Skip to main content
Erschienen in: Cardiovascular Diabetology 1/2023

Open Access 01.12.2023 | Review

Cardiovascular complications of diabetes: role of non-coding RNAs in the crosstalk between immune and cardiovascular systems

verfasst von: Gaia Spinetti, Martina Mutoli, Simona Greco, Federica Riccio, Soumaya Ben-Aicha, Franziska Kenneweg, Amela Jusic, David de Gonzalo-Calvo, Anne Yaël Nossent, Susana Novella, Georgios Kararigas, Thomas Thum, Costanza Emanueli, Yvan Devaux, Fabio Martelli

Erschienen in: Cardiovascular Diabetology | Ausgabe 1/2023

Abstract

Diabetes mellitus, a group of metabolic disorders characterized by high levels of blood glucose caused by insulin defect or impairment, is a major risk factor for cardiovascular diseases and related mortality. Patients with diabetes experience a state of chronic or intermittent hyperglycemia resulting in damage to the vasculature, leading to micro- and macro-vascular diseases. These conditions are associated with low-grade chronic inflammation and accelerated atherosclerosis. Several classes of leukocytes have been implicated in diabetic cardiovascular impairment. Although the molecular pathways through which diabetes elicits an inflammatory response have attracted significant attention, how they contribute to altering cardiovascular homeostasis is still incompletely understood. In this respect, non-coding RNAs (ncRNAs) are a still largely under-investigated class of transcripts that may play a fundamental role. This review article gathers the current knowledge on the function of ncRNAs in the crosstalk between immune and cardiovascular cells in the context of diabetic complications, highlighting the influence of biological sex in such mechanisms and exploring the potential role of ncRNAs as biomarkers and targets for treatments. The discussion closes by offering an overview of the ncRNAs involved in the increased cardiovascular risk suffered by patients with diabetes facing Sars-CoV-2 infection.

Graphical Abstract

Hinweise
Gaia Spinetti and Martina Mutoli contributed equally.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Diabetes mellitus (DM) is an important cause of functional impairment, morbidity, and mortality, representing a heavy economic burden for global healthcare systems. About 537 million adults (20–79 years) were estimated to be living with DM worldwide in 2021, and most of them were affected by Type 2 DM (T2DM). This number has been predicted to rise to 643 million by 2030 and to 783 million by 2045 [1]. Notably, DM-related mortality and reduction in life expectancy are greater in women than men [2, 3]. DM is characterized by high levels of blood glucose caused by impairment in the insulin production and/or signalling. Hyperglycemia, directly and indirectly, alters cells of the cardiovascular system leading to microvascular and macrovascular diseases in a sex-dependent manner. In particular, microvascular complications include diabetic retinopathy and nephropathy while macrovascular complications mainly refer to accelerated atherosclerosis in different areas including cerebrovascular atherosclerotic disease manifesting as stroke, coronary artery disease (CAD) that can result in an acute myocardial infarction (MI) and peripheral arterial disease (PAD) that can evolve into critical limb ischemia (CLI), resulting in wound healing inhibition and diabetic foot ulcers. Other macrovascular complications refer to cardiac dysfunction, including cardiomyopathy and heart failure (HF) [49]. A state of low-grade inflammation characterizes both DM and atherosclerosis together with a decrease/impairment in cardiovascular protective bone marrow (BM)-derived hematopoietic stem/progenitor cells (HSPCs) and a pro-inflammatory shift of mature immune cells, resulting in the impairment of the repair processes needed to maintain tissue homeostasis [1014]. Different classes of leukocytes belonging to both innate and adaptive immunity, e.g. monocytes, macrophages, dendritic cells (DCs), T-lymphocytes, B-lymphocytes, and natural killer (NK) cells regulate the DM-associated inflammatory response, in a time- and tissue-specific manner [15]. However, the molecular pathways through which DM elicits an immune response and how this contributes to altering cardiovascular homeostasis have yet to be fully characterized. Hence, understanding the complex mechanisms of communication between immune and cardiovascular cells is pivotal for the identification of novel therapeutic targets and potential biomarkers for disease prognosis. In this respect, non-coding RNAs (ncRNAs) represent a still insufficiently studied class of transcripts that may play a fundamental role. Indeed, ncRNAs are major regulators and players of both cardiovascular and immune functions in DM and its related cardiovascular complications (Fig. 1) [1618]. Here we discuss the roles of ncRNAs in the crosstalks between immune and cardiovascular cells underlying the onset and perpetuation of DM and its related cardiovascular diseases (CVDs), highlighting the implication of aging and biological sex in such a contest. Furthermore, selected, key DM-modulated peripheral blood (PB) ncRNAs are suggested as early biomarkers of disease development and outcome, and, ultimately, as new therapeutic tools. The discussion closes by offering an overview of the ncRNAs involved in the increased cardiovascular risk suffered by patients with diabetes facing Sars-CoV-2 infection.
To identify relevant reports for this narrative review, we searched on PubMed the literature published in English up to March 2023, using as query: (“non-coding RNA” or “microRNA” or “long non-coding RNA”) and (“inflammation” or “immune system”) and (“cardiovascular”) and (“diabetes” or “diabetic”). We then excluded articles not dealing with DM, coronavirus disease 2019 (COVID-19), or immune and cardiovascular cells, and selected those providing the most robust information in terms of crosstalk between the two systems. Other articles were selected from the reference list of the retrieved publications.

The ncRNAs world: definition and classes

The vast majority of the human genome is transcribed into RNA that is not translated into proteins. It is also well recognized that these ncRNAs play a central role in human physiology and pathology. However, so far, we have only started to scratch the surface of the collective function of the non-coding part of the genome. Novel types of RNA are still being discovered and novel functions identified. A very simplistic, operational, classification subdivides ncRNAs according to their size: ncRNAs shorter than 200 nucleotides are called small ncRNAs (sncRNAs) (Table 1), ncRNAs longer than 200 nucleotides are called long ncRNAs (lncRNAs) (Table 2) [19, 20]. Of all ncRNAs, microRNAs (miRNAs) are probably the most studied and best understood so far. miRNAs are sncRNAs, with a length of approximately 22 nucleotides. miRNAs are known to bind preferentially to the 3’ untranslated region of target messenger RNAs (mRNAs), thereby inducing either mRNA degradation or repression of translation, via the RNA-induced silencing complex. In general, a single miRNA has only a slight effect on the translation of an individual target, but as single miRNAs can target many different mRNAs, often in the same pathway or gene program, the effect of a single miRNA on cell and tissue function is significant. One mRNA can be regulated by more than one miRNA, which leads to the creation of highly-interconnected large signaling networks [2123]. Moreover, variations in miRNA biogenesis generating alternative miRNA isoforms termed isomiRNAs and post-transcriptional modifications like A-to-I editing and methylation, which are altered under pathological conditions, form a new and exciting area in ncRNA research in the cardiovascular field [2426].
Table 1
Small non-coding RNA classes
Reference
Type of small non-coding RNA
Length
Reported functions
[22]
miRNA
~ 22 nucleotides
- mRNA degradation
- translational repression
[27]
piRNA
~ 24–31 nucleotides
- transposon repression
[2831, 332]
snoRNA
60–200 nucleotides
- pseudouridylation or 2’O-ribose methylation of rRNAs, snRNAs, and tRNAs
- mRNA methylation
- alternative splicing and processing
- mRNA stabilization
[37, 38]
tRF
16–35 nucleotides
- miRNA-like mRNA translational repression
- enhance translation
Table 2
Long non-coding RNA classes
Reference
Type of long non-coding RNA
Length
Reported functions
[41, 42]
lncRNA
> 200 nucleotides
- repress/activate transcription
- repress/enhance translation
- impact on RNA stability
[4345]
circRNA
> 200 nucleotides
- sponge miRNAs
- protein binding
- translation into polypeptides
Much less is known about other types of sncRNAs, like piwi-interacting RNAs (piRNAs), small nucleolar RNAs (snoRNAs), and transfer RNA (tRNA)-derived fragments (tRFs). piRNAs, typically 24–31 nucleotides long, are mostly expressed in germline cells, where they target and repress transposons. However, piRNAs are also found in somatic cells, where their function remains to be elucidated [27].
snoRNAs are principally part of the ‘housekeeping’ of the cellular machinery and consist of 60–200 nucleotides. They play a crucial role in processing and stabilizing ribosomal RNAs (rRNAs) and small nuclear RNAs (snRNAs), tRNAs via guiding important base modifications; H/ACA box snoRNAs guide pseudouridylation and C/D box snoRNAs guide 2’O-ribose methylation. However, many snoRNAs have no known rRNA or snRNA targets, which are referred to as ‘orphan’ snoRNAs and their function is mostly unknown [28, 29]. In addition, certain snoRNAs can also target and regulate mRNAs. For certain orphan C/D box snoRNAs, it has been shown that they can target mRNAs and guide mRNA methylation, direct 3’-end processing, alternative splicing, and mRNA stability [30, 31]. Of relevance, several snoRNAs gene clusters have been associated with both DM and CVDs [3234].
As the name implies, tRFs are derived from either pre-tRNAs or mature tRNAs, and it has been only recently accepted that they are more than merely tRNA degradation products [35, 36]. Their length ranges from 16 to 35 nucleotides and they either enhance translation or have miRNA-like functions such as mRNA translational repression, but many other mechanisms of action likely remain to be uncovered [37, 38]. Importantly, tRFs are associated with cardiovascular events, such as stroke [39].
lncRNA is the collective term for all ncRNAs longer than 200 nucleotides. Roughly, lncRNAs are subdivided into two classes, namely linear lncRNAs and circular RNAs (circRNAs). The latter is produced by back-splicing of precursor coding and lncRNA transcripts [40]. A broad range of functions and mechanisms of action has been described for lncRNAs. Linear lncRNAs can act as molecular scaffolds or decoys, for example, having an effect on transcription (activating or repressing it), modifying chromatin structure, or guiding pre-mRNA processing, but they can also influence protein translation (enhancing or repressing it), or affect RNA turnover rates [41, 42]. CircRNAs are mostly known for their ability to ‘sponge’ miRNAs but may have other important functions such as interaction with RNA-binding proteins, directing their interaction with downstream target proteins or mRNAs. Moreover, a small subset of circRNAs is translated [4346]. lncRNAs are involved in all forms of human physiology and pathology [47], but due to lower conservation, their study using animal models is methodologically more challenging.

ncRNAs in the immune-cardiovascular cell interaction

Innate immune cells, i.e. neutrophils, monocytes/macrophages, NK, and DCs, are involved in the early steps of the inflammatory response and are associated with atherosclerotic diseases [13, 48]. In recent years, ncRNAs are emerging as activators or inhibitors of the innate inflammatory response due to their role in the regulation of inflammatory gene expression. Furthermore, they are involved in the differentiation and function of innate immune cells [49, 50]. Their altered expression in innate immune cells is linked to diabetic complications. These disorders display a chronic activation of pro-inflammatory pathways, as well as abnormal activation of innate and adaptive immunity. The diabetic environment strongly facilitates chronic inflammation of the endothelium, as it is characterized by high levels of low-density lipoproteins (LDLs), glucose, advanced glycation end-products, advanced lipoxidation end-products, and reactive oxygen species (ROS) [5155]. Long-term exposure to the diabetic environment can damage the vascular endothelium by inducing post-translational modifications of biological molecules, which damage the endothelium and trigger the innate and adaptive immune system. Leucocytes contribute to the pro-oxidant and pro-inflammatory milieu by continuously infiltrating the endothelium, where they accumulate and are activated in an uncontrolled manner, producing pro-inflammatory molecules [56, 57]. Moreover, many studies reported an increased number of circulating leucocytes in T2DM [58, 59]. In the context of atherosclerosis, monocytes differentiate into macrophages, which internalize LDL thus forming foam cells. Both monocytes and macrophages are characterized by a pro-inflammatory phenotype in patients with DM [60, 61]. Inflammatory signaling causes an additional release of cytokines, vasoactive molecules, and proteases from macrophages and foam cells, generating an additional T-helper-1 (Th1) pro-inflammatory response with the release of pro-inflammatory cytokines, which contributes to local inflammation and plaque growth. Th1 cells have a pro-atherosclerotic function, unlike regulatory T cells (T-regs), which have a protective effect [62]. Later phases of the plaque’s development include local proteolysis, plaque rupture, and ultimately thrombus formation, all of which can result in life-threatening events, such as MI or stroke [8, 60]. It has been shown that the vascular wall of humans and mice include high levels of DCs [63, 64]. It is known that the DC subset can play pro- and anti-atherogenic roles at all phases of atherogenesis through their wide range of functions, including lipid uptake, efferocytosis, inflammation resolution, and antigen presentation [65]. Also, neutrophils contribute to the advancement of atherosclerosis by releasing neutrophils extracellular traps (NETs), which have been found in atherosclerotic plaques [66, 67]. NETs have been linked to plasmacytoid DC-driven autoimmune activation and the production of anti-double strand DNA antibodies, both of which have the potential to significantly worsen the development of atherosclerosis lesions [68, 69]. Recent evidence supports the role also for NK in atherosclerosis, as recently discussed [13]. These findings reveal how the diabetic milieu activates the immune system resulting in chronic inflammation and ultimately leading to the onset of atherosclerosis. How ncRNAs are intertwisted in these processes is the object of the following sections.

Hematopoietic stem and progenitor cells

HSPCs contribute to the homeostasis of the cardiovascular system [70], especially the subpopulation of CD34 + cells [71]. Indeed, CD34 + HSPCs contribute to vessel repair and their function is known to be altered in DM [72]. Both mobilization of CD34 + HSPCs from BM and their angiogenic function are impaired in patients with DM, a condition often referred to as ‘mobilopathy’ at least in part associated with the deregulation of ncRNAs [10, 70, 7376]. BM CD34 + HSPCs show an altered expression of miRNAs in DM, including miR-155 and miR-21 [14, 75] associated with lower cell survival and induction of apoptosis. A recent study from our group showed that, compared to healthy controls, BM-derived CD34 + HSPCs bear lower levels of miR-21 and higher levels of one of miR-21 targets, the tumor suppressor programmed cell death 4. Of note, these changes can be transferred to endothelial cells (ECs) via taurine upregulated gene 1, a lncRNA responsible for the sequestration of miR-21, delivering a death signal to ECs [77]. In addition, serum and PB circulating HSPCs from diabetic patients with CLI contain increased levels of miR-15a and miR-16 with consequential impairment in HSPCs migration and adhesion capabilities [78]. On the other hand, miR-210 overexpression improves HSPC hypoxia-induced, SDF-1-driven migration [79]. Interestingly, restoring the expression levels of miR-92a in CD34 + cells from patients with diabetic retinopathy has been associated with an anti-inflammatory effect [80]. Lastly, angiogenic miR-126 is modulated in DM in CD34 + cells and its transfer can impact the endothelium [81].

Neutrophils

Neutrophils are continuously recruited to chronic inflammation sites in patients with DM, promoting inflammation through an excessive NET formation and impaired apoptosis which contribute to accelerated atherosclerosis and delayed wound healing respectively [8286]. miRNAs have emerged as important regulators of inflammation-modulating neutrophils functions [87, 88]. In particular, miR146a and miR-129-2-3p are two inflammatory miRNAs in neutrophils involved in DM complications. Ana BA et al. showed that miR-146a deficiency promotes NET formations in an atherosclerosis mouse model (Fig. 2a) [89]. Previous studies demonstrated that miR-146a also has an anti-inflammatory role in monocytes, by downregulating the production of pro-inflammatory cytokines [90]. Umehara et al. observed that diabetic mice display an increase of wound neutrophils in which the downregulation of miR-129-2-3p contributes to prolonged inflammation and impaired apoptosis through upregulation of Caspase (CASP)-6 and C-C chemokine receptor type 2, thus delaying diabetic wound healing (Fig. 2b). Accordingly, overexpression of miR-129-2-3p in the skin wound site of diabetic mice enhanced the wound healing process [86]. These observations point to miRNAs as crucial elements regulating the role of neutrophils in DM complications.

Mono/macrophages

Like neutrophils, monocytes\macrophages are excessively recruited into chronic inflammation sites in patients with DM [91]. Two important lncRNAs, anti-inflammatory Diabetes Regulated anti-inflammatory RNA (DRAIR) and pro-inflammatory dynamin 3 opposite strand (Dnm3os) are involved in DM. DRAIR was found to be downregulated in monocytes of patients with DM compared with non-DM controls (Fig. 2c) [92]. DRAIR modulation was confirmed in vitro by culturing THP1 monocytes in normal glucose versus high glucose conditions. It was also shown that DRAIR silencing inhibited the expression of anti-inflammatory genes in THP1 monocytes. Furthermore, DRAIR silencing enhances monocyte-endothelial cell (EC) adhesion and expression of pro-inflammatory genes, such as IL-1β. Conversely, DRAIR overexpression in THP-1 cells increases monocytes/macrophage differentiation and the expression of anti-inflammatory target genes, such as cytoplasmic polyadenylation element-binding protein 2 gene and interleukin 1 receptor antagonist; at the same time, it also inhibits pro-inflammatory genes, such as tumor necrosis factor-α (TNF-α) and fc gamma receptor IIIb. Therefore, DRAIR has an anti-inflammatory role, and it has been observed that it modulates the inflammatory phenotype of monocytes via epigenetic mechanisms in humans. Indeed, its downregulation is associated with an inflammatory phenotype of monocytes in DM thus promoting chronic inflammation [92]. The nuclear lncRNA Dnm3os is significantly upregulated in CD14 + monocytes from patients with DM compared with healthy controls (Fig. 2c). A similar regulation of Dnm3os was observed in macrophages from high-fat diet-induced insulin-resistant mice, streptozotocin (STZ)-induced type 1 diabetic mice, and STZ-induced diabetic apolipoprotein E deficient mice (a model of accelerated atherosclerosis) compared with controls (Fig. 3a). Dnm3os overexpression increases the pro-inflammatory gene expression and phagocytosis in macrophages, while its silencing results in the opposite response. Dnm3os interacts with nucleolin which has a protective role in macrophages. Nucleolin expression levels are decreased under diabetic conditions, and this leads to Dnm3os increase, promoting inflammatory gene expression. This suggest that Dnm3os play a role in DM and accelerated atherosclerosis [93]. Emerging evidence indicates that NETs trigger NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) assembly in macrophages, thereby sustaining inflammatory response with cytokine production, e.g. interleukin (IL)-1β, IL-18 [94]. An excessive NLRP3 inflammasome activation causes tissue damage increasing the risk of developing atherosclerosis [95, 96]. Interestingly, miRNAs control the activity of NLRP3 inflammasome [97]. In particular, miR-146a and miR-155 are two of the most studied miRNAs in the field of diabetes and atherosclerosis which control NLRP3 inflammasome activity in macrophages. In intrarenal macrophages from diabetic mice, miR-146a deficiency increased NLRP3, IL-1 β, and IL-18 expression levels leading to accelerated diabetic nephropathy, ultimately confirming the protective role of miR-146a within the inflammatory signaling pathway (Fig. 3b) [98]. Also, miR-155 is involved in NLPR3 activation by regulating the mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK)/nuclear factor kappa B (NFκB) pathway in oxidized LDLs (ox-LDL)-induced-macrophages. Lentivirus-mediated overexpression of miR155 in apolipoprotein E deficient mice accelerated atherosclerosis (Fig. 3b). Therefore, miR-155 may promote accelerated atherosclerosis via NLPR3 activation [99]. These two miRNAs appear to be an attractive target to attenuate diabetic inflammation by suppressing the NLRP3 inflammasome.

Natural killer

Several ncRNAs are important regulators of NK cell functionality. In particular, two ncRNAs are involved in the NK interferon-gamma (IFN-γ) production and activation respectively. Specifically, IFN-γ antisense 1 is overexpressed in NK cells upon activation, leading to increased IFN-γ secretion [100]. Moreover, Ni et al. found that NK cells in human PB express high levels of miR-362-5p [101]. This miRNA can up-regulate the expression of CD107a (a cell surface marker exhibited by NK-activated cells), promoting perforin and granzyme B secretion. Importantly, the production of IFN-γ and the activation of NK cells have a crucial role in vascular homeostasis and the occurrence of vascular diseases [102].

Dendritic cells

Several miRNAs, like miR-182, -148, and − 152, are known to affect dendritic cell activation. Worth mentioning, miR-181a controls the inflammatory response of DCs. Indeed, it has been observed that upon hyperlipidemia, the increase of miR-181a attenuated ox-LDL-induced DC immune inflammatory response. In addition, miR-181a inhibits the expression of an important inflammatory transcription factor, c-Fos, further corroborating miR-181a anti-inflammatory role, especially during vascular diabetic complications, like atherosclerosis [103]. Other studies confirm the importance of ncRNAs in DCs function. miR-148 and miR-152 regulate the antigen presentation of toll-like receptors (TLR) to T cells triggered by DCs [104], an important process that defends against infection, autoimmunity, and CVDs [105, 106].

Adaptive immune cells

T lymphocytes are major components of the adaptive immune system [107]. Several studies have shown that the immune-pathological effects mediated by ncRNAs on T lymphocytes play a role in the development of diabetic complications. In particular, four ncRNAs (miR-155, ENST00000411554, miR-29b, miR-10a, or -10b) are involved in T lymphocyte deregulation. In the PB of T2DM patients with retinopathy or proliferative retinopathy, intracellular miR-155, and serum transforming growth factor-β (TGF-β) levels are significantly upregulated while the percentage of CD4 + CD25 + forkhead box (Fox) P3 + T-regs was lower compared to non-diabetic retinopathy and healthy control group (Fig. 4a). It was shown that the expression of miR-155 was negatively related to the T-regs and TGF-β levels emphasizing that miR-155 may play an important role in the pathogenesis of T2DM retinopathy by regulating the Tregs via TGF-β [108]. Another study reported the altered expression of the lncRNA ENST00000411554 in the wound surface of foot ulcers from patients with T2DM and found an association with T lymphocytes impaired functions and immune regulation imbalance (Fig. 4b). In particular, Xu et al. observed that the diabetic wound was infiltrated with higher levels of CD3, CD8 lymphocytes, and inflammatory factors, i.e. IL-2, IL-10, IFN-γ, and TNF-α, compared to non-diabetic control. They performed a microarray on the tissue of diabetic foot ulcers to investigate the molecular mechanisms involved in immune dysfunction. Analyses showed the downregulation of 13 lncRNAs, and, among them, of lncRNA ENST00000411554, which is located in a key upstream regulatory region of the target gene Mitogen-Activated Protein Kinase (MAPK) 1. Interestingly, an inverse correlation between the lncRNA and MAPK1 was observed. Therefore, it was hypothesized that ENST00000411554 could be involved in immune-regulation imbalance of T2DM, thus regulating the expression of inflammatory factors in T-cell-induced responses by downregulating MAPK1 [109]. Several studies indicate miRNAs as regulators of T lymphocytes activities in diabetic nephropathy. As an example, renal miR-29b downregulation contributed to the upregulation of T-Box Expressed in T cells and IFN-γ at both mRNA and protein levels, promoting CD4 + IFN-γ + Th1 cell infiltration in db/db mice (Fig. 4c). Overexpression of miR-29b inhibited Th1-associated immune injury [110]. Similarly, miR-10a or -10b downregulation promoted the infiltration of T cells (CD3+), Th cells (CD3+/CD4+), and killer T cells (CD3+/CD8+) in the kidney from STZ-treated diabetic mice (Fig. 4c) [111]. Although several studies have investigated the role of B lymphocytes, more investigations are needed to understand the role of B-lymphocyte ncRNAs in this context.

