA. J Thromb Thrombolysis. 54:411–419. 2022. View Article : Google Scholar : PubMed/NCBI

81 

Yang X, Song Y, Sun Y, Wang M and Xiang Y: Down-regulation of miR-361-5p promotes the viability, migration and tube formation of endothelial progenitor cells via targeting FGF1. Biosci Rep. 40:BSR202005572020. View Article : Google Scholar : PubMed/NCBI

82 

Li HQ, Pan ZY, Yang Z, Zhang DB and Chen Q: Overexpression of MicroRNA-122 resists oxidative stress-induced human umbilical vascular endothelial cell injury by inhibition of p53. Biomed Res Int. 2020:97916082020.PubMed/NCBI

83 

He X, Liu Y, Li Y and Wu K: Long non-coding RNA crnde promotes deep vein thrombosis by sequestering miR-181a-5p away from thrombogenic Pcyox1l. Thromb J. 21:442023. View Article : Google Scholar : PubMed/NCBI

84 

Jin J, Wang C, Ouyang Y and Zhang D: Elevated miR-195-5p expression in deep vein thrombosis and mechanism of action in the regulation of vascular endothelial cell physiology. Exp Ther Med. 18:4617–4624. 2019.PubMed/NCBI

85 

Li Y, Ge J, Yin Y, Yang R, Kong J and Gu J: Upregulated miR-206 aggravates deep vein thrombosis by regulating GJA1-mediated autophagy of endothelial progenitor cells. Cardiovasc Ther. 2022:99663062022. View Article : Google Scholar : PubMed/NCBI

86 

Zhang Y, Zhang Z, Wei R, Miao X, Sun S, Liang G, Chu C, Zhao L, Zhu X, Guo Q, et al: IL (interleukin)-6 contributes to deep vein thrombosis and is negatively regulated by miR-338-5p. Arterioscler Thromb Vasc Biol. 40:323–334. 2020. View Article : Google Scholar : PubMed/NCBI

87 

Wang H, Lin S, Yang Y, Zhao M, Li X and Zhang L: Significant role of long non-coding RNA MALAT1 in deep vein thrombosis via the regulation of vascular endothelial cell physiology through the microRNA-383-5p/BCL2L11 axis. Bioengineered. 13:13728–13738. 2022. View Article : Google Scholar : PubMed/NCBI

88 

Wang P, Dai J and Li D: Peripheral blood levels of miR-448 and SIRT1 in patients with deep venous thrombosis and their relationship. Clin Lab. 68:2022. View Article : Google Scholar

89 

Kong L, Hu N, Du X, Wang W, Chen H, Li W, Wei S, Zhuang H, Li X and Li C: Upregulation of miR-483-3p contributes to endothelial progenitor cells dysfunction in deep vein thrombosis patients via SRF. J Transl Med. 14:232016. View Article : Google Scholar : PubMed/NCBI

90 

Zhu X, Chen B and Xu H: By modulating miR-525-5p/Bax axis, LINC00659 promotes vascular endothelial cell apoptosis. Immun Inflamm Dis. 11:e7642023. View Article : Google Scholar : PubMed/NCBI

91 

Kong L, Du X, Hu N, Li W, Wang W, Wei S, Zhuang H, Li X and Li C: Downregulation of let-7e-5p contributes to endothelial progenitor cell dysfunction in deep vein thrombosis via targeting FASLG. Thromb Res. 138:30–36. 2016. View Article : Google Scholar : PubMed/NCBI

92 

Mulder FI, Horváth-Puhó E, van Es N, van Laarhoven HWM, Pedersen L, Moik F, Ay C, Büller HR and Sørensen HT: Venous thromboembolism in cancer patients: A population-based cohort study. Blood. 137:1959–1969. 2021. View Article : Google Scholar : PubMed/NCBI

93 

Tavares V, Neto BV, Marques IS, Assis J, Pereira D and Medeiros R: Cancer-associated thrombosis: What about microRNAs targeting the tissue factor coagulation pathway? Biochim Biophys Acta Rev Cancer. 1879:1890532024. View Article : Google Scholar : PubMed/NCBI

94 

Lazar S and Goldfinger LE: Platelet microparticles and miRNA transfer in cancer progression: Many targets, modes of action, and effects across cancer stages. Front Cardiovasc Med. 5:132018. View Article : Google Scholar : PubMed/NCBI

95 

Liu S, Guo W, Shi J, Li N, Yu X, Xue J, Fu X, Chu K, Lu C, Zhao J, et al: MicroRNA-135a contributes to the development of portal vein tumor thrombus by promoting metastasis in hepatocellular carcinoma. J Hepatol. 56:389–396. 2012. View Article : Google Scholar : PubMed/NCBI

