Abstract

The haemostatic system is pivotal to maintaining vascular integrity. Multiple components involved in blood coagulation have central functions in inflammation and immunity. A derailed haemostasis is common in prevalent pathologies such as sepsis, cardiovascular disorders, and lately, COVID-19. Physiological mechanisms limit the deleterious consequences of a hyperactivated haemostatic system through adaptive changes in gene expression. While this is mainly regulated at the level of transcription, co- and posttranscriptional mechanisms are increasingly perceived as central hubs governing multiple facets of the haemostatic system. This layer of regulation modulates the biogenesis of haemostatic components, for example in situations of increased turnover and demand. However, they can also be ‘hijacked’ in disease processes, thereby perpetuating and even causally entertaining associated pathologies. This review summarizes examples and emerging concepts that illustrate the importance of posttranscriptional mechanisms in haemostatic control and crosstalk with the immune system. It also discusses how such regulatory principles can be used to usher in new therapeutic concepts to combat global medical threats such as sepsis or cardiovascular disorders.

1. Introduction

In light of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) pandemic, the mechanisms underlying the crosstalk between the haemostatic system and the immune system have received unprecedented attention. This interplay plays a central role in many pathological processes, ranging from sepsis to cardiovascular disease.

Perturbations of the haemostatic system are common in sepsis, the leading cause of death in critically ill patients worldwide.1 As a systemic inflammatory response to severe infections, sepsis involves excessive activation of the coagulation system.2 This can result in severe complications such as disseminated intravascular coagulation, which eventually leads to tissue necrosis, multiple organ failure, and death, illustrating that inappropriate amplification of protective host-defense mechanisms can become a devastating alliance of harm.3

Cardiovascular disorders (CVDs) including myocardial infarction, ischaemic stroke, and venous thromboembolism are the leading global cause of mortality with over 17 million deaths annually.4 The incidence of CVDs increases markedly with age, starting in the late 40 s, with a dramatic increase occurring at 60 years of age.5 They account for approximately 32% of all deaths worldwide, underscoring the need of illuminating underlying mechanisms and devising therapeutic interventions to treat and prevent CVDs.6

The immune system and the haemostatic system are closely linked,7 and their responses tend to reinforce each other.8,9 Activation of coagulation and fibrin deposition in response to inflammation is well known. This led to the emergence of the concept of immunothrombosis, a defence mechanism in which inflammatory cells participate in thrombotic processes, and thrombosis in turn acts as an intravascular effector of innate immunity by limiting the spread of invading pathogens.10 However, a derailed haemostatic response can lead to a situation where coagulation, fibrin deposition, and thrombosis contribute to disease, as evidenced by the propagation and exacerbation of atherosclerotic plaques.11 Another example is the systemic activation of coagulation combined with microvascular failure resulting from the systemic inflammatory response to severe infection or sepsis, which eventually contributes to multiple organ dysfunction, such as in septicaemia3 or COVID-19.12

The multifaceted and intricate link between haemostasis and inflammation involves crosstalk between both systems at multiple levels,3,7–11 including coordinated changes in gene expression in megakaryocytes, immune cells, the vessel wall, and/or the liver. A notable example is the acute phase response, in which central haemostatic components such as fibrinogen,13,14 Von Willebrand factor,15,16 and factor VIII17–21 are induced in response to inflammatory signals. Such changes in gene expression are primarily regulated at the level of transcription, and the transcriptional regulation of haemostasis-related genes in physiological and pathological conditions has been well studied.14,22–26

In the present review, we focus on emerging concepts of posttranscriptional mechanisms underlying the control of haemostasis and its crosstalk to other systems. In doing so, we discuss examples of the complexity of the transcriptome architecture arising from the use of alternative transcription start sites, exons, and polyadenylation sites, as well as gene regulation by non-coding RNAs [microRNAs (miRNAs), long-non-coding RNAs (lncRNAs), circular RNAs (circRNAs)], RNA-binding proteins (RBPs), and mechanisms of RNA modification. Remarkably, many of these regulatory principles also play an important functional role in tuning the immune system,27–31 suggesting conserved regulatory links between both systems. Finally, we also illustrate the emerging therapeutic opportunities on the cusp of a new era of targeted therapeutic approaches,32 exemplified by the recent introduction of novel RNA therapeutics in the haemostatic system.33

2. Role of splicing regulation in the haemostatic system

With the completion of the human genome project in 2003, it became apparent that the human genome comprises around 22.000 protein-coding genes, far less than actually required for the functional complexity in higher eukaryotes.34 On the other hand, next generation RNA sequencing and particularly the recently introduced long-read sequencing technologies35,36 are uncovering a perplexingly complex transcriptome architecture that arises from the use of alternative transcription start sites, exons, and polyadenylation sites.37,38 The combinatorial use of such elements considerably expands genomic information and is subject to dynamic spatial and temporal modulation during development and adaptation (Figure 1).

The functional complexity encoded by approximately 22.000 genes is substantially diversified by co- and post-transcriptional mechanisms involving alternative transcription initiation, alternative splicing and alternative polyadenylation (APA). Regulation by non-coding RNAs such as miRNAs, lncRNAs, circRNAs, as well as RBPs and RNA modifications, further tunes the functional output of the transcriptome. Modulation of the biogenesis and the post-transcriptional fate of RNAs (RNA localization, transport, translation, stability or decay, RNA modifications) are emerging therapeutic principles (further details see text; *39 **40).
Figure 1

The functional complexity encoded by approximately 22.000 genes is substantially diversified by co- and post-transcriptional mechanisms involving alternative transcription initiation, alternative splicing and alternative polyadenylation (APA). Regulation by non-coding RNAs such as miRNAs, lncRNAs, circRNAs, as well as RBPs and RNA modifications, further tunes the functional output of the transcriptome. Modulation of the biogenesis and the post-transcriptional fate of RNAs (RNA localization, transport, translation, stability or decay, RNA modifications) are emerging therapeutic principles (further details see text; *39 **40).

Pre-messenger RNA (mRNA) splicing, i.e. the accurate removal of introns and ligation of exons, is a pivotal step in the co- and posttranscriptional regulation of gene expression.41 Depending on how the exon/intron structure of the pre-mRNA is decoded by the spliceosome, the same primary transcript may be processed into different mature mRNAs (alternative splicing), encoding different isoforms of the same protein. In fact, the recognition of exon/intron boundaries in the pre-mRNA is critically dependent on the engagement of nearby splicing enhancer and silencer sequences by trans-acting proteins (splicing factors) whose availability varies in different cell types and disease states. As a consequence, splicing patterns are typically regulated in a tissue-specific manner and may change according to the developmental stage or in response to pathological processes. Moreover, they can be disrupted by genetic variants that weaken (or strengthen) the consensus sequences recognized by the spliceosome on the pre-mRNA. This is a well-known mechanism of disease in mendelian disorders,42 but it is increasingly appreciated that much of the genetic variation associated with complex traits also acts by altering splicing patterns.43,44

Like most human genes,45 many genes encoding proteins of the haemostatic system are alternatively spliced.46–58 This often results in isoforms with distinct structural and functional characteristics, as exemplified by two major components of the extrinsic coagulation pathway (Figure 2).

Alternative splicing in components of the haemostatic system, resulting in distinct structural and biochemical characteristics. Of note, the 5’UTR of TFPI contains several non-coding exons (not to scale), a regulatory feature found in many genes.59,60
Figure 2

Alternative splicing in components of the haemostatic system, resulting in distinct structural and biochemical characteristics. Of note, the 5’UTR of TFPI contains several non-coding exons (not to scale), a regulatory feature found in many genes.59,60

Tissue factor (TF), the main trigger of blood coagulation, acts as cofactor of the circulating serine protease factor VIIa (FVIIa) and comes in two isoforms: as membrane-bound (full-length) protein and as a shorter, alternatively spliced variant that is secreted in soluble form (Figure 2).61 The two isoforms are identical at the N-terminal end, but the soluble form, which arises from exon 5 skipping, lacks the transmembrane and cytoplasmic domains, and has a completely different C-terminal sequence.61 Just as full-length TF, alternatively spliced TF is produced by a variety of cell types,61,62 is induced by pro-inflammatory stimuli62,63 and enhances factor X (FX) activation by FVIIa, albeit less potently than full-length TF.61 However, while membrane-bound TF is essential for normal haemostasis, elevated intravascular levels of TF have been proposed to contribute to venous as well as arterial thrombosis.64 Despite conflicting data, it has been suggested that soluble TF, which is most likely dispensable for normal haemostasis, may represent a preferential target for antithrombotic therapy than full-length TF, due to a lower risk of bleeding.65

TF pathway inhibitor (TFPI) is a glycoprotein that functions as an inhibitor of coagulation and of TF-dependent signalling.66 The TFPI gene encodes two main splicing isoforms that are generated by the alternative inclusion of exon 8 (TFPIβ) or exons 9–10 (TFPIα) in the mature mRNA (Figure 2). Both isoforms are expressed in endothelial cells, but TFPIα is also found in plasma, platelets, and the extracellular matrix.67 Structurally, TFPIα comprises an acidic N-terminus, three Kunitz domains, and a basic C-terminus, whereas TFPIβ lacks the third Kunitz domain and the basic C-terminus, which are replaced by a glycosylphosphatidylinositol-anchor that tethers the protein to the cell membrane.68 Both TFPI isoforms inhibit TF/FVIIa and FXa with their Kunitz-1 and Kunitz-2 domains, respectively, but TFPIα has additional properties by virtue of its Kunitz-3 domain (which binds protein S) and basic C-terminus (which binds FV/FV-short). Binding to protein S and FV/FV-short prevents the clearance of plasma TFPIα from the circulation52,69,70 and promotes its association with biological membranes, enhancing its anti-FXa activity.71–73 Moreover, the interaction with FV/FV-short allows TFPIα to inhibit FV activation74 and early prothrombinase activity,75,76 while TFPIβ lacks these anticoagulant functions.

These and other52,77 examples illustrate how alternative splicing can change the structural and hence functional properties of central components in the haemostatic system.78 Extracellular signals, such as pro-inflammatory cytokines, can modify global patterns of alternative splicing79 and it will be interesting to explore how this plays out in different (disease) contexts, including COVID-19.80 Moreover, since alternative splicing is pervasive and there are increasingly new therapeutic means to (re)direct splicing,81,82 modulation of alternative splicing may become relevant for the therapeutic manipulation of the haemostatic system. In particular, many studies support the utility of antisense oligonucleotides (ASOs) to mask specific splicing signals on the pre-mRNA and thus prevent the recognition of these sequences by spliceosomal components, thereby re-directing splicing.83 Alternatively, ad hoc engineered U1snRNA can be employed to promote the usage of donor splice sites that are naturally weak or have been disrupted by mutation.84

Apart from diversifying the transcriptome and proteome, alternative splicing has been proposed to contribute to the overall regulation of gene expression through its coupling with non-sense mediated decay (NMD), a surveillance pathway that degrades mRNAs containing premature stop codons. In fact, it has been observed that up to one third of all human transcripts are normally spliced into non-viable mRNAs that are substrates for NMD. This phenomenon, known as ‘regulated unproductive splicing and translation’ (RUST), has been interpreted as a mechanism for the post-transcriptional temporal and spatial fine-tuning of gene expression.85 Evidence that this control mechanism may apply within the realm of haemostasis has been provided for the F11 gene, encoding coagulation FXI.48 Interestingly, targeting non-productive splicing by ASOs can be exploited for the upregulation of gene expression from wild-type or hypomorphic alleles in disease states.86

3. Role of polyadenylation in the haemostatic system

In addition to capping and splicing, almost all eukaryotic transcripts undergo further processing at the RNA 3′-end (Figure 1). For most genes, this involves endonucleolytic cleavage and non-templated polyadenylation (CPA) before the mature RNA can be exported to the cytoplasm.87 As CPA controls almost all genes, regulation of CPA has evolved as an important layer of gene expression regulation. CPA is carried out by a multi-subunit complex involving over 80 trans-acting proteins organized in four core protein subcomplexes.88 The recruitment of these multimeric complexes to dedicated, but largely poorly conserved, RNA sequence elements89 ensures that 3′-end processing of the nascent transcript occurs timely and at the right position.90,91 Perturbations of this process—due to mutations in RNA sequence elements or defects in the RNA processing machinery—have drastic consequences, as exemplified by numerous diseases.92,93

The common thrombophilia mutation in the prothrombin (F2) gene (F2 G20210A) is a prime example of how mutations in noncoding regions can become pathogenic.87 This mutation affects the last nucleotide of the 3′-untranslated region (UTR), where the pre-mRNA is cleaved and polyadenylated. As a result of the mutation, the efficiency of endonucleolytic cleavage is increased, leading to more prothrombin mRNA and protein expression. Although this mutation merely increases the amount of the precursor of a central haemostatic component (i.e. thrombin), it already shifts the balance of the haemostatic system towards a procoagulant condition.94–96 Consequently, the expression of F2 must be tightly controlled: even small changes (1.5- to 1.7-fold) in gene expression due to mutations at this and other nearby positions (F2 C20209T and F2 G20221T)96,97 can result in clinically relevant thrombophilia.97–100

Compared to other genes, the architecture of sequence determinants directing 3′-end processing in F2 is unconventional.99 It consists of weak signals, which explains the unusual susceptibility to thrombophilic gain-of-function mutations.97,100 At the same time, this configuration allows for mechanisms that enhance processing and thereby upregulate F2 expression when needed.101 This is achieved through complex, mutually exclusive binding of suppressive and stimulatory RNA-binding proteins (RBPs), and is regulated by activation of the p38 mitogen-activated protein kinase (MAPK) signal transduction pathway (Figure 3).102 After phosphorylation by p38 MAPK, inhibitory RBPs (FBP2, FBP3) can no longer bind to the processing sites in the F2 pre-mRNA, allowing 3′-end processing to proceed. Thus, virtually all types of ‘environmental’ conditions that lead to activation of p38 MAPK103,104 can induce F2 expression.

Modulated 3′end processing as a principle to rapidly adjust protein output. Example shown for the prothrombin (F2) gene, where mutually exclusive binding of inhibitory (red) and stimulatory (green) RNA-binding proteins modulates cleavage and polyadenylation of the F2 pre-mRNA. Upon induction of p38 MAPK signalling, the abundance of cleavage and polyadenylation (CPA) factors (grey) is induced, and the inhibitory (-) proteins (FBP2 and FBP3, shown in red) are phosphorylated. This impairs RNA binding of these proteins, and allows for binding of stimulatory (+) components (green), which eventually enhances RNA maturation and protein output (modified from102).
Figure 3

Modulated 3′end processing as a principle to rapidly adjust protein output. Example shown for the prothrombin (F2) gene, where mutually exclusive binding of inhibitory (red) and stimulatory (green) RNA-binding proteins modulates cleavage and polyadenylation of the F2 pre-mRNA. Upon induction of p38 MAPK signalling, the abundance of cleavage and polyadenylation (CPA) factors (grey) is induced, and the inhibitory (-) proteins (FBP2 and FBP3, shown in red) are phosphorylated. This impairs RNA binding of these proteins, and allows for binding of stimulatory (+) components (green), which eventually enhances RNA maturation and protein output (modified from102).

Inflammatory conditions are known to trigger F2 expression.7,105–109 Consistently, the mechanism described here was found to account for the induction of F2 expression under inflammatory conditions, including septicaemia.102,110 While this may contribute to the initial onset and undesirable propagation of haemostatic perturbances during septicaemia, such mechanisms may also play a compensatory role.3 After an initial hypercoagulable state, septicaemia is often followed by a haemorrhagic phase, in part due to consumption of procoagulant components.111 Such conditions of increased turnover and demand require mechanisms to restore the haemostatic balance and stockpile haemostatic components.112

In addition to the critical function in haemostasis, the role of thrombin in angiogenesis113 suggests that regulatory mechanisms have evolved a sensor for low oxygen pressure. This could explain why F2 is overexpressed due to ischaemic events114 or in the tumour micromilieu.102 Consistent with its role in oxygen pressure sensing,103,104 activation of p38 MAPK also drives F2 overexpression in the tumour microenvironment. This activates protease-activated receptors that induce genes with a role in angiogenesis and tumour dissemination.102

Thus, regulated 3′-end processing emerged as an important mechanism of gene regulation in the control of the haemostatic system. While such mechanisms are desirable under physiological conditions (to replenish the amount of blood coagulation factors under high turnover, see above), they can be ‘hijacked’ under pathological conditions (such as inflammation or cancer), thereby leading to a thrombophilic state.110,115Since prothrombin is expressed in a wide variety of organs and cells,110this type of regulation may become relevant to numerous other thrombin-mediated diseases.115However, it also appears that tissue-specific mechanisms can be used to selectively target deleterious prothrombin expression without altering essential prothrombin expression in the liver.110

Targeted interference with cleavage and polyadenylation is increasingly perceived as an important therapeutic means. This involves either redirection of aberrant RNA processing (through ASOs, U1snRNP interference or trans-splicing) or the elimination of faulty transcripts92 to prevent the fatal consequences of aberrant 3′-end processing.116,117 Perturbations of 3′-end processing can, for example, act as nongenomic oncogenic drivers of tumourigenesis,117 but they also play important roles in inflammatory conditions.118 Deciphering the underlying mechanisms is of paramount importance for establishing targets with therapeutic selectivity and specificity.

RNA-protein interactome studies119 and transcriptome-wide profiling of polyadenylation120 are thus central to defining new therapeutic targets, their specificity and downstream consequences.121 Since most miRNA-binding sites are localized in the 3′-UTR, when and where a pre-mRNA is polyadenylated has a critical impact on the regulatory properties of the resulting mRNA molecule (see below). A significant proportion of genetic variants in 3′-UTRs, often dismissed as ‘non-functional’ polymorphisms, are therefore likely to disrupt important regulatory mechanisms, ultimately leading to pathologies including a dysbalanced haemostatic system.92 This is supported by the thrombophilia variants discovered in the F2 gene. However, this also extends to other coagulation factor 3′-UTR variants that affect, for example, miRNA regulation.122,123

4. Role of miRNAs in the haemostatic system

miRNAs are small single-stranded non-coding RNAs (17–25 nucleotides in length) that post-transcriptionally down-regulate target gene expression by RNA silencing.124 After transcription, miRNAs are processed in the nucleus by the microprocessor complex consisting of Drosha and DGCR8 to produce a pre-miRNA.125 After export to the cytoplasm and further processing by Dicer,126 the mature miRNA duplex is incorporated into the RNA-induced silencing complex (RISC).127 This complex is guided by miRNA base pairing to a target gene mRNA resulting in translational inhibition and/or transcript degradation.128 Generally, miRNAs target mRNAs via the 3'-UTR. In a few cases, miRNAs can also carry out their inhibitory function by binding to the coding region or the 5′-UTR of target mRNAs.129

Over 2600 human miRNAs have been identified,130 regulating the majority of human genes.131 Thus almost every biological process is modulated through miRNAs.132 Although miRNAs generally fine-tune gene expression,133 they can also function as master regulators.134 For example, multiple miRNAs can cooperatively silence a single gene to gain regulatory specificity, with the targeting of particular network hub genes enabling the regulation of entire pathways.135 In addition, a single miRNA can target multiple genes, allowing broad regulation of molecular networks.129,135 Perturbations of miRNA expression are observed in most disorders, with some of them even causally contributing to the development and progression of disease.132

A growing number of studies document a contribution of miRNAs to the regulation of haemostatic135–139 and thrombotic123,135,138–141 functions. miRNAs directly regulate multiple haemostatic factors through interactions with the 3'-UTR (Table 1). Additionally, miRNAs can tune haemostatic factors indirectly, for example fibrinogen via interleukin-6-mediated signalling,187 factor IX by repressing NMD,188 plasminogen activator inhibitor 1 (PAI-1) via SMAD2 signalling189 and CXCL12 to reduce inflammatory response and thrombosis, altering the expression of multiple factors including TF, PAI-1, and VWF.190