Extracellular vesicles shuttle ncRNAs between immune and cardiovascular cells

Cell-to-cell communication mediated by extracellular vesicles

The cells in the adult human heart and vasculature are continuously engaged in cell-to-cell communications (CCC) to regulate both single-cell functions and coordinated responses needed for homeostasis [112]. CCC is ensured by a variety of biochemical mediators and electrical impulses [113, 114]. Diseases disrupt CCC, and cells might not interact properly or decode molecular messages properly. The study of altered cellular crosstalk might facilitate an understanding of disease pathogenesis and progression. Although protein-mediated interactions, such as ligand-receptor, receptor–receptor, and receptor-extracellular matrix interactions, have been better studied so far [114, 115], CCC can also take place via ncRNAs, and miRNAs in particular. miRNA-based CCC takes advantage of the interaction between miRNAs released by “message sender cells” and the miRNAs “target genes” present in the “message recipient cells”. miRNA exchanges are mostly realized via extracellular vesicles (EVs) (Fig. 5), although several alternative extracellular carriers of miRNAs, such as lipoproteins and argonaute-2, have been described [115]. Moreover, miRNAs can also be exchanged via connexin gap junctions, without being released extracellularly.
The term EVs denotes a highly diverse family of membrane vesicles of different biogenesis and sizes: apoptotic bodies (> 1000 nm), microvesicles (MVs) (150 nm − 1 μm), and small EVs (sEVs, circa 30–150 nm). sEVs, also called “exosomes”, are currently of particular interest to the biomedical community. sEVs are actively secreted and contain endosomal membrane markers, such as tetraspanins, plus a composite molecular cargo; that can include RNAs and proteins [116118], as well as mitochondria fragments and cytosol-derived small molecules (< 1500 Da), including metabolites as sugars, amino acids, lipids, nucleotides, and N-glycans [119, 120]. Once released by their parent cells, sEVs protect their molecular cargo from degradation until they pass it in a functionally active status to neighboring cells through binding, fusion, or endocytosis [117]. Alternatively, sEVs are transported in biological fluids to reach distant organs [121] or be excreted [118]. sEVs shuttle biologically active miRNAs from their parent cell to recipient cells, thus spreading the miRNA regulatory actions and profoundly influencing biological functions, including pro and anti-angiogenic responses [122124].
The adult human heart comprises billions of cells and approximately a third of them are cardiomyocytes, whilst the remainder comprises ECs and other vascular cell types, fibroblasts, resident macrophages, and neural cells [125, 126]. Recent evidence shows that different types of cardiovascular cells use sEVs as a means to communicate with each other as well as with other cells present in the cardiac microenvironment, such as immune cells and adipocytes [122, 124, 127, 128]. Moreover, evidence is accumulating for different types of sEVs being simultaneously released by a single cell type [129]. sEVs can differ substantially in their cargo and membrane composition [130, 131]. Consequently, the pool of sEVs that populate the heart’s extracellular space and contribute to cell-to-cell communications is diverse and variable. The coordination of sEV-mediated responses is important for maintaining physiology, but it can also mutate to support disease propagation [132, 133]. This can be understood considering that the amount and quality of sEVs secreted from a cell are influenced by both its activation status and the external environment to which the cell is exposed [134, 135], including hypoxia and D-glucose concentration [136]. As an example, Wang et al. showed that rat diabetic cardiomyocytes produce anti-angiogenic sEVs in vitro. This is in net contrast with the proangiogenic program activated in recipient ECs by their non-diabetic sEV counterparts [124]. Also, macrophages and ECs communicate via EVs, and changes in the macrophage phenotype contribute to reduced microvascular repair and impaired myocardial perfusion by promoting atherosclerosis [132], chronic inflammation and fibrosis [130, 137, 138].

Extracellular vesicles mediated transfer of ncRNAs in diabetes

The chronic inflammation induced by DM dysregulates the constant interaction of immune cells and cardiovascular cells [57]. CCC in DM has been reported to be driven mainly by sEVs RNA cargo [139142]. Indeed, higher levels of EVs have been found in human and animal models of type 1 and 2 DM [141]. miRNAs are the most studied ncRNA cargo of sEVs with a demonstrated functional role. sEVs-miRNAs have been shown to act on pancreatic β-cells, the insulin-producing cells. In detail, Zhang et al. showed that miR-223-3p and − 5p were two to three times increased in the medium of cultured pancreatic β-cells after high glucose stimulation, which modulated Glucose transporter 4, favoring insulin sensitivity [142]. Of note, several studies revealed bidirectional crosstalk between β-cells and immune cells via EVs, both in physiological and pathological conditions [143146]. As such, metabolic regulatory miRNAs, such as miR-29a-3p [147], miR-155-5p [148], miR-503 [149], and miR-210-3p and − 5p [150], can be carried by EVs and delivered into insulin-responsive cells and organs via paracrine or endocrine routes.
Macrophage-derived sEVs also play a role in diabetic complications. It has been reported that high glucose levels in pro-inflammatory macrophages induce the production of EVs carrying high levels of IL-1β, inducible nitric oxide synthase, human antigen R (HuR), miR-21-5p, miR-486-5p, and TGF-β mRNA [151155]. These biomolecules can be transferred to target cells and subsequently induce renal and cardiac injury and dysfunction. In particular, two miRNAs closely related to cardiac fibrosis and diastolic dysfunction, miR-122-5p [156158] and miR-21-5p are sorted specifically into EVs by the retinol-binding protein HuR [159]. Additionally, sEVs can effectively transmit pathogenic miRNAs to target cells, including vascular smooth muscle cells (VSMCs), ECs, neutrophils, and macrophages, leading to vascular stenosis, dysfunction, and inflammation, promoting atherosclerosis and thrombosis [157]. Likewise, recent mouse studies suggest that T cells might also have a role in DM by communicating with β-cells through the transfer of pro-apoptotic sEVs-miR-142-3p/5p and miR-155 [158]. As mentioned above, CD34 + HSPCs can transfer a negative pro-apoptotic signal including ncRNAs to the ECs via sEVs [14].
Exposure to numerous noxious stimuli, such as other cardiovascular risk factors, is also considered an important pathogenic event in DM [160]. ECs activation entails the expression of cell adhesion molecules like intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1) that serve as triggering signalling to the leukocyte adhesion on the endothelium. The uptake of LDLs into the intima and its oxidation, with the enhanced infiltration of monocytes/macrophages, promotes the formation of foam cells that induce cytokine and chemokine production and the additional recruitment of circulating immune cells [61]. As an example, miR-205-3p/-5p can regulate the adhesive properties of ECs, most likely by targeting the tissue metallopeptidase inhibitor 3, thereby triggering the release of soluble TNF-α, increasing monocyte adhesion [161]. Similarly, numerous miRNAs (miR-181b-5p, miR-103, miR-126-3p, miR-10a-5p, miR-92a-3p, let-7 g-3p/-5p, miR-195-5p) regulate expression in ECs of cytokines with pro or anti-inflammatory potential by indirectly targeting NF-kB and consequently affecting the immunophenotyping [162164].
In addition to miRNAs, lncRNAs, and circRNAs can also be exchanged between cells via EVs, and play a regulatory role in DM complications. For example, lncRNA Nexilin F-actin binding protein antisense RNA 1 (NEXN-AS1) circulates in sEV and suppresses the TLR4/NF-kB pathway, reducing endothelial activation and monocyte recruitment in human vascular ECs [165]. Further, its overexpression inhibits pro-inflammatory pyroptosis-related biomarkers known to drive atherosclerosis, such as NLRP3, CASP-1, IL-1β, IL-18, Gasdermin D [166]. Moreover, lncRNA growth arrest specific 5 (GAS5), long intergenic non-protein coding RNA 1005 (LINC01005), and metastasis associated lung adenocarcioma transcript-1 (MALAT-1) have also been detected in sEVs and may play a role in DM pathogenesis [167169]. In detail, silencing lncRNA GAS5 alleviates apoptosis and fibrosis in diabetic cardiomyopathy by targeting miR-26a/b-5p [170]. Besides, sEVs-LINC01005 from ECs has been reported to promote VSMC phenotype switch, proliferation, and migration by regulating the miR-128-3p/Kruppel-like factor (KLF) 4 axis [168]. On the contrary, sEVs-MALAT-1 derived from ECs promoted anti-inflammatory macrophage polarization [169, 171].
Of relevance, the circRNA homeodomain-interacting protein kinase 3 (circHIPK3) released by ECs-sEVs has been reported to promote the proliferation of VSMCs induced by high glucose via the miR-106a-5p/FoxO1/VCAM1 pathway [172]. Moreover, sEVs- circRNA euchromatic histone-lysine-N-methyltransferases 1 from pericytes could suppress the generation of NLRP3 inflammasomes in high glucose-treated ECs by upregulating the levels of the transcription factor nuclear factor I A, which in turn inhibited the apoptosis of ECs and reduced its angiogenic activity, ultimately attenuating high glucose-induced retinal vascular dysfunction [173].

Effect of aging and sex on diabetes-related CVDs: the role of ncRNAs

A large body of evidence shows sex-based differences in the pathophysiology of CVDs and the response to cardiovascular treatments [174181], which also implies underlying ncRNA-mediated regulatory mechanisms in a sex-dependent manner [182]. In this context, there are pronounced sex differences in the impairment of glucose metabolism, with men presenting more frequently elevated fasting glucose levels, while women more frequently develop impaired glucose tolerance [183186]. In addition, the menopause transition accelerates the risk for CVDs [187, 188]. The influence of estrogen decline with menopause on risk factor clustering increases, thereby leading to hormonal regulation of body fat distribution and related metabolic abnormalities [189, 190]. In turn, together with aging, these elements can account for CVDs in postmenopausal women [191, 192]. Along this line, altered glucose homeostasis leading to metabolic changes [193] may predispose postmenopausal women to the development of T2DM [194]. In this regard, in human ECs estradiol modulates regulatory networks composed of miRNAs, transcription factors, and downstream genes.

miRNAs associated with diabetes mellitus, aging, and sex

Interestingly, aging and DM share common pro-inflammatory pathways, such as the NF-κB and NLRP3 signaling pathways, which are regulated by several miRNAs. Among the most remarkable miRNAs regulating age-mediated inflammation, miR-146a stands out, as it can reduce NF-κB DNA binding activity [195] and has been proposed as a biomarker of healthy aging [196]. Inflammation is a common ground in several CVDs and sex differences in the transcriptomic regulation of inflammatory genes and pathways in CVDs have been previously reported [181, 197, 198]. Circulating levels of miR-146a decline in aged people, and this phenomenon is particularly evident in men [196]. In turn, miR-146a levels decrease after estrogen treatment in mouse-isolated splenic lymphocytes [199], and it could act as a sex-biased inflammation-associated biomarker, modulating T2DM and other aging-associated diseases [200]. Also, miR-34a, a key promoter of vascular aging related to DM-associated vascular complications, shows sex differences in its expression [201]. Of note, it induces a systemic inflammatory state [202]. Indeed, miR-34a expression is elevated in several cardiac pathologies, and its therapeutic inhibition in a mouse model of dilated cardiomyopathy provides more protection in females than in males [203]. Particularly, in association with DM, sex was found to be a determinant of plasma miR-34a levels in patients with left ventricular diastolic dysfunction [204].
Other miRNAs, such as let-7 g and miR-221 have been related to glucose metabolism [205207]. Thus, different studies support a sex-dependent regulation of physiological functions by ncRNAs in proinflammatory states that underlie cardiovascular or metabolic diseases, among others, also show sex differences, as women with metabolic syndrome display higher circulating levels of let-7 g and miR-221 than men [207]. In addition, mouse studies have revealed a male-specific induction of miR-23a-3p, miR-27b-3p, miR-130a-3p, miR-133a-3p, miR-143-3p, and let-7e-5p, together with a corresponding downregulation of downstream protein targets involved in mitochondrial metabolism, thereby probably contributing to sex-biased maladaptive cardiac remodeling [208].

Intrauterine/placental RNA modulation and gestational DM

Some of the sex differences in transcriptomics are evident from fetal age. In particular, analysis of miRNA expression patterns in healthy placentas performed in the Environmental Influences on Early Ageing (ENVIRONAGE) study shows a different miRNA expression pattern in the placenta of mothers of girls and boys, and enrichment analyses showed sex-dependent differences in the regulation of biological processes, including metabolic and immunological pathways [209]. In newborn girls, the higher expression of placental miRNA-210, miR-146a, miR-34a, and miR-222, among others, is critical for specific cellular processes in aging and inflammation [210]. It is important to note that some of the alterations that occur during pregnancy can affect the future development of pathologies in both the mother and the newborn [211]. In particular, women with gestational DM have a higher risk of developing future T2DM [212]. In this regard, during gestational DM, the miRNA expression profile changes in human placental ECs [213]. Thus, the hyperglycemic intrauterine environment could affect placental function through elevated levels of proinflammatory cytokines, as well as through transcriptional modifications, including those mediated by ncRNAs.

Circulating immune-associated ncRNAs as biomarkers of diabetic CVDs

Extracellular ncRNA levels are dynamically regulated in response to stress and diseases providing an opportunity for the identification of biological markers [214]. Indeed, circulating ncRNA biomarkers may reflect the physiological or pathological status of a subject. The analysis of ncRNA patterns may lead to the development of non-invasive tools for clinical decision-making in the fields of diagnosis, prognosis, and therapeutic guidance [215, 216].
Several studies have already provided relevant data on the potential use of immune-associated ncRNAs in the context of the cardiovascular complications of DM, the miRNAs being the main subclass investigated (Table 3). For instance, Zampetaki et al. [217] provided indirect evidence on the role of these transcripts in the link between the pathophysiology of DM and the manifestation of its vascular complications. The authors demonstrated the alteration in the plasma levels of miRNAs implicated in vascular diseases, e.g., miR-126, in prevalent DM. Supporting these results, Jansen et al. [218] reported the deregulated levels of miR-126 in circulating MVs from patients with DM and that patients with reduced miR-126 levels are at higher risk for the occurrence of concomitant coronary CAD. Another remarkable example of immune-associated miRNAs as indicators of vascular complications is given by miR-15a and miR-16a. The serum levels of both miRNAs are upregulated in patients with CLI and positively correlated with amputation after restenosis at 12 months post-revascularization in T2DM patients with CLI [78].
Table 3
Examples of the potential role of ncRNAs as biomarkers of cardiovascular complications of diabetes
miRNAs
Reference
Study Population
Source
Main findings
[78]
122 patients with CLI and T2DM, 20 patients with CLI & 43 non-ischemic and non-diabetic subjects
Serum
miR-15a and miR-16 positively correlate with the risk of amputation after restenosis
Serum miR-15a is positively associated with post-revascularization restenosis considered as the first event
[221]
90 non-HF, 90 HFpEF & 90 HFrEF
Serum
Reduced levels of miR-146a-5p in HF patients compared with the control group
Combination with contemporaneous laboratory parameters provides an optimal discriminative value
[218]
55 patients with DM & 80 non-DM patients
Circulating microparticles
miR-26a and miR-126 are significantly reduced in DM patients compared to controls
Patients with reduced miR-26a and miR-126 expression levels are at higher risk for the occurrence of a concomitant CAD
[219]
86 patients with well-controlled T2DM
Serum
miR-1 and miR-133a levels are independent predictors of myocardial steatosis
[222]
28 healthy controls, 26 patients with DM, 22 patients with chronic HF & 15 patients with both DM and chronic HF
Plasma
miR-30c is reduced in patients with DM and HF
miR-30c levels are negatively correlated with plasma glucose levels in patients with chronic HF
[223]
63 patients with DM with and without cardiac dysfunction
Plasma
miR-144 is significantly decreased in the plasma of patients with DM with cardiac dysfunction
Plasma miR-144 could serve as a specific predictor of patients with DM developing cardiac dysfunction
Long non-coding RNAs
[232]
414 patients with acute MI
Whole Blood
MIAT is a predictor of LV dysfunction at 4 months after MI
 
56 patients with diabetic cardiomyopathy, 44 patients with DM but without cardiomyopathy & 42 healthy controls
Serum
HOTAIR is a diagnostic biomarker of diabetic cardiomyopathy
[234]
6 patients with DMs and 6 healthy subjects
Serum
KCNQ1OT1 is elevated in patients with DM
Circular RNAs
[234]
45 patients with T2DM and 45 healthy subjects
Peripheral white blood cells
circANKRD36 is associated with chronic inflammation in T2DM patients
DM: diabetes mellitus, CAD: coronary artery disease, circANKRD36: circular ankyrin repeat domain 36, CLI: critical limb ischemia, HF: heart failure, HFpEF: heart failure with preserved ejection fraction, HFrEF: heart failure with reduced ejection fraction, HOTAIR: homeobox transcript antisense RNA, KCNQ1OT1: potassium voltage-gated channel subfamily Q member 1 opposite strand/antisense transcript 1, LV: left ventricle, MI: myocardial infarction, MIAT: myocardial infarction-associated transcript, T2: type 2.
Concerning diabetic cardiomyopathy, serum miR-1, and miR-133a are independent predictors of myocardial steatosis in patients with well-controlled T2DM patients [219]. Both miRNAs may be useful biomarkers for the evaluation of myocardial steatosis in T2DM, an early manifestation in the pathogenesis of cardiac-related complications which quantification is currently impractical for large-scale population screening. In line with these findings, miR-146a-5p is a key regulator of the immune response, inflammation, and fibrosis in diabetic cardiomyopathy [220] and its circulating levels have been proposed as a biomarker of HF [221]. The plasma levels of miR-30c are reduced in patients with DM and HF and its levels are negatively correlated with plasma glucose concentration in chronic HF [222]. Additionally, Tao et al. [223] reported that plasma miR-144 is downregulated in the plasma of patients with DM with cardiac dysfunction, compared to patients with DM without cardiac dysfunction, and proposed this miRNA as a predictor of cardiac dysfunction in DM patients. Also, given the involvement of KLF5 in diabetic cardiomyopathy, it is also relevant that, miR-145, and miR-375 target KLF5, thereby repressing its levels [224226]. While the role of these miRNAs as potential circulatory biomarkers is yet to be explored, there are indications of the mechanistic involvement of at least miR-145 in the pathological process contributing to the development of diabetic cardiomyopathy [227229].
More recently, other ncRNA classes, such as lncRNAs, have been proposed as tools for disease management [230], including immune-associated lncRNAs as indicators of DM-induced CVDs. The long-non-coding myocardial infarction–associated transcript (MIAT), which is upregulated in serum and PB-mononuclear cells from T2DM patients [231] and predominantly expressed in lymphocytes, has been described as a predictor of left ventricle dysfunction at 4-month follow-up in patients that suffered an acute MI [232]. The expression of the lncRNA Homeobox transcript antisense RNA (HOTAIR) is downregulated in myocardial tissues and the serum of patients with diabetic cardiomyopathy compared with both patients with DM but without cardiomyopathy and healthy controls. Interestingly, the serum levels of this lncRNA discriminate between patients with diabetic cardiomyopathy and healthy controls [233]. In addition, the lncRNA Potassium Voltage-Gated Channel Subfamily Q Member 1 Opposite Strand/Antisense Transcript 1 (KCNQ1OT1), associated with pyroptosis and fibrosis in diabetic cardiomyopathy, is significantly elevated in the serum of patients with DM compared with healthy subjects [234].
Advances in RNA sequencing have also drawn attention to circRNAs. The evidence on immune-associated circRNAs as biomarkers of the cardiovascular complications induced by DM is still scarce. Nevertheless, some data point to a role as an indicator of pathological mechanisms linked to these conditions. Fang et al. [235] identified circular ankyrin repeat domain 36 (circANKRD36) as a mediator of inflammation-associated pathways in T2DM patients. Interestingly, circANKRD36 was upregulated in PB leucocytes and correlated with inflammatory cytokines involved in CVDs such as IL-6 and TNF-α.
Therefore, immune-associated ncRNAs emerge as an interesting tool to evaluate the cardiovascular complications associated with DM. The translation of these transcripts to clinical practice may improve patient management and decision-making. Of note, circulating ncRNA quantification does not require any special infrastructure, since they can be quantified in clinical specimens (plasma and serum) with techniques (quantitative PCR) currently used in clinical laboratories. Nevertheless, their incorporation into clinical laboratories needs to overcome several challenges, including the improvement of reproducibility, which is beyond the scope of the current article and discussed here.

ncRNAs as therapeutic targets in diabetic cardiovascular complications

Strategies to modulate ncRNAs-ncRNA therapeutics: experiences in diabetic CVDs

The epidemic of diabetic cardiovascular complications underlines the need for the translation of current preclinical knowledge and novel technologies into actual clinical practice. Recently, ncRNAs were shown to be powerful therapeutic tools for different disease settings by affecting the onset and progression of several human disorders [236]. Therefore, researchers focused on the development of innovative next-generation technologies to exploit these ncRNAs to treat such diseases, especially those not susceptible to pharmaceutical interventions [237].
The use of ncRNA-based therapies has several advantages compared to other compounds. NcRNAs, and in particular miRNAs, can naturally modulate different specific targets within one pathway thereby exerting a broader biological effect. Thus, ncRNA therapies might have the potential to boost therapeutic effects compared to those drugs acting on one single target gene. Additionally, chemical modifications can easily be adapted to prolong the stability and half-life of ncRNA drugs in circulation, facilitating the effective delivery to the site of interest without degradation (Fig. 6).
Strategies to target the level of ncRNAs can be classified into two approaches: suppressing upregulated ncRNAs or enhancing levels of downregulated ncRNAs. Technologies to modulate miRNA expression include miRNA mimics to elevate their expression; concerning miRNA inhibition, miRNA sponges exert their function by sequestration of endogenous miRNAs, while inhibitory chemical compounds can compromise miRNA action through the interaction with components of miRNA biogenesis pathways. Moreover, the most pursued strategy relies on base pairing of complementary antisense oligonucleotides (ASOs) with ncRNA sequences leading to duplex formation or subsequent degradation (e.g. anti-miRNAs). LncRNAs can be targeted through modified ASOs such as locked nucleic acids (LNAs) with modified phosphorothioate backbones, gapmers composed of two LNA flanking regions and a central DNA stretch that activates ribonuclease H cleavage, or small interfering RNAs inducing their degradation [237, 238]. Other modulation tools include genome-editing strategies such as Clustered Regularly Interspaced Short Palindromic Repeats (CRISP)/CRISPR associated protein 9 (Cas9) or viral-based approaches [239, 240]. Although there is an increasing number of preclinical reports regarding ncRNA therapies, only a small amount have proceeded into clinical trials [241, 242]. Key challenges have to be faced to successfully translate ncRNA therapeutics into the clinic. Off-target effects result in suppressing genes other than the desired target due to sequence similarities or overdosing along with undesired immune responses. Additionally, special delivery systems are required to target the organ and cell type of interest and naked oligonucleotides have to be chemically modified to resolve issues such as degradation by nucleases or their inefficient uptake due to negative charge or size [243245]. In this respect, recently, it has been shown that lipid- or polymer-based nanoparticles, as well as exosomes, are promising delivery tools (Fig. 6) [246250].
Nonetheless, a plethora of RNA-based drugs are already under clinical development, most of them aiming to modify mRNA levels in the liver, central nervous system, or muscle and some of them have even been approved by the Food and Drug Administration or European Medicines Agency (e.g. Viltolarsen, Nusinersen, Patisiran) [251254]. In addition, tremendous progress has also been made in the field of miRNA-based drugs which entered clinical testing [238, 255] and lncRNA-based formulations are also gaining more interest as promising treatment options. One of the ncRNA compounds, which entered the clinical phase, is an inhibitor against miR-92a (MRG-110) [256, 257] that was shown to exhibit beneficial effects in wound healing and CVDs such as LI, MI (NCT03603431, NCT03494712). Another example is anti-miR-21, which was evaluated for its capability to reduce kidney fibrosis in patients with Alport syndrome (NCT03373786) and will be further examined in a clinical phase II study (NCT02855268) [258]. Moreover, CDR132L, a synthetic oligonucleotide inhibiting miR-132 indicated as a key regulator in cardiac remodeling processes [259, 260], entered recent clinical study phase II (NCT04045405, NCT05350969) for safety and efficacy assessment in ischemic HF patients. Concerning DM, AstraZeneca performed a phase I/IIa clinical trial (NCT02612662, NCT02826525) with an N-Acetylgalactosamine (GalNAc)-conjugated anti-miRNA against miR-103/107 (RG-125) [261]. miR-103/107 is an insulin sensitizer already tested in preclinical models that show promise as a compound to treat patients with T2DM and nonalcoholic fatty liver diseases.