96 

Oto J, Navarro S, Larsen AC, Solmoirago MJ, Plana E, Hervás D, Fernández-Pardo Á, España F, Kristensen SR, Thorlacius-Ussing O and Medina P: MicroRNAs and neutrophil activation markers predict venous thrombosis in pancreatic ductal adenocarcinoma and distal extrahepatic cholangiocarcinoma. Int J Mol Sci. 21:8402020. View Article : Google Scholar : PubMed/NCBI

97 

Anijs RJS, Laghmani EH, Ünlü B, Kiełbasa SM, Mei H, Cannegieter SC, Klok FA, Kuppen PJK, Versteeg HH and Buijs JT: Tumor-expressed microRNAs associated with venous thromboembolism in colorectal cancer. Res Pract Thromb Haemost. 6:e127492022. View Article : Google Scholar : PubMed/NCBI

98 

Morelli VM, Snir O, Hindberg KD, Hveem K, Brækkan SK and Hansen JB: High microRNA-145 plasma levels are associated with decreased risk of future incident venous thromboembolism-The HUNT study. Blood. blood.2023022285. 2024.(Epub ahead of print). View Article : Google Scholar

99 

Anijs RJS, Nguyen YN, Cannegieter SC, Versteeg HH and Buijs JT: MicroRNAs as prognostic biomarkers for (cancer-associated) venous thromboembolism. J Thromb Haemost. 21:7–17. 2023. View Article : Google Scholar : PubMed/NCBI

100 

Feng Y, Lei B, Zhang H, Niu L, Li X, Luo X and Zhang F: Long noncoding RNA TUG1 induces angiogenesis of endothelial progenitor cells and dissolution of deep vein thrombosis. Thromb J. 20:542022. View Article : Google Scholar : PubMed/NCBI

101 

Kuhnert F, Mancuso MR, Hampton J, Stankunas K, Asano T, Chen CZ and Kuo CJ: Attribution of vascular phenotypes of the murine Egfl7 locus to the microRNA miR-126. Development. 135:3989–3993. 2008. View Article : Google Scholar : PubMed/NCBI

102 

Feng Y, Lei B, Zhang H, Niu L, Li X, Luo X and Zhang F: MicroRNA-136-5p from endothelial progenitor cells-released extracellular vesicles mediates TXNIP to promote the dissolution of deep venous thrombosis. Shock. 57:714–721. 2022. View Article : Google Scholar : PubMed/NCBI

103 

Jin QQ, Sun JH, Du QX, Lu XJ, Zhu XY, Fan HL, Hölscher C and Wang YY: Integrating microRNA and messenger RNA expression profiles in a rat model of deep vein thrombosis. Int J Mol Med. 40:1019–1028. 2017. View Article : Google Scholar : PubMed/NCBI

104 

Edelstein LC, McKenzie SE, Shaw C, Holinstat MA, Kunapuli SP and Bray PF: MicroRNAs in platelet production and activation. J Thromb Haemost. 11 (Suppl 1):S340–S350. 2013. View Article : Google Scholar

105 

Jankowska KI, Sauna ZE and Atreya CD: Role of microRNAs in hemophilia and thrombosis in humans. Int J Mol Sci. 21:35982020. View Article : Google Scholar : PubMed/NCBI

106 

Hembrom AA, Srivastava S, Garg I and Kumar B: MicroRNAs in venous thrombo-embolism. Clin Chim Acta. 504:66–72. 2020. View Article : Google Scholar : PubMed/NCBI

107 

Marques-Rocha JL, Samblas M, Milagro FI, Bressan J, Martínez JA and Marti A: Noncoding RNAs, cytokines, and inflammation-related diseases. FASEB J. 29:3595–3611. 2015. View Article : Google Scholar : PubMed/NCBI

108 

Wang X, Sundquist K, Elf JL, Strandberg K, Svensson PJ, Hedelius A, Palmer K, Memon AA, Sundquist J and Zöller B: Diagnostic potential of plasma microRNA signatures in patients with deep-vein thrombosis. Thromb Haemost. 116:328–336. 2016. View Article : Google Scholar : PubMed/NCBI

109 

Monguió-Tortajada M, Gálvez-Montón C, Bayes-Genis A, Roura S and Borràs FE: Extracellular vesicle isolation methods: Rising impact of size-exclusion chromatography. Cell Mol Life Sci. 76:2369–2382. 2019. View Article : Google Scholar : PubMed/NCBI

110 

Felekkis K and Papaneophytou C: Challenges in using circulating Micro-RNAs as biomarkers for cardiovascular diseases. Int J Mol Sci. 21:5612020. View Article : Google Scholar : PubMed/NCBI