Table 1

Haemostatic components under miRNA control and relation to thromboinflammation

ProcoagulantMain miRNAs (functionally validated)References
Fibrinogen alphaFGAmiR-193b-3p
miR-194-5p
miR-759
135,142
135
142
Fibrinogen betaFGBmiR-409-3p (miR-29 family)143
Fibrinogen gammaFGGmiR-99b-3p
miR-193a-5p
135
Coagulation factor III, tissue factorF3miR-19b
 • Anti-thrombotic protector in patients with unstable angina
144,145,146,147,148,150
144
miR-19b, miR-20a
 • Down-regulation contributes to a hypercoagulable state in SLE and APS
145
miR-126
 • Reduces thrombogenicity in diabetes mellitus
146
miR-145
 • Impedes thrombus formation in venous thrombosis
147
miR-223
 • Partially blocks TNF-α-induced increase of TF activity in endothelial cells
148
miR-365a-3p
 • Interacts with TF 3’-UTR to modulate TF-initiated thrombin generation
149
Coagulation factor VIIF7miR-19a-3p
miR-19b-3p
135
Coagulation factor VIIIF8miR-7-5p
miR 454-3p
miR-532-5p
miR-1246
135,151
135
135
151
Coagulation factor XIF11miR-15b-5p
 • Biomarker for PAD
 • Influences platelet reactivity and clopidogrel response
135
152
153
miR-24-3p
 • Biomarker for acute cerebral infarction, arteriosclerosis obliterans, atherosclerosis and severe trauma
135
154,155,156,157
miR-30a-3p
 • Biomarker for AMI and ischaemic stroke
135
158,159
miR-30d-3p135
miR-96-5p
 • Biomarker for DVT and DIC
135
160,161
miR-103a-3p
 • Involved in atherosclerosis and vascular inflammation by suppression of KLF4
 • Biomarker for VTE
135
162
163
miR-145-5p
 • Biomarker for CAD, AMI, stroke, long-term outcome
 • Impedes thrombus formation in atherosclerosis by targeting tissue factor and influencing platelet reactivity
164
165,166,167,168,169,170,171,172
147,153
miR-148b-3p135
miR-151a-3p135
miR-181a-5p
 • Biomarker for AMI and PAD
164,173
152,174
miR-181b-5p135
miR-191b-5p173
miR-544a175
miR-1255a
 • Biomarker for stroke
135
176
(pre)kallikreinKLKB1miR-24-3p135
Von Willebrand factorVWFmiR-24177,178
ADAM metallopeptidase with thrombospondin type 1 motif 13ADAMTS13miR-525-5p179
Anticoagulant
Tissue factor pathway inhibitorTFPImiR-27a/b
miR-494
miR-27a/b-3p
180,181
AntithrombinSERPINC1miR-19b-3p
miR-186-5p
135
Protein CPROCmiR-494
let-7 family
135
Protein SPROS1miR-494182
Protein ZPROZmiR-30a-5p
miR-128-3p
miR-148a-3p
miR-148b-3p
miR-375
miR-671-3p
135
Protein Z-dependent ;?>protease inhibitorSERPINA10miR-15b-5p
miR-16-5p
miR-17-3p
miR-197-3p
135
Heparin cofactor 2SERPIND1miR-183-5p
miR-210-3p
miR-218-5p
miR-1296-5p
135
Fibrinolytic
PlasminogenPLGmiR-148a-3p
miR-148b-3p
miR-181a-5p
miR-181b-5p
miR-483-3p
135
Tissue-type plasminogen activatorPLATmiR-340183
Plasminogen activator inhibitorSERPINE1miR-3°c
 • Biomarker for inflammatory and thrombotic disorders
184,185,186
184
miR-421
 • Biomarker for inflammatory and thrombotic disorders

184
miR-301a
ProcoagulantMain miRNAs (functionally validated)References
Fibrinogen alphaFGAmiR-193b-3p
miR-194-5p
miR-759
135,142
135
142
Fibrinogen betaFGBmiR-409-3p (miR-29 family)143
Fibrinogen gammaFGGmiR-99b-3p
miR-193a-5p
135
Coagulation factor III, tissue factorF3miR-19b
 • Anti-thrombotic protector in patients with unstable angina
144,145,146,147,148,150
144
miR-19b, miR-20a
 • Down-regulation contributes to a hypercoagulable state in SLE and APS
145
miR-126
 • Reduces thrombogenicity in diabetes mellitus
146
miR-145
 • Impedes thrombus formation in venous thrombosis
147
miR-223
 • Partially blocks TNF-α-induced increase of TF activity in endothelial cells
148
miR-365a-3p
 • Interacts with TF 3’-UTR to modulate TF-initiated thrombin generation
149
Coagulation factor VIIF7miR-19a-3p
miR-19b-3p
135
Coagulation factor VIIIF8miR-7-5p
miR 454-3p
miR-532-5p
miR-1246
135,151
135
135
151
Coagulation factor XIF11miR-15b-5p
 • Biomarker for PAD
 • Influences platelet reactivity and clopidogrel response
135
152
153
miR-24-3p
 • Biomarker for acute cerebral infarction, arteriosclerosis obliterans, atherosclerosis and severe trauma
135
154,155,156,157
miR-30a-3p
 • Biomarker for AMI and ischaemic stroke
135
158,159
miR-30d-3p135
miR-96-5p
 • Biomarker for DVT and DIC
135
160,161
miR-103a-3p
 • Involved in atherosclerosis and vascular inflammation by suppression of KLF4
 • Biomarker for VTE
135
162
163
miR-145-5p
 • Biomarker for CAD, AMI, stroke, long-term outcome
 • Impedes thrombus formation in atherosclerosis by targeting tissue factor and influencing platelet reactivity
164
165,166,167,168,169,170,171,172
147,153
miR-148b-3p135
miR-151a-3p135
miR-181a-5p
 • Biomarker for AMI and PAD
164,173
152,174
miR-181b-5p135
miR-191b-5p173
miR-544a175
miR-1255a
 • Biomarker for stroke
135
176
(pre)kallikreinKLKB1miR-24-3p135
Von Willebrand factorVWFmiR-24177,178
ADAM metallopeptidase with thrombospondin type 1 motif 13ADAMTS13miR-525-5p179
Anticoagulant
Tissue factor pathway inhibitorTFPImiR-27a/b
miR-494
miR-27a/b-3p
180,181
AntithrombinSERPINC1miR-19b-3p
miR-186-5p
135
Protein CPROCmiR-494
let-7 family
135
Protein SPROS1miR-494182
Protein ZPROZmiR-30a-5p
miR-128-3p
miR-148a-3p
miR-148b-3p
miR-375
miR-671-3p
135
Protein Z-dependent ;?>protease inhibitorSERPINA10miR-15b-5p
miR-16-5p
miR-17-3p
miR-197-3p
135
Heparin cofactor 2SERPIND1miR-183-5p
miR-210-3p
miR-218-5p
miR-1296-5p
135
Fibrinolytic
PlasminogenPLGmiR-148a-3p
miR-148b-3p
miR-181a-5p
miR-181b-5p
miR-483-3p
135
Tissue-type plasminogen activatorPLATmiR-340183
Plasminogen activator inhibitorSERPINE1miR-3°c
 • Biomarker for inflammatory and thrombotic disorders
184,185,186
184
miR-421
 • Biomarker for inflammatory and thrombotic disorders

184
miR-301a

For full Hemostatic miRNA Targetome Atlas, see reference Nourse et al.135

Table 1

Haemostatic components under miRNA control and relation to thromboinflammation

ProcoagulantMain miRNAs (functionally validated)References
Fibrinogen alphaFGAmiR-193b-3p
miR-194-5p
miR-759
135,142
135
142
Fibrinogen betaFGBmiR-409-3p (miR-29 family)143
Fibrinogen gammaFGGmiR-99b-3p
miR-193a-5p
135
Coagulation factor III, tissue factorF3miR-19b
 • Anti-thrombotic protector in patients with unstable angina
144,145,146,147,148,150
144
miR-19b, miR-20a
 • Down-regulation contributes to a hypercoagulable state in SLE and APS
145
miR-126
 • Reduces thrombogenicity in diabetes mellitus
146
miR-145
 • Impedes thrombus formation in venous thrombosis
147
miR-223
 • Partially blocks TNF-α-induced increase of TF activity in endothelial cells
148
miR-365a-3p
 • Interacts with TF 3’-UTR to modulate TF-initiated thrombin generation
149
Coagulation factor VIIF7miR-19a-3p
miR-19b-3p
135
Coagulation factor VIIIF8miR-7-5p
miR 454-3p
miR-532-5p
miR-1246
135,151
135
135
151
Coagulation factor XIF11miR-15b-5p
 • Biomarker for PAD
 • Influences platelet reactivity and clopidogrel response
135
152
153
miR-24-3p
 • Biomarker for acute cerebral infarction, arteriosclerosis obliterans, atherosclerosis and severe trauma
135
154,155,156,157
miR-30a-3p
 • Biomarker for AMI and ischaemic stroke
135
158,159
miR-30d-3p135
miR-96-5p
 • Biomarker for DVT and DIC
135
160,161
miR-103a-3p
 • Involved in atherosclerosis and vascular inflammation by suppression of KLF4
 • Biomarker for VTE
135
162
163
miR-145-5p
 • Biomarker for CAD, AMI, stroke, long-term outcome
 • Impedes thrombus formation in atherosclerosis by targeting tissue factor and influencing platelet reactivity
164
165,166,167,168,169,170,171,172
147,153
miR-148b-3p135
miR-151a-3p135
miR-181a-5p
 • Biomarker for AMI and PAD
164,173
152,174
miR-181b-5p135
miR-191b-5p173
miR-544a175
miR-1255a
 • Biomarker for stroke
135
176
(pre)kallikreinKLKB1miR-24-3p135
Von Willebrand factorVWFmiR-24177,178
ADAM metallopeptidase with thrombospondin type 1 motif 13ADAMTS13miR-525-5p179
Anticoagulant
Tissue factor pathway inhibitorTFPImiR-27a/b
miR-494
miR-27a/b-3p
180,181
AntithrombinSERPINC1miR-19b-3p
miR-186-5p
135
Protein CPROCmiR-494
let-7 family
135
Protein SPROS1miR-494182
Protein ZPROZmiR-30a-5p
miR-128-3p
miR-148a-3p
miR-148b-3p
miR-375
miR-671-3p
135
Protein Z-dependent ;?>protease inhibitorSERPINA10miR-15b-5p
miR-16-5p
miR-17-3p
miR-197-3p
135
Heparin cofactor 2SERPIND1miR-183-5p
miR-210-3p
miR-218-5p
miR-1296-5p
135
Fibrinolytic
PlasminogenPLGmiR-148a-3p
miR-148b-3p
miR-181a-5p
miR-181b-5p
miR-483-3p
135
Tissue-type plasminogen activatorPLATmiR-340183
Plasminogen activator inhibitorSERPINE1miR-3°c
 • Biomarker for inflammatory and thrombotic disorders
184,185,186
184
miR-421
 • Biomarker for inflammatory and thrombotic disorders

184
miR-301a
ProcoagulantMain miRNAs (functionally validated)References
Fibrinogen alphaFGAmiR-193b-3p
miR-194-5p
miR-759
135,142
135
142
Fibrinogen betaFGBmiR-409-3p (miR-29 family)143
Fibrinogen gammaFGGmiR-99b-3p
miR-193a-5p
135
Coagulation factor III, tissue factorF3miR-19b
 • Anti-thrombotic protector in patients with unstable angina
144,145,146,147,148,150
144
miR-19b, miR-20a
 • Down-regulation contributes to a hypercoagulable state in SLE and APS
145
miR-126
 • Reduces thrombogenicity in diabetes mellitus
146
miR-145
 • Impedes thrombus formation in venous thrombosis
147
miR-223
 • Partially blocks TNF-α-induced increase of TF activity in endothelial cells
148
miR-365a-3p
 • Interacts with TF 3’-UTR to modulate TF-initiated thrombin generation
149
Coagulation factor VIIF7miR-19a-3p
miR-19b-3p
135
Coagulation factor VIIIF8miR-7-5p
miR 454-3p
miR-532-5p
miR-1246
135,151
135
135
151
Coagulation factor XIF11miR-15b-5p
 • Biomarker for PAD
 • Influences platelet reactivity and clopidogrel response
135
152
153
miR-24-3p
 • Biomarker for acute cerebral infarction, arteriosclerosis obliterans, atherosclerosis and severe trauma
135
154,155,156,157
miR-30a-3p
 • Biomarker for AMI and ischaemic stroke
135
158,159
miR-30d-3p135
miR-96-5p
 • Biomarker for DVT and DIC
135
160,161
miR-103a-3p
 • Involved in atherosclerosis and vascular inflammation by suppression of KLF4
 • Biomarker for VTE
135
162
163
miR-145-5p
 • Biomarker for CAD, AMI, stroke, long-term outcome
 • Impedes thrombus formation in atherosclerosis by targeting tissue factor and influencing platelet reactivity
164
165,166,167,168,169,170,171,172
147,153
miR-148b-3p135
miR-151a-3p135
miR-181a-5p
 • Biomarker for AMI and PAD
164,173
152,174
miR-181b-5p135
miR-191b-5p173
miR-544a175
miR-1255a
 • Biomarker for stroke
135
176
(pre)kallikreinKLKB1miR-24-3p135
Von Willebrand factorVWFmiR-24177,178
ADAM metallopeptidase with thrombospondin type 1 motif 13ADAMTS13miR-525-5p179
Anticoagulant
Tissue factor pathway inhibitorTFPImiR-27a/b
miR-494
miR-27a/b-3p
180,181
AntithrombinSERPINC1miR-19b-3p
miR-186-5p
135
Protein CPROCmiR-494
let-7 family
135
Protein SPROS1miR-494182
Protein ZPROZmiR-30a-5p
miR-128-3p
miR-148a-3p
miR-148b-3p
miR-375
miR-671-3p
135
Protein Z-dependent ;?>protease inhibitorSERPINA10miR-15b-5p
miR-16-5p
miR-17-3p
miR-197-3p
135
Heparin cofactor 2SERPIND1miR-183-5p
miR-210-3p
miR-218-5p
miR-1296-5p
135
Fibrinolytic
PlasminogenPLGmiR-148a-3p
miR-148b-3p
miR-181a-5p
miR-181b-5p
miR-483-3p
135
Tissue-type plasminogen activatorPLATmiR-340183
Plasminogen activator inhibitorSERPINE1miR-3°c
 • Biomarker for inflammatory and thrombotic disorders
184,185,186
184
miR-421
 • Biomarker for inflammatory and thrombotic disorders

184
miR-301a

For full Hemostatic miRNA Targetome Atlas, see reference Nourse et al.135

Further evidence implicating miRNAs in the haemostatic system comes from the important roles that miRNAs play in the development of bleeding disorders and thrombosis. Blood miRNA levels are associated with haemostatic perturbations, suggesting their potential use as prognostic or diagnostic tools in VTE191 and beyond.192 These include aberrant coagulation in sepsis,193 venous thromboembolism,141,194–204 trauma-induced coagulopathy,154 atherosclerosis,205–209 coronary artery disease,210–212 ischaemic stroke,213,214 and autoimmune inflammatory conditions such as systemic lupus erythematosus.215–217

Recently, using an unbiased systematic search based on a biophysical miRNA interaction study coupled to high-throughput sequencing, the Atlas of the Hemostatic miRNA Targetome was released.135 This screening identified more than 1500 miRNA/3'-UTR interactions with potential function in the haemostatic system from nearly 4500 miRNA/3'-UTR biophysical interactions.135 A proof-of-concept, rigorous filtering combined with loss-of-function studies (limited to 96 of the 1500 miRNA/3′-UTR interactions with a potential function) identified dozens of miRNAs targeting 27 haemostasis-associated gene 3′-UTRs globally or in a gene-specific manner (Figure 4). This highlights the global importance of miRNAs in controlling the haemostatic system and suggests that many more functional miRNAs will be discovered in this system.

Snapshot on the human haemostatic miRNA targetome (for the full miRNA atlas, see supplementary Table S4 of reference135; https://onlinelibrary.wiley.com/doi/full/10.1111/jth.14290). Heatmap of miRNA/3′UTR interactions (only highly stringent interactions are depicted). Results of miTRAP assays were divided into three functional categories of procoagulant, anticoagulant and fibrinolytic components, and for miRNAs targeting multiple 3′UTRs each category subjected to unsupervised hierarchical clustering as indicated by tree on the left (modified from reference135). For further information of miRNA-mediated regulation of haemostatic components, see Table 1.
Figure 4

Snapshot on the human haemostatic miRNA targetome (for the full miRNA atlas, see supplementary Table S4 of reference135; https://onlinelibrary.wiley.com/doi/full/10.1111/jth.14290). Heatmap of miRNA/3′UTR interactions (only highly stringent interactions are depicted). Results of miTRAP assays were divided into three functional categories of procoagulant, anticoagulant and fibrinolytic components, and for miRNAs targeting multiple 3′UTRs each category subjected to unsupervised hierarchical clustering as indicated by tree on the left (modified from reference135). For further information of miRNA-mediated regulation of haemostatic components, see Table 1.

The unbiased view on miRNAs regulating the haemostatic system also sheds light on hitherto functionally poorly characterized connections between different physiological systems and diseases. These include the link between tumour formation and haemostatic perturbations135 or the intricate relationship between the haemostatic system and inflammatory processes (Table 1). For example, miR-181 family members that target the 3'-UTR of F11 mRNA135 are involved in several aspects of haemostasis, including vascular inflammation198,218–220 and platelet activation.221 Another example is miR-24 which controls the expression of VWF.177 Here, hyperglycaemia-induced repression of miR-24 increases VWF expression and secretion in diabetes mellitus, linking metabolic dysfunction to a miRNA-mediated mechanism of haemostatic deregulation.

On the other hand, polymorphisms affecting miRNA binding sites in haemostatic genes can be associated with disease. For example, deletion of the miR-759 binding site of FGA is associated with susceptibility to chronic thromboembolic pulmonary hypertension,222 and SNPs in the 3′-UTR of the F2, F8, and F11 genes are associated with increased activity levels of these haemostatic components.122,223–226

The importance of miRNAs in haemostasis is further corroborated by their role in platelet biology.137 Here miRNAs modulate the expression of target mRNAs important for haemostatic and thrombotic function.227–231 For example, miRNA levels are altered in platelets from patients with essential thrombocythaemia and this in turn is associated with elevated platelet counts and an increased risk of thromboembolic events.232 Additionally altered miRNA expression is often observed in atherosclerotic plaques233 (and references therein).

In light of the functional importance of miRNAs in the haemostatic system135 and the increasingly recognized role of miRNA therapeutics234 currently conquering the cardiovascular system,235 it is tempting to turn this knowledge into new therapeutics (see targeting section below).139 In support of this, miRNA treatment has been demonstrated to result in therapeutic response in thrombosis and haemostasis. In murine models of venous thrombosis, overexpression of miRNAs contributes to thrombus resolution,236 reduces thrombogenesis,147 enhances endothelial progenitor cell migration and tubulogenic activity,237 angiogenesis and thrombosis recanalization.238 Furthermore, the use of antagomirs (i.e. molecules that silence miRNAs) has been shown to block miR-19b-3p-mediated silencing of SERPINC1 (antithrombin), resulting in increased antithrombin expression and activity in vivo.135 This documents the in-principle druggability of the haemostatic system in a miRNA-directed manner and opens opportunities to target other haemostatic components such as coagulation FXI.139

5. Other means of posttranscriptional regulation of the haemostatic system

5.1 RBPs beyond their function in the biogenesis of mRNAs

In addition to co- and posttranscriptional processing, much of the fate of RNAs from synthesis to decay depends on RNA-binding proteins (RBPs).239 RBPs regulate RNA localization, transport, translation, stabilization and degradation of bound RNA molecules. In fact, much of the rapid adjustment of gene expression in inflammation and the immune system29–31 is executed via modulation of RNA stability and decay. The same is likely to apply to the haemostatic system as well, and the adaptation of prothrombin expression (Figure 3) may be a prototype for analogous occurrences.240 It is interesting to note that even in apparently non-polar cells such as hepatocytes, the major source of most haemostatic components, localization of transcripts and thus protein output critically depends on UTR-RBP interactions.241 This suggests that dynamic changes of 5′ and 3′-UTR structures of mRNAs, due to the use of alternative transcription start sites and alternative splicing/polyadenylation, may have a critical impact on protein output and ultimately function. This is corroborated, for example, by the role of 5′-UTR variants that alter upstream open reading frames in CVDs.242

5.2 RNA modification and networks of competitive RNA–RBP binding

As soon as the nascent RNA molecules emerge from the RNA polymerase during transcription, they are instantly decorated with RBPs. While this ensures that co-transcriptional processing takes place effectively and at the right position, RBP loading also prevents the hybridization of the nascent RNA molecule with the DNA strand. This helps to avoid the formation of reactive RNA:DNA hybrids (so called R-loops),243,244 which can lead to genomic instability.245,246 Most importantly, binding of RBPs and non-coding RNAs to (pre-)mRNAs can occur in a complex, sometimes mutually exclusive manner, thereby determining the posttranscriptional fate of mRNAs selectively87 or in a global manner.247,248 This is supported by the observation that the density of RBP and miRNA binding to the UTRs of coagulation factor mRNAs is very high,139 and that numerous RBP and miRNA binding sites are in close proximity (Figure 5).