Anti-diabetic and anti-inflammatory drugs affecting ncRNAs

Antidiabetic drugs and ncRNAs

Antidiabetic drugs can impact the cellular epigenetic landscape, providing possible interactors of these drugs. In this respect, metformin is one of the most studied drugs. Metformin is a biguanide derivative and a mainstay prescribed drug for the treatment of T2DM [262]. Metformin’s primary effect is mediated by the inhibition of hepatic gluconeogenesis, but it can also ameliorate the “inflammaging” process, increase vascular function, and improve lipid profiles affecting the outcomes of CVDs [263, 264]. Interestingly, metformin can reduce ROS generation and contrast oxidative stress-induced apoptosis and inflammation in cardiomyocytes and ECs via AMP-activated protein kinase activation [265]. In particular, metformin treatment increased the levels of miR-20a-5p, miR-34a-5p, miR-130a-3p, miR-106b-5p, miR-125b-3p, and let-7c [266], and of the miR-200s family (miR-200a-3p, miR-141-3p, and miR-429), previously associated to senescence and aging (Table 4) [267, 268]. Moreover, Giuliani et al. reported that metformin can revert the senescence-associated modulation of miR-216-3p, -216-5p, and − 217-5p, which were previously identified among the most upregulated miRNAs in senescent ECs [269].
Table 4
Anti-diabetic, anti-inflammatory/anti-aggregation drugs impacting on ncRNAs
Antidiabetic drugs and ncRNAs
Reference
Study Population/experimental system
Treatment
Main findings
[333]
Mouse liver
Mice treated with metformin
Metformin induced the expression of miR-20a-5p, miR-34a-5p, miR-130a-3p, miR-106b-5p, miR-125b-3p, and let-7c
[334]
BJ-1s human neonatal foreskin fibroblasts
Cells treated with metformin at different passages
Metformin induced the expression of miR-200a, miR-141, miR-429 and of miR-205 in senescent BJ-1s cells
[335]
Human umbilical vein ECs
Cells treated with metformin at different passages
Metformin induced the expression of miR-100-5p, miR-125b-5p, miR-654-3p, miR-217 and miR-216a-3p/5p in senescent ECs
[270]
Mouse bone marrow-derived endothelial precursor cells
Cells treated with palmitic acid and metformin
Metformin reverted angiogenesis impairment caused by palmitic acid by attenuating miR-130a/p-AKT axis and increasing PTEN expression
[336]
Mouse microvascular ECs
Cells exposed to HG and treated with metformin
Metformin reduced and increased miR-34a-5p and SIRT1 expression levels, respectively, attenuating HG-induced angiogenesis impairment
[337]
Neonatal rat ventricular cells
Cells exposed to H2O2 and treated with metformin
Metformin reverted H2O2- and ischemia/reperfusion-induced miR-1a-3p expression, reducing cell death
Mouse ischemia/reperfusion
Hearts of mice that underwent the I/R injury and treated with metformin
[273]
Mouse ischemia/reperfusion
Hearts of mice that underwent the I/R injury and treated with metformin
Metformin reduced I/R induced-miR-34a-5p expression
H9C2 rat cardiomyocyte cells
Oxygen-glucose deprivation/recovery and treatment with metformin
Metformin reduced miR-34a-5p levels through decreasing SIRT1-p53 activity
90 ACS (STEMI) patients
Metformin 3-months treatment pre-infarction
Metformin reduced serum miR-34a levels and CKMB activity and mitigated PCI-induced reperfusion injury
 
Thoracic aortas of diabetic rats
Liraglutide treatment
Liraglutide reduced miR-34a-5p and increased the anti-apoptotic protein Bcl2 and SIRT1, contrasting cell death
[280]
25 patients with T2DM
Serum from patients with DM treated with liraglutide
Liraglutide induced the expression of miR-130a-3p, miR-27b-3p, and miR-210-3p
[338]
Mouse MCAO
Brain after MCAO and metformin treatment
Metformin reduced H19-induced oxidative stress injury
[282]
10 frail old adults with HFpEF and DM
Whole blood after 3 months-treatment with empagliflozin or metformin or insulin
Empagliflozin specifically reduced miR-21 and miR-92 levels compared to metformin- or insulin-treated HFpEF patients and to controls
Anti-inflammatory/anti-aggregation drugs and non-coding RNAs.
Reference
Study Population/experimental system
Treatment
Main findings
[339]
46 ischemic stroke patients
PBMNCs from stroke patients treated with ASA for 10 days
ASA induced miR-145-5p levels in stroke patients
VSMCs
Cells treated with ASA
ASA increased miR-145-5p and decreased CD40 levels, respectively, reducing VSMCs proliferation
[340]
Platelets of 12 ASA -treated CVD patients (6 with low and 6 high platelet reactivity)
ASA treatment
miR-135a-5p and miR-204-5p levels correlated with platelet reactivity
[341]
Platelets of 945 acute coronary syndrome patients
ASA treatment
-Lower miR-19b‐1‐5p expression was associated to ASA insensitivity and to a higher risk of MACCE
- Low miR-223 was a predictor of responsiveness to antiplatelet therapies
ACS: acute coronary syndrome, ASA: acetylsalicylic acid, Bcl2: B-cell lymphoma 2, CKMB: creatine kinase MB, CVD: cardiovascular disease, DM: diabetes mellitus, ECs: endothelial cells HFpEF: heart failure with preserved ejection fraction, HG: high glucose, MCAO: middle cerebral artery occlusion, I/R: Ischemia-reperfusion, MACCE: major adverse cardiac and cerebrovascular events, PBMNCs: peripheral blood mononuclear cells, PCI: percutaneous coronary intervention, PTEN: phosphatase and tensin homolog, SIRT1: Silent information regulator 1, STEMI: ST-segment elevation myocardial infarction, VSMCs: vascular smooth muscle cells.
Recent data showed that metformin reverted impaired angiogenesis caused by palmitic acid treatment of HSPCs by the miR‑130a-3p/phosphatase and tensin homolog (PTEN)/ phosphoinositide 3-kinases/protein kinase B axis [270] in mouse microvascular ECs by reducing miR-34a-5p and increasing silent information regulator 1 (SIRT1), a miR-34a target [271].
Metformin is also able to reduce oxidative stress, which is a crucial mechanism of cardiac injury caused by ischemia-reperfusion (I/R), a major side effect of the reperfusion treatment of the ischemic heart [272]. This beneficial effect is associated with the regulation of a p53-miR-34a-5p-Bcl-2 pathway axis [273]. Moreover, Zhang et al. demonstrated that metformin prevented hydrogen peroxide (H2O2)-induced neonatal rat ventricular cell death by reducing miR-1a-3p via AMP phosphorylation and inducing the expression of its target glucose-regulated protein 94, resulting in decreased cell death signaling activation [274, 275]. Recently, it has been reported that metformin exerts an epigenetic effect in T2DM South African patients. Indeed, several DNA regions were differentially methylated and 57 lncRNAs-associated DNA methylation regions were identified in the genomic DNA of metformin-treated T2DM patients.
The expression of lncRNA-H19 is closely related to the progression of cerebral ischemia [276]. Zeng et al., in a stroke mouse model [277], reported that lncRNA-H19 expression was elevated and that it participated in the ischemic stroke-induced oxidative stress injury via inhibition of miR-148a-3p and upregulation of its target Rho associated coiled-coil containing protein kinase 2, which increases oxidative stress [278]. These responses were reduced by metformin treatment that ameliorated stroke-related brain injury [277].
Among the new generation of anti-diabetic drugs, liraglutide, a glucagon-like peptide-1 analog, in diabetic rats improved significantly aortic endothelial dysfunction by downregulating miR-34a-5p and increasing the expression of anti-apoptotic protein B-cell lymphoma 2 (Bcl2) and SIRT1 [279]. Moreover, recently it has been shown that liraglutide therapy increased serum levels of miR-130a-3p, miR-27b-3p, and miR-210-3p in patients with T2DM [280].
Empagliflozin is a novel selective inhibitor of sodium glucose cotransporter 2 associated with benefits on mortality, HF hospitalizations, and quality of life [281]. In a preliminary study with a small group of frail old adults with HF with preserved ejection fraction (HFpEF) and DM patients compared to controls, circulating levels of miR-21 and miR-92 were found to be significantly increased. The levels of these miRNAs decreased after 3-month treatment with empagliflozin, but not in response to metformin or insulin [282].

Anti-inflammatory/anti-aggregation drugs and ncRNAs

Acetylsalicylic acid (ASA) is a widely used nonsteroidal anti-inflammatory drug. Low doses of ASA inhibit cyclo-oxygenase-(COX)-1 and oppose platelet aggregation inhibiting the release of thromboxane A2 (TXA2) [283, 284], while higher ASA doses inhibit COX-2 and decrease prostaglandin E levels, inducing antipyretic and pain-relieving effects [285]. Accordingly, low ASA doses are prescribed for primary and secondary CVDs prevention [285287].
Several reports indicate that ASA treatment increased miRNAs expression and that several miRNAs can be useful biomarkers of ASA resistance. For instance, Guo et al. observed that miR-145-5p levels in PB mononuclear cells of ischemic stroke patients are increased by ASA treatment. Moreover, an ASA-mediated increase of miR-145-5p inhibited CD40 mRNA translation, reducing vascular smooth muscle cell proliferation in vitro. The interplay between ASA and miR-145-5p improved also the stability of atherosclerotic plaques.
ASA resistance is linked to the variability of platelet reactivity among cardiovascular patients [288]. MiR-135a-5p and miR-204-5p are correlated with platelet reactivity [289], while lower expression of miR-19b-1-5p is correlated with ASA insensitivity [290, 291]. Moreover, decreased levels of circulating miR-223, which is among the highest expressed miRNAs in platelets, is an independent predictor of responsiveness to antiplatelet therapies in patients suffering from CAD [292].
The P2Y12 inhibitors clopidogrel, prasugrel and ticagrelor are antiplatelet agents alternative to ASA. They are characterized, unlike ASA, by a prolonged presence in the plasma [293] and the combination of clopidogrel and ASA has a potentiated effect in the secondary prevention in patients with T2DM [294]. Moreover, ticagrelor and prasugrel when given in combination with ASA have beneficial effects in patients after MI [295]. The potential of clopidogrel or prasugrel antiplatelet therapy versus ASA therapy on platelet function, fibrin network characteristics, inflammation, and expression of miRNAs was investigated in a clinical trial in T2DM patients [296]. Prasugrel monotherapy in T2DM showed to have the strongest effect on platelet inhibition and in reducing the levels of several miRNAs relevant for platelet function and CVDs (miR-24, miR-191, miR-197, and miR-223) [297, 298]. In addition, reduced levels of miR-197 have been identified as a marker of CVDs in T2DM patients treated with prasugrel [296].

ncRNAs in coronavirus disease 2019 associated inflammation and cardiovascular damage

Inflammatory response in coronavirus disease 2019: diabetes as a risk factor

Clinical outcomes of COVID-19 are worse in the presence of CVDs and risk factors, such as DM (Fig. 7). Severe COVID-19, DM, and CVDs share a common denominator, an increased inflammatory status, which is also thought to underlie male-biased cardiovascular complications in COVID-19 [299, 300]. Assessing cardiovascular inflammatory status through evaluation of ncRNAs levels appears as a promising strategy to aid in personalized healthcare, prevent fatal cardiovascular damage, and thus improve clinical outcomes of COVID-19 patients. SARS-CoV-2 can affect multiple organ systems directly or indirectly through cytokine storm and the adaptive immune system activation [301, 302], dysregulation of the renin-angiotensin-aldosterone system (RAAS) as a result of virus-mediated suppression of angiotensin-converting enzyme (ACE) 2 expression [303], damage to ECs [304], vascular inflammation and sepsis [305]. Cardiovascular damage is frequently observed in severe COVID-19 patients and it is dictated directly by the amount of viral load or indirectly by the extent of the patient’s immune response. Cardiovascular damage is also dependent on the presence of co-morbidities associated with RAAS deregulation (Fig. 7) [306]. Although severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has shown major tropism to the pulmonary cells, recent reports confirmed its presence in myocardial biopsies of COVID-19 patients with myocarditis [307309]. Cardiac tissues of COVID-19 patients with high viral invasion had increased expression of pro-inflammatory cytokines [307]. However, this finding was not related to the prominent invasion of the cells, thereby indicating that the presence of SARS-CoV-2 in the heart may not fundamentally cause an inflammatory response, which is in line with clinical myocarditis. The long-term consequences of SARS-CoV-2 infection and associated cardiovascular outcomes require advanced examination.
ACE2 is a part of RAAS and plays a critical role in the development of DM, hypertension, and HF. Disturbance of RAAS caused by loss of ACE2 in diabetic patients results in declined vascular function and systolic dysfunction that are dependent on the angiotensin II/AT1 receptor [310]. Since SARS-CoV-2 uses human ACE2 as cell entry receptors and human proteases as entry activators, RAAS is an important component of COVID-19 pathogenesis. Detailed mechanisms of SARS-CoV-2 entry into host cells have been recently reviewed in [311]. Imbalanced ACE/ACE2 and RAAS activation are responsible, at least in part, for COVID-19 progression which can lead to cardiac damage by inducing a hyperinflammatory response characterized by high levels of IL-6, IL-1, and TNF-α [312]. A recent meta-analysis that pooled 76,993 patients from 10 studies has demonstrated that hypertension, renal, liver and cerebrovascular diseases, obesity, smoking history, and current smoking are associated with the severity and poor prognosis of COVID-19 [313]. The mechanisms which increment vulnerability to COVID-19 infection in patients with DM may occur through ACE inhibitors or angiotensin receptor blockers which modulate ACE2 expression or through compromised immunity and elevated furin levels [314]. However, the Council on Hypertension of the European Society of Cardiology emphasized that there is no evidence supporting harmful effect of ACE inhibitors (ACE-I) or angiotensin receptor blockers (ARBs) during the COVID-19 outbreak. The Council strongly advise physicians and patients to continue their anti-hypertensive therapy treatment since there is no scientific or clinical basis for stopping ACEi or ARBs due to Covid-19 1.
A study by Abe et al. has shown that COVID-19 patients with DM had an increased incidence of acute myocarditis, acute HF, acute MI, and atrial fibrillation [315, 316]. Increased serum levels of IL-6, IL-8, and IP-10 were associated with a high mortality rate in COVID-19 patients [315]. The interplay between COVID-19 inflammatory response and DM may help to explain the poor cardiovascular prognosis of patients after SARS-CoV-2 infection, suggesting that interventions targeting innate immune pathways could potentially benefit COVID-19 patients with DM and other cardiovascular risk factors. Overall, applying therapeutic strategies to improve cardiovascular health may improve outcomes in COVID-19 patients, including patients with diabetes [317319].

ncRNAs in COVID-19

Several studies highlighted that altered levels of circulating cardiovascular, immunological, and inflammatory ncRNAs could be potential biomarkers of COVID-19 severity and myocardial damage. As an example, Garg et al. observed that inflammation-associated miR-155-5p, profibrogenic miRNA miR-21-5p, and cardiomyocyte-associated miRNAs miR-499-5p and miR-208a-3p were upregulated in the serum of severe COVID-19 patients requiring invasive ventilation in comparison to healthy controls and influenza-acute respiratory distress syndrome patients [320]. Another study showed that circulating myomiRNA miR-133a was upregulated in the plasma of severe compared to mild COVID-19 patients [321]. In particular, miR-133a overexpression was associated with intensive care unit (ICU) mortality within 28 days. Interestingly, myocardial injury correlated with an increase in circulating miR-133a levels [321]. In this setting, the main source of circulating miR-133a is most likely to be the cardiomyocytes, since they express high levels of this miRNA. However, miR-133a is also expressed in other cell types, such as neutrophils [319], that may be contributing sources of plasmatic miR-133a levels, considering that neutrophil degranulation and extravasation play a role in myocyte damage. Indeed, it was observed that circulating levels of miR-133a inversely correlate to neutrophil counts and directly correlate to myeloperoxidase, a neutrophil activation marker associated with cardiovascular outcomes in COVID-19 patients [322]. Other inflammation and cardiovascular-related miRNAs were modulated, e.g. miR-21-5p, miR-142‐3p, and miR‐146a‐5p were downregulated, and miR-15b-5p was upregulated, in red blood cell‐depleted whole blood of severe COVID-19 patients compared to moderate COVID-19 patients.
De Gonzalo-Calvo et al. [323] showed that miRNAs implicated in immune/inflammatory response and coagulation were altered in the plasma of COVID-19 patients. In particular, miR-27a-3p, miR-27b-3p, miR-148a-3p, miR-199a-5p and miR-491-5p were upregulated and miR-16-5p, miR-92a-3p, miR-150-5p, miR-451a and miR-486-5p were downregulated in ICU patients compared to ward patients. Moreover, three miRNAs (miR-148a-3p, miR-451a, and miR-486-5p) discriminated between ICU and ward patients. miR-16-5p, miR-92a-3p, miR-98-5p, miR-132-3p, miR-192-5p, and miR-323a-3p were downregulated between non-survivors and survivors to ICU stay. Two miRNAs (miR-192-5p and miR-323a-3p) distinguished between ICU non-survivors and survivors. Of note, a recent investigation has shown the potential of miRNAs, including miR-145a-5p and miR-181-5p, to define endotypes in critically ill COVID-19 patients [324], which suggests their usefulness in personalized medicine.
Recently, our investigations indicated that decreased plasma levels of miR-144-3p and − 5p were associated with both mortality and disease severity grade in three independent cohorts of hospitalized and non-hospitalized COVID-19 patients [325]. Thus, circulating miR-144 emerges as a noninvasive tool for early risk-based stratification and mortality prediction in COVID-19. This result is noteworthy in light of the reduced plasma levels of miR-144 observed in patients with DM with cardiac dysfunction [223].
Additional investigations have proposed that the disease not only impacts the circulating miRNA profile during the acute phase but also in “long-COVID-19”. Plasma levels miRNAs implicated in diverse immune mechanisms, e.g. miR-9-5p or miR-146a-5p, or miR-221-5p, have been associated with pulmonary dysfunction and radiologic abnormalities after hospital discharge in survivors of severe COVID-19 [326].
Collectively, the results show the association between COVID-19 severity and the alterations in the circulating miRNA profile, which may also contribute to the inflammatory effects of the disease. Indeed, circulating miRNAs could serve as biomarkers for clinical decision-making, but also as targets for developing specific therapies. For instance, Meidert et al. has proposed different EV-derived miRNAs, including let-7e-5p, miR-146a-5p, and miR-3168, as key regulators of signaling pathways implicated in inflammation and immune function in COVID-19 patients [327]. Results from independent publications support the participation of immune- and inflammatory-related miRNAs in the modulation of these mechanisms [328, 329]. In the context of metabolic disease, Wang et al. identified three antiviral miRNAs, miR-7-5p, miR-24-3p, and miR-223-3p, showing anti-inflammatory and cardio-protective roles, and remarkably decreased levels in serum exosomes of patients with DM compared to non-diabetic controls. The altered levels of these exosomal miRNAs in circulating exosomes weakened the inhibition of SARS-CoV-2 replication in patients with DM [49, 330, 331]. Therefore, miRNAs could serve as potential therapeutic candidates to prevent the progression from mild to severe COVID-19 disease in patients with DM by reducing viral replication.

Conclusions

Although the field of ncRNAs is still largely unexplored, it is now clear that ncRNAs play a crucial role in inflammation and DM. As described in this article, indeed, many ncRNAs are modulated in their expression in DM patients, can be transferred between inflammatory cells and cardiovascular cells inducing changes in their respective functions, representing in many cases viable therapeutic targets. Moreover, ncRNAs can be detected in the circulation representing valid biomarkers for the evolution of the disease.
Limitations to our understanding of ncRNAs in DM and inflammation are still present, due to incomplete sequence annotation (both structural and functional) and only partial conservation (more structural than sequence conservation). Publically available databases are dispersed and each one of them is incomplete in some aspect. In this respect, transcriptomic datasets are often only restricted to poly (A+), thus limiting the information to the mature form of mRNAs and a subset of lncRNAs.
Despite these difficulties, the current knowledge about ncRNAs has already allowed us to unveil many complex networks, shedding light on the multiple interactions governing the inflammatory response in the context of DM, and many more are likely to be identified in the upcoming years. We are beginning to understand that the potential of RNA therapy is enormous and results have started coming (ncRNAs clinical trials, development of RNA-based COVID-19 vaccines). Therefore, we believe that new larger cohort studies and further insights into the mechanisms of action will allow the clinical translation of prevention, prognosis, and cure of DM cardiovascular complications via ncRNA-based control of immune cell regulation.

Acknowledgements

This article is based upon work from EU-CardioRNA COST Action CA17129 (www.​cardiorna.​eu) supported by COST (European Cooperation in Science and Technology). Graphical abstract was created with BioRender.com online tool.