111 

He Y, Lin J, Kong D, Huang M, Xu C, Kim TK, Etheridge A, Luo Y, Ding Y and Wang K: Current state of circulating MicroRNAs as cancer biomarkers. Clin Chem. 61:1138–1155. 2015. View Article : Google Scholar : PubMed/NCBI

112 

Ban E and Song EJ: Considerations and suggestions for the reliable analysis of miRNA in plasma using qRT-PCR. Genes (Basel). 13:3282022. View Article : Google Scholar : PubMed/NCBI

113 

Masubuchi T, Endo M, Iizuka R, Iguchi A, Yoon DH, Sekiguchi T, Qi H, Iinuma R, Miyazono Y, Shoji S, et al: Construction of integrated gene logic-chip. Nat Nanotechnol. 13:933–940. 2018. View Article : Google Scholar : PubMed/NCBI

114 

Andrews WJ, Brown ED, Dellett M, Hogg RE and Simpson DA: Rapid quantification of microRNAs in plasma using a fast real-time PCR system. Biotechniques. 58:244–252. 2015. View Article : Google Scholar : PubMed/NCBI

115 

Chandrasekaran AR, Punnoose JA, Zhou L, Dey P, Dey BK and Halvorsen K: DNA nanotechnology approaches for microRNA detection and diagnosis. Nucleic Acids Res. 47:10489–10505. 2019. View Article : Google Scholar : PubMed/NCBI

116 

Qin J, Liang H, Shi D, Dai J, Xu Z, Chen D, Chen X and Jiang Q: A panel of microRNAs as a new biomarkers for the detection of deep vein thrombosis. J Thromb Thrombolysis. 39:215–221. 2015. View Article : Google Scholar : PubMed/NCBI

117 

Xu L, Ji C, Miao X, Ge J, Li F and Xu C: Combination of circulating miR-125a-5p, miR-223-3p and D-dimer as a novel biomarker for deep vein thrombosis. Am J Med Sci. 364:601–611. 2022. View Article : Google Scholar : PubMed/NCBI

118 

Xie X, Liu C, Lin W, Zhan B, Dong C, Song Z, Wang S, Qi Y, Wang J and Gu Z: Deep vein thrombosis is accurately predicted by comprehensive analysis of the levels of microRNA-96 and plasma D-dimer. Exp Ther Med. 12:1896–1900. 2016. View Article : Google Scholar : PubMed/NCBI

119 

Jiang Z, Ma J, Wang Q, Wu F, Ping J and Ming L: Combination of circulating miRNA-320a/b and D-dimer improves diagnostic accuracy in deep vein thrombosis patients. Med Sci Monit. 24:2031–2037. 2018. View Article : Google Scholar : PubMed/NCBI

120 

Sun LL, Liu Z, Ran F, Huang D, Zhang M, Li XQ and Li WD: Non-coding RNAs regulating endothelial progenitor cells for venous thrombosis: Promising therapy and innovation. Stem Cell Res Ther. 15:72024. View Article : Google Scholar : PubMed/NCBI

121 

Gareri C, De Rosa S and Indolfi C: MicroRNAs for restenosis and thrombosis after vascular injury. Circ Res. 118:1170–1184. 2016. View Article : Google Scholar : PubMed/NCBI

122 

Lu Y, Thavarajah T, Gu W, Cai J and Xu Q: Impact of miRNA in atherosclerosis. Arterioscler Thromb Vasc Biol. 38:e159–e170. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2024
Volume 29 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Fang C, Huang F, Yao M, Wang Z, Ma J, Wu D, Guo T, Zhang F and Mo J: Advances in microRNA regulation of deep vein thrombosis through venous vascular endothelial cells (Review). Mol Med Rep 29: 96, 2024
APA
Fang, C., Huang, F., Yao, M., Wang, Z., Ma, J., Wu, D. ... Mo, J. (2024). Advances in microRNA regulation of deep vein thrombosis through venous vascular endothelial cells (Review). Molecular Medicine Reports, 29, 96. https://doi.org/10.3892/mmr.2024.13220
MLA
Fang, C., Huang, F., Yao, M., Wang, Z., Ma, J., Wu, D., Guo, T., Zhang, F., Mo, J."Advances in microRNA regulation of deep vein thrombosis through venous vascular endothelial cells (Review)". Molecular Medicine Reports 29.6 (2024): 96.
Chicago
Fang, C., Huang, F., Yao, M., Wang, Z., Ma, J., Wu, D., Guo, T., Zhang, F., Mo, J."Advances in microRNA regulation of deep vein thrombosis through venous vascular endothelial cells (Review)". Molecular Medicine Reports 29, no. 6 (2024): 96. https://doi.org/10.3892/mmr.2024.13220