FXI 3′-UTR interactome. The graph depicts the density of sites for miRNA and RBPs across the FXI 3'-UTR (based on 125 FXI 3'-UTR/miRNA interactions identified by miTRAP/RNA-seq135 with 41 mapped to the FXI 3'-UTR using miRWalk target site prediction, and 392 FXI 3'-UTR/RBP interactions identified by miTRAP/MS and of which 66 are mapped to the FXI 3'-UTR using RBPDB target site prediction. Site density calculated by number of sites present in 50 nt windows over length of the FXI 3'-UTR; modified from ref. 139 (https://www.sciencedirect.com/science/article/pii/S0163725820302060?via%3Dihub).
Figure 5

FXI 3′-UTR interactome. The graph depicts the density of sites for miRNA and RBPs across the FXI 3'-UTR (based on 125 FXI 3'-UTR/miRNA interactions identified by miTRAP/RNA-seq135 with 41 mapped to the FXI 3'-UTR using miRWalk target site prediction, and 392 FXI 3'-UTR/RBP interactions identified by miTRAP/MS and of which 66 are mapped to the FXI 3'-UTR using RBPDB target site prediction. Site density calculated by number of sites present in 50 nt windows over length of the FXI 3'-UTR; modified from ref. 139 (https://www.sciencedirect.com/science/article/pii/S0163725820302060?via%3Dihub).

Hence, there must be mechanisms that coordinate the binding of such molecules. Although not yet studied in great detail, it is likely that modifications of both RNAs249 and RBPs250 can result in remodelling of the 3′-UTR-RBP architecture and thereby change the fate of RNAs encoding coagulation factors under inflammatory conditions. In support of this notion, posttranslational modifications of RBPs have been shown to change the fate of mRNAs encoding central haemostatic components (Figure 3).102 But also variations in N6-methyladenosine (m6A), the most prevalent RNA modification with a wide biological impact,251,252 have been documented in various RNA transcripts in vascular tissues of septic rats.253 Additionally, there is growing evidence that m6A modification is closely related to the development and progression of CVD, including cardiac hypertrophy, heart failure, ischaemic heart disease and pulmonary hypertension.254,255 It is tempting to explore if therapeutic modulation of the cellular m6A machinery (for example in COVID-19256) might be useful in preserving vascular integrity and function in sepsis and/or CVD. Interestingly, the fat mass and obesity-associated protein, one of the few m6A erasers, have emerged as an important pharmaceutical target in many pathophysiological conditions.251 As many more RNA modifications are currently being discovered,257 this holds great potential for systematically uncovering their importance in human diseases and defining novel therapeutic avenues.

5.3 Long non-coding RNAs and circRNAs

Despite the unexpectedly small number of protein-coding genes identified by the human genome project, RNA sequencing has shown that up to 85% of the human genome is transcribed.258 This led to the identification of a large number of non-coding RNA molecules with regulatory functions.259 In contrast to small non-coding RNAs (such a miRNAs, smal nucleolar RNAs (snoRNAs), or PIWI interacting RNAs (piRNAs)), lncRNAs are around 200 nucleotides or more260 and often undergo alternative splicing, which further expands their repertoire. lncRNAs can bind to DNA, mRNAs, miRNAs, and proteins depending on sequence and secondary structure, thereby modulating gene expression under physiological and pathological conditions.261 Their modes of action include epigenetic, transcriptional and post-transcriptional mechanisms. Accordingly, this new class of ncRNAs is increasingly taking centre stage in the modulation of the cardiovascular system. As an example, lncRNA H19 is involved in the pathogenesis of atherosclerosis.262 The expression of lncRNA H19 is significantly increased in patients with ischaemic stroke compared to healthy controls.263 Genome-wide association studies have identified SNPs in the lncRNA ANRIL associated with CVD, such as coronary atherosclerosis and cardiac infarction,264,265 while variants in lncRNA ZFAS1 are associated with susceptibility to ischaemic stroke.266 Recently, a transcriptome wide association study on VTE also revealed further lncRNA hits (RP11-747H7.3, RP4-737E23.2),267 corroborating their function in CVD.

Unlike miRNAs or proteins, lncRNA function cannot currently be simply inferred from sequence or structure, and the diversity of lncRNAs described to date precludes simple generalizations.261 In the context of the haemostatic system, this hitherto poorly explored area deserves attention. This is also supported by the role lncRNAs have in platelets,268,269 although their role is still under active investigation. In analogy to the central regulatory function of non-coding RNAs in the immune system and because of the resulting therapeutic implications,270 it will be important to better understand the pathophysiological dimension of this class of regulators in thrombosis and its connection to inflammation.

circRNAs are another class of endogenous non-coding regulatory biomolecules. They are prevalent and arise from a non-canonical splicing event called ‘backsplicing’.271 They exert important biological functions by acting as miRNA or protein sponges, by regulating protein function or by being translated.272 As such, circRNAs regulate a plethora of biological functions including ROS formation and cardiovascular metabolic inflammation.273 Accordingly, perturbations of these process(es) can become pathogenic and result in CVD. For example, a haplotype on 9p21 that protects against coronary artery disease has been shown to be associated with the abundance of circRNA ANRIL, which in turn regulates ribosomal RNA maturation, conferring atheroprotection.274 Accordingly, circANRIL has been proposed as a potential therapeutic target for the treatment of atherosclerosis. The in-principle therapeutic utility of circRNA is also supported by recent preclinical observations demonstrating their use, for example, to attenuate cell apoptosis in cerebral ischaemia-reperfusion.275 Finally, circulating circRNA may have diagnostic potential and serve as biomarkers for acute ischaemic stroke276 and even help distinguish different etiologies (i.e. atherothrombotic, cardiothrombotic vs. undetermined stroke).277

5.4 What comes next? Alternative polyadenylation and 3′-UTR diversity as central regulatory hubs

Much of the posttranscriptional regulation of the haemostatic system depends on players that determine the fate of RNAs encoding the respective haemostatic components. The different layers of regulation are largely inter-dependent, as alternative splicing and polyadenylation are coupled to each other87 and thereby determine not only the final open reading frame, but also the 3′-UTR sequence and hence the susceptibility of the mature mRNA to posttranscriptional control by RBPs and ncRNAs.

Since much of the posttranscriptional regulation of gene expression takes place at the level of the 3′-UTR, to which RBPs and ncRNAs are abundantly recruited, the 3′-UTR architecture has an important regulatory function (Figure 6).87 Diversification of the transcriptome at the 3′-end by alternative polyadenylation (APA) has recently emerged as a pervasive and evolutionarily conserved layer of gene expression control278 (Figure 1), which affects more than 70% of all genes. APA considerably expands the diversity of the transcriptome 3′-end, affecting protein output, isoform composition and protein localization.279

Alternative polyadenylation (APA) is a pervasive gene regulatory mechanism that results in mRNA isoforms with different 3′-ends. This can result in mRNA isoforms encoding truncated proteins or in mRNA isoforms with distinct 3′-UTR properties altering RNA transport, localization, translation, and/or stability (through binding to non-coding RNAs (such as miRNAs, lncRNAs, ceRNA), through binding to RBPs and/or through complex, sometimes mutually exclusive, interactions of RNA motifs with RBPs and/or ncRNAs. Of note, modifications of RNAs (such as ‘m6A’) or post-translational modifications (PTMs) of RBPs introduce further layers of modulation). Inflammatory conditions tend to result in the generation of shorter mRNA isoforms (either lacking elements of 3′-UTR regulation or resulting in truncated proteins,118,121). APA affects numerous genes involved in blood coagulation and inflammation (Table 2).
Figure 6

Alternative polyadenylation (APA) is a pervasive gene regulatory mechanism that results in mRNA isoforms with different 3′-ends. This can result in mRNA isoforms encoding truncated proteins or in mRNA isoforms with distinct 3′-UTR properties altering RNA transport, localization, translation, and/or stability (through binding to non-coding RNAs (such as miRNAs, lncRNAs, ceRNA), through binding to RBPs and/or through complex, sometimes mutually exclusive, interactions of RNA motifs with RBPs and/or ncRNAs. Of note, modifications of RNAs (such as ‘m6A’) or post-translational modifications (PTMs) of RBPs introduce further layers of modulation). Inflammatory conditions tend to result in the generation of shorter mRNA isoforms (either lacking elements of 3′-UTR regulation or resulting in truncated proteins,118,121). APA affects numerous genes involved in blood coagulation and inflammation (Table 2).

APA is globally regulated in various conditions,121 including developmental and adaptive programmes.92 It is thus likely that APA also tunes the haemostatic system, as exemplified by alternative processing of TF and TFPI, where alternative splicing also generates different 3′-UTRs (Figure 2). In addition, a recent large scale RNAi screen based on the depletion of more than 170 putative APA regulators revealed how individual regulators affect the APA landscape,117 including the resulting impact on gene ontologies.121 Several significantly enriched GO terms suggest a critical function of UTR structures in inflammatory processes and innate and adaptive immunity.121 APA affects key components broadly involved in inflammation and blood coagulation (Table 2). This is consistent with findings that APA is a critical component in the control of inflammatory processes118,280,281 (including COVID-19282), that typically result in shorter mRNA isoforms (Figure 6).

Table 2

Alternative polyadenylation regulates components involved in blood coagulation and inflammation

Regulated by CPSF6-dependent APARegulated by NUDT21-dependent APARegulated by PCF11-dependent APA
Affected GO termBlood coagulationRegulation of inflammationComplementBlood coagulationRegulation of inflammationComplementBlood coagulationRegulation of inflammationComplement
affected genesARRB1DDX3XC7ARRB1ATMC7ACTG1ABHD12HSP90AB1
CBX5DROSHACD59CAPZBCD47ARRB1DROSHARAB27A
CD59LDLRCBX5HSPD1GNA12GPS2
GATA2LYNGATA2ISL1GNB1NDFIP1
GNA11MACIRGATA4LYNGNG2NEAT1
GNA12NDFIP1GGCXMACIRH3-3BNT5E
GNA13PBKGNA11MCPH1IRF2PRCP
GNB1PDCD4GNA12NDFIP1PRCPSTMP1
GNG2PRCPGNA13PDCD4PRKAR1AVPS35
H3-3BSETD6GNB1SETD6PRKAR2B
LMAN1SMAD3GNG2SMAD3RAB27A
LYNSTMP1H3-3BSOD1VAV2
MAPK1SYT11HPS5SYT11VPS45
PRCPVPS35LMAN1TREX1
PRKAR1ALYNVPS35
PRKAR2BPHF21A
RAB27APRCP
RAC1PRKAR1A
RAD51CRAB27A
STXBP1RAC1
YWHAZSTXBP1
Regulated by CPSF6-dependent APARegulated by NUDT21-dependent APARegulated by PCF11-dependent APA
Affected GO termBlood coagulationRegulation of inflammationComplementBlood coagulationRegulation of inflammationComplementBlood coagulationRegulation of inflammationComplement
affected genesARRB1DDX3XC7ARRB1ATMC7ACTG1ABHD12HSP90AB1
CBX5DROSHACD59CAPZBCD47ARRB1DROSHARAB27A
CD59LDLRCBX5HSPD1GNA12GPS2
GATA2LYNGATA2ISL1GNB1NDFIP1
GNA11MACIRGATA4LYNGNG2NEAT1
GNA12NDFIP1GGCXMACIRH3-3BNT5E
GNA13PBKGNA11MCPH1IRF2PRCP
GNB1PDCD4GNA12NDFIP1PRCPSTMP1
GNG2PRCPGNA13PDCD4PRKAR1AVPS35
H3-3BSETD6GNB1SETD6PRKAR2B
LMAN1SMAD3GNG2SMAD3RAB27A
LYNSTMP1H3-3BSOD1VAV2
MAPK1SYT11HPS5SYT11VPS45
PRCPVPS35LMAN1TREX1
PRKAR1ALYNVPS35
PRKAR2BPHF21A
RAB27APRCP
RAC1PRKAR1A
RAD51CRAB27A
STXBP1RAC1
YWHAZSTXBP1

Each column depicts genes belonging to the GO term “blood coagulation”, “regulation of inflammation” and “complement” that are affected by alternative polyadenylation (APA) upon depletion of central APA regulators (CPSF6, NUDT21, PCF11). Data obtained from TREND-DB121; for further APA affected genes and -effectors see: http://shiny.imbei.uni-mainz.de:3838/trend-db/

Platelet degranulation

Thrombin/G-Protein coupled receptor signaling

Complement regulation

Positive regulation of secretion by cell

Regulation of inflammatory response/cytokine production

Angiotensin conversion

Table 2

Alternative polyadenylation regulates components involved in blood coagulation and inflammation

Regulated by CPSF6-dependent APARegulated by NUDT21-dependent APARegulated by PCF11-dependent APA
Affected GO termBlood coagulationRegulation of inflammationComplementBlood coagulationRegulation of inflammationComplementBlood coagulationRegulation of inflammationComplement
affected genesARRB1DDX3XC7ARRB1ATMC7ACTG1ABHD12HSP90AB1
CBX5DROSHACD59CAPZBCD47ARRB1DROSHARAB27A
CD59LDLRCBX5HSPD1GNA12GPS2
GATA2LYNGATA2ISL1GNB1NDFIP1
GNA11MACIRGATA4LYNGNG2NEAT1
GNA12NDFIP1GGCXMACIRH3-3BNT5E
GNA13PBKGNA11MCPH1IRF2PRCP
GNB1PDCD4GNA12NDFIP1PRCPSTMP1
GNG2PRCPGNA13PDCD4PRKAR1AVPS35
H3-3BSETD6GNB1SETD6PRKAR2B
LMAN1SMAD3GNG2SMAD3RAB27A
LYNSTMP1H3-3BSOD1VAV2
MAPK1SYT11HPS5SYT11VPS45
PRCPVPS35LMAN1TREX1
PRKAR1ALYNVPS35
PRKAR2BPHF21A
RAB27APRCP
RAC1PRKAR1A
RAD51CRAB27A
STXBP1RAC1
YWHAZSTXBP1
Regulated by CPSF6-dependent APARegulated by NUDT21-dependent APARegulated by PCF11-dependent APA
Affected GO termBlood coagulationRegulation of inflammationComplementBlood coagulationRegulation of inflammationComplementBlood coagulationRegulation of inflammationComplement
affected genesARRB1DDX3XC7ARRB1ATMC7ACTG1ABHD12HSP90AB1
CBX5DROSHACD59CAPZBCD47ARRB1DROSHARAB27A
CD59LDLRCBX5HSPD1GNA12GPS2
GATA2LYNGATA2ISL1GNB1NDFIP1
GNA11MACIRGATA4LYNGNG2NEAT1
GNA12NDFIP1GGCXMACIRH3-3BNT5E
GNA13PBKGNA11MCPH1IRF2PRCP
GNB1PDCD4GNA12NDFIP1PRCPSTMP1
GNG2PRCPGNA13PDCD4PRKAR1AVPS35
H3-3BSETD6GNB1SETD6PRKAR2B
LMAN1SMAD3GNG2SMAD3RAB27A
LYNSTMP1H3-3BSOD1VAV2
MAPK1SYT11HPS5SYT11VPS45
PRCPVPS35LMAN1TREX1
PRKAR1ALYNVPS35
PRKAR2BPHF21A
RAB27APRCP
RAC1PRKAR1A
RAD51CRAB27A
STXBP1RAC1
YWHAZSTXBP1

Each column depicts genes belonging to the GO term “blood coagulation”, “regulation of inflammation” and “complement” that are affected by alternative polyadenylation (APA) upon depletion of central APA regulators (CPSF6, NUDT21, PCF11). Data obtained from TREND-DB121; for further APA affected genes and -effectors see: http://shiny.imbei.uni-mainz.de:3838/trend-db/

Platelet degranulation

Thrombin/G-Protein coupled receptor signaling

Complement regulation

Positive regulation of secretion by cell

Regulation of inflammatory response/cytokine production

Angiotensin conversion

Strikingly, several haemostatic components have alternative transcripts that differ not only in their exon composition but also in their 3′-UTR structure (see NCBI Ref seq). These include essential components of the protein C pathway (i.e. protein C and protein S) with established functions at the interface of coagulation and inflammation.283 For the protein C cofactor protein S, 3'-UTR dynamics are already documented,121which appear to be regulated by specific RBPs (RNPS1) or other components (CDKN2D). This points to a regulatory function of APA at the interface of the haemostatic and the immune system. Due to the pervasive regulatory function of APA in various processes121 (with perturbations leading to numerous diseases92) it is plausible that much of this diversity in the haemostatic system is regulated in response to inflammatory signals. This is illustrated by inflammation-triggered alternative processing of the FGG mRNA,284 resulting in gamma prime (γ’) fibrinogen.77 γ’ fibrinogen is the fibrinogen fraction that contains the γ’ chain, which arises when the FGG mRNA is polyadenylated at an APA signal, resulting in a polypeptide with a unique 20-amino acid extension encoded by intron 9.77 Thanks to the strongly negatively charged C-terminus of the γ’ chain, fibrinogen γ’ can bind with high affinity to thrombin exosite II, decreasing thrombin activity on several substrates (antithrombin I activity).285 As a consequence, low γ’ fibrinogen levels have been associated with an increased risk of venous thrombosis,77,286 while a potential role in CVD287 and ischaemic stroke288 is under debate.289 This highlights how seemingly subtle changes through alterations of APA and 3′-UTR diversity can have most significant functional effects in the haemostatic system. It also serves as an example illustrating the complex interdependency of posttranscriptional processing of RNA molecules and hence functional output.

Interrogating system-wide posttranscriptional gene regulation37,38 and transcriptome 3′-end diversity,120,121 combined with unbiased RNA interactome studies119,135 and strategies to disentangle the functional significance of genomic perturbations in non-coding elements,290 therefore holds great potential to unravel novel layers of coupling of the haemostatic system with inflammatory processes.121 This could also open entirely new therapeutic perspectives92 to combat medical threats centreing around thromboinflammation such as sepsis, which is still the leading cause of death in the Western world and in critically ill patients worldwide.1

6. Targeting post-transcriptional regulation of the haemostatic system

The multiple layers of posttranscriptional control of gene expression offer various opportunities and targets for therapeutic intervention. For example, RNA-based therapeutics can be used not only to re-direct splicing83 and polyadenylation291 but also to silence an mRNA or to prevent its interaction with other RNAs or RBPs.292,293

Compared to ‘conventional’ small therapeutic molecules, RNA-based therapeutics such as ASOs, siRNAs, and miRNAs offer the advantage of being able to act on ‘non-druggable’ targets (i.e. proteins that lack enzymatic function or whose conformation is inaccessible to traditional drug molecules), as they can be designed to affect virtually any gene of interest.294

ASOs are relatively short, chemically modified single-stranded nucleic acids that selectively pair to specific regions of mRNA resulting in endonucleolytic cleavage and degradation.292 Currently, more than 60 ASO therapies are in or have completed phase I/II trials, with a substantial number of antithrombotic ASO therapeutics currently under development.139

The recent introduction of ASOs down-regulating FXI expression exemplifies the potential of such therapeutics to modulate the haemostatic system via post-transcriptional mechanisms.33 This phase II study in patients undergoing knee surgery revealed that the FXI-targeting ASO effectively protects patients against venous thrombosis with a relatively limited risk of bleeding. However, this proof-of-concept trial was too small to assess the effect on other thrombotic end points. Other genes that are being explored as potential targets for antithrombotic therapy using silencing ASOs are FII, FVII, FXII, prekallikrein, plasmin activator inhibitor, thrombopoetin, and FMO3.139 A possible concern is that changes in platelet counts were observed in non-human primates treated with ASOs,295 which has been attributed to peripheral clearance296 and could potentially impact haemostasis.

miRNA therapeutics represent another highly versatile therapeutic means in the context of the haemostatic system.139 miRNA mimics may be employed to silence pro-coagulant genes to treat thrombosis (or alternatively, anticoagulant genes to treat bleeding). Conversely, antagomirs or target site blockers can be used to relieve silencing of anticoagulant genes to treat thrombosis. Moreover, some miRNAs target several haemostatic components at the same time (Figure 4), and silencing of such miRNAs can be intentionally used to control several haemostatic components. On the other hand, undesired pleiotropy is one of the conceptual downsides of therapeutic miRNA targeting.

miRNA therapeutics are currently at an early stage of development and not yet applicable in the clinical setting.297 In preclinical studies, several miRNA mimics and antagomirs have been shown to reduce thrombus formation139 or increase the antithrombin activity in vivo.135 One of the biggest challenges in the clinical development of miRNA-based therapeutics is the identification of key miRNA candidates and targets, their specificity and effect size. There is currently a relatively small number of experimentally validated miRNA:mRNA interactions, making knowledge of the miRNA targetome in the haemostatic system a major trove for future targeted therapeutics.135

ASOs and most siRNAs exhibit perfect complementary to their targets, which usually results in degradation of the target mRNA.298 In contrast, partial base-pairing of miRNAs prevents the cleavage activity of RISC, predominately causing translational repression, and only in some cases deadenylation, decapping, and finally mRNA degradation.299 Although the proportion of mRNA target degradation varies widely,300 a number of targets are almost exclusively repressed at the level of translation.301 How much each mechanism contributes to down-regulation depends on characteristics, such as seed-flanking nucleotides, of the individual miRNA–mRNA pair.302

In the context of the haemostatic system, it is interesting to note that miRNA regulation of transcripts encoding secretory proteins results almost exclusively in translational repression, because miRNA translational repression is stronger for mRNAs translated at the endoplasmic-reticulum compared to free cytosolic ribosomes.301 Thus, miRNA-mediated therapeutic targeting without degradation of the target mRNAs preserves physiological cell intrinsic regulatory mechanisms carried out by 3′-UTRs and their binding partners (such as RBPs, miRNAs, lncRNAs, circRNA, or miRNA sponges). This allows for ‘compensatory’ on-demand adjustments of protein output even in the presence of the miRNA therapeutic and thus may represent a conceptual advantage of miRNA therapeutics over ASO-based approaches.139

While RNA therapeutic approaches have been used in the development of new drugs and clinical trials are underway,303 there are still concerns and challenges to be overcome. These include, but are not limited to, off-target effects,304 triggering innate immune responses,305 stability of the therapeutic RNA molecule, and design of optimal delivery systems for disease-specific release with minimal toxicity.234

Finally, there are increasingly strategies to modulate other facets of the RNA biogenesis. This concerns the targeted interference with splicing83 or with cleavage and polyadenylation,291 involving either redirection of aberrant RNA processing (through ASOs, U1snRNP interference or trans-splicing) or the elimination of aberrant transcripts.82,92 The characterization of transcriptome dynamics and elucidation of the RNA interactome thus become the next milestones to exploit the untapped therapeutic opportunities arising from the increasingly available RNA therapeutics.