Declarations

Not applicable
Not applicable

Competing interest

YD holds patents related to diagnostic and therapeutic applications of RNAs. TT filed and licensed patents in the field of non-coding RNAs. TT is the founder and shareholder of Cardior Pharmaceuticals GmbH.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Sun H, Saeedi P, Karuranga S, Pinkepank M, Ogurtsova K, Duncan BB, et al. IDF Diabetes Atlas: Global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract. 2022;183:109119.PubMedCrossRef Sun H, Saeedi P, Karuranga S, Pinkepank M, Ogurtsova K, Duncan BB, et al. IDF Diabetes Atlas: Global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract. 2022;183:109119.PubMedCrossRef
2.
Zurück zum Zitat Read SH, Kerssens JJ, McAllister DA, Colhoun HM, Fischbacher CM, Lindsay RS, et al. Trends in type 2 diabetes incidence and mortality in Scotland between 2004 and 2013. Diabetologia. 2016;59(10):2106–13.PubMedPubMedCentralCrossRef Read SH, Kerssens JJ, McAllister DA, Colhoun HM, Fischbacher CM, Lindsay RS, et al. Trends in type 2 diabetes incidence and mortality in Scotland between 2004 and 2013. Diabetologia. 2016;59(10):2106–13.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Livingstone SJ, Levin D, Looker HC, Lindsay RS, Wild SH, Joss N, et al. Estimated life expectancy in a scottish cohort with type 1 diabetes, 2008–2010. JAMA. 2015;313(1):37–44.PubMedPubMedCentralCrossRef Livingstone SJ, Levin D, Looker HC, Lindsay RS, Wild SH, Joss N, et al. Estimated life expectancy in a scottish cohort with type 1 diabetes, 2008–2010. JAMA. 2015;313(1):37–44.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Negre-Salvayre A, Salvayre R, Augé N, Pamplona R, Portero-Otín M. Hyperglycemia and glycation in Diabetic Complications. Antioxidants & Redox Signaling. 2009;11(12):3071–109.CrossRef Negre-Salvayre A, Salvayre R, Augé N, Pamplona R, Portero-Otín M. Hyperglycemia and glycation in Diabetic Complications. Antioxidants & Redox Signaling. 2009;11(12):3071–109.CrossRef
5.
Zurück zum Zitat Rampin A, Carrabba M, Mutoli M, Eman CL, Testa G, Madeddu P, et al. Recent advances in KEAP1/NRF2-Targeting strategies by Phytochemical Antioxidants, Nanoparticles, and Biocompatible Scaffolds for the treatment of Diabetic Cardiovascular Complications. Antioxid Redox Signal. 2022;36(10–12):707–28.PubMedCrossRef Rampin A, Carrabba M, Mutoli M, Eman CL, Testa G, Madeddu P, et al. Recent advances in KEAP1/NRF2-Targeting strategies by Phytochemical Antioxidants, Nanoparticles, and Biocompatible Scaffolds for the treatment of Diabetic Cardiovascular Complications. Antioxid Redox Signal. 2022;36(10–12):707–28.PubMedCrossRef
6.
Zurück zum Zitat Santopaolo M, Sambataro M, Spinetti G, Madeddu P. Bone marrow as a target and accomplice of vascular complications in diabetes. Diabetes Metab Res Rev. 2020;36 Suppl 1:e3240.PubMed Santopaolo M, Sambataro M, Spinetti G, Madeddu P. Bone marrow as a target and accomplice of vascular complications in diabetes. Diabetes Metab Res Rev. 2020;36 Suppl 1:e3240.PubMed
7.
Zurück zum Zitat Wang Y, Shao T, Wang J, Huang X, Deng X, Cao Y, et al. An update on potential biomarkers for diagnosing diabetic foot ulcer at early stage. Biomed Pharmacother. 2021;133:110991.PubMedCrossRef Wang Y, Shao T, Wang J, Huang X, Deng X, Cao Y, et al. An update on potential biomarkers for diagnosing diabetic foot ulcer at early stage. Biomed Pharmacother. 2021;133:110991.PubMedCrossRef
8.
Zurück zum Zitat Forbes JM, Cooper ME. Mechanisms of diabetic complications. Physiol Rev. 2013;93(1):137–88.PubMedCrossRef Forbes JM, Cooper ME. Mechanisms of diabetic complications. Physiol Rev. 2013;93(1):137–88.PubMedCrossRef
9.
Zurück zum Zitat Tresierra-Ayala MÁ, García Rojas A. Association between peripheral arterial disease and diabetic foot ulcers in patients with diabetes mellitus type 2. Medicina Universitaria. 2017;19(76):123–6.CrossRef Tresierra-Ayala MÁ, García Rojas A. Association between peripheral arterial disease and diabetic foot ulcers in patients with diabetes mellitus type 2. Medicina Universitaria. 2017;19(76):123–6.CrossRef
10.
Zurück zum Zitat Ferraro F, Lymperi S, Mendez-Ferrer S, Saez B, Spencer JA, Yeap BY, et al. Diabetes impairs hematopoietic stem cell mobilization by altering niche function. Sci Transl Med. 2011;3(104):104ra1.CrossRef Ferraro F, Lymperi S, Mendez-Ferrer S, Saez B, Spencer JA, Yeap BY, et al. Diabetes impairs hematopoietic stem cell mobilization by altering niche function. Sci Transl Med. 2011;3(104):104ra1.CrossRef
12.
Zurück zum Zitat Berbudi A, Rahmadika N, Tjahjadi AI, Ruslami R. Type 2 diabetes and its impact on the Immune System. Curr Diabetes Rev. 2020;16(5):442–9.PubMedPubMedCentral Berbudi A, Rahmadika N, Tjahjadi AI, Ruslami R. Type 2 diabetes and its impact on the Immune System. Curr Diabetes Rev. 2020;16(5):442–9.PubMedPubMedCentral
13.
Zurück zum Zitat Palano MT, Cucchiara M, Gallazzi M, Riccio F, Mortara L, Gensini GF, et al. When a friend becomes your enemy: natural killer cells in atherosclerosis and Atherosclerosis-Associated Risk factors. Front Immunol. 2021;12:798155.PubMedCrossRef Palano MT, Cucchiara M, Gallazzi M, Riccio F, Mortara L, Gensini GF, et al. When a friend becomes your enemy: natural killer cells in atherosclerosis and Atherosclerosis-Associated Risk factors. Front Immunol. 2021;12:798155.PubMedCrossRef
14.
Zurück zum Zitat Spinetti G, Sangalli E, Tagliabue E, Maselli D, Colpani O, Ferland-McCollough D, et al. MicroRNA-21/PDCD4 Proapoptotic Signaling from circulating CD34(+) cells to vascular endothelial cells: a potential contributor to adverse Cardiovascular Outcomes in patients with critical limb ischemia. Diabetes Care. 2020;43(7):1520–9.PubMedPubMedCentralCrossRef Spinetti G, Sangalli E, Tagliabue E, Maselli D, Colpani O, Ferland-McCollough D, et al. MicroRNA-21/PDCD4 Proapoptotic Signaling from circulating CD34(+) cells to vascular endothelial cells: a potential contributor to adverse Cardiovascular Outcomes in patients with critical limb ischemia. Diabetes Care. 2020;43(7):1520–9.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Pezhman L, Tahrani A, Chimen M. Dysregulation of leukocyte trafficking in type 2 diabetes: mechanisms and potential therapeutic avenues. Front Cell Dev Biol. 2021;9:624184.PubMedPubMedCentralCrossRef Pezhman L, Tahrani A, Chimen M. Dysregulation of leukocyte trafficking in type 2 diabetes: mechanisms and potential therapeutic avenues. Front Cell Dev Biol. 2021;9:624184.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Jansen F, Li Q, Pfeifer A, Werner N. Endothelial- and Immune Cell-Derived Extracellular vesicles in the regulation of Cardiovascular Health and Disease. JACC Basic Transl Sci. 2017;2(6):790–807.PubMedPubMedCentralCrossRef Jansen F, Li Q, Pfeifer A, Werner N. Endothelial- and Immune Cell-Derived Extracellular vesicles in the regulation of Cardiovascular Health and Disease. JACC Basic Transl Sci. 2017;2(6):790–807.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Schober A, Blay RM, Saboor Maleki S, Zahedi F, Winklmaier AE, Kakar MY, et al. MicroRNA-21 controls circadian regulation of apoptosis in atherosclerotic lesions. Circulation. 2021;144(13):1059–73.PubMedCrossRef Schober A, Blay RM, Saboor Maleki S, Zahedi F, Winklmaier AE, Kakar MY, et al. MicroRNA-21 controls circadian regulation of apoptosis in atherosclerotic lesions. Circulation. 2021;144(13):1059–73.PubMedCrossRef
18.
Zurück zum Zitat Poller W, Dimmeler S, Heymans S, Zeller T, Haas J, Karakas M, et al. Non-coding RNAs in cardiovascular diseases: diagnostic and therapeutic perspectives. Eur Heart J. 2018;39(29):2704–16.PubMedCrossRef Poller W, Dimmeler S, Heymans S, Zeller T, Haas J, Karakas M, et al. Non-coding RNAs in cardiovascular diseases: diagnostic and therapeutic perspectives. Eur Heart J. 2018;39(29):2704–16.PubMedCrossRef
19.
Zurück zum Zitat Robinson EL, Baker AH, Brittan M, McCracken I, Condorelli G, Emanueli C, et al. Dissecting the transcriptome in cardiovascular disease. Cardiovasc Res. 2022;118(4):1004–19.PubMedCrossRef Robinson EL, Baker AH, Brittan M, McCracken I, Condorelli G, Emanueli C, et al. Dissecting the transcriptome in cardiovascular disease. Cardiovasc Res. 2022;118(4):1004–19.PubMedCrossRef
20.
Zurück zum Zitat Gomes CPC, Schroen B, Kuster GM, Robinson EL, Ford K, Squire IB, et al. Regulatory RNAs in Heart Failure. Circulation. 2020;141(4):313–28.PubMed Gomes CPC, Schroen B, Kuster GM, Robinson EL, Ford K, Squire IB, et al. Regulatory RNAs in Heart Failure. Circulation. 2020;141(4):313–28.PubMed
22.
23.
24.
Zurück zum Zitat van der Kwast R, van Ingen E, Parma L, Peters HAB, Quax PHA, Nossent AY. Adenosine-to-inosine editing of MicroRNA-487b alters Target Gene Selection after Ischemia and promotes neovascularization. Circ Res. 2018;122(3):444–56.PubMedCrossRef van der Kwast R, van Ingen E, Parma L, Peters HAB, Quax PHA, Nossent AY. Adenosine-to-inosine editing of MicroRNA-487b alters Target Gene Selection after Ischemia and promotes neovascularization. Circ Res. 2018;122(3):444–56.PubMedCrossRef
25.
Zurück zum Zitat van der Kwast R, Woudenberg T, Quax PHA, Nossent AY. MicroRNA-411 and its 5’-IsomiR have distinct targets and functions and are differentially regulated in the vasculature under Ischemia. Mol Ther. 2020;28(1):157–70.PubMedCrossRef van der Kwast R, Woudenberg T, Quax PHA, Nossent AY. MicroRNA-411 and its 5’-IsomiR have distinct targets and functions and are differentially regulated in the vasculature under Ischemia. Mol Ther. 2020;28(1):157–70.PubMedCrossRef
26.
Zurück zum Zitat Chamorro-Jorganes A, Sweaad WK, Katare R, Besnier M, Anwar M, Beazley-Long N, et al. METTL3 regulates angiogenesis by modulating let-7e-5p and miRNA-18a-5p expression in endothelial cells. Arterioscler Thromb Vasc Biol. 2021;41(6):e325-e37.CrossRef Chamorro-Jorganes A, Sweaad WK, Katare R, Besnier M, Anwar M, Beazley-Long N, et al. METTL3 regulates angiogenesis by modulating let-7e-5p and miRNA-18a-5p expression in endothelial cells. Arterioscler Thromb Vasc Biol. 2021;41(6):e325-e37.CrossRef
27.
Zurück zum Zitat Iwasaki YW, Siomi MC, Siomi H. PIWI-Interacting RNA: Its Biogenesis and Functions. Annu Rev Biochem. 2015;84:405–33.PubMedCrossRef Iwasaki YW, Siomi MC, Siomi H. PIWI-Interacting RNA: Its Biogenesis and Functions. Annu Rev Biochem. 2015;84:405–33.PubMedCrossRef
30.
Zurück zum Zitat Ingen E, Homberg DAL, Bent ML, Mei H, Papac-Milicevic N, Kremer V, et al. C/D box snoRNA SNORD113-6/AF357425 plays a dual role in integrin signalling and arterial fibroblast function via pre-mRNA processing and 2’O-ribose methylation. Hum Mol Genet. 2021. Ingen E, Homberg DAL, Bent ML, Mei H, Papac-Milicevic N, Kremer V, et al. C/D box snoRNA SNORD113-6/AF357425 plays a dual role in integrin signalling and arterial fibroblast function via pre-mRNA processing and 2’O-ribose methylation. Hum Mol Genet. 2021.
31.
Zurück zum Zitat Kishore S, Stamm S. The snoRNA HBII-52 regulates alternative splicing of the serotonin receptor 2 C. Science. 2006;311(5758):230–2.PubMedCrossRef Kishore S, Stamm S. The snoRNA HBII-52 regulates alternative splicing of the serotonin receptor 2 C. Science. 2006;311(5758):230–2.PubMedCrossRef
32.
Zurück zum Zitat Håkansson KEJ, Goossens EAC, Trompet S, van Ingen E, de Vries MR, van der Kwast R, et al. Genetic associations and regulation of expression indicate an independent role for 14q32 snoRNAs in human cardiovascular disease. Cardiovasc Res. 2019;115(10):1519–32.PubMedCrossRef Håkansson KEJ, Goossens EAC, Trompet S, van Ingen E, de Vries MR, van der Kwast R, et al. Genetic associations and regulation of expression indicate an independent role for 14q32 snoRNAs in human cardiovascular disease. Cardiovasc Res. 2019;115(10):1519–32.PubMedCrossRef
33.
Zurück zum Zitat Lee J, Harris AN, Holley CL, Mahadevan J, Pyles KD, Lavagnino Z, et al. Rpl13a small nucleolar RNAs regulate systemic glucose metabolism. J Clin Invest. 2016;126(12):4616–25.PubMedPubMedCentralCrossRef Lee J, Harris AN, Holley CL, Mahadevan J, Pyles KD, Lavagnino Z, et al. Rpl13a small nucleolar RNAs regulate systemic glucose metabolism. J Clin Invest. 2016;126(12):4616–25.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Burnett LC, Hubner G, LeDuc CA, Morabito MV, Carli JFM, Leibel RL. Loss of the imprinted, non-coding Snord116 gene cluster in the interval deleted in the Prader Willi syndrome results in murine neuronal and endocrine pancreatic developmental phenotypes. Hum Mol Genet. 2017;26(23):4606–16.PubMedPubMedCentralCrossRef Burnett LC, Hubner G, LeDuc CA, Morabito MV, Carli JFM, Leibel RL. Loss of the imprinted, non-coding Snord116 gene cluster in the interval deleted in the Prader Willi syndrome results in murine neuronal and endocrine pancreatic developmental phenotypes. Hum Mol Genet. 2017;26(23):4606–16.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Keam SP, Hutvagner G. tRNA-Derived fragments (tRFs): emerging New Roles for an ancient RNA in the regulation of Gene expression. Life (Basel). 2015;5(4):1638–51.PubMed Keam SP, Hutvagner G. tRNA-Derived fragments (tRFs): emerging New Roles for an ancient RNA in the regulation of Gene expression. Life (Basel). 2015;5(4):1638–51.PubMed
36.
Zurück zum Zitat Fu H, Feng J, Liu Q, Sun F, Tie Y, Zhu J, et al. Stress induces tRNA cleavage by angiogenin in mammalian cells. FEBS Lett. 2009;583(2):437–42.PubMedCrossRef Fu H, Feng J, Liu Q, Sun F, Tie Y, Zhu J, et al. Stress induces tRNA cleavage by angiogenin in mammalian cells. FEBS Lett. 2009;583(2):437–42.PubMedCrossRef
38.
39.
Zurück zum Zitat Winek K, Lobentanzer S, Nadorp B, Dubnov S, Dames C, Jagdmann S, et al. Transfer RNA fragments replace microRNA regulators of the cholinergic poststroke immune blockade. Proc Natl Acad Sci U S A. 2020;117(51):32606–16.PubMedPubMedCentralCrossRef Winek K, Lobentanzer S, Nadorp B, Dubnov S, Dames C, Jagdmann S, et al. Transfer RNA fragments replace microRNA regulators of the cholinergic poststroke immune blockade. Proc Natl Acad Sci U S A. 2020;117(51):32606–16.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Braun S, Domdey H, Wiebauer K. Inverse splicing of a discontinuous pre-mRNA intron generates a circular exon in a HeLa cell nuclear extract. Nucleic Acids Res. 1996;24(21):4152–7.PubMedPubMedCentralCrossRef Braun S, Domdey H, Wiebauer K. Inverse splicing of a discontinuous pre-mRNA intron generates a circular exon in a HeLa cell nuclear extract. Nucleic Acids Res. 1996;24(21):4152–7.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Mercer TR, Mattick JS. Structure and function of long noncoding RNAs in epigenetic regulation. Nat Struct Mol Biol. 2013;20(3):300–7.PubMedCrossRef Mercer TR, Mattick JS. Structure and function of long noncoding RNAs in epigenetic regulation. Nat Struct Mol Biol. 2013;20(3):300–7.PubMedCrossRef
43.
Zurück zum Zitat Hansen TB, Jensen TI, Clausen BH, Bramsen JB, Finsen B, Damgaard CK, et al. Natural RNA circles function as efficient microRNA sponges. Nature. 2013;495(7441):384–8.PubMedCrossRef Hansen TB, Jensen TI, Clausen BH, Bramsen JB, Finsen B, Damgaard CK, et al. Natural RNA circles function as efficient microRNA sponges. Nature. 2013;495(7441):384–8.PubMedCrossRef
44.
Zurück zum Zitat Memczak S, Jens M, Elefsinioti A, Torti F, Krueger J, Rybak A, et al. Circular RNAs are a large class of animal RNAs with regulatory potency. Nature. 2013;495(7441):333–8.PubMedCrossRef Memczak S, Jens M, Elefsinioti A, Torti F, Krueger J, Rybak A, et al. Circular RNAs are a large class of animal RNAs with regulatory potency. Nature. 2013;495(7441):333–8.PubMedCrossRef
45.
Zurück zum Zitat Carrara M, Fuschi P, Ivan C, Martelli F. Circular RNAs: methodological challenges and perspectives in cardiovascular diseases. J Cell Mol Med. 2018;22(11):5176–87.PubMedPubMedCentralCrossRef Carrara M, Fuschi P, Ivan C, Martelli F. Circular RNAs: methodological challenges and perspectives in cardiovascular diseases. J Cell Mol Med. 2018;22(11):5176–87.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Voellenkle C, Fuschi P, Mutoli M, Carrara M, Righini P, Nano G, et al. CircANKRD12 is Induced in endothelial cell response to oxidative stress. Cells. 2022;11(22).CrossRef Voellenkle C, Fuschi P, Mutoli M, Carrara M, Righini P, Nano G, et al. CircANKRD12 is Induced in endothelial cell response to oxidative stress. Cells. 2022;11(22).CrossRef
47.
Zurück zum Zitat Fernandes JCR, Acuna SM, Aoki JI, Floeter-Winter LM, Muxel SM. Long non-coding RNAs in the regulation of Gene expression: physiology and disease. Noncoding RNA. 2019;5(1). Fernandes JCR, Acuna SM, Aoki JI, Floeter-Winter LM, Muxel SM. Long non-coding RNAs in the regulation of Gene expression: physiology and disease. Noncoding RNA. 2019;5(1).
48.
Zurück zum Zitat Gong T, Liu L, Jiang W, Zhou R. DAMP-sensing receptors in sterile inflammation and inflammatory diseases. Nat Rev Immunol. 2020;20(2):95–112.PubMedCrossRef Gong T, Liu L, Jiang W, Zhou R. DAMP-sensing receptors in sterile inflammation and inflammatory diseases. Nat Rev Immunol. 2020;20(2):95–112.PubMedCrossRef
49.
Zurück zum Zitat Tahamtan A, Teymoori-Rad M, Nakstad B, Salimi V. Anti-inflammatory MicroRNAs and their potential for inflammatory Diseases treatment. Front Immunol. 2018;9:1377.PubMedPubMedCentralCrossRef Tahamtan A, Teymoori-Rad M, Nakstad B, Salimi V. Anti-inflammatory MicroRNAs and their potential for inflammatory Diseases treatment. Front Immunol. 2018;9:1377.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Xie Y, Wei Y. A Novel Regulatory Player in the Innate Immune System: long non-coding RNAs. Int J Mol Sci. 2021;22(17). Xie Y, Wei Y. A Novel Regulatory Player in the Innate Immune System: long non-coding RNAs. Int J Mol Sci. 2021;22(17).
51.
Zurück zum Zitat Tanwar VS, Reddy MA, Natarajan R. Emerging role of long non-coding RNAs in Diabetic Vascular Complications. Front Endocrinol (Lausanne). 2021;12:665811-.PubMedCrossRef Tanwar VS, Reddy MA, Natarajan R. Emerging role of long non-coding RNAs in Diabetic Vascular Complications. Front Endocrinol (Lausanne). 2021;12:665811-.PubMedCrossRef
52.
Zurück zum Zitat Jakubik D, Fitas A, Eyileten C, Jarosz-Popek J, Nowak A, Czajka P, et al. MicroRNAs and long non-coding RNAs in the pathophysiological processes of diabetic cardiomyopathy: emerging biomarkers and potential therapeutics. Cardiovasc Diabetol. 2021;20(1):55-.PubMedPubMedCentralCrossRef Jakubik D, Fitas A, Eyileten C, Jarosz-Popek J, Nowak A, Czajka P, et al. MicroRNAs and long non-coding RNAs in the pathophysiological processes of diabetic cardiomyopathy: emerging biomarkers and potential therapeutics. Cardiovasc Diabetol. 2021;20(1):55-.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Tang N, Jiang S, Yang Y, Liu S, Ponnusamy M, Xin H, et al. Noncoding RNAs as therapeutic targets in atherosclerosis with diabetes mellitus. Cardiovascular Therapeutics. 2018;36(4):e12436.PubMedCrossRef Tang N, Jiang S, Yang Y, Liu S, Ponnusamy M, Xin H, et al. Noncoding RNAs as therapeutic targets in atherosclerosis with diabetes mellitus. Cardiovascular Therapeutics. 2018;36(4):e12436.PubMedCrossRef
54.
Zurück zum Zitat Vistoli G, De Maddis D, Cipak A, Zarkovic N, Carini M, Aldini G. Advanced glycoxidation and lipoxidation end products (AGEs and ALEs): an overview of their mechanisms of formation. Free Radic Res. 2013;47 Suppl 1:3–27.PubMedCrossRef Vistoli G, De Maddis D, Cipak A, Zarkovic N, Carini M, Aldini G. Advanced glycoxidation and lipoxidation end products (AGEs and ALEs): an overview of their mechanisms of formation. Free Radic Res. 2013;47 Suppl 1:3–27.PubMedCrossRef
55.
Zurück zum Zitat Flynn MC, Kraakman MJ, Tikellis C, Lee MKS, Hanssen NMJ, Kammoun HL, et al. Transient intermittent hyperglycemia accelerates atherosclerosis by promoting Myelopoiesis. Circ Res. 2020;127(7):877–92.PubMedPubMedCentralCrossRef Flynn MC, Kraakman MJ, Tikellis C, Lee MKS, Hanssen NMJ, Kammoun HL, et al. Transient intermittent hyperglycemia accelerates atherosclerosis by promoting Myelopoiesis. Circ Res. 2020;127(7):877–92.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Di Marco E, Gray SP, Jandeleit-Dahm K. Diabetes alters activation and repression of pro- and anti-inflammatory signaling pathways in the vasculature. Front Endocrinol (Lausanne). 2013;4:68.PubMedCrossRef Di Marco E, Gray SP, Jandeleit-Dahm K. Diabetes alters activation and repression of pro- and anti-inflammatory signaling pathways in the vasculature. Front Endocrinol (Lausanne). 2013;4:68.PubMedCrossRef
57.
Zurück zum Zitat Girard D, Vandiedonck C. How dysregulation of the immune system promotes diabetes mellitus and cardiovascular risk complications. Front Cardiovasc Med. 2022;9:991716.PubMedPubMedCentralCrossRef Girard D, Vandiedonck C. How dysregulation of the immune system promotes diabetes mellitus and cardiovascular risk complications. Front Cardiovasc Med. 2022;9:991716.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Palella E, Cimino R, Pullano SA, Fiorillo AS, Gulletta E, Brunetti A, et al. Laboratory Parameters of Hemostasis, Adhesion Molecules, and inflammation in type 2 diabetes Mellitus: correlation with Glycemic Control. Int J Environ Res Public Health. 2020;17(1). Palella E, Cimino R, Pullano SA, Fiorillo AS, Gulletta E, Brunetti A, et al. Laboratory Parameters of Hemostasis, Adhesion Molecules, and inflammation in type 2 diabetes Mellitus: correlation with Glycemic Control. Int J Environ Res Public Health. 2020;17(1).
59.
Zurück zum Zitat Huang J, Xiao Y, Zheng P, Zhou W, Wang Y, Huang G, et al. Distinct neutrophil counts and functions in newly diagnosed type 1 diabetes, latent autoimmune diabetes in adults, and type 2 diabetes. Diabetes Metab Res Rev. 2019;35(1):e3064.PubMedCrossRef Huang J, Xiao Y, Zheng P, Zhou W, Wang Y, Huang G, et al. Distinct neutrophil counts and functions in newly diagnosed type 1 diabetes, latent autoimmune diabetes in adults, and type 2 diabetes. Diabetes Metab Res Rev. 2019;35(1):e3064.PubMedCrossRef
60.
Zurück zum Zitat Poznyak A, Grechko AV, Poggio P, Myasoedova VA, Alfieri V, Orekhov AN. The diabetes Mellitus-Atherosclerosis connection: the role of lipid and glucose metabolism and chronic inflammation. Int J Mol Sci. 2020;21(5). Poznyak A, Grechko AV, Poggio P, Myasoedova VA, Alfieri V, Orekhov AN. The diabetes Mellitus-Atherosclerosis connection: the role of lipid and glucose metabolism and chronic inflammation. Int J Mol Sci. 2020;21(5).
61.
Zurück zum Zitat Kanter JE, Hsu CC, Bornfeldt KE. Monocytes and macrophages as Protagonists in Vascular Complications of Diabetes. Front Cardiovasc Med. 2020;7:10.PubMedPubMedCentralCrossRef Kanter JE, Hsu CC, Bornfeldt KE. Monocytes and macrophages as Protagonists in Vascular Complications of Diabetes. Front Cardiovasc Med. 2020;7:10.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Subramanian M, Tabas I. Dendritic cells in atherosclerosis. Semin Immunopathol. 2014;36(1):93–102.PubMedCrossRef Subramanian M, Tabas I. Dendritic cells in atherosclerosis. Semin Immunopathol. 2014;36(1):93–102.PubMedCrossRef
64.
Zurück zum Zitat Choi JH, Do Y, Cheong C, Koh H, Boscardin SB, Oh YS, et al. Identification of antigen-presenting dendritic cells in mouse aorta and cardiac valves. J Exp Med. 2009;206(3):497–505.PubMedPubMedCentralCrossRef Choi JH, Do Y, Cheong C, Koh H, Boscardin SB, Oh YS, et al. Identification of antigen-presenting dendritic cells in mouse aorta and cardiac valves. J Exp Med. 2009;206(3):497–505.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Megens RT, Vijayan S, Lievens D, Doring Y, van Zandvoort MA, Grommes J, et al. Presence of luminal neutrophil extracellular traps in atherosclerosis. Thromb Haemost. 2012;107(3):597–8.PubMedCrossRef Megens RT, Vijayan S, Lievens D, Doring Y, van Zandvoort MA, Grommes J, et al. Presence of luminal neutrophil extracellular traps in atherosclerosis. Thromb Haemost. 2012;107(3):597–8.PubMedCrossRef
67.
Zurück zum Zitat Johnson J, Jaggers RM, Gopalkrishna S, Dahdah A, Murphy AJ, Hanssen NMJ, et al. Oxidative stress in neutrophils: implications for Diabetic Cardiovascular Complications. Antioxid Redox Signal. 2022;36(10–12):652–66.PubMedPubMedCentralCrossRef Johnson J, Jaggers RM, Gopalkrishna S, Dahdah A, Murphy AJ, Hanssen NMJ, et al. Oxidative stress in neutrophils: implications for Diabetic Cardiovascular Complications. Antioxid Redox Signal. 2022;36(10–12):652–66.PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Doring Y, Manthey HD, Drechsler M, Lievens D, Megens RT, Soehnlein O, et al. Auto-antigenic protein-DNA complexes stimulate plasmacytoid dendritic cells to promote atherosclerosis. Circulation. 2012;125(13):1673–83.PubMedCrossRef Doring Y, Manthey HD, Drechsler M, Lievens D, Megens RT, Soehnlein O, et al. Auto-antigenic protein-DNA complexes stimulate plasmacytoid dendritic cells to promote atherosclerosis. Circulation. 2012;125(13):1673–83.PubMedCrossRef
69.
70.
71.
Zurück zum Zitat Silvestri F, Banavali S, Baccarani M, Preisler HD. The CD34 hemopoietic progenitor cell associated antigen: biology and clinical applications. Haematologica. 1992;77(3):265–73.PubMed Silvestri F, Banavali S, Baccarani M, Preisler HD. The CD34 hemopoietic progenitor cell associated antigen: biology and clinical applications. Haematologica. 1992;77(3):265–73.PubMed
72.
Zurück zum Zitat Peichev M, Naiyer AJ, Pereira D, Zhu Z, Lane WJ, Williams M, et al. Expression of VEGFR-2 and AC133 by circulating human CD34(+) cells identifies a population of functional endothelial precursors. Blood. 2000;95(3):952–8.PubMedCrossRef Peichev M, Naiyer AJ, Pereira D, Zhu Z, Lane WJ, Williams M, et al. Expression of VEGFR-2 and AC133 by circulating human CD34(+) cells identifies a population of functional endothelial precursors. Blood. 2000;95(3):952–8.PubMedCrossRef
73.
Zurück zum Zitat Fadini GP, Boscaro E, de Kreutzenberg S, Agostini C, Seeger F, Dimmeler S, et al. Time course and mechanisms of circulating progenitor cell reduction in the natural history of type 2 diabetes. Diabetes Care. 2010;33(5):1097–102.PubMedPubMedCentralCrossRef Fadini GP, Boscaro E, de Kreutzenberg S, Agostini C, Seeger F, Dimmeler S, et al. Time course and mechanisms of circulating progenitor cell reduction in the natural history of type 2 diabetes. Diabetes Care. 2010;33(5):1097–102.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Fadini GP, Miorin M, Facco M, Bonamico S, Baesso I, Grego F, et al. Circulating endothelial progenitor cells are reduced in peripheral vascular complications of type 2 diabetes mellitus. J Am Coll Cardiol. 2005;45(9):1449–57.PubMedCrossRef Fadini GP, Miorin M, Facco M, Bonamico S, Baesso I, Grego F, et al. Circulating endothelial progenitor cells are reduced in peripheral vascular complications of type 2 diabetes mellitus. J Am Coll Cardiol. 2005;45(9):1449–57.PubMedCrossRef
75.
Zurück zum Zitat Spinetti G, Cordella D, Fortunato O, Sangalli E, Losa S, Gotti A, et al. Global remodeling of the vascular stem cell niche in bone marrow of diabetic patients: implication of the microRNA-155/FOXO3a signaling pathway. Circ Res. 2013;112(3):510–22.PubMedCrossRef Spinetti G, Cordella D, Fortunato O, Sangalli E, Losa S, Gotti A, et al. Global remodeling of the vascular stem cell niche in bone marrow of diabetic patients: implication of the microRNA-155/FOXO3a signaling pathway. Circ Res. 2013;112(3):510–22.PubMedCrossRef
76.
Zurück zum Zitat Tepper OM, Galiano RD, Capla JM, Kalka C, Gagne PJ, Jacobowitz GR, et al. Human endothelial progenitor cells from type II diabetics exhibit impaired proliferation, adhesion, and incorporation into vascular structures. Circulation. 2002;106(22):2781–6.PubMedCrossRef Tepper OM, Galiano RD, Capla JM, Kalka C, Gagne PJ, Jacobowitz GR, et al. Human endothelial progenitor cells from type II diabetics exhibit impaired proliferation, adhesion, and incorporation into vascular structures. Circulation. 2002;106(22):2781–6.PubMedCrossRef
77.
Zurück zum Zitat Spinetti G SE, Tagliabue E, Maselli D, Colpani O, Ferland-McCollough D, Carnelli F, Orlando P,Paccagnella A, Furlan A, Stefani PM, Sambado L, Sambataro M, Paolo Madeddu P. MicroRNA-21/PDCD4 proapoptotic signaling from circulating CD34 + cells to vascular endothelial cells: a potential contributor to adverse cardiovascular outcomes in patients with critical limb ischemia. Diabetes Care. 2020. Spinetti G SE, Tagliabue E, Maselli D, Colpani O, Ferland-McCollough D, Carnelli F, Orlando P,Paccagnella A, Furlan A, Stefani PM, Sambado L, Sambataro M, Paolo Madeddu P. MicroRNA-21/PDCD4 proapoptotic signaling from circulating CD34 + cells to vascular endothelial cells: a potential contributor to adverse cardiovascular outcomes in patients with critical limb ischemia. Diabetes Care. 2020.
78.
Zurück zum Zitat Spinetti G, Fortunato O, Caporali A, Shantikumar S, Marchetti M, Meloni M, et al. MicroRNA-15a and microRNA-16 impair human circulating proangiogenic cell functions and are increased in the proangiogenic cells and serum of patients with critical limb ischemia. Circ Res. 2013;112(2):335–46.PubMedCrossRef Spinetti G, Fortunato O, Caporali A, Shantikumar S, Marchetti M, Meloni M, et al. MicroRNA-15a and microRNA-16 impair human circulating proangiogenic cell functions and are increased in the proangiogenic cells and serum of patients with critical limb ischemia. Circ Res. 2013;112(2):335–46.PubMedCrossRef
79.
Zurück zum Zitat Besnier M, Gasparino S, Vono R, Sangalli E, Facoetti A, Bollati V, et al. miR-210 enhances the therapeutic potential of bone-marrow-derived circulating Proangiogenic cells in the setting of Limb Ischemia. Mol Ther. 2018;26(7):1694–705.PubMedPubMedCentralCrossRef Besnier M, Gasparino S, Vono R, Sangalli E, Facoetti A, Bollati V, et al. miR-210 enhances the therapeutic potential of bone-marrow-derived circulating Proangiogenic cells in the setting of Limb Ischemia. Mol Ther. 2018;26(7):1694–705.PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Bhatwadekar AD, Yan Y, Stepps V, Hazra S, Korah M, Bartelmez S, et al. miR-92a Corrects CD34 + cell dysfunction in diabetes by modulating core circadian genes involved in Progenitor differentiation. Diabetes. 2015;64(12):4226–37.PubMedPubMedCentralCrossRef Bhatwadekar AD, Yan Y, Stepps V, Hazra S, Korah M, Bartelmez S, et al. miR-92a Corrects CD34 + cell dysfunction in diabetes by modulating core circadian genes involved in Progenitor differentiation. Diabetes. 2015;64(12):4226–37.PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Mocharla P, Briand S, Giannotti G, Dorries C, Jakob P, Paneni F, et al. AngiomiR-126 expression and secretion from circulating CD34(+) and CD14(+) PBMCs: role for proangiogenic effects and alterations in type 2 diabetics. Blood. 2013;121(1):226–36.PubMedCrossRef Mocharla P, Briand S, Giannotti G, Dorries C, Jakob P, Paneni F, et al. AngiomiR-126 expression and secretion from circulating CD34(+) and CD14(+) PBMCs: role for proangiogenic effects and alterations in type 2 diabetics. Blood. 2013;121(1):226–36.PubMedCrossRef
83.
Zurück zum Zitat Njeim R, Azar WS, Fares AH, Azar ST, Kfoury Kassouf H, Eid AA. NETosis contributes to the pathogenesis of diabetes and its complications. J Mol Endocrinol. 2020;65(4):R65-R76.PubMedCrossRef Njeim R, Azar WS, Fares AH, Azar ST, Kfoury Kassouf H, Eid AA. NETosis contributes to the pathogenesis of diabetes and its complications. J Mol Endocrinol. 2020;65(4):R65-R76.PubMedCrossRef
84.
85.
Zurück zum Zitat Klopf J, Brostjan C, Eilenberg W, Neumayer C. Neutrophil Extracellular Traps and their implications in Cardiovascular and Inflammatory Disease. Int J Mol Sci. 2021;22(2). Klopf J, Brostjan C, Eilenberg W, Neumayer C. Neutrophil Extracellular Traps and their implications in Cardiovascular and Inflammatory Disease. Int J Mol Sci. 2021;22(2).
86.
Zurück zum Zitat Umehara T, Mori R, Mace KA, Murase T, Abe Y, Yamamoto T, et al. Identification of specific miRNAs in neutrophils of type 2 Diabetic mice: overexpression of miRNA-129-2-3p accelerates Diabetic Wound Healing. Diabetes. 2018;68(3):617–30.PubMedCrossRef Umehara T, Mori R, Mace KA, Murase T, Abe Y, Yamamoto T, et al. Identification of specific miRNAs in neutrophils of type 2 Diabetic mice: overexpression of miRNA-129-2-3p accelerates Diabetic Wound Healing. Diabetes. 2018;68(3):617–30.PubMedCrossRef
87.
Zurück zum Zitat Águila S, de los Reyes-García AM, Fernández-Pérez MP, Reguilón-Gallego L, Zapata-Martínez L, Ruiz-Lorente I, et al. MicroRNAs as new regulators of Neutrophil Extracellular trap formation. International Journal of Molecular Sciences. 2021;22(4). Águila S, de los Reyes-García AM, Fernández-Pérez MP, Reguilón-Gallego L, Zapata-Martínez L, Ruiz-Lorente I, et al. MicroRNAs as new regulators of Neutrophil Extracellular trap formation. International Journal of Molecular Sciences. 2021;22(4).
88.
Zurück zum Zitat Yuan X, Berg N, Lee JW, Le TT, Neudecker V, Jing N, et al. MicroRNA miR-223 as regulator of innate immunity. J Leukoc Biol. 2018;104(3):515–24.PubMedCrossRef Yuan X, Berg N, Lee JW, Le TT, Neudecker V, Jing N, et al. MicroRNA miR-223 as regulator of innate immunity. J Leukoc Biol. 2018;104(3):515–24.PubMedCrossRef
89.
Zurück zum Zitat Ana BA, María PF-P, Alberto del M, Sonia Á, Raúl M, Rebecca H-A, et al. miR-146a is a pivotal regulator of neutrophil extracellular trap formation promoting thrombosis. Haematologica. 2020;106(6):1636–46.CrossRef Ana BA, María PF-P, Alberto del M, Sonia Á, Raúl M, Rebecca H-A, et al. miR-146a is a pivotal regulator of neutrophil extracellular trap formation promoting thrombosis. Haematologica. 2020;106(6):1636–46.CrossRef
90.
Zurück zum Zitat Zhou C, Zhao L, Wang K, Qi Q, Wang M, Yang L, et al. MicroRNA-146a inhibits NF-kappaB activation and pro-inflammatory cytokine production by regulating IRAK1 expression in THP-1 cells. Exp Ther Med. 2019;18(4):3078–84.PubMedPubMedCentral Zhou C, Zhao L, Wang K, Qi Q, Wang M, Yang L, et al. MicroRNA-146a inhibits NF-kappaB activation and pro-inflammatory cytokine production by regulating IRAK1 expression in THP-1 cells. Exp Ther Med. 2019;18(4):3078–84.PubMedPubMedCentral
91.
Zurück zum Zitat Prame Kumar K, Nicholls AJ, Wong CHY. Partners in crime: neutrophils and monocytes/macrophages in inflammation and disease. Cell and Tissue Research. 2018;371(3):551–65.PubMedPubMedCentralCrossRef Prame Kumar K, Nicholls AJ, Wong CHY. Partners in crime: neutrophils and monocytes/macrophages in inflammation and disease. Cell and Tissue Research. 2018;371(3):551–65.PubMedPubMedCentralCrossRef
92.
Zurück zum Zitat Reddy MA, Amaram V, Das S, Tanwar VS, Ganguly R, Wang M, et al. lncRNA DRAIR is downregulated in diabetic monocytes and modulates the inflammatory phenotype via epigenetic mechanisms. JCI Insight. 2021;6(11). Reddy MA, Amaram V, Das S, Tanwar VS, Ganguly R, Wang M, et al. lncRNA DRAIR is downregulated in diabetic monocytes and modulates the inflammatory phenotype via epigenetic mechanisms. JCI Insight. 2021;6(11).
93.
Zurück zum Zitat Das S, Reddy MA, Senapati P, Stapleton K, Lanting L, Wang M, et al. Diabetes Mellitus-Induced Long Noncoding RNA Dnm3os regulates macrophage functions and inflammation via Nuclear Mechanisms. Arteriosclerosis, thrombosis, and vascular biology. 2018;38(8):1806–20. Das S, Reddy MA, Senapati P, Stapleton K, Lanting L, Wang M, et al. Diabetes Mellitus-Induced Long Noncoding RNA Dnm3os regulates macrophage functions and inflammation via Nuclear Mechanisms. Arteriosclerosis, thrombosis, and vascular biology. 2018;38(8):1806–20.
94.
Zurück zum Zitat Grebe A, Hoss F, Latz E. NLRP3 inflammasome and the IL-1 pathway in atherosclerosis. Circ Res. 2018;122(12):1722–40.PubMedCrossRef Grebe A, Hoss F, Latz E. NLRP3 inflammasome and the IL-1 pathway in atherosclerosis. Circ Res. 2018;122(12):1722–40.PubMedCrossRef
96.
Zurück zum Zitat Kelley N, Jeltema D, Duan Y, He Y. The NLRP3 inflammasome: an overview of mechanisms of activation and regulation. International journal of molecular sciences. 2019;20(13):3328.PubMedPubMedCentralCrossRef Kelley N, Jeltema D, Duan Y, He Y. The NLRP3 inflammasome: an overview of mechanisms of activation and regulation. International journal of molecular sciences. 2019;20(13):3328.PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat Zamani P, Oskuee RK, Atkin SL, Navashenaq JG, Sahebkar A. MicroRNAs as important regulators of the NLRP3 inflammasome. Progress in Biophysics and Molecular Biology. 2020;150:50–61.PubMedCrossRef Zamani P, Oskuee RK, Atkin SL, Navashenaq JG, Sahebkar A. MicroRNAs as important regulators of the NLRP3 inflammasome. Progress in Biophysics and Molecular Biology. 2020;150:50–61.PubMedCrossRef
98.
Zurück zum Zitat Bhatt K, Lanting LL, Jia Y, Yadav S, Reddy MA, Magilnick N, et al. Anti-inflammatory role of MicroRNA-146a in the Pathogenesis of Diabetic Nephropathy. Journal of the American Society of Nephrology: JASN. 2016;27(8):2277–88.PubMedCrossRef Bhatt K, Lanting LL, Jia Y, Yadav S, Reddy MA, Magilnick N, et al. Anti-inflammatory role of MicroRNA-146a in the Pathogenesis of Diabetic Nephropathy. Journal of the American Society of Nephrology: JASN. 2016;27(8):2277–88.PubMedCrossRef
99.
Zurück zum Zitat Yin R, Zhu X, Wang J, Yang S, Ma A, Xiao Q, et al. MicroRNA-155 promotes the ox-LDL-induced activation of NLRP3 inflammasomes via the ERK1/2 pathway in THP-1 macrophages and aggravates atherosclerosis in ApoE-/- mice. (2224–5839 (Electronic)). Yin R, Zhu X, Wang J, Yang S, Ma A, Xiao Q, et al. MicroRNA-155 promotes the ox-LDL-induced activation of NLRP3 inflammasomes via the ERK1/2 pathway in THP-1 macrophages and aggravates atherosclerosis in ApoE-/- mice. (2224–5839 (Electronic)).
100.
Zurück zum Zitat Stein N, Berhani O, Schmiedel D, Duev-Cohen A, Seidel E, Kol I, et al. IFNG-AS1 enhances Interferon Gamma production in human natural killer cells. iScience. 2019;11:466–73.PubMedPubMedCentralCrossRef Stein N, Berhani O, Schmiedel D, Duev-Cohen A, Seidel E, Kol I, et al. IFNG-AS1 enhances Interferon Gamma production in human natural killer cells. iScience. 2019;11:466–73.PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Ni F, Guo C, Sun R, Fu B, Yang Y, Wu L, et al. MicroRNA transcriptomes of distinct human NK cell populations identify mir-362-5p as an essential regulator of NK cell function. Scientific Reports. 2015;5(1):9993.PubMedPubMedCentralCrossRef Ni F, Guo C, Sun R, Fu B, Yang Y, Wu L, et al. MicroRNA transcriptomes of distinct human NK cell populations identify mir-362-5p as an essential regulator of NK cell function. Scientific Reports. 2015;5(1):9993.PubMedPubMedCentralCrossRef
102.
Zurück zum Zitat Palano MT, Cucchiara M, Gallazzi M, Riccio F, Mortara L, Gensini GF, et al. When a friend becomes your enemy: natural killer cells in atherosclerosis and Atherosclerosis-Associated Risk factors. Front Immunol. 2022;12. Palano MT, Cucchiara M, Gallazzi M, Riccio F, Mortara L, Gensini GF, et al. When a friend becomes your enemy: natural killer cells in atherosclerosis and Atherosclerosis-Associated Risk factors. Front Immunol. 2022;12.
103.
Zurück zum Zitat Wu C, Gong Y, Yuan J, Zhang W, Zhao G, Li H, et al. microRNA-181a represses ox-LDL-stimulated inflammatory response in dendritic cell by targeting c-Fos. J Lipid Res. 2012;53(11):2355–63.PubMedPubMedCentralCrossRef Wu C, Gong Y, Yuan J, Zhang W, Zhao G, Li H, et al. microRNA-181a represses ox-LDL-stimulated inflammatory response in dendritic cell by targeting c-Fos. J Lipid Res. 2012;53(11):2355–63.PubMedPubMedCentralCrossRef
104.
Zurück zum Zitat Liu X, Zhan Z, Xu L, Ma F, Li D, Guo Z, et al. MicroRNA-148/152 impair innate response and antigen presentation of TLR-triggered dendritic cells by targeting CaMKIIalpha. J Immunol. 2010;185(12):7244–51.PubMedCrossRef Liu X, Zhan Z, Xu L, Ma F, Li D, Guo Z, et al. MicroRNA-148/152 impair innate response and antigen presentation of TLR-triggered dendritic cells by targeting CaMKIIalpha. J Immunol. 2010;185(12):7244–51.PubMedCrossRef
105.
Zurück zum Zitat Steinman RM, Banchereau J. Taking dendritic cells into medicine. Nature. 2007;449(7161):419–26.PubMedCrossRef Steinman RM, Banchereau J. Taking dendritic cells into medicine. Nature. 2007;449(7161):419–26.PubMedCrossRef
106.
Zurück zum Zitat Niessner A, Weyand CM. Dendritic cells in atherosclerotic disease. Clinical Immunology. 2010;134(1):25–32.PubMedCrossRef Niessner A, Weyand CM. Dendritic cells in atherosclerotic disease. Clinical Immunology. 2010;134(1):25–32.PubMedCrossRef
107.
Zurück zum Zitat Rabb H. The T cell as a bridge between innate and adaptive immune systems: implications for the kidney. Kidney Int. 2002;61(6):1935–46.PubMedCrossRef Rabb H. The T cell as a bridge between innate and adaptive immune systems: implications for the kidney. Kidney Int. 2002;61(6):1935–46.PubMedCrossRef
108.
Zurück zum Zitat Yang TT, Song SJ, Xue HB, Shi DF, Liu CM, Liu H. Regulatory T cells in the pathogenesis of type 2 diabetes mellitus retinopathy by miR-155. Eur Rev Med Pharmacol Sci. 2015;19(11):2010–5.PubMed Yang TT, Song SJ, Xue HB, Shi DF, Liu CM, Liu H. Regulatory T cells in the pathogenesis of type 2 diabetes mellitus retinopathy by miR-155. Eur Rev Med Pharmacol Sci. 2015;19(11):2010–5.PubMed
109.
Zurück zum Zitat Xu S, Weng X, Wang Y, Lv D, Zeng M, Zhao F, et al. Screening and preliminary validation of T lymphocyte immunoregulation–associated long non–coding RNAs in diabetic foot ulcers. Mol Med Rep. 2019;19(3):2368–76.PubMed Xu S, Weng X, Wang Y, Lv D, Zeng M, Zhao F, et al. Screening and preliminary validation of T lymphocyte immunoregulation–associated long non–coding RNAs in diabetic foot ulcers. Mol Med Rep. 2019;19(3):2368–76.PubMed
110.