7. Summary

Besides transcriptional control, post-transcriptional regulation of gene expression is taking centre stage in the modulation of the haemostatic system. The highly regulated use of alternative transcription start sites, exons, and polyadenylation sites makes the transcriptome highly dynamic in time, space, and in response to pathological processes. Additional post-transcriptional regulation by non-coding RNAs, RNA-binding proteins and RNA modification mechanisms further modulate the functional output of numerous biological processes, including the haemostatic system. Many of these regulatory principles also play an important functional role in tuning the immune system,27–31 suggesting conserved regulatory links between both systems. It will be critical to characterize these links to identify rational targets for the emerging repertoire of RNA therapeutics to effectively combat the dangerous alliance of the haemostatic and the immune system.

Acknowledgements

The authors would like to express their gratitude to current and former members of the Danckwardt and the Castoldi lab. Work in the Danckwardt lab is kindly supported by the German Research Foundation Priority Programme SPP 1935, German Research Foundation grants DA 1189/2-1 and DA 1189/5-1, Germany Research Foundation Major Research Instrumentation Programme INST 371/33-1, the German Research Foundation graduate school GRK 1591, Federal Ministry of Education and Research (BMBF01EO1003), German Society of Clinical and Laboratory Medicine, and the Hella Bühler Award for Cancer Research. Work in the labs of S.D. and E.C. is supported by the EU Horizon 2020 Innovative training network ‘Thromboinflammation in Cardiovascular disorders’ (TICARDIO, Marie Skłodowska-Curie grant agreement No 813409). D.-A. T. is supported by the EPIDEMIOM-VT Senior Chair from the University of Bordeaux initiative of excellence IdEX.

References

1

Angus
DC
,
van der Poll
T
.
Severe sepsis and septic shock
.
N Engl J Med
2013
;
369
:
840
851
.

2

Hotchkiss
RS
,
Karl
IE
.
The pathophysiology and treatment of sepsis
.
N Engl J Med
2003
;
348
:
138
150
.

3

Rittirsch
D
,
Flierl
MA
,
Ward
PA
.
Harmful molecular mechanisms in sepsis
.
Nat Rev Immunol
2008
;
8
:
776
787
.

4

Lozano
R
,
Naghavi
M
,
Foreman
K
,
Lim
S
,
Shibuya
K
,
Aboyans
V
,
Abraham
J
,
Adair
T
,
Aggarwal
R
,
Ahn
SY
,
AlMazroa
MA
,
Alvarado
M
,
Anderson
HR
,
Anderson
LM
,
Andrews
KG
,
Atkinson
C
,
Baddour
LM
,
Barker-Collo
S
,
Bartels
DH
,
Bell
ML
,
Benjamin
EJ
,
Bennett
D
,
Bhalla
K
,
Bikbov
B
,
Abdulhak
AB
,
Birbeck
G
,
Blyth
F
,
Bolliger
I
,
Boufous
S
,
Bucello
C
,
Burch
M
,
Burney
P
,
Carapetis
J
,
Chen
H
,
Chou
D
,
Chugh
SS
,
Coffeng
LE
,
Colan
SD
,
Colquhoun
S
,
Colson
KE
,
Condon
J
,
Connor
MD
,
Cooper
LT
,
Corriere
M
,
Cortinovis
M
, de
Vaccaro
KC
,
Couser
W
,
Cowie
BC
,
Criqui
MH
,
Cross
M
,
Dabhadkar
KC
,
Dahodwala
N
, De
Leo
D
,
Degenhardt
L
,
Delossantos
A
,
Denenberg
J
, Des
Jarlais
DC
,
Dharmaratne
SD
,
Dorsey
ER
,
Driscoll
T
,
Duber
H
,
Ebel
B
,
Erwin
PJ
,
Espindola
P
,
Ezzati
M
,
Feigin
V
,
Flaxman
AD
,
Forouzanfar
MH
,
Fowkes
FGR
,
Franklin
R
,
Fransen
M
,
Freeman
MK
,
Gabriel
SE
,
Gakidou
E
,
Gaspari
F
,
Gillum
RF
,
Gonzalez-Medina
D
,
Halasa
YA
,
Haring
D
,
Harrison
JE
,
Havmoeller
R
,
Hay
RJ
,
Hoen
B
,
Hotez
PJ
,
Hoy
D
,
Jacobsen
KH
,
James
SL
,
Jasrasaria
R
,
Jayaraman
S
,
Johns
N
,
Karthikeyan
G
,
Kassebaum
N
,
Keren
A
, Khoo J-P,
Knowlton
LM
,
Kobusingye
O
,
Koranteng
A
,
Krishnamurthi
R
,
Lipnick
M
,
Lipshultz
SE
,
Ohno
SL
,
Mabweijano
J
,
MacIntyre
MF
,
Mallinger
L
,
March
L
,
Marks
GB
,
Marks
R
,
Matsumori
A
,
Matzopoulos
R
,
Mayosi
BM
,
McAnulty
JH
,
McDermott
MM
,
McGrath
J
,
Memish
ZA
,
Mensah
GA
,
Merriman
TR
,
Michaud
C
,
Miller
M
,
Miller
TR
,
Mock
C
,
Mocumbi
AO
,
Mokdad
AA
,
Moran
A
,
Mulholland
K
,
Nair
MN
,
Naldi
L
,
Narayan
KMV
,
Nasseri
K
,
Norman
P
, O'
Donnell
M
,
Omer
SB
,
Ortblad
K
,
Osborne
R
,
Ozgediz
D
,
Pahari
B
,
Pandian
JD
,
Rivero
AP
,
Padilla
RP
,
Perez-Ruiz
F
,
Perico
N
,
Phillips
D
,
Pierce
K
,
Pope
CA
,
Porrini
E
,
Pourmalek
F
,
Raju
M
,
Ranganathan
D
,
Rehm
JT
,
Rein
DB
,
Remuzzi
G
,
Rivara
FP
,
Roberts
T
, De Leó
n
FR
,
Rosenfeld
LC
,
Rushton
L
,
Sacco
RL
,
Salomon
JA
,
Sampson
U
,
Sanman
E
,
Schwebel
DC
,
Segui-Gomez
M
,
Shepard
DS
,
Singh
D
,
Singleton
J
,
Sliwa
K
,
Smith
E
,
Steer
A
,
Taylor
JA
,
Thomas
B
,
Tleyjeh
IM
,
Towbin
JA
,
Truelsen
T
,
Undurraga
EA
,
Venketasubramanian
N
,
Vijayakumar
L
,
Vos
T
,
Wagner
GR
,
Wang
M
,
Wang
W
,
Watt
K
,
Weinstock
MA
,
Weintraub
R
,
Wilkinson
JD
,
Woolf
AD
,
Wulf
S
, Yeh P-H,
Yip
P
,
Zabetian
A
, Zheng Z-J,
Lopez
AD
,
Murray
CJL.
Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the global burden of disease study 2010
.
Lancet
2012
;
380
:
2095
2128
.

5

Berry
JD
,
Dyer
A
,
Cai
X
,
Garside
DB
,
Ning
H
,
Thomas
A
,
Greenland
P
, Van
Horn
L
,
Tracy
RP
,
Lloyd-Jones
DM.
Lifetime risks of cardiovascular disease
.
N Engl J Med
2012
;
366
:
321
329
.

6

Nabel
EG
.
Cardiovascular disease
.
N Engl J Med
2003
;
349
:
60
72
.

7

Levi
M
,
van der Poll
T
,
Buller
HR
.
Bidirectional relation between inflammation and coagulation
.
Circulation
2004
;
109
:
2698
2704
.

8

Borissoff
JI
,
Spronk
HMH
,
ten Cate
H
.
The hemostatic system as a modulator of atherosclerosis
.
N Engl J Med
2011
;
364
:
1746
1760
.

9

Antoniak
S
.
The coagulation system in host defense
.
Res Pract Thromb Haemost
2018
;
2
:
549
557
.

10

Engelmann
B
,
Massberg
S
.
Thrombosis as an intravascular effector of innate immunity
.
Nat Rev Immunol
2013
;
13
:
34
45
.

11

Libby
P
.
Inflammation in atherosclerosis
.
Nature
2002
;
420
:
868
874
.

12

Jose
RJ
,
Manuel
A
.
COVID-19 cytokine storm: the interplay between inflammation and coagulation
.
Lancet Resp Med
2020
;
8
:
e46
e47
.

13

Gruys
E
,
Toussaint
MJ
,
Niewold
TA
,
Koopmans
SJ
.
Acute phase reaction and acute phase proteins
.
J Zhej Univ Sci B
2005
;
6
:
1045
1056
.

14

Fish
RJ
,
Neerman-Arbez
M
.
Fibrinogen gene regulation
.
Thromb Haemost
2012
;
108
:
419
426
.

15

Pottinger
BE
,
Read
RC
,
Paleolog
EM
,
Higgins
PG
,
Pearson
JD
.
Von Willebrand factor is an acute phase reactant in man
.
Thromb Res
1989
;
53
:
387
394
.

16

Chen
J
,
Chung
DW
.
Inflammation, von Willebrand factor, and adamts13
.
Blood
2018
;
132
:
141
147
.

17

Rennie
JA
,
Ogston
D
.
Changes in coagulation factors following acute myocardial infarction in man
.
Haemost
1976
;
5
:
258
264
.

18

Vaziri
ND
,
Branson
HE
,
Ness
R
.
Changes of coagulation factors IX, VIII, VII, X and V in nephrotic syndrome
.
Am J Med Sci
1980
;
280
:
167
171
.

19

Wyshock
EG
,
Suffredini
AF
,
Parrillo
JE
,
Colman
RW
.
Cofactors V and VIII after endotoxin administration to human volunteers
.
Thromb Res
1995
;
80
:
377
389
.

20

Stirling
D
,
Hannant
WA
,
Ludlam
CA
.
Transcriptional activation of the factor VIII gene in liver cell lines by interleukin-6
.
Thromb Haemost
1998
;
79
:
74
78
.

21

Begbie
M
,
Notley
C
,
Tinlin
S
,
Sawyer
L
,
Lillicrap
D
.
The factor VIII acute phase response requires the participation of NFkappab and c/ebp
.
Thromb Haemost
2000
;
84
:
216
222
.

22

Bancroft
JD
,
McDowell
SA
,
Degen
SJ
.
The human prothrombin gene: transcriptional regulation in HEPG2 cells
.
Biochem
1992
;
31
:
12469
12476
.

23

Parry
GC
,
Mackman
N
.
Transcriptional regulation of tissue factor expression in human endothelial cells
.
Arterioscler Thromb Vasc Biol
1995
;
15
:
612
621
.

24

Zhang
K
,
Kurachi
S
,
Kurachi
K
.
Genetic mechanisms of age regulation of protein c and blood coagulation
.
J Biol Chem
2002
;
277
:
4532
4540
.

25

Hung
H-L
,
Pollak
ES
,
Kudaravalli
RD
,
Arruda
V
,
Chu
K
,
High
KA
.
Regulation of human coagulation factor X gene expression by GATA-4 and the SP family of transcription factors
.
Blood
2001
;
97
:
946
951
.

26

Peghaire
C
,
Dufton
NP
,
Lang
M
,
Salles-Crawley
II
,
Ahnström
J
,
Kalna
V
,
Raimondi
C
,
Pericleous
C
,
Inuabasi
L
,
Kiseleva
R
,
Muzykantov
VR
,
Mason
JC
,
Birdsey
GM
,
Randi
AM.
The transcription factor ERG regulates a low shear stress-induced anti-thrombotic pathway in the microvasculature
.
Nat Commun
2019
;
10
:
5014
.

27

Chen
YG
,
Satpathy
AT
,
Chang
HY
.
Gene regulation in the immune system by long noncoding RNAs
.
Nat Immunol
2017
;
18
:
962
972
.

28

Shulman
Z
,
Stern-Ginossar
N
.
The RNA modification n6-methyladenosine as a novel regulator of the immune system
.
Nat Immunol
2020
;
21
:
501
512
.

29

Chen
C-YA
,
Shyu
A-B
.
AU-rich elements: characterization and importance in mRNA degradation
.
Trends Biochem Sci
1995
;
20
:
465
470
.

30

Anderson
P
.
Post-transcriptional control of cytokine production
.
Nat Immunol
2008
;
9
:
353
359
.

31

Carpenter
S
,
Ricci
EP
,
Mercier
BC
,
Moore
MJ
,
Fitzgerald
KA
.
Post-transcriptional regulation of gene expression in innate immunity
.
Nat Rev Immunol
2014
;
14
:
361
376
.

32

Neil
CR
,
Seiler
MW
,
Reynolds
DJ
,
Smith
JJ
,
Vaillancourt
FH
,
Smith
PG
,
Agrawal
AA.
Reprogramming RNA processing: an emerging therapeutic landscape
.
Trends Pharmacol Sci
2022
;
43
(
5
):
437
454
.

33

Büller
HR
,
Bethune
C
,
Bhanot
S
,
Gailani
D
,
Monia
BP
,
Raskob
GE
,
Segers
A
,
Verhamme
P
,
Weitz
JI.
Factor XI antisense oligonucleotide for prevention of venous thrombosis
.
N Engl J Med
2015
;
372
:
232
240
.

34

International Human Genome Sequencing Consortium
.
Initial sequencing and analysis of the human genome
.
Nature
2001
;
409
:
860
921
.

35

De Paoli-Iseppi
R
,
Gleeson
J
,
Clark
MB
.
Isoform age—splice isoform profiling using long-read technologies
.
Front Mol Biosci
2021
;
8
:
711733
.

36

Glinos
DA
,
Garborcauskas
G
,
Hoffman
P
,
Ehsan
N
,
Jiang
L
,
Gokden
A
,
Dai
X
,
Aguet
F
,
Brown
KL
,
Garimella
K
,
Bowers
T
,
Costello
M
,
Ardlie
K
,
Jian
R
,
Tucker
NR
,
Ellinor
PT
,
Harrington
ED
,
Tang
H
,
Snyder
M
,
Juul
S
,
Mohammadi
P
,
MacArthur
DG
,
Lappalainen
T
,
Cummings
BB.
Transcriptome variation in human tissues revealed by long-read sequencing
.
Nature
2022
;
608
:
353
359
.

37

Carninci
P
,
Kasukawa
T
,
Katayama
S
,
Gough
J
,
Frith
MC
,
Maeda
N
,
Oyama
R
,
Ravasi
T
,
Lenhard
B
,
Wells
C
,
Kodzius
R
,
Shimokawa
K
,
Bajic
VB
,
Brenner
SE
,
Batalov
S
,
Forrest
AR
,
Zavolan
M
,
Davis
MJ
,
Wilming
LG
,
Aidinis
V
,
Allen
JE
,
Ambesi-Impiombato
A
,
Apweiler
R
,
Aturaliya
RN
,
Bailey
TL
,
Bansal
M
,
Baxter
L
,
Beisel
KW
,
Bersano
T
,
Bono
H
,
Chalk
AM
,
Chiu
KP
,
Choudhary
V
,
Christoffels
A
,
Clutterbuck
DR
,
Crowe
ML
,
Dalla
E
,
Dalrymple
BP
, de
Bono
B
, Della
Gatta
G
, di
Bernardo
D
,
Down
T
,
Engstrom
P
,
Fagiolini
M
,
Faulkner
G
,
Fletcher
CF
,
Fukushima
T
,
Furuno
M
,
Futaki
S
,
Gariboldi
M
,
Georgii-Hemming
P
,
Gingeras
TR
,
Gojobori
T
,
Green
RE
,
Gustincich
S
,
Harbers
M
,
Hayashi
Y
,
Hensch
TK
,
Hirokawa
N
,
Hill
D
,
Huminiecki
L
,
Iacono
M
,
Ikeo
K
,
Iwama
A
,
Ishikawa
T
,
Jakt
M
,
Kanapin
A
,
Katoh
M
,
Kawasawa
Y
,
Kelso
J
,
Kitamura
H
,
Kitano
H
,
Kollias
G
,
Krishnan
SP
,
Kruger
A
,
Kummerfeld
SK
,
Kurochkin
IV
,
Lareau
LF
,
Lazarevic
D
,
Lipovich
L
,
Liu
J
,
Liuni
S
,
McWilliam
S
, Madan
Babu
M
,
Madera
M
,
Marchionni
L
,
Matsuda
H
,
Matsuzawa
S
,
Miki
H
,
Mignone
F
,
Miyake
S
,
Morris
K
,
Mottagui-Tabar
S
,
Mulder
N
,
Nakano
N
,
Nakauchi
H
,
Ng
P
,
Nilsson
R
,
Nishiguchi
S
,
Nishikawa
S
,
Nori
F
,
Ohara
O
,
Okazaki
Y
,
Orlando
V
,
Pang
KC
,
Pavan
WJ
,
Pavesi
G
,
Pesole
G
,
Petrovsky
N
,
Piazza
S
,
Reed
J
,
Reid
JF
,
Ring
BZ
,
Ringwald
M
,
Rost
B
,
Ruan
Y
,
Salzberg
SL
,
Sandelin
A
,
Schneider
C
,
Schonbach
C
,
Sekiguchi
K
,
Semple
CA
,
Seno
S
,
Sessa
L
,
Sheng
Y
,
Shibata
Y
,
Shimada
H
,
Shimada
K
,
Silva
D
,
Sinclair
B
,
Sperling
S
,
Stupka
E
,
Sugiura
K
,
Sultana
R
,
Takenaka
Y
,
Taki
K
,
Tammoja
K
,
Tan
SL
,
Tang
S
,
Taylor
MS
,
Tegner
J
,
Teichmann
SA
,
Ueda
HR
, van
Nimwegen
E
,
Verardo
R
,
Wei
CL
,
Yagi
K
,
Yamanishi
H
,
Zabarovsky
E
,
Zhu
S
,
Zimmer
A
,
Hide
W
,
Bult
C
,
Grimmond
SM
,
Teasdale
RD
,
Liu
ET
,
Brusic
V
,
Quackenbush
J
,
Wahlestedt
C
,
Mattick
JS
,
Hume
DA
,
Kai
C
,
Sasaki
D
,
Tomaru
Y
,
Fukuda
S
,
Kanamori-Katayama
M
,
Suzuki
M
,
Aoki
J
,
Arakawa
T
,
Iida
J
,
Imamura
K
,
Itoh
M
,
Kato
T
,
Kawaji
H
,
Kawagashira
N
,
Kawashima
T
,
Kojima
M
,
Kondo
S
,
Konno
H
,
Nakano
K
,
Ninomiya
N
,
Nishio
T
,
Okada
M
,
Plessy
C
,
Shibata
K
,
Shiraki
T
,
Suzuki
S
,
Tagami
M
,
Waki
K
,
Watahiki
A
,
Okamura-Oho
Y
,
Suzuki
H
,
Kawai
J
,
Hayashizaki
Y.
The transcriptional landscape of the mammalian genome
.
Science
2005
;
309
:
1559
1563
.

38

Wang
ET
,
Sandberg
R
,
Luo
S
,
Khrebtukova
I
,
Zhang
L
,
Mayr
C
,
Kingsmore
SF
,
Schroth
GP
,
Burge
CB.
Alternative isoform regulation in human tissue transcriptomes
.
Nature
2008
;
456
:
470
476
.

39

Aebersold
R
,
Agar
JN
,
Amster
IJ
,
Baker
MS
,
Bertozzi
CR
,
Boja
ES
,
Costello
CE
,
Cravatt
BF
,
Fenselau
C
,
Garcia
BA
,
Ge
Y
,
Gunawardena
J
,
Hendrickson
RC
,
Hergenrother
PJ
,
Huber
CG
,
Ivanov
AR
,
Jensen
ON
,
Jewett
MC
,
Kelleher
NL
,
Kiessling
LL
,
Krogan
NJ
,
Larsen
MR
,
Loo
JA
, Ogorzalek
Loo
RR
,
Lundberg
E
,
MacCoss
MJ
,
Mallick
P
,
Mootha
VK
,
Mrksich
M
,
Muir
TW
,
Patrie
SM
,
Pesavento
JJ
,
Pitteri
SJ
,
Rodriguez
H
,
Saghatelian
A
,
Sandoval
W
, Schlü
ter
H
,
Sechi
S
,
Slavoff
SA
,
Smith
LM
,
Snyder
MP
,
Thomas
PM
, Uhlé
n
M
, Van
Eyk
JE
,
Vidal
M
,
Walt
DR
,
White
FM
,
Williams
ER
,
Wohlschlager
T
,
Wysocki
VH
,
Yates
NA
,
Young
NL
,
Zhang
B.
How many human proteoforms are there?
Nat Chem Biol
2018
;
14
:
206
214
.

40

Yates
AD
,
Achuthan
P
,
Akanni
W
,
Allen
J
,
Allen
J
,
Alvarez-Jarreta
J
,
Amode
MR
,
Armean
IM
,
Azov
AG
,
Bennett
R
,
Bhai
J
,
Billis
K
,
Boddu
S
, Marugá
n
JC
,
Cummins
C
,
Davidson
C
,
Dodiya
K
,
Fatima
R
,
Gall
A
,
Giron
CG
,
Gil
L
,
Grego
T
,
Haggerty
L
,
Haskell
E
,
Hourlier
T
,
Izuogu
OG
,
Janacek
SH
,
Juettemann
T
,
Kay
M
,
Lavidas
I
,
Le
T
,
Lemos
D
,
Martinez
JG
,
Maurel
T
,
McDowall
M
,
McMahon
A
,
Mohanan
S
,
Moore
B
,
Nuhn
M
,
Oheh
DN
,
Parker
A
,
Parton
A
,
Patricio
M
,
Sakthivel
MP
, Abdul
Salam
AI
,
Schmitt
BM
,
Schuilenburg
H
,
Sheppard
D
,
Sycheva
M
,
Szuba
M
,
Taylor
K
,
Thormann
A
,
Threadgold
G
,
Vullo
A
,
Walts
B
,
Winterbottom
A
,
Zadissa
A
,
Chakiachvili
M
,
Flint
B
,
Frankish
A
,
Hunt
SE
,
Iisley
G
,
Kostadima
M
,
Langridge
N
,
Loveland
JE
,
Martin
FJ
,
Morales
J
,
Mudge
JM
,
Muffato
M
,
Perry
E
,
Ruffier
M
,
Trevanion
SJ
,
Cunningham
F
,
Howe
KL
,
Zerbino
DR
,
Flicek
P.
Ensembl 2020
.
Nucl Acids Res
2020
;
48
:
D682
D688
.

41

Lee
Y
,
Rio
DC
.
Mechanisms and regulation of alternative pre-mRNA splicing
.
Ann Rev Biochem
2015
;
84
:
291
323
.

42

Danckwardt
S
,
Neu-Yilik
G
,
Thermann
R
,
Frede
U
,
Hentze
MW
,
Kulozik
AE
.
Abnormally spliced beta-globin mRNAs: a single point mutation generates transcripts sensitive and insensitive to nonsense-mediated mRNA decay
.
Blood
2002
;
99
:
1811
1816
.

43

Li
YI
,
van de Geijn
B
,
Raj
A
,
Knowles
DA
,
Petti
AA
,
Golan
D
,
Gilad
Y
,
Pritchard
JK.
RNA Splicing is a primary link between genetic variation and disease
.
Science
2016
;
352
:
600
604
.

44

Castaldi
PJ
,
Abood
A
,
Farber
CR
,
Sheynkman
GM
.
Bridging the splicing gap in human genetics with long-read RNA sequencing: finding the protein isoform drivers of disease
.
Hum Mol Genet
2022
;
31
:
R123
R136
.

45

Reyes
A
,
Huber
W
.
Alternative start and termination sites of transcription drive most transcript isoform differences across human tissues
.
Nucl Acids Res
2017
;
46
:
582
592
.

46

Yuan
L
,
Janes
L
,
Beeler
D
,
Spokes
KC
,
Smith
J
,
Li
D
, Jaminet S-C,
Oettgen
P
,
Aird
WC.
Role of RNA splicing in mediating lineage-specific expression of the von Willebrand factor gene in the endothelium
.
Blood
2013
;
121
:
4404
4412
.

47

Hsu
T-C
,
Shore
SK
,
Seshsmma
T
,
Bagasra
O
,
Walsh
PN
.
Molecular cloning of platelet factor XI, an alternative splicing product of the plasma factor XI gene
.
J Biol Chem
1998
;
273
:
13787
13793
.

48

Asselta
R
,
Rimoldi
V
,
Guella
I
,
Soldà
G
,
De Cristofaro
R
,
Peyvandi
F
,
Duga
S.
Molecular characterization of in-frame and out-of-frame alternative splicings in coagulation factor XI pre-mRNA
.
Blood
2010
;
115
:
2065
2072
.

49

Podmore
A
,
Smith
M
,
Savidge
G
,
Alhaq
A
.
Real-time quantitative PCR analysis of factor XI mRNA variants in human platelets
.
J Thromb Haemost
2004
;
2
:
1713
1719
.

50

Fu
Y
,
Weissbach
L
,
Plant
PW
,
Oddoux
C
,
Cao
Y
,
Liang
TJ
,
Roy
SN
,
Redman
CM
,
Grieninger
G.
Carboxy-terminal-extended variant of the human fibrinogen alpha subunit: a novel exon conferring marked homology to beta and gamma subunits
.
Biochemistry
1992
;
31
:
11968
11972
.

51

Chung
DW
,
Davie
EW
.
Gamma and gamma’ chains of human fibrinogen are produced by alternative mRNA processing
.
Biochemistry
1984
;
23
:
4232
4236
.

52

Vincent
LM
,
Tran
S
,
Livaja
R
,
Bensend
TA
,
Milewicz
DM
,
Dahlbäck
B
.
Coagulation factor v(a2440g) causes east Texas bleeding disorder via TFPIiα
.
J Clin Invest
2013
;
123
:
3777
3787
.

53

Hagen
FS
,
Gray
CL
,
O'Hara
P
,
Grant
FJ
,
Saari
GC
,
Woodbury
RG
,
Hart
CE
,
Insley
M
,
Kisiel
W
,
Kurachi
K
.
Characterization of a cDNA coding for human factor VII
.
Proc Natl Acad Sci
1986
;
83
:
2412
2416
.

54

O'Hara
PJ
,
Grant
FJ
,
Haldeman
BA
,
Gray
CL
,
Insley
MY
,
Hagen
FS
,
Murray
MJ
.
Nucleotide sequence of the gene coding for human factor VII, a vitamin k-dependent protein participating in blood coagulation
.
Proc Natl Acad Sci
1987
;
84
:
5158
5162
.

55

Shovlin
CL
,
Angus
G
,
Manning
RA
,
Okoli
GN
,
Govani
FS
,
Elderfield
K
,
Birdsey
GM
,
Mollet
IG
,
Laffan
MA
,
Mauri
FA.
Endothelial cell processing and alternatively spliced transcripts of factor VIII: potential implications for coagulation cascades and pulmonary hypertension
.
PloS one
2010
;
5
:
e9154
.

56

Zamolodchikov
D
,
Bai
Y
,
Tang
Y
,
McWhirter
JR
,
Macdonald
LE
,
Alessandri-Haber
N
.
A short isoform of coagulation factor XII mRNA is expressed by neurons in the human brain
.
Neuroscience
2019
;
413
:
294
307
.

57

Rehman
SU
,
Ashraf
S
,
Ahamad
S
,
Sarwar
T
,
Husain
MA
,
Ahmad
P
,
Ahmad
P
,
Tabish
M
,
Jairajpuri
MA.
Identification of a novel alternatively spliced isoform of antithrombin containing an additional RCL-like loop
.
Biochem Biophysical Res Commun
2019
;
517
:
421
426
.

58

Bano
S
,
Fatima
S
,
Ahamad
S
,
Ansari
S
,
Gupta
D
,
Tabish
M
,
Rehman
SU
and
Jairajpuri
MA.
Identification and characterization of a novel isoform of heparin cofactor II in human liver
.
IUBMB Life
2020
;
72
:
2180
2193
.

59

Mignone
F
,
Gissi
C
,
Liuni
S
,
Pesole
G
.
Untranslated regions of mRNAs
.
Genome Biol
2002
;
3
:
reviews0004.0001
.

60

Araujo
PR
,
Yoon
K
,
Ko
D
,
Smith
AD
,
Qiao
M
,
Suresh
U
,
Burns
SC
,
Penalva
LOF.
Before it gets started: regulating translation at the 5′ UTR
.
Comp Funct Gen
2012
;
2012
:
475731
.

61

Bogdanov
VY
,
Balasubramanian
V
,
Hathcock
J
,
Vele
O
,
Lieb
M
,
Nemerson
Y
.
Alternatively spliced human tissue factor: a circulating, soluble, thrombogenic protein
.
Nat Med
2003
;
9
:
458
462
.

62

Witkowski
M
,
Landmesser
U
,
Rauch
U
.
Tissue factor as a link between inflammation and coagulation
.
Trends Cardiovasc Med
2016
;
26
:
297
303
.

63

Szotowski
B
,
Antoniak
S
,
Poller
W
,
Schultheiss
HP
,
Rauch
U
.
Procoagulant soluble tissue factor is released from endothelial cells in response to inflammatory cytokines
.
Circ Res
2005
;
96
:
1233
1239
.

64

Owens
AP
,
Mackman
N
.
Tissue factor and thrombosis: the clot starts here
.
Thromb Haemost
2010
;
104
:
432
439
.

65

Bogdanov
VY
,
Versteeg
HH
.
“Soluble tissue factor” in the 21st century: definitions, biochemistry, and pathophysiological role in thrombus formation
.
Sem Thromb Hemost
2015
;
41
:
700
707
.

66

Mast
AE
,
Ruf
W
.
Regulation of coagulation by tissue factor pathway inhibitor: implications for hemophilia therapy
.
J Thromb Haemost
2022
;
20
:
1290
1300
.

67

Peterson
JA
,
Maroney
SA
,
Martinez
ND
,
Mast
AE
.
Major reservoir for heparin-releasable TFPIα (tissue factor pathway inhibitor α) is extracellular matrix
.
Arterioscler Thromb Vasc Biol
2021
;
41
:
1942
1955
.

68

Wood
JP
,
Ellery
PE
,
Maroney
SA
,
Mast
AE
.
Biology of tissue factor pathway inhibitor
.
Blood
2014
;
123
:
2934
2943
.

69

Castoldi
E
,
Simioni
P
,
Tormene
D
,
Rosing
J
,
Hackeng
TM
.
Hereditary and acquired protein s deficiencies are associated with low TFPI levels in plasma
.
J Thromb Haemost
2010
;
8
:
294
300
.

70

Duckers
C
,
Simioni
P
,
Spiezia
L
,
Radu
C
,
Gavasso
S
,
Rosing
J
,
Castoldi
E.
Low plasma levels of tissue factor pathway inhibitor in patients with congenital factor V deficiency
.
Blood
2008
;
112
:
3615
3623
.

71

Hackeng
TM
,
Seré
KM
,
Tans
G
,
Rosing
J
.
Protein s stimulates inhibition of the tissue factor pathway by tissue factor pathway inhibitor
.
Proc Natl Acad Sci
2006
;
103
:
3106
3111
.

72

Santamaria
S
,
Reglińska-Matveyev
N
,
Gierula
M
,
Camire
RM
,
Crawley
JTB
,
Lane
DA
,
Ahnström
J.
Factor V has an anticoagulant cofactor activity that targets the early phase of coagulation
.
J Biol Chem
2017
;
292
:
9335
9344
.

73

Dahlbäck
B
,
Guo
LJ
,
Livaja-Koshiar
R
,
Tran
S
.
Factor v-short and protein s as synergistic tissue factor pathway inhibitor (TFPIα) cofactors
.
Rese Pract Thromb Haemost
2018
;
2
:
114
124
.

74

van Doorn
P
,
Rosing
J
,
Wielders
SJ
,
Hackeng
TM
,
Castoldi
E
.
The c-terminus of tissue factor pathway inhibitor-α inhibits factor V activation by protecting the arg(1545) cleavage site
.
J Thromb Haemost
2017
;
15
:
140
149
.

75

Wood
JP
,
Bunce
MW
,
Maroney
SA
,
Tracy
PB
,
Camire
RM
,
Mast
AE
.
Tissue factor pathway inhibitor-alpha inhibits prothrombinase during the initiation of blood coagulation
.
Proc Natl Acad Sci
2013
;
110
:
17838
17843
.

76

Wood
JP
,
Petersen
HH
,
Yu
B
,
Wu
X
,
Hilden
I
,
Mast
AE
.
Tfpiα interacts with FVa and FXa to inhibit prothrombinase during the initiation of coagulation
.
Blood Adv
2017
;
1
:
2692
2702
.

77

Uitte de Willige
S
,
Standeven
KF
,
Philippou
H
,
Ariëns
RA
.
The pleiotropic role of the fibrinogen gamma’ chain in hemostasis
.
Blood
2009
;
114
:
3994
4001
.

78

Girard
TJ
,
Grunz
K
,
Lasky
NM
,
Malone
JP
,
Broze
GJ
.
Re-evaluation of mouse tissue factor pathway inhibitor and comparison of mouse and human tissue factor pathway inhibitor physiology
.
J Thromb Haemost
2018
;
16
:
2246
2257
.

79

Louis
JM
,
Vaz
C
,
Balaji
A
,
Tanavde
V
,
Talukdar
I
.
TNF-alpha regulates alternative splicing of genes participating in pathways of crucial metabolic syndromes; a transcriptome wide study
.
Cytokine
2020
;
125
:
154815
.

80

Wang
C
,
Chen
L
,
Chen
Y
,
Jia
W
,
Cai
X
,
Liu
Y
,
Ji
F
,
Xiong
P
,
Liang
A
,
Liu
R
,
Guan
Y
,
Cheng
Z
,
Weng
Y
,
Wang
W
,
Duan
Y
,
Kuang
D
,
Xu
S
,
Cai
H
,
Xia
Q
,
Yang
D
,
Wang
MW
,
Yang
X
,
Zhang
J
,
Cheng
C
,
Liu
L
,
Liu
Z
,
Liang
R
,
Wang
G
,
Li
Z
,
Xia
H
,
Xia
T.
Abnormal global alternative RNA splicing in COVID-19 patients
.
PLoS Genet
2022
;
18
:
e1010137
.

81

Lee
SC
,
Abdel-Wahab
O
.
Therapeutic targeting of splicing in cancer
.
Nat Med
2016
;
22
:
976
986
.

82

Desterro
J
,
Bak-Gordon
P
,
Carmo-Fonseca
M
.
Targeting mRNA processing as an anticancer strategy
.
Nat Rev Drug Discov
2020
;
19
:
112
129
.

83

Havens
MA
,
Hastings
ML
.
Splice-switching antisense oligonucleotides as therapeutic drugs
.
Nucl Acids Res
2016
;
44
:
6549
6563
.

84

Fernandez Alanis
E
,
Pinotti
M
,
Dal Mas
A
,
Balestra
D
,
Cavallari
N
,
Rogalska
ME
,
Bernardi
F
,
Pagani
F
.
An exon-specific U1 small nuclear RNA (snRNA) strategy to correct splicing defects
.
Hum Mol Genet
2012
;
21
:
2389
2398
.

85

Lewis
BP
,
Green
RE
,
Brenner
SE
.
Evidence for the widespread coupling of alternative splicing and nonsense-mediated mRNA decay in humans
.
Proc Natl Acad Sci
2003
;
100
:
189
192
.

86

Lim
KH
,
Han
Z
,
Jeon
HY
,
Kach
J
,
Jing
E
,
Weyn-Vanhentenryck
S
,
Weyn-Vanhentenryck
S
,
Downs
M
,
Corrionero
A
,
Oh
R
,
Scharner
J
,
Venkatesh
A
,
Ji
S
,
Liau
G
,
Ticho
B
,
Nash
H
,
Aznarez
I
.
Antisense oligonucleotide modulation of non-productive alternative splicing upregulates gene expression
.
Nat Commun
2020
;
11
:
3501
.

87

Danckwardt
S
,
Hentze
MW
,
Kulozik
AE
.
3’ end mRNA processing: molecular mechanisms and implications for health and disease
.
EMBO J
2008
;
27
:
482
498
.

88

Shi
Y
,
Manley
JL
.
The end of the message: multiple protein-RNA interactions define the mRNA polyadenylation site
.
Genes Dev
2015
;
29
:
889
897
.

89

Gruber
AJ
,
Schmidt
R
,
Gruber
AR
,
Martin
G
,
Ghosh
S
,
Belmadani
M
,
Keller
W
,
Zavolan
MA
.
comprehensive analysis of 3′ end sequencing data sets reveals novel polyadenylation signals and the repressive role of heterogeneous ribonucleoprotein C on cleavage and polyadenylation
.
Genome Res
2016
;
26
:
1145
1159
.

90

Proudfoot
N
.
Transcriptional termination in mammals: stopping the RNA polymerase II juggernaut
.
Science
2016
;
352
:
6291
.

91

Reimer
KA
,
Mimoso
CA
,
Adelman
K
,
Neugebauer
KM
.
Co-transcriptional splicing regulates 3’ end cleavage during mammalian erythropoiesis
.
Mol Cell
2021
;
81
:
998
1012.e1017
.

92

Nourse
J
,
Spada
S
,
Danckwardt
S
.
Emerging roles of RNA 3′-end cleavage and polyadenylation in pathogenesis, diagnosis and therapy of human disorders
.
Biomol
2020
;
10
:
915
.

93

Hollerer
I
,
Grund
K
,
Hentze
MW
,
Kulozik
AE
.
mRNA 3'end processing: a tale of the tail reaches the clinic
.
EMBO Mol Med
2014
;
6
:
16
26
.

94

Poort
SR
,
Rosendaal
FR
,
Reitsma
PH
,
Bertina
RM
.
A common genetic variation in the 3'-untranslated region of the prothrombin gene is associated with elevated plasma prothrombin levels and an increase in venous thrombosis
.
Blood
1996
;
88
:
3698
3703
.

95

Ceelie
H
,
Bertina
RM
,
van Hylckama Vlieg
A
,
Rosendaal
FR
,
Vos
HL
.
Polymorphisms in the prothrombin gene and their association with plasma prothrombin levels
.
Thromb Haemost
2001
;
85
:
1066
1070
.

96

Gehring
NH
,
Frede
U
,
Neu-Yilik
G
,
Hundsdoerfer
P
,
Vetter
B
,
Hentze
MW
,
Kulozik
AE.
Increased efficiency of mRNA 3’ end formation: a new genetic mechanism contributing to hereditary thrombophilia
.
Nat Genet
2001
;
28
:
389
392
.

97

Danckwardt
S
,
Gehring
NH
,
Neu-Yilik
G
,
Hundsdoerfer
P
,
Pforsich
M
,
Frede
U
,
Hentze
MW
,
Kulozik
AE
.
The prothrombin 3'end formation signal reveals a unique architecture that is sensitive to thrombophilic gain-of-function mutations
.
Blood
2004
;
104
:
428
435
.

98

Soria
JM
,
Almasy
L
,
Souto
JC
,
Tirado
I
,
Borell
M
,
Mateo
J
,
Mateo
J
,
Slifer
S
,
Stone
W
,
Blangero
J
,
Fontcuberta
J
.
Linkage analysis demonstrates that the prothrombin g20210a mutation jointly influences plasma prothrombin levels and risk of thrombosis
.
Blood
2000
;
95
:
2780
2785
.

99

Danckwardt
S
,
Hartmann
K
,
Gehring
NH
,
Hentze
MW
,
Kulozik
AE
.
3’ end processing of the prothrombin mRNA in thrombophilia
.
Acta Haematol
2006
;
115
:
192
197
.

100

Danckwardt
S
,
Hartmann
K
,
Katz
B
,
Hentze
M
,
Levy
Y
,
Eichele
R
,
Deutsch
V
,
Kulozik
A
,
Ben-Tal
O.
The prothrombin 20209 c > t mutation in Jewish-Moroccan caucasians: molecular analysis of gain-of-function of 3'end processing
.
J Thromb Haemost
2006
;
4
:
1078
1085
.