Zurück zum Zitat Chen HY, Zhong X, Huang XR, Meng XM, You Y, Chung AC, et al. MicroRNA-29b inhibits diabetic nephropathy in db/db mice. Mol Ther. 2014;22(4):842–53.PubMedPubMedCentralCrossRef Chen HY, Zhong X, Huang XR, Meng XM, You Y, Chung AC, et al. MicroRNA-29b inhibits diabetic nephropathy in db/db mice. Mol Ther. 2014;22(4):842–53.PubMedPubMedCentralCrossRef
111.
Zurück zum Zitat Ding H, Li J, Li Y, Yang M, Nie S, Zhou M, et al. MicroRNA-10 negatively regulates inflammation in diabetic kidney via targeting activation of the NLRP3 inflammasome. Mol Ther. 2021;29(7):2308–20.PubMedPubMedCentralCrossRef Ding H, Li J, Li Y, Yang M, Nie S, Zhou M, et al. MicroRNA-10 negatively regulates inflammation in diabetic kidney via targeting activation of the NLRP3 inflammasome. Mol Ther. 2021;29(7):2308–20.PubMedPubMedCentralCrossRef
113.
Zurück zum Zitat Adams DS, Levin M. Endogenous voltage gradients as mediators of cell-cell communication: strategies for investigating bioelectrical signals during pattern formation. Cell Tissue Res. 2013;352(1):95–122.PubMedCrossRef Adams DS, Levin M. Endogenous voltage gradients as mediators of cell-cell communication: strategies for investigating bioelectrical signals during pattern formation. Cell Tissue Res. 2013;352(1):95–122.PubMedCrossRef
114.
Zurück zum Zitat Armingol E, Officer A, Harismendy O, Lewis NE. Deciphering cell-cell interactions and communication from gene expression. Nat Rev Genet. 2021;22(2):71–88.PubMedCrossRef Armingol E, Officer A, Harismendy O, Lewis NE. Deciphering cell-cell interactions and communication from gene expression. Nat Rev Genet. 2021;22(2):71–88.PubMedCrossRef
115.
Zurück zum Zitat Makarova J, Turchinovich A, Shkurnikov M, Tonevitsky A. Extracellular miRNAs and cell–cell communication: problems and prospects. Trends in Biochemical Sciences. 2021;46(8):640–51.PubMedCrossRef Makarova J, Turchinovich A, Shkurnikov M, Tonevitsky A. Extracellular miRNAs and cell–cell communication: problems and prospects. Trends in Biochemical Sciences. 2021;46(8):640–51.PubMedCrossRef
117.
Zurück zum Zitat Di Bella MA. Overview and update on Extracellular vesicles: considerations on Exosomes and their application in Modern Medicine. Biology (Basel). 2022;11(6). Di Bella MA. Overview and update on Extracellular vesicles: considerations on Exosomes and their application in Modern Medicine. Biology (Basel). 2022;11(6).
118.
Zurück zum Zitat Rodrigues D, Rowland A. From endogenous compounds as biomarkers to plasma-derived nanovesicles as Liquid Biopsy; has the Golden Age of translational Pharmacokinetics-Absorption, distribution, metabolism, excretion-drug–drug Interaction Science finally arrived? Clinical Pharmacology & Therapeutics. 2019;105(6):1407–20.CrossRef Rodrigues D, Rowland A. From endogenous compounds as biomarkers to plasma-derived nanovesicles as Liquid Biopsy; has the Golden Age of translational Pharmacokinetics-Absorption, distribution, metabolism, excretion-drug–drug Interaction Science finally arrived? Clinical Pharmacology & Therapeutics. 2019;105(6):1407–20.CrossRef
119.
Zurück zum Zitat Coumans FAW, Brisson AR, Buzas EI, Dignat-George F, Drees EEE, El-Andaloussi S, et al. Methodological guidelines to study Extracellular vesicles. Circulation Research. 2017;120(10):1632–48.PubMedCrossRef Coumans FAW, Brisson AR, Buzas EI, Dignat-George F, Drees EEE, El-Andaloussi S, et al. Methodological guidelines to study Extracellular vesicles. Circulation Research. 2017;120(10):1632–48.PubMedCrossRef
120.
Zurück zum Zitat Zhang H, Freitas D, Kim HS, Fabijanic K, Li Z, Chen H, et al. Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation. Nat Cell Biol. 2018;20(3):332–43.PubMedPubMedCentralCrossRef Zhang H, Freitas D, Kim HS, Fabijanic K, Li Z, Chen H, et al. Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation. Nat Cell Biol. 2018;20(3):332–43.PubMedPubMedCentralCrossRef
121.
Zurück zum Zitat Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Tesic Mark M, et al. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527(7578):329–35.PubMedPubMedCentralCrossRef Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Tesic Mark M, et al. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527(7578):329–35.PubMedPubMedCentralCrossRef
122.
Zurück zum Zitat Shanmuganathan M, Vughs J, Noseda M, Emanueli C. Exosomes: Basic Biology and Technological advancements suggesting their potential as ischemic heart disease therapeutics. Front Physiol. 2018;9:1159-.PubMedPubMedCentralCrossRef Shanmuganathan M, Vughs J, Noseda M, Emanueli C. Exosomes: Basic Biology and Technological advancements suggesting their potential as ischemic heart disease therapeutics. Front Physiol. 2018;9:1159-.PubMedPubMedCentralCrossRef
123.
Zurück zum Zitat Beltrami C, Besnier M, Shantikumar S, Shearn AIU, Rajakaruna C, Laftah A, et al. Human pericardial fluid contains Exosomes enriched with Cardiovascular-Expressed MicroRNAs and promotes therapeutic angiogenesis. Mol Ther. 2017;25(3):679–93.PubMedPubMedCentralCrossRef Beltrami C, Besnier M, Shantikumar S, Shearn AIU, Rajakaruna C, Laftah A, et al. Human pericardial fluid contains Exosomes enriched with Cardiovascular-Expressed MicroRNAs and promotes therapeutic angiogenesis. Mol Ther. 2017;25(3):679–93.PubMedPubMedCentralCrossRef
124.
Zurück zum Zitat Wang X, Huang W, Liu G, Cai W, Millard RW, Wang Y, et al. Cardiomyocytes mediate anti-angiogenesis in type 2 diabetic rats through the exosomal transfer of miR-320 into endothelial cells. J Mol Cell Cardiol. 2014;74:139–50.PubMedPubMedCentralCrossRef Wang X, Huang W, Liu G, Cai W, Millard RW, Wang Y, et al. Cardiomyocytes mediate anti-angiogenesis in type 2 diabetic rats through the exosomal transfer of miR-320 into endothelial cells. J Mol Cell Cardiol. 2014;74:139–50.PubMedPubMedCentralCrossRef
125.
Zurück zum Zitat Litvinukova M, Talavera-Lopez C, Maatz H, Reichart D, Worth CL, Lindberg EL, et al. Cells of the adult human heart. Nature. 2020;588(7838):466–72.PubMedPubMedCentralCrossRef Litvinukova M, Talavera-Lopez C, Maatz H, Reichart D, Worth CL, Lindberg EL, et al. Cells of the adult human heart. Nature. 2020;588(7838):466–72.PubMedPubMedCentralCrossRef
127.
Zurück zum Zitat Bang C, Batkai S, Dangwal S, Gupta SK, Foinquinos A, Holzmann A, et al. Cardiac fibroblast-derived microRNA passenger strand-enriched exosomes mediate cardiomyocyte hypertrophy. J Clin Invest. 2014;124(5):2136–46.PubMedPubMedCentralCrossRef Bang C, Batkai S, Dangwal S, Gupta SK, Foinquinos A, Holzmann A, et al. Cardiac fibroblast-derived microRNA passenger strand-enriched exosomes mediate cardiomyocyte hypertrophy. J Clin Invest. 2014;124(5):2136–46.PubMedPubMedCentralCrossRef
128.
Zurück zum Zitat Kita S, Maeda N, Shimomura I. Interorgan communication by exosomes, adipose tissue, and adiponectin in metabolic syndrome. J Clin Invest. 2019;129(10):4041–9.PubMedPubMedCentralCrossRef Kita S, Maeda N, Shimomura I. Interorgan communication by exosomes, adipose tissue, and adiponectin in metabolic syndrome. J Clin Invest. 2019;129(10):4041–9.PubMedPubMedCentralCrossRef
129.
Zurück zum Zitat Lässer C, Jang SC, Lötvall J. Subpopulations of extracellular vesicles and their therapeutic potential. Molecular Aspects of Medicine. 2018;60:1–14.PubMedCrossRef Lässer C, Jang SC, Lötvall J. Subpopulations of extracellular vesicles and their therapeutic potential. Molecular Aspects of Medicine. 2018;60:1–14.PubMedCrossRef
130.
Zurück zum Zitat Baer C, Squadrito ML, Iruela-Arispe ML, De Palma M. Reciprocal interactions between endothelial cells and macrophages in angiogenic vascular niches. Exp Cell Res. 2013;319(11):1626–34.PubMedPubMedCentralCrossRef Baer C, Squadrito ML, Iruela-Arispe ML, De Palma M. Reciprocal interactions between endothelial cells and macrophages in angiogenic vascular niches. Exp Cell Res. 2013;319(11):1626–34.PubMedPubMedCentralCrossRef
131.
Zurück zum Zitat Akbar N, Digby JE, Cahill TJ, Tavare AN, Corbin AL, Saluja S, et al. Endothelium-derived extracellular vesicles promote splenic monocyte mobilization in myocardial infarction. JCI Insight. 2017;2(17):e93344.PubMedPubMedCentralCrossRef Akbar N, Digby JE, Cahill TJ, Tavare AN, Corbin AL, Saluja S, et al. Endothelium-derived extracellular vesicles promote splenic monocyte mobilization in myocardial infarction. JCI Insight. 2017;2(17):e93344.PubMedPubMedCentralCrossRef
132.
Zurück zum Zitat Stratmann B, Tschoepe D. The diabetic heart: sweet, fatty and stressed. Expert Review of Cardiovascular Therapy. 2011;9(9):1093–6.PubMedCrossRef Stratmann B, Tschoepe D. The diabetic heart: sweet, fatty and stressed. Expert Review of Cardiovascular Therapy. 2011;9(9):1093–6.PubMedCrossRef
133.
Zurück zum Zitat Barile L, Lionetti V, Cervio E, Matteucci M, Gherghiceanu M, Popescu LM, et al. Extracellular vesicles from human cardiac progenitor cells inhibit cardiomyocyte apoptosis and improve cardiac function after myocardial infarction. Cardiovasc Res. 2014;103(4):530–41.PubMedCrossRef Barile L, Lionetti V, Cervio E, Matteucci M, Gherghiceanu M, Popescu LM, et al. Extracellular vesicles from human cardiac progenitor cells inhibit cardiomyocyte apoptosis and improve cardiac function after myocardial infarction. Cardiovasc Res. 2014;103(4):530–41.PubMedCrossRef
134.
Zurück zum Zitat Loyer X, Zlatanova I, Devue C, Yin M, Howangyin KY, Klaihmon P, et al. Intra-Cardiac Release of Extracellular vesicles shapes inflammation following myocardial infarction. Circ Res. 2018;123(1):100–6.PubMedPubMedCentralCrossRef Loyer X, Zlatanova I, Devue C, Yin M, Howangyin KY, Klaihmon P, et al. Intra-Cardiac Release of Extracellular vesicles shapes inflammation following myocardial infarction. Circ Res. 2018;123(1):100–6.PubMedPubMedCentralCrossRef
135.
Zurück zum Zitat Sanz-Ruiz R, Climent AM, Fernandez-Santos ME, Villa Arranz A, Gutierrez Ibanes E, Vazquez-Alvarez ME, et al. General Overview of the 14th International Symposium on Stem Cell Therapy and Cardiovascular Innovations: Working Progress of a Global Initiative in 2017. Circ Res. 2017;121(9):1040-3. Sanz-Ruiz R, Climent AM, Fernandez-Santos ME, Villa Arranz A, Gutierrez Ibanes E, Vazquez-Alvarez ME, et al. General Overview of the 14th International Symposium on Stem Cell Therapy and Cardiovascular Innovations: Working Progress of a Global Initiative in 2017. Circ Res. 2017;121(9):1040-3.
136.
Zurück zum Zitat Mathiyalagan P, Liang Y, Kim D, Misener S, Thorne T, Kamide CE, et al. Angiogenic mechanisms of human CD34(+) stem cell exosomes in the repair of ischemic hindlimb. Circ Res. 2017;120(9):1466–76.PubMedPubMedCentralCrossRef Mathiyalagan P, Liang Y, Kim D, Misener S, Thorne T, Kamide CE, et al. Angiogenic mechanisms of human CD34(+) stem cell exosomes in the repair of ischemic hindlimb. Circ Res. 2017;120(9):1466–76.PubMedPubMedCentralCrossRef
137.
Zurück zum Zitat Kanter JE, Kramer F, Barnhart S, Averill MM, Vivekanandan-Giri A, Vickery T, et al. Diabetes promotes an inflammatory macrophage phenotype and atherosclerosis through acyl-CoA synthetase 1. Proc Natl Acad Sci U S A. 2012;109(12):E715-E24.CrossRef Kanter JE, Kramer F, Barnhart S, Averill MM, Vivekanandan-Giri A, Vickery T, et al. Diabetes promotes an inflammatory macrophage phenotype and atherosclerosis through acyl-CoA synthetase 1. Proc Natl Acad Sci U S A. 2012;109(12):E715-E24.CrossRef
139.
Zurück zum Zitat Sahoo S, Emanueli C. Exosomes in Diabetic Cardiomyopathy: the Next-Generation Therapeutic Targets? Diabetes. 2016;65(10):2829–31.PubMed Sahoo S, Emanueli C. Exosomes in Diabetic Cardiomyopathy: the Next-Generation Therapeutic Targets? Diabetes. 2016;65(10):2829–31.PubMed
140.
Zurück zum Zitat Caporali A, Meloni M, Völlenkle C, Bonci D, Sala-Newby GB, Addis R, et al. Deregulation of microRNA-503 contributes to Diabetes Mellitus–Induced impairment of endothelial function and reparative angiogenesis after limb ischemia. Circulation. 2011;123(3):282–91.PubMedCrossRef Caporali A, Meloni M, Völlenkle C, Bonci D, Sala-Newby GB, Addis R, et al. Deregulation of microRNA-503 contributes to Diabetes Mellitus–Induced impairment of endothelial function and reparative angiogenesis after limb ischemia. Circulation. 2011;123(3):282–91.PubMedCrossRef
141.
Zurück zum Zitat Andriantsitohaina R. Microparticles as a means of cell-to-cell communication in endothelial dysfunction associated with type 1 diabetes. Acta physiologica (Oxford, England). 2015;216. Andriantsitohaina R. Microparticles as a means of cell-to-cell communication in endothelial dysfunction associated with type 1 diabetes. Acta physiologica (Oxford, England). 2015;216.
142.
Zurück zum Zitat Zhang A, Li D, Liu Y, Li J, Zhang Y, Zhang C-Y. Islet β cell: an endocrine cell secreting miRNAs. Biochemical and Biophysical Research Communications. 2018;495(2):1648–54.PubMedCrossRef Zhang A, Li D, Liu Y, Li J, Zhang Y, Zhang C-Y. Islet β cell: an endocrine cell secreting miRNAs. Biochemical and Biophysical Research Communications. 2018;495(2):1648–54.PubMedCrossRef
143.
Zurück zum Zitat Vomund AN, Zinselmeyer BH, Hughes J, Calderon B, Valderrama C, Ferris ST, et al. Beta cells transfer vesicles containing insulin to phagocytes for presentation to T cells. Proc Natl Acad Sci U S A. 2015;112(40):E5496-E502.CrossRef Vomund AN, Zinselmeyer BH, Hughes J, Calderon B, Valderrama C, Ferris ST, et al. Beta cells transfer vesicles containing insulin to phagocytes for presentation to T cells. Proc Natl Acad Sci U S A. 2015;112(40):E5496-E502.CrossRef
144.
Zurück zum Zitat Cianciaruso C, Phelps EA, Pasquier M, Hamelin R, Demurtas D, Alibashe Ahmed M, et al. Primary human and rat β-Cells release the Intracellular Autoantigens GAD65, IA-2, and Proinsulin in Exosomes together with Cytokine-Induced Enhancers of Immunity. Diabetes. 2016;66(2):460–73.PubMedCrossRef Cianciaruso C, Phelps EA, Pasquier M, Hamelin R, Demurtas D, Alibashe Ahmed M, et al. Primary human and rat β-Cells release the Intracellular Autoantigens GAD65, IA-2, and Proinsulin in Exosomes together with Cytokine-Induced Enhancers of Immunity. Diabetes. 2016;66(2):460–73.PubMedCrossRef
145.
Zurück zum Zitat Hasilo CP, Negi S, Allaeys I, Cloutier N, Rutman AK, Gasparrini M, et al. Presence of diabetes autoantigens in extracellular vesicles derived from human islets. Scientific Reports. 2017;7(1):5000.PubMedPubMedCentralCrossRef Hasilo CP, Negi S, Allaeys I, Cloutier N, Rutman AK, Gasparrini M, et al. Presence of diabetes autoantigens in extracellular vesicles derived from human islets. Scientific Reports. 2017;7(1):5000.PubMedPubMedCentralCrossRef
146.
Zurück zum Zitat Giri KR, de Beaurepaire L, Jegou D, Lavy M, Mosser M, Dupont A, et al. Molecular and functional diversity of distinct subpopulations of the stressed insulin-secreting cell’s Vesiculome. Front Immunol. 2020;11:1814-.PubMedPubMedCentralCrossRef Giri KR, de Beaurepaire L, Jegou D, Lavy M, Mosser M, Dupont A, et al. Molecular and functional diversity of distinct subpopulations of the stressed insulin-secreting cell’s Vesiculome. Front Immunol. 2020;11:1814-.PubMedPubMedCentralCrossRef
147.
Zurück zum Zitat Liu T, Sun Y-C, Cheng P, Shao H-G. Adipose tissue macrophage-derived exosomal miR-29a regulates obesity-associated insulin resistance. Biochemical and Biophysical Research Communications. 2019;515(2):352–8.PubMedCrossRef Liu T, Sun Y-C, Cheng P, Shao H-G. Adipose tissue macrophage-derived exosomal miR-29a regulates obesity-associated insulin resistance. Biochemical and Biophysical Research Communications. 2019;515(2):352–8.PubMedCrossRef
148.
Zurück zum Zitat Ying W, Riopel M, Bandyopadhyay G, Dong Y, Birmingham A, Seo JB, et al. Adipose tissue macrophage-derived exosomal miRNAs can modulate in vivo and in Vitro insulin sensitivity. Cell. 2017;171(2):372 – 84.e12.PubMedCrossRef Ying W, Riopel M, Bandyopadhyay G, Dong Y, Birmingham A, Seo JB, et al. Adipose tissue macrophage-derived exosomal miRNAs can modulate in vivo and in Vitro insulin sensitivity. Cell. 2017;171(2):372 – 84.e12.PubMedCrossRef
149.
Zurück zum Zitat Caporali A, Meloni M, Nailor A, Mitić T, Shantikumar S, Riu F, et al. p75NTR-dependent activation of NF-κB regulates microRNA-503 transcription and pericyte–endothelial crosstalk in diabetes after limb ischaemia. Nature Communications. 2015;6(1):8024.PubMedCrossRef Caporali A, Meloni M, Nailor A, Mitić T, Shantikumar S, Riu F, et al. p75NTR-dependent activation of NF-κB regulates microRNA-503 transcription and pericyte–endothelial crosstalk in diabetes after limb ischaemia. Nature Communications. 2015;6(1):8024.PubMedCrossRef
150.
Zurück zum Zitat Tian F, Tang P, Sun Z, Zhang R, Zhu D, He J, et al. miR-210 in Exosomes Derived from Macrophages under high glucose promotes Mouse Diabetic obesity pathogenesis by suppressing NDUFA4 expression. J Diabetes Res. 2020;2020:6894684-.PubMedPubMedCentralCrossRef Tian F, Tang P, Sun Z, Zhang R, Zhu D, He J, et al. miR-210 in Exosomes Derived from Macrophages under high glucose promotes Mouse Diabetic obesity pathogenesis by suppressing NDUFA4 expression. J Diabetes Res. 2020;2020:6894684-.PubMedPubMedCentralCrossRef
151.
Zurück zum Zitat Zhu Q-J, Zhu M, Xu X-X, Meng X-M, Wu Y-G. Exosomes from high glucose–treated macrophages activate glomerular mesangial cells via TGF-β1/Smad3 pathway in vivo and in vitro. The FASEB Journal. 2019;33(8):9279–90.PubMedCrossRef Zhu Q-J, Zhu M, Xu X-X, Meng X-M, Wu Y-G. Exosomes from high glucose–treated macrophages activate glomerular mesangial cells via TGF-β1/Smad3 pathway in vivo and in vitro. The FASEB Journal. 2019;33(8):9279–90.PubMedCrossRef
152.
Zurück zum Zitat Zhu M, Sun X, Qi X, Xia L, Wu Y. Exosomes from high glucose-treated macrophages activate macrophages andinduce inflammatory responses via NF-κB signaling pathway in vitro and in vivo. International Immunopharmacology. 2020;84:106551.PubMedCrossRef Zhu M, Sun X, Qi X, Xia L, Wu Y. Exosomes from high glucose-treated macrophages activate macrophages andinduce inflammatory responses via NF-κB signaling pathway in vitro and in vivo. International Immunopharmacology. 2020;84:106551.PubMedCrossRef
153.
Zurück zum Zitat Kawakami R, Katsuki S, Travers R, Romero DC, Becker-Greene D, Passos LSA, et al. S100A9-RAGE Axis Accelerates Formation of Macrophage-Mediated Extracellular Vesicle Microcalcification in Diabetes Mellitus. Arteriosclerosis, Thrombosis, and Vascular Biology. 2020;40(8):1838-53. Kawakami R, Katsuki S, Travers R, Romero DC, Becker-Greene D, Passos LSA, et al. S100A9-RAGE Axis Accelerates Formation of Macrophage-Mediated Extracellular Vesicle Microcalcification in Diabetes Mellitus. Arteriosclerosis, Thrombosis, and Vascular Biology. 2020;40(8):1838-53.
154.
Zurück zum Zitat Ding X, Jing N, Shen A, Guo F, Song Y, Pan M, et al. MiR-21-5p in macrophage-derived extracellular vesicles affects podocyte pyroptosis in diabetic nephropathy by regulating A20. Journal of Endocrinological Investigation. 2021;44(6):1175–84.PubMedCrossRef Ding X, Jing N, Shen A, Guo F, Song Y, Pan M, et al. MiR-21-5p in macrophage-derived extracellular vesicles affects podocyte pyroptosis in diabetic nephropathy by regulating A20. Journal of Endocrinological Investigation. 2021;44(6):1175–84.PubMedCrossRef
155.
Zurück zum Zitat Bouchareychas L, Duong P, Phu TA, Alsop E, Meechoovet B, Reiman R, et al. High glucose macrophage exosomes enhance atherosclerosis by driving cellular proliferation & hematopoiesis. iScience. 2021;24(8):102847-.PubMedPubMedCentralCrossRef Bouchareychas L, Duong P, Phu TA, Alsop E, Meechoovet B, Reiman R, et al. High glucose macrophage exosomes enhance atherosclerosis by driving cellular proliferation & hematopoiesis. iScience. 2021;24(8):102847-.PubMedPubMedCentralCrossRef
156.
Zurück zum Zitat Liu Y, Song J-W, Lin J-Y, Miao R, Zhong J-C. Roles of MicroRNA-122 in Cardiovascular Fibrosis and Related Diseases. Cardiovasc Toxicol. 2020;20(5):463–73.PubMedPubMedCentralCrossRef Liu Y, Song J-W, Lin J-Y, Miao R, Zhong J-C. Roles of MicroRNA-122 in Cardiovascular Fibrosis and Related Diseases. Cardiovasc Toxicol. 2020;20(5):463–73.PubMedPubMedCentralCrossRef
157.
Zurück zum Zitat Li K, Cui M, Zhang K, Wang G, Zhai S. M1 macrophages-derived extracellular vesicles elevate microRNA-185-3p to aggravate the development of atherosclerosis in ApoE-/- mice by inhibiting small mothers against decapentaplegic 7. International Immunopharmacology. 2021;90:107138.PubMedCrossRef Li K, Cui M, Zhang K, Wang G, Zhai S. M1 macrophages-derived extracellular vesicles elevate microRNA-185-3p to aggravate the development of atherosclerosis in ApoE-/- mice by inhibiting small mothers against decapentaplegic 7. International Immunopharmacology. 2021;90:107138.PubMedCrossRef
158.
Zurück zum Zitat Guay C, Kruit JK, Rome S, Menoud V, Mulder NL, Jurdzinski A, et al. Lymphocyte-derived exosomal MicroRNAs promote pancreatic β cell death and may contribute to type 1 Diabetes Development. Cell Metabolism. 2019;29(2):348 – 61.e6.PubMedCrossRef Guay C, Kruit JK, Rome S, Menoud V, Mulder NL, Jurdzinski A, et al. Lymphocyte-derived exosomal MicroRNAs promote pancreatic β cell death and may contribute to type 1 Diabetes Development. Cell Metabolism. 2019;29(2):348 – 61.e6.PubMedCrossRef