101

Danckwardt
S
,
Kaufmann
I
,
Gentzel
M
,
Foerstner
KU
,
Gantzert
AS
,
Gehring
NH
,
Neu-Yilik
G
,
Bork
P
,
Keller
W
,
Wilm
M
,
Hentze
MW
,
Kulozik
AE.
Splicing factors stimulate polyadenylation via uses at non-canonical 3’ end formation signals
.
EMBO J
2007
;
26
:
2658
2669
.

102

Danckwardt
S
,
Gantzert
AS
,
Macher-Goeppinger
S
,
Probst
HC
,
Gentzel
M
,
Wilm
M
,
Grone
HJ
,
Schirmacher
P
,
Hentze
MW
and
Kulozik
AE.
P38 MAPK controls prothrombin expression by regulated RNA 3’ end processing
.
Mol Cell
2011
;
41
:
298
310
.

103

Roux
PP
,
Blenis
J
.
Erk and p38 mapk-activated protein kinases: a family of protein kinases with diverse biological functions
.
Microbiol Mol Biol Rev
2004
;
68
:
320
344
.

104

Wagner
EF
,
Nebreda
AR
.
Signal integration by jnk and p38 mapk pathways in cancer development
.
Nat Rev Cancer
2009
;
9
:
537
549
.

105

Murakami
H
,
Okazaki
M
,
Amagasa
H
,
Oguchi
K
.
Increase in hepatic mRNA expression of coagulant factors in type 2 diabetic model mice
.
Thromb Res
2003
;
111
:
81
87
.

106

Boven
LA
,
Vergnolle
N
,
Henry
SD
,
Silva
C
,
Imai
Y
,
Holden
J
,
Warren
K
,
Hollenberg
MD
,
Power
C.
Up-regulation of proteinase-activated receptor 1 expression in astrocytes during hiv encephalitis
.
J Immunol
2003
;
170
:
2638
2646
.

107

Huber-Lang
M
,
Sarma
JV
,
Zetoune
FS
,
Rittirsch
D
,
Neff
TA
,
McGuire
SR
,
Lambris
JD
,
Warner
RL
,
Flierl
MA
,
Hoesel
LM
,
Gebhard
F
,
Younger
JG
,
Drouin
SM
,
Wetsel
RA
,
Ward
PA.
Generation of c5a in the absence of c3: a new complement activation pathway
.
Nat Med
2006
;
12
:
682
687
.

108

Yin
X
,
Wright
J
,
Wall
T
,
Grammas
P
.
Brain endothelial cells synthesize neurotoxic thrombin in Alzheimer's disease
.
Am J Pathol
2010
;
176
:
1600
1606
.

109

Esmon
CT
.
The interactions between inflammation and coagulation
.
B J Haematol
2005
;
131
:
417
430
.

110

Nourse
J
,
Tokalov
S
,
Khokhar
S
,
Khan
E
,
Schott
LK
,
Hinz
L
,
Eder
L
,
Arnold-Schild
D
,
Satrapa
J
,
Lackner
KJ
, ten
Cate
H
,
Probst
HC
,
Danckwardt
S.
Non-invasive imaging of gene expression and protein secretion dynamics in living mice: identification of ectopic prothrombin expression as driver of thrombosis in cancer
.
bioRxiv
2021
;
2021
:
2007.2008.451623
.

111

Cohen
J
.
The immunopathogenesis of sepsis
.
Nature
2002
;
420
:
885
891
.

112

Russell
JA
.
Management of sepsis
.
N Engl J Med
2006
;
355
:
1699
1713
.

113

Coughlin
SR
.
Thrombin signalling and protease-activated receptors
.
Nature
2000
;
407
:
258
264
.

114

Riek-Burchardt
M
,
Striggow
F
,
Henrich-Noack
P
,
Reiser
G
,
Reymann
KG
.
Increase of prothrombin-mRNA after global cerebral ischemia in rats, with constant expression of protease nexin-1 and protease-activated receptors
.
Neurosci Lett
2002
;
329
:
181
184
.

115

Danckwardt
S
,
Hentze
MW
,
Kulozik
AE
.
Pathologies at the nexus of blood coagulation and inflammation: thrombin in hemostasis, cancer and beyond
.
J Mol Med
2013
;
91
:
1257
1271
.

116

Vorlova
S
,
Rocco
G
,
Lefave
CV
,
Jodelka
FM
,
Hess
K
,
Hastings
ML
,
Henke
E
,
Cartegni
L.
Induction of antagonistic soluble decoy receptor tyrosine kinases by intronic polya activation
.
Mol Cell
2011
;
43
:
927
939
.

117

Ogorodnikov
A
,
Levin
M
,
Tattikota
S
,
Tokalov
S
,
Hoque
M
,
Scherzinger
D
,
Marini
F
,
Poetsch
A
,
Binder
H
, Macher-Gö
ppinger
S
,
Probst
HC
,
Tian
B
,
Schaefer
M
,
Lackner
KJ
,
Westermann
F
,
Danckwardt
S.
Transcriptome 3′end organization by pcf11 links alternative polyadenylation to formation and neuronal differentiation of neuroblastoma
.
Nat Commun
2018
;
9
:
5331
.

118

Jia
X
,
Yuan
S
,
Wang
Y
,
Fu
Y
,
Ge
Y
,
Ge
Y
,
Lan
X
,
Feng
Y
,
Qiu
F
,
Li
P
,
Chen
S
and
Xu
A.
The role of alternative polyadenylation in the antiviral innate immune response
.
Nat Commun
2017
;
8
:
14605
.

119

Kargapolova
Y
,
Levin
M
,
Lackner
K
,
Danckwardt
S
.
Sclip—an integrated platform to study RNA–protein interactomes in biomedical research: identification of cstf2tau in alternative processing of small nuclear RNAs
.
Nucl Acid Res
2017
;
45
:
6074
6086
.

120

Ogorodnikov
A
,
Danckwardt
S
.
Trendseq-a highly multiplexed high throughput RNA 3’ end sequencing for mapping alternative polyadenylation
.
Meth Enzymol
2021
;
655
:
37
72
.

121

Marini
F
,
Scherzinger
D
,
Danckwardt
S
.
Trend-db—a transcriptome-wide atlas of the dynamic landscape of alternative polyadenylation
.
Nucl Acids Res
2021
;
49
:
D243
D253
.

122

Vossen
CY
,
van Hylckama Vlieg
A
,
Teruel-Montoya
R
,
Salloum-Asfar
S
,
de Haan
H
,
Corral
J
,
Reitsma
P
,
Koeleman
BPC
,
Martínez
C.
Identification of coagulation gene 3′utr variants that are potentially regulated by microRNAs
.
Brit J Haematol
2017
;
177
:
782
790
.

123

Arroyo
AB
,
de Los Reyes-García
AM
,
Teruel-Montoya
R
,
Vicente
V
,
González-Conejero
R
,
Martínez
C
.
MicroRNAs in the haemostatic system: more than witnesses of thromboembolic diseases?
Thromb Res
2018
;
166
:
1
9
.

124

Chen
K
,
Rajewsky
N
.
The evolution of gene regulation by transcription factors and microRNAs
.
Nat Rev Genet
2007
;
8
:
93
103
.

125

Li
S
,
Patel
DJ
.
Drosha and dicer: slicers cut from the same cloth
.
Cell Res
2016
;
26
:
511
512
.

126

Krol
J
,
Loedige
I
,
Filipowicz
W
.
The widespread regulation of microRNA biogenesis, function and decay
.
Nat Rev Genet
2010
;
11
:
597
610
.

127

Meister
G
.
Argonaute proteins: functional insights and emerging roles
.
Nat Rev Genet
2013
;
14
:
447
.

128

Fabian
MR
,
Sonenberg
N
,
Filipowicz
W
.
Regulation of mRNA translation and stability by microRNAs
.
Ann Rev Biochem
2010
;
79
:
351
379
.

129

Bartel
DP
.
MicroRNAs: target recognition and regulatory functions
.
Cell
2009
;
136
:
215
233
.

130

Kozomara
A
,
Birgaoanu
M
,
Griffiths-Jones
S
.
Mirbase: from microRNA sequences to function
.
Nucl Acids Res
2018
;
47
:
D155
D162
.

131

Friedman
RC
,
Farh
KK-H
,
Burge
CB
,
Bartel
DP
.
Most mammalian mRNAs are conserved targets of microRNAs
.
Genome Res
2009
;
19
:
92
105
.

132

Saliminejad
K
,
Khorram Khorshid
HR
,
Soleymani Fard
S
,
Ghaffari
SH
.
An overview of microRNAs: biology, functions, therapeutics, and analysis methods
.
J Cell Physiol
2019
;
234
:
5451
5465
.

133

Mukherji
S
,
Ebert
MS
,
Zheng
GXY
,
Tsang
JS
,
Sharp
PA
,
van Oudenaarden
A
.
MicroRNAs can generate thresholds in target gene expression
.
Nat Genet
2011
;
43
:
854
859
.

134

Lui
P-Y
,
Jin
D-Y
,
Stevenson
NJ
.
MicroRNA: master controllers of intracellular signaling pathways
.
Cell Mol Life Sci
2015
;
72
:
3531
3542
.

135

Nourse
J
,
Braun
J
,
Lackner
K
,
Hüttelmaier
S
,
Danckwardt
S
.
Large-scale identification of functional microRNA targeting reveals cooperative regulation of the hemostatic system
.
J Thromb Haemost
2018
;
16
:
2233
2245
.

136

Teruel-Montoya
R
,
Rosendaal
FR
,
Martínez
C
.
MicroRNAs in hemostasis
.
J Thromb Haemost
2015
;
13
:
170
181
.

137

Reyes-García AM
DL
,
Arroyo
AB
,
Teruel-Montoya
R
,
Vicente
V
,
Lozano
ML
,
González-Conejero
R
,
Martínez
C
.
MicroRNAs as potential regulators of platelet function and bleeding diatheses
.
Platelets
2019
;
30
:
803
808
.

138

Jankowska
KI
,
Sauna
ZE
,
Atreya
CD
.
Role of microRNAs in hemophilia and thrombosis in humans
.
Int J Mol Sci
2020
;
21
:
3598
.

139

Nourse
J
,
Danckwardt
S
.
A novel rationale for targeting FXI: insights from the hemostatic microRNA targetome for emerging anticoagulant strategies
.
Pharmacol Therap
2021
;
218
:
107676
.

140

Morelli
VM
,
Brækkan
SK
,
Hansen
J-B
.
Role of microRNAs in venous thromboembolism
.
Int J Mol Sci
2020
;
21
:
2602
.

141

Hembrom
AA
,
Srivastava
S
,
Garg
I
,
Kumar
B
.
MicroRNAs in venous thrombo-embolism
.
Clin Chim Acta; Int J Clin Chem
2020
;
504
:
66
72
.

142

Chen
Z
,
Nakajima
T
,
Tanabe
N
,
Hinohara
K
,
Sakao
S
,
Kasahara
Y
,
Tatsumi
K
,
Inoue
Y
,
Kimura
A.
Susceptibility to chronic thromboembolic pulmonary hypertension may be conferred by mir-759 via its targeted interaction with polymorphic fibrinogen alpha gene
.
Hum Genetics
2010
;
128
:
443
452
.

143

Fort
A
,
Borel
C
,
Migliavacca
E
,
Antonarakis
SE
,
Fish
RJ
,
Neerman-Arbez
M
.
Regulation of fibrinogen production by microRNAs
.
Blood
2010
;
116
:
2608
2615
.

144

Li
S
,
Ren
J
,
Xu
N
,
Zhang
J
,
Geng
Q
,
Cao
C
,
Lee
C
,
Song
J
,
Li
J
,
Chen
H.
MicroRNA-19b functions as potential anti-thrombotic protector in patients with unstable angina by targeting tissue factor
.
J Mol Cell Cardiol
2014
;
75
:
49
57
.

145

Teruel
R
,
Pérez-Sánchez
C
,
Corral
J
,
Herranz
MT
,
Pérez-Andreu
V
,
Saiz
E
,
GarcÍa-BarberÁ
N
,
MartÍnez-MartÍnez
I
,
RoldÁn
V
,
Vicente
V
,
LÓpez-Pedrera
C
,
MartÍnez
C.
Identification of miRNAs as potential modulators of tissue factor expression in patients with systemic lupus erythematosus and antiphospholipid syndrome
.
J Thromb Haemost
2011
;
9
:
1985
1992
.

146

Witkowski
M
,
Weithauser
A
,
Tabaraie
T
,
Steffens
D
,
Kränkel
N
,
Witkowski
M
,
Stratmann
B
,
Tschoepe
D
,
Landmesser
U
,
Rauch-Kroehnert
Ul.
MicroRNA-126 reduces the blood thrombogenicity in diabetes mellitus via targeting of tissue factor
.
Arterioscler Thromb Vasc Biol
2016
;
36
:
1263
1271
.

147

Sahu
A
,
Jha
PK
,
Prabhakar
A
,
Singh
HD
,
Gupta
N
,
Chatterjee
T
,
Tyagi
T
,
Sharma
S
,
Kumari
B
and
Singh
S.
MicroRNA-145 impedes thrombus formation via targeting tissue factor in venous thrombosis
.
EBioMedicine
2017
;
26
:
175
186
.

148

Li
S
,
Chen
H
,
Ren
J
,
Geng
Q
,
Song
J
,
Lee
C
,
Cao
C
,
Zhang
J
,
Xu
N.
MicroRNA-223 inhibits tissue factor expression in vascular endothelial cells
.
Atheroscler
2014
;
237
:
514
520
.

149

Tian
J
,
Adams
MJ
,
Tay
JWT
,
James
I
,
Powell
S
,
Hughes
QW
,
Gilmore
G
,
Baker
RI
,
Tiao
JY.
Estradiol-responsive mir-365a-3p interacts with tissue factor 3'UTR to modulate tissue factor-initiated thrombin generation
.
Thromb Haemost
2021
;
121
:
1483
1496
.

150

Zhang
X
,
Yu
H
,
Lou
JR
,
Zheng
J
,
Zhu
H
,
Popescu
N-I
,
Lupu
F
,
Lind
SE
,
Ding
W-Q.
MicroRNA-19 (mir-19) regulates tissue factor expression in breast cancer cells
.
J Biol Chem
2011
;
286
:
1429
1435
.

151

Jankowska
KI
,
McGill
J
,
Pezeshkpoor
B
,
Oldenburg
J
,
Atreya
CD
,
Sauna
ZE
.
Clinical manifestation of hemophilia A in the absence of mutations in the F8 gene that encodes FVIII: role of microRNAs
.
Transfusion
2020
;
60
:
401
413
.

152

Stather
PW
,
Sylvius
N
,
Wild
JB
,
Choke
E
,
Sayers
RD
,
Bown
MJ
.
Differential microRNA expression profiles in peripheral arterial disease
.
Circulation Cardiovasc Genet
2013
;
6
:
490
497
.

153

de Freitas
RCC
,
Bortolin
RH
,
Lopes
MB
,
Tamborlin
L
,
Meneguello
L
,
Silbiger
VN
,
Hirata
RDC
,
Hirata
MH
,
Luchessi
AD
,
Luchessi
AD.
Modulation of mir-26a-5p and mir-15b-5p exosomal expression associated with clopidogrel-induced hepatotoxicity in HEPG2 cells
.
Front Pharmacol
2017
;
8
:
906
.

154

Chen
L-J
,
Yang
L
,
Cheng
X
,
Xue
Y-K
,
Chen
L-B
.
Overexpression of mir-24 is involved in the formation of hypocoagulation state after severe trauma by inhibiting the synthesis of coagulation factor X
.
Dis Markers
2017
;
2017
:
3649693
.

155

Zheng
Y
,
Li
Y
,
Liu
G
,
Qi
X
,
Cao
X
.
MicroRNA-24 inhibits the proliferation and migration of endothelial cells in patients with atherosclerosis by targeting importin-α3 and regulating inflammatory responses
.
Exp Ther Med
2018
;
15
:
338
344
.

156

Zhou
J
,
Zhang
J
.
Identification of miRNA-21 and miRNA-24 in plasma as potential early stage markers of acute cerebral infarction
.
Mol Med Rep
2014
;
10
:
971
976
.

157

Zhu
X
,
Shan
Z
,
Ma
J
,
Wang
M
,
Zhang
C
,
Liu
R
,
Wu
W
,
Shi
Y
,
Li
W
,
Wang
S.
Investigating the role of the post-transcriptional gene regulator mir-24-3p in the proliferation, migration and apoptosis of human arterial smooth muscle cells in arteriosclerosis obliterans
.
Cell Physiol Biochem
2015
;
36
:
1359
1370
.

158

Long
G
,
Wang
F
,
Duan
Q
,
Yang
S
,
Chen
F
,
Gong
W
,
Yang
X
,
Wang
Y
,
Chen
C
,
Wang
DW.
Circulating mir-30a, mir-195 and let-7b associated with acute myocardial infarction
.
PloS one
2012
;
7
:
e50926
.

159

Long
G
,
Wang
F
,
Li
H
,
Yin
Z
,
Sandip
C
,
Lou
Y
,
Wang
Y
,
Chen
C
,
Wang
DW.
Circulating mir-30a, mir-126 and let-7b as biomarker for ischemic stroke in humans
.
BMC Neurol
2013
;
13
:
178
.

160

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

161

Zhang
Y
,
Chen
M
,
Zhang
Y
,
Peng
P
,
Li
J
,
Xin
X
.
Mir-96 and mir-330 overexpressed and targeted AQP5 in lipopolysaccharide-induced rat lung damage of disseminated intravascular coagulation
.
Blood Coag Fibrinol
2014
;
25
:
731
737
.

162

Hartmann
P
,
Zhou
Z
,
Natarelli
L
,
Wei
Y
,
Nazari-Jahantigh
M
,
Zhu
M
,
Grommes
J
,
Steffens
S
,
Weber
C
,
Schober
A.
Endothelial dicer promotes atherosclerosis and vascular inflammation by miRNA-103-mediated suppression of KLF4
.
Nat Commun
2016
;
7
:
1
16
.

163

Starikova
I
,
Jamaly
S
,
Sorrentino
A
,
Blondal
T
,
Latysheva
N
,
Sovershaev
M
,
Hansen
JB.
Differential expression of plasma miRNAs in patients with unprovoked venous thromboembolism and healthy control individuals
.
Thromb Res
2015
;
136
:
566
572
.

164

Sennblad
B
,
Basu
S
,
Mazur
J
,
Suchon
P
,
Martinez-Perez
A
,
van Hylckama Vlieg
A
,
Truong
V
,
Li
Y
, Gå
din
JR
,
Tang
W
,
Grossman
V
, de
Haan
HG
,
Handin
N
,
Silveira
A
,
Souto
JC
,
Franco-Cereceda
A
,
Morange
PE
,
Gagnon
F
,
Soria
JM
,
Eriksson
P
,
Hamsten
A
,
Maegdefessel
L
,
Rosendaal
FR
,
Wild
P
,
Folsom
AR
, Trégouë
t
DA
,
Sabater-Lleal
M.
Genome-wide association study with additional genetic and post-transcriptional analyses reveals novel regulators of plasma factor XI levels
.
Hum Mol Genet
2017
;
26
:
637
649
.

165

Dong
Y-M
,
Liu
X-X
,
Wei
G-Q
,
Da
Y-N
,
Cha
L
,
Ma
C-S
.
Prediction of long-term outcome after acute myocardial infarction using circulating mir-145
.
Scand J Clin Lab Invest
2015
;
75
:
85
91
.

166

Faccini
J
,
Ruidavets
J-B
,
Cordelier
P
,
Martins
F
,
Maoret
J-J
,
Bongard
V
, Ferriè
res
J
,
Roncalli
J
,
Elbaz
M
,
Vindis
C.
Circulating mir-155, mir-145 and let-7c as diagnostic biomarkers of the coronary artery disease
.
Sci Rep
2017
;
7
:
1
10
.

167

Gao
H
,
Guddeti
RR
,
Matsuzawa
Y
,
Liu
L-P
,
Su
L-X
,
Guo
D
, Nie S-P,
Du
J
,
Zhang
M.
Plasma levels of microRNA-145 are associated with severity of coronary artery disease
.
PloS one
2015
;
10
:
e0123477
.

168

Jia
L
,
Hao
F
,
Wang
W
,
Qu
Y
.
Circulating mir-145 is associated with plasma high-sensitivity c-reactive protein in acute ischemic stroke patients
.
Cell Biochem Funct
2015
;
33
:
314
319
.

169

Maitrias
P
,
Metzinger-Le Meuth
V
,
Massy
ZA
,
M'Baya-Moutoula
E
,
Reix
T
,
Caus
T
,
Metzinger
L.
MicroRNA deregulation in symptomatic carotid plaque
.
J Vasc Surg
2015
;
62
:
1245
1250. e1241
.

170

Meder
B
,
Keller
A
,
Vogel
B
,
Haas
J
,
Sedaghat-Hamedani
F
,
Kayvanpour
E
,
Kayvanpour
E
,
Just
S
,
Borries
A
,
Rudloff
J
and
Leidinger
P.
MicroRNA signatures in total peripheral blood as novel biomarkers for acute myocardial infarction
.
Basic Res Cardiol
2011
;
106
:
13
23
.

171

Sala
F
,
Aranda
JF
,
Rotllan
N
,
Ramírez
CM
,
Aryal
B
,
Elia
L
,
Condorelli
G
,
Catapano
AL
,
Fernández-Hernando
C
,
Norata
GD.
Mir-143/145 deficiency attenuates the progression of atherosclerosis in ldlr-/-mice
.
Thromb Haemost
2014
;
112
:
796
802
.

172

Zhang
M
,
Cheng
Y-J
,
Sara
JD
,
Liu
L-J
,
Liu
L-P
,
Zhao
X
,
Gao
H.
Circulating microRNA-145 is associated with acute myocardial infarction and heart failure
.
Chin Med Jour
2017
;
130
:
51
56
.

173

Salloum-Asfar
S
,
Teruel-Montoya
R
,
Arroyo
AB
,
García-Barberá
N
,
Chaudhry
A
,
Schuetz
E
,
Luengo-Gil
G
,
Vicente
V
,
González-Conejero
R
,
Martínez
C.
Regulation of coagulation factor XI expression by microrRNAs in the human liver
.
PloS one
2014
;
9
:
e111713
.

174

Zhu
J
,
Yao
K
,
Wang
Q
,
Guo
J
,
Shi
H
,
Ma
L
,
Liu
H
,
Gao
W
,
Zou
Y
,
Ge
J.
Circulating mir-181a as a potential novel biomarker for diagnosis of acute myocardial infarction
.
Cell Physiol Biochem
2016
;
40
:
1591
1602
.

175

Vossen
CY
,
van Hylckama Vlieg
A
,
Teruel-Montoya
R
,
Salloum-Asfar
S
,
de Haan
H
,
Corral
J
,
Reitsma
P
,
Koeleman
BP
,
Martínez
C.
Identification of coagulation gene 3′ UTR variants that are potentially regulated by microRNAs
.
Brit J Haematol
2017
;
177
:
782
790
.

176

Edwardson
MA
,
Zhong
X
,
Fiandaca
MS
,
Federoff
HJ
,
Cheema
AK
,
Dromerick
AW
.
Plasma microRNA markers of upper limb recovery following human stroke
.
Sci Rep
2018
;
8
:
1
7
.

177

Xiang
Y
,
Cheng
J
,
Wang
D
,
Hu
X
,
Xie
Y
,
Stitham
J
,
Atteya
G
,
Du
J
,
Tang
WH
,
Lee
SH
,
Leslie
K
,
Spollett
G
,
Liu
Z
,
Herzog
E
,
Herzog
RI
,
Lu
J
,
Martin
KA
,
Hwa
J.
Hyperglycemia repression of mir-24 coordinately upregulates endothelial cell expression and secretion of von Willebrand factor
.
Blood
2015
;
125
:
3377
3387
.

178

Liu
L
,
Pan
J
,
Wang
H
,
Ma
Z
,
Yin
J
,
Yuan
F
,
Yuan
Q
,
Zhou
L
,
Liu
X
,
Zhang
Y
,
Bao
Z
,
Yang
H
,
Ling
J.
Von Willebrand factor rescued by mir-24 inhibition facilitates the proliferation and migration of osteosarcoma cells in vitro
.
Biosci Rep
2018
;
38
:
BSR20180372
.

179

Zhao
L
,
Hua
C
,
Li
Y
,
Sun
Q
,
Wu
W
.
Mir-525-5p inhibits ADAMTS13 and is correlated with ischemia/reperfusion injury-induced neuronal cell death
.
Int J Clin Exp Med
2015
;
8
:
18115
18122
.

180

Ali
HO
,
Arroyo
AB
,
González-Conejero
R
,
Stavik
B
,
Iversen
N
,
Sandset
PM
, Martí
nez
C
,
Skretting
G.
The role of microRNA-27a/b and microRNA-494 in estrogen-mediated downregulation of tissue factor pathway inhibitor α
.
J Thromb Haemost
2016
;
14
:
1226
1237
.

181

Salloum-Asfar S
ABA
,
Pérez-Sánchez
C
,
Teruel-Montoya
R
,
Navarro
S
,
García-Barberá
N
,
Luengo-Gil
G
, Roldá
n
V
,
Hansen
JB
,
López-Pedrera
C
,
Vicente
V
,
González-Conejero
R
,
Martínez
C.
Regulation of TFPIα expression by mir-27a/b-3p in human endothelial cells under normal conditions and in response to androgens
.
Sci Rep
2017
;
7
:
43500
.

182

Tay
JW
,
Romeo
G
,
Hughes
QW
,
Baker
RI
.
Micro-ribonucleic acid 494 regulation of protein S expression
.
J Thromb Haemost
2013
;
11
:
1547
1555
.

183

Li
S
,
Zhang
R
,
Yuan
Y
,
Yi
S
,
Chen
Q
,
Gong
L
,
Liu
J
,
Ding
F
,
Cao
Z
,
Gu
X.
Mir-340 regulates fibrinolysis and axon regrowth following sciatic nerve injury
.
Mol Neurobiol
2017
;
54
:
4379
4389
.

184

Marchand
A
,
Proust
C
,
Morange
P-E
,
Lompré
A-M
,
Trégouët
D-A
.
Mir-421 and mir-3°c inhibit serpine 1 gene expression in human endothelial cells
.
PloS one
2012
;
7
:
e44532
.

185

Patel
N
,
Tahara
SM
,
Malik
P
,
Kalra
VK
.
Involvement of mir-30c and mir-301a in immediate induction of plasminogen activator inhibitor-1 by placental growth factor in human pulmonary endothelial cells
.
Biochem J
2011
;
434
:
473
482
.

186

Luo
M
,
Li
R
,
Ren
M
,
Chen
N
,
Deng
X
,
Tan
X
,
Li
Y
,
Zeng
M
,
Yang
Y
,
Wan
Q
,
Wu
J
.
Hyperglycaemia-induced reciprocal changes in mir-30c and pai-1 expression in platelets
.
Sci Rep
2016
;
6
:
36687
.

187

Brock
M
,
Trenkmann
M
,
Gay
RE
,
Gay
S
,
Speich
R
,
Huber
LC
.
MicroRNA-18a enhances the interleukin-6-mediated production of the acute-phase proteins fibrinogen and haptoglobin in human hepatocytes
.
J Biol Chem
2011
;
286
:
40142
40150
.

188

Wang
G
,
Chai
B
,
Yang
L
.
Mir-128 and mir-125 regulate expression of coagulation factor IX gene with nonsense mutation by repressing nonsense-mediated mRNA decay
.
Biomed Pharmacother
2016
;
80
:
331
337
.

189

Liao
YC
,
Wang
YS
,
Guo
YC
,
Lin
WL
,
Chang
MH
,
Juo
SH
.
Let-7 g improves multiple endothelial functions through targeting transforming growth factor-beta and SIRT-1 signaling
.
J Am Coll Cardiol
2014
;
63
:
1685
1694
.

190

Sun
S
,
Chai
S
,
Zhang
F
,
Lu
L
.
Overexpressed microRNA-103a-3p inhibits acute lower-extremity deep venous thrombosis via inhibition of CXCL12
.
IUBMB Life
2020
;
72
:
492
504
.

191

Thibord
F
,
Munsch
G
,
Perret
C
,
Suchon
P
,
Roux
M
,
Ibrahim-Kosta
M
,
Goumidi
L
,
Deleuze
JF
,
Morange
PE
,
Trégouët
DA.
Bayesian Network analysis of plasma microRNA sequencing data in patients with venous thrombosis
.
Europ Heart Jour
2020
;
22
:
C34
c45
.

192

Wang
J
,
Chen
J
,
Sen
S
.
MicroRNA as biomarkers and diagnostics
.
J Cell Physiol
2016
;
231
:
25
30
.

193

Wang
HT
,
Chang
YJ
,
Xu
LP
,
Liu
DH
,
Wang
Y
,
Liu
KY
,
Huang
XJ.
Ebmt risk score can predict the outcome of leukaemia after unmanipulated haploidentical blood and marrow transplantation
.
Bone Marrow Transplant
2014
;
49
:
927
933
.

194

Rodriguez-Rius
A
,
Lopez
S
,
Martinez-Perez
A
,
Souto
JC
,
Soria
JM
.
Identification of a plasma microRNA profile associated with venous thrombosis
.
Arterioscler Thromb Vasc Biol
2020
;
40
:
1392
1399
.

195

Wang
X
,
Sundquist
K
,
Elf
JL
,
Strandberg
K
,
Svensson
PJ
,
Hedelius
A
,
Diagnostic potential of plasma microRNA signatures in patients with deep-vein thrombosis
.
Thromb Haemost
2016
;
116
:
328
336
.

196

Wang
X
,
Sundquist
K
,
Svensson
PJ
,
Rastkhani
H
,
Palmér
K
,
Memon
AA
,
Association of recurrent venous thromboembolism and circulating microRNAs
.
Clin Epigenet
2019
;
11
:
28
.

197

Xiang
Q
,
Zhang
HX
,
Wang
Z
,
Liu
ZY
,
Xie
QF
,
Hu
K
,
Zhang
Z
,
Mu
GY
,
Ma
LY
,
Jiang
J
,
Cui
YM
.
The predictive value of circulating microRNAs for venous thromboembolism diagnosis: a systematic review and diagnostic meta-analysis
.
Thromb Res
2019
;
181
:
127
134
.

198

Lin
J
,
He
S
,
Sun
X
,
Franck
G
,
Deng
Y
,
Yang
D
,
MicroRNA-181b inhibits thrombin-mediated endothelial activation and arterial thrombosis by targeting caspase recruitment domain family member 10
.
FASEB J
2016
;
30
:
3216
3226
.

199

Xiao
J
,
Jing
ZC
,
Ellinor
PT
,
Liang
D
,
Zhang
H
,
Liu
Y
,
Chen
X
,
Pan
L
,
Lyon
R
,
Liu
Y
,
Peng
LY
,
Liang
X
,
Sun
Y
,
Popescu
LM
,
Condorelli
G
and
Chen
YH.
MicroRNA-134 as a potential plasma biomarker for the diagnosis of acute pulmonary embolism
.
J Transl Med
2011
;
9
:
159
.

200

Wang
Q
,
Ma
J
,
Jiang
Z
,
Wu
F
,
Ping
J
,
Ming
L
.
Diagnostic value of circulating microRNA-27a/b in patients with acute pulmonary embolism
.
Int Angiol
2018
;
37
:
19
25
.

201

Guo
L
,
Yang
Y
,
Liu
J
,
Wang
L
,
Li
J
,
Wang
Y
,
Liu
Y
,
Gu
S
,
Gan
H
,
Cai
J
,
Yuan
JX
,
Wang
J
,
Wang
C.
Differentially expressed plasma microRNAs and the potential regulatory function of let-7b in chronic thromboembolic pulmonary hypertension
.
PloS one
2014
;
9
:
e101055
.

202

Liu
T
,
Kang
J
,
Liu
F
.
Plasma levels of microRNA-221 (mir-221) are increased in patients with acute pulmonary embolism
.
Int Med J Exp Clin Res
2018
;
24
:
8621
8626
.

203

Zhou
X
,
Wen
W
,
Shan
X
,
Qian
J
,
Li
H
,
Jiang
T
,
Wang
W
,
Cheng
W
,
Wang
F
,
Qi
L
,
Ding
Y
,
Liu
P
,
Zhu
W
,
Chen
Y.
Mir-28-3p as a potential plasma marker in diagnosis of pulmonary embolism
.
Thromb Res
2016
;
138
:
91
95
.

204

Kessler
T
,
Erdmann
J
,
Vilne
B
,
Bruse
P
,
Kurowski
V
,
Diemert
P
,
Schunkert
H
,
Sager
HB.
Serum microRNA-1233 is a specific biomarker for diagnosing acute pulmonary embolism
.
J Transl Med
2016
;
14
:
120
.

205

Hosin
AA
,
Prasad
A
,
Viiri
LE
,
Davies
AH
,
Shalhoub
J
.
MicroRNAs in atherosclerosis
.
J Vasc Res
2014
;
51
:
338
349
.

206

Menghini
R
,
Stöhr
R
,
Federici
M
.
MicroRNAs in vascular aging and atherosclerosis
.
Ageing Res Rev
2014
;
17
:
68
78
.

207

Feinberg
MW
,
Moore
KJ
.
MicroRNA regulation of atherosclerosis
.
Circ Res
2016
;
118
:
703
720
.

208

Dolz
S
,
Górriz
D
,
Tembl
JI
,
Sánchez
D
,
Fortea
G
,
Parkhutik
V
,
Lago
,
A.
Circulating microRNAs as novel biomarkers of stenosis progression in asymptomatic carotid stenosis
.
Stroke
2017
;
48
:
10
16
.

209

Jansen
F
,
Schäfer
L
,
Wang
H
,
Schmitz
T
,
Flender
A
,
Schueler
R
,
Hammerstingl
C
,
Nickenig
G
,
Sinning
JM
,
Werner
N.
Kinetics of circulating microRNAs in response to cardiac stress in patients with coronary artery disease
.
J Am Heart Assoc
2017
;
6
:
e005270
.

210

Fujii
S
,
Sugiura
T
,
Dohi
Y
,
Ohte
N
.
MicroRNA in atherothromobosis: is it useful as a disease marker?
Thromb J
2016
;
14
:
21
.

211

Sun
X
,
Zhang
M
,
Sanagawa
A
,
Mori
C
,
Ito
S
,
Iwaki
S
,
Satoh
H
,
Fujii
S.
Circulating microRNA-126 in patients with coronary artery disease: correlation with LDL cholesterol
.
Thromb J
2012
;
10
:
16
.

212

Fichtlscherer
S
,
De Rosa
S
,
Fox
H
,
Schwietz
T
,
Fischer
A
,
Liebetrau
C
,
Weber
M
,
Hamm
CW
,
RDPC:\Users\uzoo\RDP6|xe
T
,
Müller-Ardogan
M
,
Bonauer
A
,
Zeiher
AM
,
Dimmeler
S.
Circulating microRNAs in patients with coronary artery disease
.
Circ Res
2010
;
107
:
677
684
.

213

Sørensen
SS
,
Nygaard
AB
,
Christensen
T
.
MiRNA expression profiles in cerebrospinal fluid and blood of patients with Alzheimer's disease and other types of dementia—an exploratory study
.
Transl Neurodeg
2016
;
5
:
6
.

214

Wang
Y
,
Ma
Z
,
Kan
P
,
Zhang
B
.
The diagnostic value of serum miRNA-221-3p, miRNA-382-5p, and miRNA-4271 in ischemic stroke
.
J Stroke Cerebrovasc Dis
2017
;
26
:
1055
1060
.

215

Chen
JQ
,
Papp
G
,
Póliska
S
,
Szabó
K
,
Tarr
T
,
Bálint
BL
,
Szodoray
P
,
Zeher
M.
MicroRNA expression profiles identify disease-specific alterations in systemic lupus erythematosus and primary sjögren's syndrome
.
PloS one
2017
;
12
:
e0174585
.

216

Kim
BS
,
Jung
JY
,
Jeon
JY
,
Kim
HA
,
Suh
CH
.
Circulating hsa-mir-30e-5p, hsa-mir-92a-3p, and hsa-mir-223-3p may be novel biomarkers in systemic lupus erythematosus
.
Hla
2016
;
88
:
187
193
.

217

Pérez-Sánchez
C
,
Aguirre
MA
,
Ruiz-Limón
P
,
Barbarroja
N
,
Jiménez-Gómez
Y
,
de la Rosa
IA
,
Rodriguez-Ariza
A
, Collantes-Esté
vez
E
,
Segui
P
,
Velasco
F
,
Cuadrado
MJ
,
Teruel
R
,
González-Conejero
R
, Martí
nez
C
,
López-Pedrera
C.
Atherothrombosis-associated microRNAs in antiphospholipid syndrome and systemic lupus erythematosus patients
.
Sci Rep
2016
;
6
:
31375
.

218

Sun
X
,
Icli
B
,
Wara
AK
,
Belkin
N
,
He
S
,
Kobzik
L
.
MicroRNA-181b regulates NF-κb-mediated vascular inflammation
.
J Clin Invest
2012
;
122
:
1973
1990
.

219

Witkowski
M
,
Witkowski
M
,
Saffarzadeh
M
,
Friebel
J
,
Tabaraie
T
,
Ta Bao
L
.
Vascular mir-181b controls tissue factor-dependent thrombogenicity and inflammation in type 2 diabetes
.
Cardiovasc Diabetol
2020
;
19
:
20
.

220

Hartmann
P
,
Zhou
Z
,
Natarelli
L
,
Wei
Y
,
Nazari-Jahantigh
M
,
Zhu
M
,
Grommes
J
,
Steffens
S
,
Weber
C
,
Schober
A.
Endothelial dicer promotes atherosclerosis and vascular inflammation by miRNA-103-mediated suppression of klf4
.
Nat Commun
2016
;
7
:
10521
.

221

Dahiya
N
,
Atreya
CD
.
Mir-181a reduces platelet activation via the inhibition of endogenous rap1b
.
MicroRNA
2020
;
9
:
240
246
.

222

Chen
Z
,
Nakajima
T
,
Tanabe
N
,
Hinohara
K
,
Sakao
S
,
Kasahara
Y
,
Tatsumi
K
,
Inoue
Y
,
Kimura
A.
Susceptibility to chronic thromboembolic pulmonary hypertension may be conferred by mir-759 via its targeted interaction with polymorphic fibrinogen alpha gene
.
Hum Genet
2010
;
128
:
443
452
.

223

Rosset
C
,
Vieira
IA
,
Salzano
FM
,
Bandinelli
E
.
A germline variant affects putative miRNA-binding sites at the F8 3'UTR and acts as a potential haemophilia a phenotype modifier in southern Brazilian patients
.
Haemophilia
2016
;
22
:
e327
e329
.

224

Voetsch
B
,
Loscalzo
J
.
Genetic determinants of arterial thrombosis
.
Arterioscler Thromb Vasc Biol
2004
;
24
:
216
229
.

225

Sabater-Lleal
M
,
Martinez-Perez
A
,
Buil
A
,
Folkersen
L
,
Souto
JC
,
Bruzelius
M
,
Borrell
M
,
Odeberg
J
,
Silveira
A
,
Eriksson
P
,
Almasy
L
,
Hamsten
A
,
Soria
JM.
A genome-wide association study identifies KNG1 as a genetic determinant of plasma factor XI level and activated partial thromboplastin time
.
Arterioscler Thromb Vasc Biol
2012
;
32
:
2008
2016
.

226

Sennblad
B
,
Basu
S
,
Mazur
J
,
Suchon
P
,
Martinez-Perez
A
,
van Hylckama Vlieg
A
,
Truong
V
,
Li
Y
, Gå
din
JR
,
Tang
W
,
Grossman
V
, de
Haan
HG
,
Handin
N
,
Silveira
A
,
Souto
JC
,
Franco-Cereceda
A
,
Morange
PE
,
Gagnon
F
,
Soria
JM
,
Eriksson
P
,
Hamsten
A
,
Maegdefessel
L
,
Rosendaal
FR
,
Wild
P
,
Folsom
AR
, Trégouë
t
DA
,
Sabater-Lleal
M.
Genome-wide association study with additional genetic and post-transcriptional analyses reveals novel regulators of plasma factor XI levels
.
Hum Mol Genet
2017
;
26
:
637
649
.

227

Edelstein
LC
,
McKenzie
SE
,
Shaw
C
,
Holinstat
MA
,
Kunapuli
SP
,
Bray
PF
.
MicroRNAs in platelet production and activation
.
J Thromb Haemost
2013
;
11
:
340
350
.

228

Dahiya
N
,
Sarachana
T
,
Vu
L
,
Becker
KG
,
Wood
WH
III
,
Zhang
Y
,
Atreya
CD.
Platelet microRNAs: an overview
.
Transfusion Med Rev
2015
;
29
:
215
219
.

229

Rowley
JW
,
Chappaz
S
,
Corduan
A
,
Chong
MM
,
Campbell
R
,
Khoury
A
,
Manne
BK
,
Wurtzel
JG
,
Michael
JV
,
Goldfinger
LE
,
Mumaw
MM
,
Nieman
MT
,
Kile
BT
,
Provost
P
,
Weyrich
AS.
Dicer1-mediated miRNA processing shapes the mRNA profile and function of murine platelets
.
Blood
2016
;
127
:
1743
1751
.

230

Elgheznawy
A
,
Shi
L
,
Hu
J
,
Wittig
I
,
Laban
H
,
Pircher
J
,
Mann
A
,
Provost
P
,
Randriamboavonjy
V
,
Fleming
I.
Dicer cleavage by calpain determines platelet microRNA levels and function in diabetes
.
Circ Res
2015
;
117
:
157
165
.