159.
Zurück zum Zitat Mukherjee K, Ghoshal B, Ghosh S, Chakrabarty Y, Shwetha S, Das S, et al. Reversible HuR-microRNA binding controls extracellular export of miR-122 and augments stress response. EMBO Rep. 2016;17(8):1184–203.PubMedPubMedCentralCrossRef Mukherjee K, Ghoshal B, Ghosh S, Chakrabarty Y, Shwetha S, Das S, et al. Reversible HuR-microRNA binding controls extracellular export of miR-122 and augments stress response. EMBO Rep. 2016;17(8):1184–203.PubMedPubMedCentralCrossRef
160.
Zurück zum Zitat Fu Z, Zhou E, Wang X, Tian M, Kong J, Li J, et al. Oxidized low-density lipoprotein-induced microparticles promote endothelial monocyte adhesion via intercellular adhesion molecule 1. American Journal of Physiology-Cell Physiology. 2017;313(5):C567-C74.CrossRef Fu Z, Zhou E, Wang X, Tian M, Kong J, Li J, et al. Oxidized low-density lipoprotein-induced microparticles promote endothelial monocyte adhesion via intercellular adhesion molecule 1. American Journal of Physiology-Cell Physiology. 2017;313(5):C567-C74.CrossRef
161.
Zurück zum Zitat Fan D, Kassiri Z. Biology of tissue inhibitor of metalloproteinase 3 (TIMP3), and its therapeutic implications in Cardiovascular Pathology. Front Physiol. 2020;11. Fan D, Kassiri Z. Biology of tissue inhibitor of metalloproteinase 3 (TIMP3), and its therapeutic implications in Cardiovascular Pathology. Front Physiol. 2020;11.
164.
Zurück zum Zitat Bras JP, Silva AM, Calin GA, Barbosa MA, Santos SG, Almeida MI. miR-195 inhibits macrophages pro-inflammatory profile and impacts the crosstalk with smooth muscle cells. PLoS One. 2017;12(11):e0188530-e.CrossRef Bras JP, Silva AM, Calin GA, Barbosa MA, Santos SG, Almeida MI. miR-195 inhibits macrophages pro-inflammatory profile and impacts the crosstalk with smooth muscle cells. PLoS One. 2017;12(11):e0188530-e.CrossRef
165.
Zurück zum Zitat Hu Y-W, Guo F-X, Xu Y-J, Li P, Lu Z-F, McVey DG, et al. Long noncoding RNA NEXN-AS1 mitigates atherosclerosis by regulating the actin-binding protein NEXN. J Clin Invest. 2019;129(3):1115–28.PubMedPubMedCentralCrossRef Hu Y-W, Guo F-X, Xu Y-J, Li P, Lu Z-F, McVey DG, et al. Long noncoding RNA NEXN-AS1 mitigates atherosclerosis by regulating the actin-binding protein NEXN. J Clin Invest. 2019;129(3):1115–28.PubMedPubMedCentralCrossRef
166.
Zurück zum Zitat Wu L-M, Wu S-G, Chen F, Wu Q, Wu C-M, Kang C-M, et al. Atorvastatin inhibits pyroptosis through the lncRNA NEXN-AS1/NEXN pathway in human vascular endothelial cells. Atherosclerosis. 2020;293:26–34.PubMedCrossRef Wu L-M, Wu S-G, Chen F, Wu Q, Wu C-M, Kang C-M, et al. Atorvastatin inhibits pyroptosis through the lncRNA NEXN-AS1/NEXN pathway in human vascular endothelial cells. Atherosclerosis. 2020;293:26–34.PubMedCrossRef
167.
Zurück zum Zitat Zhan R, Leng X, Liu X, Wang X, Gong J, Yan L, et al. Heat shock protein 70 is secreted from endothelial cells by a non-classical pathway involving exosomes. Biochemical and Biophysical Research Communications. 2009;387(2):229–33.PubMedCrossRef Zhan R, Leng X, Liu X, Wang X, Gong J, Yan L, et al. Heat shock protein 70 is secreted from endothelial cells by a non-classical pathway involving exosomes. Biochemical and Biophysical Research Communications. 2009;387(2):229–33.PubMedCrossRef
168.
Zurück zum Zitat Zhang Z, Yi D, Zhou J, Zheng Y, Gao Z, Hu X, et al. Exosomal LINC01005 derived from oxidized low-density lipoprotein-treated endothelial cells regulates vascular smooth muscle cell phenotypic switch. BioFactors. 2020;46(5):743–53.PubMedCrossRef Zhang Z, Yi D, Zhou J, Zheng Y, Gao Z, Hu X, et al. Exosomal LINC01005 derived from oxidized low-density lipoprotein-treated endothelial cells regulates vascular smooth muscle cell phenotypic switch. BioFactors. 2020;46(5):743–53.PubMedCrossRef
169.
Zurück zum Zitat Huang C, Han J, Wu Y, Li S, Wang Q, Lin W, et al. Exosomal MALAT1 derived from oxidized low-density lipoprotein-treated endothelial cells promotes M2 macrophage polarization. Mol Med Rep. 2018;18(1):509–15.PubMed Huang C, Han J, Wu Y, Li S, Wang Q, Lin W, et al. Exosomal MALAT1 derived from oxidized low-density lipoprotein-treated endothelial cells promotes M2 macrophage polarization. Mol Med Rep. 2018;18(1):509–15.PubMed
170.
Zurück zum Zitat Zhu C, Zhang H, Wei D, Sun Z. Silencing lncRNA GAS5 alleviates apoptosis and fibrosis in diabetic cardiomyopathy by targeting miR-26a/b-5p. Acta Diabetologica. 2021;58(11):1491–501.PubMedCrossRef Zhu C, Zhang H, Wei D, Sun Z. Silencing lncRNA GAS5 alleviates apoptosis and fibrosis in diabetic cardiomyopathy by targeting miR-26a/b-5p. Acta Diabetologica. 2021;58(11):1491–501.PubMedCrossRef
171.
Zurück zum Zitat Wang L, Qi Y, Wang Y, Tang H, Li Z, Wang Y, et al. LncRNA MALAT1 suppression protects endothelium against oxLDL-Induced inflammation via inhibiting expression of MiR-181b target gene TOX. Oxidative Medicine and Cellular Longevity. 2019;2019:8245810.PubMedPubMedCentralCrossRef Wang L, Qi Y, Wang Y, Tang H, Li Z, Wang Y, et al. LncRNA MALAT1 suppression protects endothelium against oxLDL-Induced inflammation via inhibiting expression of MiR-181b target gene TOX. Oxidative Medicine and Cellular Longevity. 2019;2019:8245810.PubMedPubMedCentralCrossRef
172.
Zurück zum Zitat Wang S, Shi M, Li J, Zhang Y, Wang W, Xu P, et al. Endothelial cell-derived exosomal circHIPK3 promotes the proliferation of vascular smooth muscle cells induced by high glucose via the miR-106a-5p/Foxo1/Vcam1 pathway. Aging (Albany NY). 2021;13(23):25241–55.PubMedCrossRef Wang S, Shi M, Li J, Zhang Y, Wang W, Xu P, et al. Endothelial cell-derived exosomal circHIPK3 promotes the proliferation of vascular smooth muscle cells induced by high glucose via the miR-106a-5p/Foxo1/Vcam1 pathway. Aging (Albany NY). 2021;13(23):25241–55.PubMedCrossRef
173.
Zurück zum Zitat Ye L, Guo H, Wang Y, Peng Y, Zhang Y, Li S, et al. Exosomal circEhmt1 released from Hypoxia-Pretreated Pericytes regulates high Glucose-Induced Microvascular Dysfunction via the NFIA/NLRP3 pathway. Oxid Med Cell Longev. 2021;2021:8833098.PubMedPubMedCentralCrossRef Ye L, Guo H, Wang Y, Peng Y, Zhang Y, Li S, et al. Exosomal circEhmt1 released from Hypoxia-Pretreated Pericytes regulates high Glucose-Induced Microvascular Dysfunction via the NFIA/NLRP3 pathway. Oxid Med Cell Longev. 2021;2021:8833098.PubMedPubMedCentralCrossRef
174.
Zurück zum Zitat Gaignebet L, Kararigas G. En route to precision medicine through the integration of biological sex into pharmacogenomics. Clin Sci (Lond). 2017;131(4):329–42.PubMedCrossRef Gaignebet L, Kararigas G. En route to precision medicine through the integration of biological sex into pharmacogenomics. Clin Sci (Lond). 2017;131(4):329–42.PubMedCrossRef
175.
Zurück zum Zitat Horvath C, Kararigas G. Sex-dependent mechanisms of cell death modalities in Cardiovascular Disease. Can J Cardiol. 2022;38(12):1844–53.PubMedCrossRef Horvath C, Kararigas G. Sex-dependent mechanisms of cell death modalities in Cardiovascular Disease. Can J Cardiol. 2022;38(12):1844–53.PubMedCrossRef
176.
Zurück zum Zitat Siokatas G, Papatheodorou I, Daiou A, Lazou A, Hatzistergos KE, Kararigas G. Sex-related Effects on Cardiac Development and Disease. J Cardiovasc Dev Dis. 2022;9(3). Siokatas G, Papatheodorou I, Daiou A, Lazou A, Hatzistergos KE, Kararigas G. Sex-related Effects on Cardiac Development and Disease. J Cardiovasc Dev Dis. 2022;9(3).
177.
Zurück zum Zitat Kararigas G, Seeland U, Barcena de Arellano ML, Dworatzek E, Regitz-Zagrosek V. Why the study of the effects of biological sex is important. Commentary. Ann Ist Super Sanita. 2016;52(2):149–50.PubMed Kararigas G, Seeland U, Barcena de Arellano ML, Dworatzek E, Regitz-Zagrosek V. Why the study of the effects of biological sex is important. Commentary. Ann Ist Super Sanita. 2016;52(2):149–50.PubMed
178.
Zurück zum Zitat Cui C, Huang C, Liu K, Xu G, Yang J, Zhou Y, et al. Large-scale in silico identification of drugs exerting sex-specific effects in the heart. J Transl Med. 2018;16(1):236.PubMedPubMedCentralCrossRef Cui C, Huang C, Liu K, Xu G, Yang J, Zhou Y, et al. Large-scale in silico identification of drugs exerting sex-specific effects in the heart. J Transl Med. 2018;16(1):236.PubMedPubMedCentralCrossRef
179.
Zurück zum Zitat Ruiz-Meana M, Boengler K, Garcia-Dorado D, Hausenloy DJ, Kaambre T, Kararigas G, et al. Ageing, sex, and cardioprotection. Br J Pharmacol. 2020;177(23):5270–86.PubMedPubMedCentralCrossRef Ruiz-Meana M, Boengler K, Garcia-Dorado D, Hausenloy DJ, Kaambre T, Kararigas G, et al. Ageing, sex, and cardioprotection. Br J Pharmacol. 2020;177(23):5270–86.PubMedPubMedCentralCrossRef
180.
Zurück zum Zitat Franconi F, Campesi I. Pharmacogenomics, pharmacokinetics and pharmacodynamics: interaction with biological differences between men and women. Br J Pharmacol. 2014;171(3):580–94.PubMedPubMedCentralCrossRef Franconi F, Campesi I. Pharmacogenomics, pharmacokinetics and pharmacodynamics: interaction with biological differences between men and women. Br J Pharmacol. 2014;171(3):580–94.PubMedPubMedCentralCrossRef
181.
Zurück zum Zitat Gaignebet L, Kańduła MM, Lehmann D, Knosalla C, Kreil DP, Kararigas G. Sex-Specific Human Cardiomyocyte Gene Regulation in Left Ventricular Pressure Overload. Mayo Clinic Proceedings. 2020;95(4):688 – 97. Gaignebet L, Kańduła MM, Lehmann D, Knosalla C, Kreil DP, Kararigas G. Sex-Specific Human Cardiomyocyte Gene Regulation in Left Ventricular Pressure Overload. Mayo Clinic Proceedings. 2020;95(4):688 – 97.
182.
Zurück zum Zitat Jusic A, Salgado-Somoza A, Paes AB, Stefanizzi FM, Martinez-Alarcon N, Pinet F, et al. Approaching sex differences in Cardiovascular non-coding RNA research. Int J Mol Sci. 2020;21(14). Jusic A, Salgado-Somoza A, Paes AB, Stefanizzi FM, Martinez-Alarcon N, Pinet F, et al. Approaching sex differences in Cardiovascular non-coding RNA research. Int J Mol Sci. 2020;21(14).
183.
Zurück zum Zitat Decode Study Group obotEDESG. Will new diagnostic criteria for diabetes mellitus change phenotype of patients with diabetes? Reanalysis of european epidemiological data. DECODE Study Group on behalf of the european diabetes Epidemiology Study Group. BMJ. 1998;317(7155):371–5.CrossRef Decode Study Group obotEDESG. Will new diagnostic criteria for diabetes mellitus change phenotype of patients with diabetes? Reanalysis of european epidemiological data. DECODE Study Group on behalf of the european diabetes Epidemiology Study Group. BMJ. 1998;317(7155):371–5.CrossRef
184.
Zurück zum Zitat Qiao Q, Nakagami T, Tuomilehto J, Borch-Johnsen K, Balkau B, Iwamoto Y, et al. Comparison of the fasting and the 2-h glucose criteria for diabetes in different asian cohorts. Diabetologia. 2000;43(12):1470–5.PubMedCrossRef Qiao Q, Nakagami T, Tuomilehto J, Borch-Johnsen K, Balkau B, Iwamoto Y, et al. Comparison of the fasting and the 2-h glucose criteria for diabetes in different asian cohorts. Diabetologia. 2000;43(12):1470–5.PubMedCrossRef
185.
Zurück zum Zitat Qiao Q, Hu G, Tuomilehto J, Nakagami T, Balkau B, Borch-Johnsen K, et al. Age- and sex-specific prevalence of diabetes and impaired glucose regulation in 11 asian cohorts. Diabetes Care. 2003;26(6):1770–80.PubMedCrossRef Qiao Q, Hu G, Tuomilehto J, Nakagami T, Balkau B, Borch-Johnsen K, et al. Age- and sex-specific prevalence of diabetes and impaired glucose regulation in 11 asian cohorts. Diabetes Care. 2003;26(6):1770–80.PubMedCrossRef
186.
Zurück zum Zitat García-Llorca A, Kararigas G. Sex-related Effects of Gut Microbiota in Metabolic Syndrome-Related Diabetic Retinopathy. Microorganisms [Internet]. 2023; 11(2). García-Llorca A, Kararigas G. Sex-related Effects of Gut Microbiota in Metabolic Syndrome-Related Diabetic Retinopathy. Microorganisms [Internet]. 2023; 11(2).
187.
Zurück zum Zitat El Khoudary SR, Aggarwal B, Beckie TM, Hodis HN, Johnson AE, Langer RD, et al. Menopause transition and Cardiovascular Disease Risk: implications for timing of early Prevention: A Scientific Statement from the American Heart Association. Circulation. 2020;142(25):e506-e32.CrossRef El Khoudary SR, Aggarwal B, Beckie TM, Hodis HN, Johnson AE, Langer RD, et al. Menopause transition and Cardiovascular Disease Risk: implications for timing of early Prevention: A Scientific Statement from the American Heart Association. Circulation. 2020;142(25):e506-e32.CrossRef
188.
Zurück zum Zitat Maas A, Rosano G, Cifkova R, Chieffo A, van Dijken D, Hamoda H, et al. Cardiovascular health after menopause transition, pregnancy disorders, and other gynaecologic conditions: a consensus document from european cardiologists, gynaecologists, and endocrinologists. Eur Heart J. 2021;42(10):967–84.PubMedPubMedCentralCrossRef Maas A, Rosano G, Cifkova R, Chieffo A, van Dijken D, Hamoda H, et al. Cardiovascular health after menopause transition, pregnancy disorders, and other gynaecologic conditions: a consensus document from european cardiologists, gynaecologists, and endocrinologists. Eur Heart J. 2021;42(10):967–84.PubMedPubMedCentralCrossRef
189.
Zurück zum Zitat Sabbatini AR, Kararigas G. Menopause-related estrogen decrease and the Pathogenesis of HFpEF: JACC Review topic of the Week. J Am Coll Cardiol. 2020;75(9):1074–82.PubMedCrossRef Sabbatini AR, Kararigas G. Menopause-related estrogen decrease and the Pathogenesis of HFpEF: JACC Review topic of the Week. J Am Coll Cardiol. 2020;75(9):1074–82.PubMedCrossRef
190.
Zurück zum Zitat Sabbatini AR, Kararigas G. Estrogen-related mechanisms in sex differences of hypertension and target organ damage. Biol Sex Differ. 2020;11(1):31.PubMedPubMedCentralCrossRef Sabbatini AR, Kararigas G. Estrogen-related mechanisms in sex differences of hypertension and target organ damage. Biol Sex Differ. 2020;11(1):31.PubMedPubMedCentralCrossRef
191.
Zurück zum Zitat Zhu D, Chung HF, Dobson AJ, Pandeya N, Giles GG, Bruinsma F, et al. Age at natural menopause and risk of incident cardiovascular disease: a pooled analysis of individual patient data. Lancet Public Health. 2019;4(11):e553-e64.CrossRef Zhu D, Chung HF, Dobson AJ, Pandeya N, Giles GG, Bruinsma F, et al. Age at natural menopause and risk of incident cardiovascular disease: a pooled analysis of individual patient data. Lancet Public Health. 2019;4(11):e553-e64.CrossRef
192.
Zurück zum Zitat Matthews KA, Crawford SL, Chae CU, Everson-Rose SA, Sowers MF, Sternfeld B, et al. Are changes in cardiovascular disease risk factors in midlife women due to chronological aging or to the menopausal transition? J Am Coll Cardiol. 2009;54(25):2366–73.PubMedPubMedCentralCrossRef Matthews KA, Crawford SL, Chae CU, Everson-Rose SA, Sowers MF, Sternfeld B, et al. Are changes in cardiovascular disease risk factors in midlife women due to chronological aging or to the menopausal transition? J Am Coll Cardiol. 2009;54(25):2366–73.PubMedPubMedCentralCrossRef
193.
Zurück zum Zitat Mauvais-Jarvis F, Clegg DJ, Hevener AL. The role of estrogens in control of energy balance and glucose homeostasis. Endocr Rev. 2013;34(3):309–38.PubMedPubMedCentralCrossRef Mauvais-Jarvis F, Clegg DJ, Hevener AL. The role of estrogens in control of energy balance and glucose homeostasis. Endocr Rev. 2013;34(3):309–38.PubMedPubMedCentralCrossRef
194.
Zurück zum Zitat Anagnostis P, Christou K, Artzouchaltzi AM, Gkekas NK, Kosmidou N, Siolos P, et al. Early menopause and premature ovarian insufficiency are associated with increased risk of type 2 diabetes: a systematic review and meta-analysis. Eur J Endocrinol. 2019;180(1):41–50.PubMedCrossRef Anagnostis P, Christou K, Artzouchaltzi AM, Gkekas NK, Kosmidou N, Siolos P, et al. Early menopause and premature ovarian insufficiency are associated with increased risk of type 2 diabetes: a systematic review and meta-analysis. Eur J Endocrinol. 2019;180(1):41–50.PubMedCrossRef
195.
Zurück zum Zitat Bhaumik D, Scott GK, Schokrpur S, Patil CK, Orjalo AV, Rodier F, et al. MicroRNAs miR-146a/b negatively modulate the senescence-associated inflammatory mediators IL-6 and IL-8. Aging (Albany NY). 2009;1(4):402–11.PubMedCrossRef Bhaumik D, Scott GK, Schokrpur S, Patil CK, Orjalo AV, Rodier F, et al. MicroRNAs miR-146a/b negatively modulate the senescence-associated inflammatory mediators IL-6 and IL-8. Aging (Albany NY). 2009;1(4):402–11.PubMedCrossRef
196.
Zurück zum Zitat Mensa E, Giuliani A, Matacchione G, Gurau F, Bonfigli AR, Romagnoli F, et al. Circulating miR-146a in healthy aging and type 2 diabetes: age- and gender-specific trajectories. Mech Ageing Dev. 2019;180:1–10.PubMedCrossRef Mensa E, Giuliani A, Matacchione G, Gurau F, Bonfigli AR, Romagnoli F, et al. Circulating miR-146a in healthy aging and type 2 diabetes: age- and gender-specific trajectories. Mech Ageing Dev. 2019;180:1–10.PubMedCrossRef
197.
Zurück zum Zitat Hartman RJG, Owsiany K, Ma L, Koplev S, Hao K, Slenders L, et al. Sex-stratified Gene Regulatory Networks reveal female key driver genes of atherosclerosis involved in smooth muscle cell phenotype switching. Circulation. 2021;143(7):713–26.PubMedPubMedCentralCrossRef Hartman RJG, Owsiany K, Ma L, Koplev S, Hao K, Slenders L, et al. Sex-stratified Gene Regulatory Networks reveal female key driver genes of atherosclerosis involved in smooth muscle cell phenotype switching. Circulation. 2021;143(7):713–26.PubMedPubMedCentralCrossRef
198.
Zurück zum Zitat Kararigas G, Dworatzek E, Petrov G, Summer H, Schulze TM, Baczko I, et al. Sex-dependent regulation of fibrosis and inflammation in human left ventricular remodelling under pressure overload. European Journal of Heart Failure. 2014;16(11):1160–7.PubMedCrossRef Kararigas G, Dworatzek E, Petrov G, Summer H, Schulze TM, Baczko I, et al. Sex-dependent regulation of fibrosis and inflammation in human left ventricular remodelling under pressure overload. European Journal of Heart Failure. 2014;16(11):1160–7.PubMedCrossRef
199.
Zurück zum Zitat Dai R, Phillips RA, Zhang Y, Khan D, Crasta O, Ahmed SA. Suppression of LPS-induced Interferon-gamma and nitric oxide in splenic lymphocytes by select estrogen-regulated microRNAs: a novel mechanism of immune modulation. Blood. 2008;112(12):4591–7.PubMedPubMedCentralCrossRef Dai R, Phillips RA, Zhang Y, Khan D, Crasta O, Ahmed SA. Suppression of LPS-induced Interferon-gamma and nitric oxide in splenic lymphocytes by select estrogen-regulated microRNAs: a novel mechanism of immune modulation. Blood. 2008;112(12):4591–7.PubMedPubMedCentralCrossRef
200.
Zurück zum Zitat Alipoor B, Ghaedi H, Meshkani R, Torkamandi S, Saffari S, Iranpour M, et al. Association of MiR-146a expression and type 2 diabetes Mellitus: a Meta-analysis. Int J Mol Cell Med. 2017;6(3):156–63.PubMedPubMedCentral Alipoor B, Ghaedi H, Meshkani R, Torkamandi S, Saffari S, Iranpour M, et al. Association of MiR-146a expression and type 2 diabetes Mellitus: a Meta-analysis. Int J Mol Cell Med. 2017;6(3):156–63.PubMedPubMedCentral
201.
Zurück zum Zitat Raucci A, Macri F, Castiglione S, Badi I, Vinci MC, Zuccolo E. MicroRNA-34a: the bad guy in age-related vascular diseases. Cell Mol Life Sci. 2021;78(23):7355–78.PubMedPubMedCentralCrossRef Raucci A, Macri F, Castiglione S, Badi I, Vinci MC, Zuccolo E. MicroRNA-34a: the bad guy in age-related vascular diseases. Cell Mol Life Sci. 2021;78(23):7355–78.PubMedPubMedCentralCrossRef
202.
Zurück zum Zitat Pan Y, Hui X, Hoo RLC, Ye D, Chan CYC, Feng T, et al. Adipocyte-secreted exosomal microRNA-34a inhibits M2 macrophage polarization to promote obesity-induced adipose inflammation. J Clin Invest. 2019;129(2):834–49.PubMedPubMedCentralCrossRef Pan Y, Hui X, Hoo RLC, Ye D, Chan CYC, Feng T, et al. Adipocyte-secreted exosomal microRNA-34a inhibits M2 macrophage polarization to promote obesity-induced adipose inflammation. J Clin Invest. 2019;129(2):834–49.PubMedPubMedCentralCrossRef
203.
Zurück zum Zitat Bernardo BC, Ooi JY, Matsumoto A, Tham YK, Singla S, Kiriazis H, et al. Sex differences in response to miRNA-34a therapy in mouse models of cardiac disease: identification of sex-, disease- and treatment-regulated miRNAs. J Physiol. 2016;594(20):5959–74.PubMedPubMedCentralCrossRef Bernardo BC, Ooi JY, Matsumoto A, Tham YK, Singla S, Kiriazis H, et al. Sex differences in response to miRNA-34a therapy in mouse models of cardiac disease: identification of sex-, disease- and treatment-regulated miRNAs. J Physiol. 2016;594(20):5959–74.PubMedPubMedCentralCrossRef
204.
Zurück zum Zitat Florijn BW, Valstar GB, Duijs J, Menken R, Cramer MJ, Teske AJ, et al. Sex-specific microRNAs in women with diabetes and left ventricular diastolic dysfunction or HFpEF associate with microvascular injury. Sci Rep. 2020;10(1):13945.PubMedPubMedCentralCrossRef Florijn BW, Valstar GB, Duijs J, Menken R, Cramer MJ, Teske AJ, et al. Sex-specific microRNAs in women with diabetes and left ventricular diastolic dysfunction or HFpEF associate with microvascular injury. Sci Rep. 2020;10(1):13945.PubMedPubMedCentralCrossRef