231

Sunderland
N
,
Skroblin
P
,
Barwari
T
,
Huntley
RP
,
Lu
R
,
Joshi
A
,
Lovering
RC
,
Mayr
M.
MicroRNA biomarkers and platelet reactivity: the clot thickens
.
Circ Res
2017
;
120
:
418
435
.

232

Tran
JQD
,
Pedersen
OH
,
Larsen
ML
,
Grove
EL
,
Kristensen
SD
,
Hvas
AM
,
Nissen
PH.
Platelet microRNA expression and association with platelet maturity and function in patients with essential thrombocythemia
.
Platelets
2020
;
31
:
365
372
.

233

Raitoharju
E
,
Lyytikäinen
LP
,
Levula
M
,
Oksala
N
,
Mennander
A
,
Tarkka
M
,
Klopp
N
,
Illig
T
, Kähö
nen
M
,
Karhunen
PJ
,
Laaksonen
R
and
Lehtimäki
T.
Mir-21, mir-210, mir-34a, and mir-146a/b are up-regulated in human atherosclerotic plaques in the tampere vascular study
.
Atherosclerosis
2011
;
219
:
211
217
.

234

Rupaimoole
R
,
Slack
FJ
.
MicroRNA therapeutics: towards a new era for the management of cancer and other diseases
.
Nat Rev Drug Discov
2017
;
16
:
203
.

235

Lu
D
,
Thum
T
.
RNA-based diagnostic and therapeutic strategies for cardiovascular disease
.
Nature reviews. Cardiol
2019
;
16
:
661
674
.

236

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
2019
;
123
:
35
41
.

237

Meng
Q
,
Wang
W
,
Yu
X
,
Li
W
,
Kong
L
,
Qian
A
,
Li
C
,
Li
X.
Upregulation of microRNA-126 contributes to endothelial progenitor cell function in deep vein thrombosis via its target pik3r2
.
J Cell Biochem
2015
;
116
:
1613
1623
.

238

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
2019
;
23
:
8493
8504
.

239

Hentze
MW
,
Castello
A
,
Schwarzl
T
,
Preiss
T
.
A brave new world of RNA-binding proteins
.
Nat Rev Mol Cell Biol
2018
;
19
:
327
341
.

240

Subramaniam
S
,
Kanse
SM
,
Kothari
H
,
Reinhardt
C
,
Fletcher
C
.
Post-transcriptional, post-translational and pharmacological regulation of tissue factor pathway inhibitor
.
Blood Coag Fibrinol
2018
;
29
:
668
682
.

241

Goering
R
,
Engel
KL
,
Gillen
AE
,
Fong
N
,
Bentley
DL
,
Taliaferro
JM
.
Labrat reveals association of alternative polyadenylation with transcript localization, RNA binding protein expression, transcription speed, and cancer survival
.
BMC Genomics
2021
;
22
:
476
476
.

242

Soukarieh
O
,
Meguerditchian
C
,
Proust
C
,
Aïssi
D
,
Eyries
M
,
Goyenvalle
A
,
Trégouët
DA.
Common and rare 5'UTR variants altering upstream open Reading frames in cardiovascular genomics
.
Front Cardiovasc Med
2022
;
9
:
841032
.

243

Crossley
MP
,
Bocek
M
,
Cimprich
KA
.
R-loops as cellular regulators and genomic threats
.
Mol Cell
2019
;
73
:
398
411
.

244

García-Muse
T
,
Aguilera
A
.
R loops: from physiological to pathological roles
.
Cell
2019
;
179
:
604
618
.

245

Spada
S
,
Luke
B
,
Danckwardt
S
.
The bidirectional link between RNA cleavage and polyadenylation and genome stability: recent insights from a systematic screen
.
Front Genet
2022
;
13
:
854907
.

246

Khan
ES
,
Danckwardt
S
.
Pathophysiological role and diagnostic potential of R-loops in cancer and beyond
.
Genes (Basel)
2022
;
13
:
2181
.

247

Lu
Y-C
,
Chang
S-H
,
Hafner
M
,
Li
X
,
Tuschl
T
,
Elemento
O
,
Hla
T.
Elavl1 modulates transcriptome-wide miRNA binding in murine macrophages
.
Cell Rep
2014
;
9
:
2330
2343
.

248

Jens
M
,
Rajewsky
N
.
Competition between target sites of regulators shapes post-transcriptional gene regulation
.
Nat Rev Genet
2014
;
16
:
113
.

249

Luo
J
,
Xu
T
,
Sun
K
.
N6-methyladenosine RNA modification in inflammation: roles, mechanisms, and applications
.
Front Cell Dev Biol
2021
;
9
:
670711
.

250

Velázquez-Cruz
A
,
Baños-Jaime
B
,
Díaz-Quintana
A
,
De la Rosa
MA
,
Díaz-Moreno
I
.
Post-translational control of RNA-binding proteins and disease-related dysregulation
.
Front Mol Biosci
2021
;
8
:
658852
658852
.

251

Jiang
X
,
Liu
B
,
Nie
Z
,
Duan
L
,
Xiong
Q
,
Jin
Z
,
Yang
C
,
Chen
Y.
The role of m6a modification in the biological functions and diseases
.
Signal Transduct Target Ther
2021
;
6
:
74
.

252

Boulias
K
,
Greer
EL
.
Biological roles of adenine methylation in RNA
.
Nat Rev Genet
2022
;
24
:
143
.

253

Shen
ZJ
,
Han
YC
,
Nie
MW
,
Wang
YN
,
Xiang
RL
,
Xie
HZ
.
Genome-wide identification of altered RNA m(6)a profiles in vascular tissue of septic rats
.
Aging
2021
;
13
:
21610
21627
.

254

Zhao
K
,
Yang
CX
,
Li
P
,
Sun
W
,
Kong
XQ
.
Epigenetic role of n6-methyladenosine (m6a) RNA methylation in the cardiovascular system
.
J Zheji Univ. Science. B
2020
;
21
:
509
523
.

255

Qin
Y
,
Li
L
,
Luo
E
,
Hou
J
,
Yan
G
,
Wang
D
,
Qiao
Y
,
Tang
C.
Role of m6a RNA methylation in cardiovascular disease
.
Int J Mol Med
2020
;
46
:
1958
1972
.

256

Zannella
C
,
Rinaldi
L
,
Boccia
G
,
Chianese
A
,
Sasso
FC
,
De Caro
F
,
Franci
G
,
Galdiero
M.
Regulation of m6a methylation as a new therapeutic option against COVID-19
.
Pharmaceuticals
2021
;
14
:
1135
.

257

Roundtree
IA
,
Evans
ME
,
Pan
T
,
He
C
.
Dynamic RNA modifications in gene expression regulation
.
Cell
2017
;
169
:
1187
1200
.

258

Berretta
J
,
Morillon
A
.
Pervasive transcription constitutes a new level of eukaryotic genome regulation
.
EMBO Rep
2009
;
10
:
973
982
.

259

Consortium
EP
.
An integrated encyclopedia of DNA elements in the human genome
.
Nature
2012
;
489
:
57
74
.

260

Esteller
M
.
Non-coding RNAs in human disease
.
Nat Rev Genet
2011
;
12
:
861
874
.

261

Mercer
TR
,
Dinger
ME
,
Mattick
JS
.
Long non-coding RNAs: insights into functions
.
Nat Rev Genet
2009
;
10
:
155
159
.

262

Pan
JX
.
LncRNA h19 promotes atherosclerosis by regulating mapk and NF-kb signaling pathway
.
Eur Rev Med Pharmacol Sci
2017
;
21
:
322
328
.

263

Wang
J
,
Zhao
H
,
Fan
Z
,
Li
G
,
Ma
Q
,
Tao
Z
,
Wang
R
,
Feng
J
,
Luo
Y.
Long noncoding RNA h19 promotes neuroinflammation in ischemic stroke by driving histone deacetylase 1-dependent M1 microglial polarization
.
Stroke
2017
;
48
:
2211
2221
.

264

Broadbent
HM
,
Peden
JF
,
Lorkowski
S
,
Goel
A
,
Ongen
H
,
Green
F
,
Clarke
R
,
Collins
R
,
Franzosi
MG
,
Tognoni
G
,
Seedorf
U
,
Rust
S
,
Eriksson
P
,
Hamsten
A
,
Farrall
M
,
Watkins
H
.
Susceptibility to coronary artery disease and diabetes is encoded by distinct, tightly linked snps in the ANRIL locus on chromosome 9p
.
Hum Mol Genet
2008
;
17
:
806
814
.

265

Holdt
LM
,
Beutner
F
,
Scholz
M
,
Gielen
S
,
Gäbel
G
,
Bergert
H
,
Schuler
G
,
Thiery
J
,
Teupser
D.
ANRIL Expression is associated with atherosclerosis risk at chromosome 9p21
.
Arterioscler Thromb Vasc Biol
2010
;
30
:
620
627
.

266

Zhang
Y
,
Zhang
Y
.
LncRNA ZFAS1 improves neuronal injury and inhibits inflammation, oxidative stress, and apoptosis by sponging mir-582 and upregulating nos3 expression in cerebral ischemia/reperfusion injury
.
Inflammation
2020
;
43
:
1337
1350
.

267

Lindström
S
,
Wang
L
,
Smith
EN
,
Gordon
W
,
van Hylckama Vlieg
A
,
de Andrade
M
,
Genomic and transcriptomic association studies identify 16 novel susceptibility loci for venous thromboembolism
.
Blood
2019
;
134
:
1645
1657
.

268

Sun
Y
,
Liu
R
,
Xia
X
,
Xing
L
,
Jiang
J
,
Bian
W
,
Zhang
W
,
Wang
C
,
Zhang
C.
Large-scale profiling on lncRNAs in human platelets: correlation with platelet reactivity
.
Cells
2022
;
11
:
2256
.

269

Gutmann
C
,
Joshi
A
,
Zampetaki
A
,
Mayr
M
.
The landscape of coding and noncoding RNAs in platelets
.
Antiox Redox Signal
2021
;
34
:
1200
1216
.

270

He
AT
,
Liu
J
,
Li
F
,
Yang
BB
.
Targeting circular RNAs as a therapeutic approach: current strategies and challenges
.
Signal Trans Targ Ther
2021
;
6
:
185
.

271

Memczak
S
,
Jens
M
,
Elefsinioti
A
,
Torti
F
,
Krueger
J
,
Rybak
A
,
Maier
L
,
Mackowiak
SD
,
Gregersen
LH
,
Munschauer
M
,
Loewer
A
,
Ziebold
U
,
Landthaler
M
,
Kocks
C
,
le Noble
F
and
Rajewsky
N.
Circular RNAs are a large class of animal RNAs with regulatory potency
.
Nature
2013
;
495
:
333
338
.

272

Kristensen
LS
,
Andersen
MS
,
Stagsted
LVW
,
Ebbesen
KK
,
Hansen
TB
,
Kjems
J
.
The biogenesis, biology and characterization of circular RNAs
.
Nat Rev Genet
2019
;
20
:
675
691
.

273

Saaoud
F
,
Drummer
IVC
,
Shao
Y
,
Sun
Y
,
Lu
Y
,
Xu
K
,
Ni
D
,
Jiang
X
,
Wang
H
,
Yang
X.
Circular RNAs are a novel type of non-coding RNAs in ROS regulation, cardiovascular metabolic inflammations and cancers
.
Pharmacol Ther
2021
;
220
:
107715
.

274

Holdt
LM
,
Stahringer
A
,
Sass
K
,
Pichler
G
,
Kulak
NA
,
Wilfert
W
,
Kohlmaier
A
,
Herbst
A
,
Northoff
BH
,
Nicolaou
A
, Gä
bel
G
,
Beutner
F
,
Scholz
M
,
Thiery
J
,
Musunuru
K
,
Krohn
K
,
Mann
M
,
Teupser
D
.
Circular non-coding RNA ANRIL modulates ribosomal RNA maturation and atherosclerosis in humans
.
Nat Commun
2016
;
7
:
12429
.

275

Chen
W
,
Wang
H
,
Feng
J
,
Chen
L
.
Overexpression of circRNA circuck2 attenuates cell apoptosis in cerebral ischemia-reperfusion injury via mir-125b-5p/gdf11 signaling
.
Mol Ther Nucl Acids
2020
;
22
:
673
683
.

276

Lu
D
,
Ho
ES
,
Mai
H
,
Zang
J
,
Liu
Y
,
Li
Y
,
Yang
B
,
Ding
Y
,
Tsang
CK
,
Xu
A.
Identification of blood circular RNAs as potential biomarkers for acute ischemic stroke
.
Front Neurosci
2020
;
14
:
81
81
.

277

Ostolaza
A
,
Blanco-Luquin
I
,
Urdánoz-Casado
A
,
Rubio
I
,
Labarga
A
,
Zandio
B
, Roldá
n
M
,
Martínez-Cascales
J
,
Mayor
S
,
Herrera
M
,
Aymerich
N
,
Gallego
J
, Muñ
oz
R
,
Mendioroz
M.
Circular RNA expression profile in blood according to ischemic stroke etiology
.
Cell Biosci
2020
;
10
:
34
.

278

Derti
A
,
Garrett-Engele
P
,
Macisaac
KD
,
Stevens
RC
,
Sriram
S
,
Chen
R
,
Rohl
CA
,
Johnson
JM
and
Babak
T.
A quantitative atlas of polyadenylation in five mammals
.
Genome Res
2012
;
22
:
1173
1183
.

279

Ogorodnikov
A
,
Kargapolova
Y
,
Danckwardt
S
.
Processing and transcriptome expansion at the mRNA 3′ end in health and disease: finding the right end
.
Eur J Physiol
2016
;
468
:
993
1012
.

280

Takagaki
Y
,
Seipelt
RL
,
Peterson
ML
,
Manley
JL
.
The polyadenylation factor cstf-64 regulates alternative processing of IgM heavy chain pre-mRNA during B cell differentiation
.
Cell
1996
;
87
:
941
952
.

281

Gruber
AR
,
Martin
G
,
Müller
P
,
Schmidt
A
,
Gruber
AJ
,
Gumienny
R
,
Mittal
N
,
Jayachandran
R
,
Pieters
J
,
Keller
W
, van
Nimwegen
E
,
Zavolan
M.
Global 3′ UTR shortening has a limited effect on protein abundance in proliferating T cells
.
Nat Commun
2014
;
5
:
5465
.

282

An
S
,
Li
Y
,
Lin
Y
,
Chu
J
,
Su
J
,
Chen
Q
,
Wang
H
,
Pan
P
,
Zheng
R
,
Li
J
,
Jiang
J
,
Ye
L
and
Liang
H.
Genome-wide profiling reveals alternative polyadenylation of innate immune-related mRNA in patients with COVID-19
.
Front Immunol
2021
;
12
:
756288
.

283

Esmon
CT
.
The protein c pathway
.
Chest
2003
;
124
:
26s
32s
.

284

Rein-Smith
CM
,
Anderson
NW
,
Farrell
DH
.
Differential regulation of fibrinogen γ chain splice isoforms by interleukin-6
.
Thromb Res
2013
;
131
:
89
93
.

285

Mosesson
MW
.
Update on antithrombin I
.
Thromb Haemost
2007
;
98
:
105
108
.

286

Mosesson
MW
,
Cooley
BC
,
Hernandez
I
,
Diorio
JP
,
Weiler
H
.
Thrombosis risk modification in transgenic mice containing the human fibrinogen thrombin-binding gamma’ chain sequence
.
J Thromb Haemost
2009
;
7
:
102
110
.

287

Appiah
D
,
Schreiner
PJ
,
MacLehose
RF
,
Folsom
AR
.
Association of plasma γ’ fibrinogen with incident cardiovascular disease: the atherosclerosis risk in communities (aric) study
.
Arterioscler Thromb Vasc Biol
2015
;
35
:
2700
2706
.

288

Maners
J
,
Gill
D
,
Pankratz
N
,
Laffan
MA
,
Wolberg
AS
,
de Maat
MPM
,
Ligthart
S
,
Tang
W
,
Ward-Caviness
CK
,
Fornage
M
,
Debette
S
,
Dichgans
M
,
McKnight
B
,
Boerwinkle
E
,
Smith
NL
,
Morrison
AC
,
Dehghan
A
,
de Vries
PS.
A Mendelian randomization of γ’ and total fibrinogen levels in relation to venous thromboembolism and ischemic stroke
.
Blood
2020
;
136
:
3062
3069
.

289

Wolberg
AS
.
Primed to understand fibrinogen in cardiovascular disease
.
Arterioscler Thromb Vasc Biol
2016
;
36
:
4
6
.

290

Thibord
F
,
Perret
C
,
Roux
M
,
Suchon
P
,
Germain
M
,
Deleuze
JF
,
Morange
PE
,
Trégouët
DA.
Optimir, a novel algorithm for integrating available genome-wide genotype data into miRNA sequence alignment analysis
.
RNA
2019
;
25
:
657
668
.

291

Vickers
TA
,
Wyatt
JR
,
Burckin
T
,
Bennett
CF
,
Freier
SM
.
Fully modified 2’ MOE oligonucleotides redirect polyadenylation
.
Nucl Acids Res
2001
;
29
:
1293
1299
.

292

Kole
R
,
Krainer
AR
,
Altman
S
.
RNA therapeutics: beyond RNA interference and antisense oligonucleotides
.
Nat Rev Drug Discov
2012
;
11
:
125
140
.

293

Mohibi
S
,
Chen
X
,
Zhang
J
.
Cancer the ‘RBP'eutics—RNA-binding proteins as therapeutic targets for cancer
.
Pharmacol Ther
2019
;
203
:
107390
.

294

Daka
A
,
Peer
D
.
RNAi-based nanomedicines for targeted personalized therapy
.
Adv Drug Deliv Rev
2012
;
64
:
1508
1521
.

295

Henry
SP
,
Narayanan
P
,
Shen
L
,
Bhanot
S
,
Younis
HS
,
Burel
SA
.
Assessment of the effects of 2'-methoxyethyl antisense oligonucleotides on platelet count in cynomolgus nonhuman primates
.
Nucl Acid Therap
2017
;
27
:
197
208
.

296

Narayanan
P
,
Shen
L
,
Curtis
BR
,
Bourdon
MA
,
Nolan
JP
,
Gupta
S
,
Hoffmaster
C
,
Zhou
F
,
Christian
B
,
Schaubhut
JL
,
Greenlee
S
,
Burel
SA
,
Witztum
JL
,
Engelhardt
JA
,
Henry
SP.
Investigation into the mechanism(s) that leads to platelet decreases in cynomolgus monkeys during administration of ISIS 104838, a 2'-moe-modified antisense oligonucleotide
.
Toxicol Sci
2018
;
164
:
613
626
.

297

Beierlein
JM
,
McNamee
LM
,
Ledley
FD
.
As technologies for nucleotide therapeutics mature, products emerge
.
Mol Ther Nucleic Acids
2017
;
9
:
379
386
.

298

Haley
B
,
Zamore
PD
.
Kinetic analysis of the RNAi enzyme complex
.
Nat Struct Mol Biol
2004
;
11
:
599
606
.

299

Fabian
MR
,
Sonenberg
N
.
The mechanics of miRNA-mediated gene silencing: a look under the hood of miRisc
.
Nat Struct Mol Biol
2012
;
19
:
586
593
.

300

Jonas
S
,
Izaurralde
E
.
Towards a molecular understanding of microRNA-mediated gene silencing
.
Nat Rev Genet
2015
;
16
:
421
433
.

301

Selbach
M
,
Schwanhausser
B
,
Thierfelder
N
,
Fang
Z
,
Khanin
R
,
Rajewsky
N
.
Widespread changes in protein synthesis induced by microRNAs
.
Nature
2008
;
455
:
58
63
.

302

Nielsen
CB
,
Shomron
N
,
Sandberg
R
,
Hornstein
E
,
Kitzman
J
,
Burge
CB
.
Determinants of targeting by endogenous and exogenous microRNAs and siRNAs
.
RNA
2007
;
13
:
1894
1910
.

303

Titze-de-Almeida
R
,
David
C
,
Titze-de-Almeida
SS
.
The race of 10 synthetic RNAi-based drugs to the pharmaceutical market
.
Pharm Res
2017
;
34
:
1339
1363
.

304

Yu
AM
,
Jian
C
,
Yu
AH
,
Tu
MJ
.
RNA Therapy: are we using the right molecules?
Pharmacol Ther
2019
;
196
:
91
104
.

305

Zhao
X
,
Pan
F
,
Holt
CM
,
Lewis
AL
,
Lu
JR
.
Controlled delivery of antisense oligonucleotides: A brief review of current strategies
.
Exp Opin Drug Deliv
2009
;
6
:
673
686
.

Author notes

Conflict of interest: None declared.

This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (https://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact [email protected]