205.
Zurück zum Zitat Frost RJ, Olson EN. Control of glucose homeostasis and insulin sensitivity by the Let-7 family of microRNAs. Proc Natl Acad Sci U S A. 2011;108(52):21075–80.PubMedPubMedCentralCrossRef Frost RJ, Olson EN. Control of glucose homeostasis and insulin sensitivity by the Let-7 family of microRNAs. Proc Natl Acad Sci U S A. 2011;108(52):21075–80.PubMedPubMedCentralCrossRef
206.
Zurück zum Zitat Li Y, Song YH, Li F, Yang T, Lu YW, Geng YJ. MicroRNA-221 regulates high glucose-induced endothelial dysfunction. Biochem Biophys Res Commun. 2009;381(1):81–3.PubMedPubMedCentralCrossRef Li Y, Song YH, Li F, Yang T, Lu YW, Geng YJ. MicroRNA-221 regulates high glucose-induced endothelial dysfunction. Biochem Biophys Res Commun. 2009;381(1):81–3.PubMedPubMedCentralCrossRef
207.
Zurück zum Zitat Wang YT, Tsai PC, Liao YC, Hsu CY, Juo SH. Circulating microRNAs have a sex-specific association with metabolic syndrome. J Biomed Sci. 2013;20:72.PubMedPubMedCentralCrossRef Wang YT, Tsai PC, Liao YC, Hsu CY, Juo SH. Circulating microRNAs have a sex-specific association with metabolic syndrome. J Biomed Sci. 2013;20:72.PubMedPubMedCentralCrossRef
208.
Zurück zum Zitat Sanchez-Ruderisch H, Queiros AM, Fliegner D, Eschen C, Kararigas G, Regitz-Zagrosek V. Sex-specific regulation of cardiac microRNAs targeting mitochondrial proteins in pressure overload. Biol Sex Differ. 2019;10(1):8.PubMedPubMedCentralCrossRef Sanchez-Ruderisch H, Queiros AM, Fliegner D, Eschen C, Kararigas G, Regitz-Zagrosek V. Sex-specific regulation of cardiac microRNAs targeting mitochondrial proteins in pressure overload. Biol Sex Differ. 2019;10(1):8.PubMedPubMedCentralCrossRef
209.
Zurück zum Zitat Tsamou M, Vrijens K, Wang C, Winckelmans E, Neven KY, Madhloum N, et al. Genome-wide microRNA expression analysis in human placenta reveals sex-specific patterns: an ENVIRONAGE birth cohort study. Epigenetics. 2021;16(4):373–88.PubMedCrossRef Tsamou M, Vrijens K, Wang C, Winckelmans E, Neven KY, Madhloum N, et al. Genome-wide microRNA expression analysis in human placenta reveals sex-specific patterns: an ENVIRONAGE birth cohort study. Epigenetics. 2021;16(4):373–88.PubMedCrossRef
210.
Zurück zum Zitat Tsamou M, Martens DS, Cox B, Madhloum N, Vrijens K, Nawrot TS. Sex-specific associations between telomere length and candidate miRNA expression in placenta. J Transl Med. 2018;16(1):254.PubMedPubMedCentralCrossRef Tsamou M, Martens DS, Cox B, Madhloum N, Vrijens K, Nawrot TS. Sex-specific associations between telomere length and candidate miRNA expression in placenta. J Transl Med. 2018;16(1):254.PubMedPubMedCentralCrossRef
211.
Zurück zum Zitat Leeson P. Long term cardiovascular outcomes for mother and child. Pregnancy Hypertens. 2013;3(2):60–1.PubMedCrossRef Leeson P. Long term cardiovascular outcomes for mother and child. Pregnancy Hypertens. 2013;3(2):60–1.PubMedCrossRef
212.
Zurück zum Zitat Bellamy L, Casas J-P, Hingorani AD, Williams D. Type 2 diabetes mellitus after gestational diabetes: a systematic review and meta-analysis. The Lancet. 2009;373(9677):1773–9.CrossRef Bellamy L, Casas J-P, Hingorani AD, Williams D. Type 2 diabetes mellitus after gestational diabetes: a systematic review and meta-analysis. The Lancet. 2009;373(9677):1773–9.CrossRef
213.
Zurück zum Zitat Strutz J, Cvitic S, Hackl H, Kashofer K, Appel HM, Thuringer A, et al. Gestational diabetes alters microRNA signatures in human feto-placental endothelial cells depending on fetal sex. Clin Sci (Lond). 2018;132(22):2437–49.PubMedCrossRef Strutz J, Cvitic S, Hackl H, Kashofer K, Appel HM, Thuringer A, et al. Gestational diabetes alters microRNA signatures in human feto-placental endothelial cells depending on fetal sex. Clin Sci (Lond). 2018;132(22):2437–49.PubMedCrossRef
214.
Zurück zum Zitat Prattichizzo F, Matacchione G, Giuliani A, Sabbatinelli J, Olivieri F, de Candia P, et al. Extracellular vesicle-shuttled miRNAs: a critical appraisal of their potential as nano-diagnostics and nano-therapeutics in type 2 diabetes mellitus and its cardiovascular complications. Theranostics. 2021;11(3):1031–45.PubMedPubMedCentralCrossRef Prattichizzo F, Matacchione G, Giuliani A, Sabbatinelli J, Olivieri F, de Candia P, et al. Extracellular vesicle-shuttled miRNAs: a critical appraisal of their potential as nano-diagnostics and nano-therapeutics in type 2 diabetes mellitus and its cardiovascular complications. Theranostics. 2021;11(3):1031–45.PubMedPubMedCentralCrossRef
215.
Zurück zum Zitat Blanco-Dominguez R, Sanchez-Diaz R, de la Fuente H, Jimenez-Borreguero LJ, Matesanz-Marin A, Relano M, et al. A Novel circulating MicroRNA for the detection of Acute Myocarditis. N Engl J Med. 2021;384(21):2014–27.PubMedPubMedCentralCrossRef Blanco-Dominguez R, Sanchez-Diaz R, de la Fuente H, Jimenez-Borreguero LJ, Matesanz-Marin A, Relano M, et al. A Novel circulating MicroRNA for the detection of Acute Myocarditis. N Engl J Med. 2021;384(21):2014–27.PubMedPubMedCentralCrossRef
216.
Zurück zum Zitat Magen I, Yacovzada NS, Yanowski E, Coenen-Stass A, Grosskreutz J, Lu CH, et al. Circulating miR-181 is a prognostic biomarker for amyotrophic lateral sclerosis. Nat Neurosci. 2021;24(11):1534–41.PubMedCrossRef Magen I, Yacovzada NS, Yanowski E, Coenen-Stass A, Grosskreutz J, Lu CH, et al. Circulating miR-181 is a prognostic biomarker for amyotrophic lateral sclerosis. Nat Neurosci. 2021;24(11):1534–41.PubMedCrossRef
217.
Zurück zum Zitat Zampetaki A, Kiechl S, Drozdov I, Willeit P, Mayr U, Prokopi M, et al. Plasma microRNA profiling reveals loss of endothelial miR-126 and other microRNAs in type 2 diabetes. Circ Res. 2010;107(6):810–7.PubMedCrossRef Zampetaki A, Kiechl S, Drozdov I, Willeit P, Mayr U, Prokopi M, et al. Plasma microRNA profiling reveals loss of endothelial miR-126 and other microRNAs in type 2 diabetes. Circ Res. 2010;107(6):810–7.PubMedCrossRef
218.
Zurück zum Zitat Jansen F, Wang H, Przybilla D, Franklin BS, Dolf A, Pfeifer P, et al. Vascular endothelial microparticles-incorporated microRNAs are altered in patients with diabetes mellitus. Cardiovasc Diabetol. 2016;15:49.PubMedPubMedCentralCrossRef Jansen F, Wang H, Przybilla D, Franklin BS, Dolf A, Pfeifer P, et al. Vascular endothelial microparticles-incorporated microRNAs are altered in patients with diabetes mellitus. Cardiovasc Diabetol. 2016;15:49.PubMedPubMedCentralCrossRef
219.
Zurück zum Zitat de Gonzalo-Calvo D, van der Meer RW, Rijzewijk LJ, Smit JW, Revuelta-Lopez E, Nasarre L, et al. Serum microRNA-1 and microRNA-133a levels reflect myocardial steatosis in uncomplicated type 2 diabetes. Sci Rep. 2017;7(1):47.PubMedPubMedCentralCrossRef de Gonzalo-Calvo D, van der Meer RW, Rijzewijk LJ, Smit JW, Revuelta-Lopez E, Nasarre L, et al. Serum microRNA-1 and microRNA-133a levels reflect myocardial steatosis in uncomplicated type 2 diabetes. Sci Rep. 2017;7(1):47.PubMedPubMedCentralCrossRef
220.
Zurück zum Zitat Feng B, Chen S, Gordon AD, Chakrabarti S. miR-146a mediates inflammatory changes and fibrosis in the heart in diabetes. J Mol Cell Cardiol. 2017;105:70–6.PubMedCrossRef Feng B, Chen S, Gordon AD, Chakrabarti S. miR-146a mediates inflammatory changes and fibrosis in the heart in diabetes. J Mol Cell Cardiol. 2017;105:70–6.PubMedCrossRef
221.
Zurück zum Zitat Watson CJ, Gupta SK, O’Connell E, Thum S, Glezeva N, Fendrich J, et al. MicroRNA signatures differentiate preserved from reduced ejection fraction heart failure. Eur J Heart Fail. 2015;17(4):405–15.PubMedCrossRef Watson CJ, Gupta SK, O’Connell E, Thum S, Glezeva N, Fendrich J, et al. MicroRNA signatures differentiate preserved from reduced ejection fraction heart failure. Eur J Heart Fail. 2015;17(4):405–15.PubMedCrossRef
222.
Zurück zum Zitat Chen C, Yang S, Li H, Yin Z, Fan J, Zhao Y, et al. Mir30c is involved in Diabetic Cardiomyopathy through Regulation of Cardiac Autophagy via BECN1. Mol Ther Nucleic Acids. 2017;7:127–39.PubMedPubMedCentralCrossRef Chen C, Yang S, Li H, Yin Z, Fan J, Zhao Y, et al. Mir30c is involved in Diabetic Cardiomyopathy through Regulation of Cardiac Autophagy via BECN1. Mol Ther Nucleic Acids. 2017;7:127–39.PubMedPubMedCentralCrossRef
223.
Zurück zum Zitat Tao L, Huang X, Xu M, Yang L, Hua F. MiR-144 protects the heart from hyperglycemia-induced injury by regulating mitochondrial biogenesis and cardiomyocyte apoptosis. FASEB J. 2020;34(2):2173–97.PubMedCrossRef Tao L, Huang X, Xu M, Yang L, Hua F. MiR-144 protects the heart from hyperglycemia-induced injury by regulating mitochondrial biogenesis and cardiomyocyte apoptosis. FASEB J. 2020;34(2):2173–97.PubMedCrossRef
224.
Zurück zum Zitat McCoy MG, Jamaiyar A, Sausen G, Cheng HS, Pérez-Cremades D, Zhuang R, et al. MicroRNA-375 repression of Kruppel-like factor 5 improves angiogenesis in diabetic critical limb ischemia. Angiogenesis. 2023;26(1):107–27.PubMedCrossRef McCoy MG, Jamaiyar A, Sausen G, Cheng HS, Pérez-Cremades D, Zhuang R, et al. MicroRNA-375 repression of Kruppel-like factor 5 improves angiogenesis in diabetic critical limb ischemia. Angiogenesis. 2023;26(1):107–27.PubMedCrossRef
225.
Zurück zum Zitat Zhang Y-n, Xie B-d, Sun L, Chen W, Jiang S-L, Liu W, et al. Phenotypic switching of vascular smooth muscle cells in the ‘normal region’ of aorta from atherosclerosis patients is regulated by miR-145. J Cell Mol Med. 2016;20(6):1049–61.PubMedPubMedCentralCrossRef Zhang Y-n, Xie B-d, Sun L, Chen W, Jiang S-L, Liu W, et al. Phenotypic switching of vascular smooth muscle cells in the ‘normal region’ of aorta from atherosclerosis patients is regulated by miR-145. J Cell Mol Med. 2016;20(6):1049–61.PubMedPubMedCentralCrossRef
226.
Zurück zum Zitat Wang Y-S, Li S-H, Guo J, Mihic A, Wu J, Sun L, et al. Role of miR-145 in cardiac myofibroblast differentiation. J Mol Cell Cardiol. 2014;66:94–105.PubMedCrossRef Wang Y-S, Li S-H, Guo J, Mihic A, Wu J, Sun L, et al. Role of miR-145 in cardiac myofibroblast differentiation. J Mol Cell Cardiol. 2014;66:94–105.PubMedCrossRef
227.
Zurück zum Zitat Fu L, Zhang J, Lin Z, Li Y, Qin G. CircularRNA circ_0071269 knockdown protects against from diabetic cardiomyopathy injury by microRNA-145/gasdermin A axis. Bioengineered. 2022;13(2):2398–411.PubMedPubMedCentralCrossRef Fu L, Zhang J, Lin Z, Li Y, Qin G. CircularRNA circ_0071269 knockdown protects against from diabetic cardiomyopathy injury by microRNA-145/gasdermin A axis. Bioengineered. 2022;13(2):2398–411.PubMedPubMedCentralCrossRef
228.
Zurück zum Zitat Zheng W, Li T, Wei J, Zhang Y, Zuo Q, Lin Y. Identification of miR–145 as a regulator of the cardiomyocyte inflammatory response and oxidative stress under hyperglycemia. Exp Ther Med. 2021;21(5):467.PubMedPubMedCentralCrossRef Zheng W, Li T, Wei J, Zhang Y, Zuo Q, Lin Y. Identification of miR–145 as a regulator of the cardiomyocyte inflammatory response and oxidative stress under hyperglycemia. Exp Ther Med. 2021;21(5):467.PubMedPubMedCentralCrossRef
229.
Zurück zum Zitat Chen Y, Zhang Z, Zhu D, Zhao W, Li F. Long non-coding RNA MEG3 serves as a ceRNA for microRNA-145 to induce apoptosis of AC16 cardiomyocytes under high glucose condition. Bioscience Reports. 2019;39(6):BSR20190444.PubMedPubMedCentralCrossRef Chen Y, Zhang Z, Zhu D, Zhao W, Li F. Long non-coding RNA MEG3 serves as a ceRNA for microRNA-145 to induce apoptosis of AC16 cardiomyocytes under high glucose condition. Bioscience Reports. 2019;39(6):BSR20190444.PubMedPubMedCentralCrossRef
230.
Zurück zum Zitat May JM, Bylicky M, Chopra S, Coleman CN, Aryankalayil MJ. Long and short non-coding RNA and radiation response: a review. Transl Res. 2021;233:162–79.PubMedPubMedCentralCrossRef May JM, Bylicky M, Chopra S, Coleman CN, Aryankalayil MJ. Long and short non-coding RNA and radiation response: a review. Transl Res. 2021;233:162–79.PubMedPubMedCentralCrossRef
231.
Zurück zum Zitat Sathishkumar C, Prabu P, Mohan V, Balasubramanyam M. Linking a role of lncRNAs (long non-coding RNAs) with insulin resistance, accelerated senescence, and inflammation in patients with type 2 diabetes. Hum Genomics. 2018;12(1):41.PubMedPubMedCentralCrossRef Sathishkumar C, Prabu P, Mohan V, Balasubramanyam M. Linking a role of lncRNAs (long non-coding RNAs) with insulin resistance, accelerated senescence, and inflammation in patients with type 2 diabetes. Hum Genomics. 2018;12(1):41.PubMedPubMedCentralCrossRef
232.
Zurück zum Zitat Vausort M, Wagner DR, Devaux Y. Long noncoding RNAs in patients with acute myocardial infarction. Circ Res. 2014;115(7):668–77.PubMedCrossRef Vausort M, Wagner DR, Devaux Y. Long noncoding RNAs in patients with acute myocardial infarction. Circ Res. 2014;115(7):668–77.PubMedCrossRef
233.
Zurück zum Zitat Qi K, Zhong J. LncRNA HOTAIR improves diabetic cardiomyopathy by increasing viability of cardiomyocytes through activation of the PI3K/Akt pathway. Exp Ther Med. 2018;16(6):4817–23.PubMedPubMedCentral Qi K, Zhong J. LncRNA HOTAIR improves diabetic cardiomyopathy by increasing viability of cardiomyocytes through activation of the PI3K/Akt pathway. Exp Ther Med. 2018;16(6):4817–23.PubMedPubMedCentral
234.
Zurück zum Zitat Yang F, Qin Y, Lv J, Wang Y, Che H, Chen X, et al. Silencing long non-coding RNA Kcnq1ot1 alleviates pyroptosis and fibrosis in diabetic cardiomyopathy. Cell Death Dis. 2018;9(10):1000.PubMedPubMedCentralCrossRef Yang F, Qin Y, Lv J, Wang Y, Che H, Chen X, et al. Silencing long non-coding RNA Kcnq1ot1 alleviates pyroptosis and fibrosis in diabetic cardiomyopathy. Cell Death Dis. 2018;9(10):1000.PubMedPubMedCentralCrossRef
235.
Zurück zum Zitat Fang Y, Wang X, Li W, Han J, Jin J, Su F, et al. Screening of circular RNAs and validation of circANKRD36 associated with inflammation in patients with type 2 diabetes mellitus. Int J Mol Med. 2018;42(4):1865–74.PubMedPubMedCentral Fang Y, Wang X, Li W, Han J, Jin J, Su F, et al. Screening of circular RNAs and validation of circANKRD36 associated with inflammation in patients with type 2 diabetes mellitus. Int J Mol Med. 2018;42(4):1865–74.PubMedPubMedCentral
236.
Zurück zum Zitat Di Mauro V, Barandalla-Sobrados M, Catalucci D. The noncoding-RNA landscape in cardiovascular health and disease. Noncoding RNA Res. 2018;3(1):12–9.PubMedPubMedCentralCrossRef Di Mauro V, Barandalla-Sobrados M, Catalucci D. The noncoding-RNA landscape in cardiovascular health and disease. Noncoding RNA Res. 2018;3(1):12–9.PubMedPubMedCentralCrossRef
237.
Zurück zum Zitat Kole R, Krainer AR, Altman S. RNA therapeutics: beyond RNA interference and antisense oligonucleotides. Nat Rev Drug Discov. 2012;11(2):125–40.PubMedPubMedCentralCrossRef Kole R, Krainer AR, Altman S. RNA therapeutics: beyond RNA interference and antisense oligonucleotides. Nat Rev Drug Discov. 2012;11(2):125–40.PubMedPubMedCentralCrossRef
238.
Zurück zum Zitat Bajan S, Hutvagner G. RNA-Based therapeutics: from antisense oligonucleotides to miRNAs. Cells. 2020;9(1). Bajan S, Hutvagner G. RNA-Based therapeutics: from antisense oligonucleotides to miRNAs. Cells. 2020;9(1).
239.
Zurück zum Zitat Rode L, Bar C, Gross S, Rossi A, Meumann N, Viereck J, et al. AAV capsid engineering identified two novel variants with improved in vivo tropism for cardiomyocytes. Mol Ther. 2022. Rode L, Bar C, Gross S, Rossi A, Meumann N, Viereck J, et al. AAV capsid engineering identified two novel variants with improved in vivo tropism for cardiomyocytes. Mol Ther. 2022.
240.
Zurück zum Zitat Liu SJ, Horlbeck MA, Cho SW, Birk HS, Malatesta M, He D, et al. CRISPRi-based genome-scale identification of functional long noncoding RNA loci in human cells. Science. 2017;355(6320). Liu SJ, Horlbeck MA, Cho SW, Birk HS, Malatesta M, He D, et al. CRISPRi-based genome-scale identification of functional long noncoding RNA loci in human cells. Science. 2017;355(6320).
241.
Zurück zum Zitat Chakraborty C, Sharma AR, Sharma G, Lee SS. Therapeutic advances of miRNAs: a preclinical and clinical update. J Adv Res. 2021;28:127–38.PubMedCrossRef Chakraborty C, Sharma AR, Sharma G, Lee SS. Therapeutic advances of miRNAs: a preclinical and clinical update. J Adv Res. 2021;28:127–38.PubMedCrossRef
242.
Zurück zum Zitat Huang CK, Kafert-Kasting S, Thum T. Preclinical and clinical development of noncoding RNA therapeutics for Cardiovascular Disease. Circ Res. 2020;126(5):663–78.PubMedPubMedCentralCrossRef Huang CK, Kafert-Kasting S, Thum T. Preclinical and clinical development of noncoding RNA therapeutics for Cardiovascular Disease. Circ Res. 2020;126(5):663–78.PubMedPubMedCentralCrossRef
243.
Zurück zum Zitat Winkle M, El-Daly SM, Fabbri M, Calin GA. Noncoding RNA therapeutics - challenges and potential solutions. Nat Rev Drug Discov. 2021;20(8):629–51.PubMedPubMedCentralCrossRef Winkle M, El-Daly SM, Fabbri M, Calin GA. Noncoding RNA therapeutics - challenges and potential solutions. Nat Rev Drug Discov. 2021;20(8):629–51.PubMedPubMedCentralCrossRef
244.
Zurück zum Zitat Hueso M, Mallen A, Sune-Pou M, Aran JM, Sune-Negre JM, Navarro E. ncRNAs in therapeutics: Challenges and Limitations in Nucleic Acid-Based drug delivery. Int J Mol Sci. 2021;22(21). Hueso M, Mallen A, Sune-Pou M, Aran JM, Sune-Negre JM, Navarro E. ncRNAs in therapeutics: Challenges and Limitations in Nucleic Acid-Based drug delivery. Int J Mol Sci. 2021;22(21).
245.
Zurück zum Zitat He C, Hu Y, Yin L, Tang C, Yin C. Effects of particle size and surface charge on cellular uptake and biodistribution of polymeric nanoparticles. Biomaterials. 2010;31(13):3657–66.PubMedCrossRef He C, Hu Y, Yin L, Tang C, Yin C. Effects of particle size and surface charge on cellular uptake and biodistribution of polymeric nanoparticles. Biomaterials. 2010;31(13):3657–66.PubMedCrossRef
246.
Zurück zum Zitat Hu J, Sheng Y, Shi J, Yu B, Yu Z, Liao G. Long circulating polymeric nanoparticles for Gene/Drug delivery. Curr Drug Metab. 2018;19(9):723–38.PubMedCrossRef Hu J, Sheng Y, Shi J, Yu B, Yu Z, Liao G. Long circulating polymeric nanoparticles for Gene/Drug delivery. Curr Drug Metab. 2018;19(9):723–38.PubMedCrossRef
248.
Zurück zum Zitat Hullinger TG, Montgomery RL, Seto AG, Dickinson BA, Semus HM, Lynch JM, et al. Inhibition of miR-15 protects against cardiac ischemic injury. Circ Res. 2012;110(1):71–81.PubMedCrossRef Hullinger TG, Montgomery RL, Seto AG, Dickinson BA, Semus HM, Lynch JM, et al. Inhibition of miR-15 protects against cardiac ischemic injury. Circ Res. 2012;110(1):71–81.PubMedCrossRef
250.
Zurück zum Zitat Wang Y, Xie Y, Kilchrist KV, Li J, Duvall CL, Oupicky D. Endosomolytic and tumor-penetrating mesoporous silica nanoparticles for siRNA/miRNA Combination Cancer Therapy. ACS Appl Mater Interfaces. 2020;12(4):4308–22.PubMedPubMedCentralCrossRef Wang Y, Xie Y, Kilchrist KV, Li J, Duvall CL, Oupicky D. Endosomolytic and tumor-penetrating mesoporous silica nanoparticles for siRNA/miRNA Combination Cancer Therapy. ACS Appl Mater Interfaces. 2020;12(4):4308–22.PubMedPubMedCentralCrossRef
251.
Zurück zum Zitat Chiriboga CA. Nusinersen for the treatment of spinal muscular atrophy. Expert Rev Neurother. 2017;17(10):955–62.PubMedCrossRef Chiriboga CA. Nusinersen for the treatment of spinal muscular atrophy. Expert Rev Neurother. 2017;17(10):955–62.PubMedCrossRef
252.
Zurück zum Zitat Dhillon S. Viltolarsen: First Approval. Drugs. 2020;80(10):1027–31.PubMed Dhillon S. Viltolarsen: First Approval. Drugs. 2020;80(10):1027–31.PubMed
255.
Zurück zum Zitat Rupaimoole R, Slack FJ. MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov. 2017;16(3):203–22.PubMedCrossRef Rupaimoole R, Slack FJ. MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov. 2017;16(3):203–22.PubMedCrossRef
256.
Zurück zum Zitat Gallant-Behm CL, Piper J, Dickinson BA, Dalby CM, Pestano LA, Jackson AL. A synthetic microRNA-92a inhibitor (MRG-110) accelerates angiogenesis and wound healing in diabetic and nondiabetic wounds. Wound Repair Regen. 2018;26(4):311–23.PubMedCrossRef Gallant-Behm CL, Piper J, Dickinson BA, Dalby CM, Pestano LA, Jackson AL. A synthetic microRNA-92a inhibitor (MRG-110) accelerates angiogenesis and wound healing in diabetic and nondiabetic wounds. Wound Repair Regen. 2018;26(4):311–23.PubMedCrossRef
257.
Zurück zum Zitat Abplanalp WT, Fischer A, John D, Zeiher AM, Gosgnach W, Darville H, et al. Efficiency and target derepression of Anti-miR-92a: results of a First in Human Study. Nucleic Acid Ther. 2020;30(6):335–45.PubMedCrossRef Abplanalp WT, Fischer A, John D, Zeiher AM, Gosgnach W, Darville H, et al. Efficiency and target derepression of Anti-miR-92a: results of a First in Human Study. Nucleic Acid Ther. 2020;30(6):335–45.PubMedCrossRef
258.
Zurück zum Zitat Rubel D, Boulanger J, Craciun F, Xu EY, Zhang Y, Phillips L, et al. Anti-microRNA-21 therapy on top of ACE inhibition delays renal failure in Alport Syndrome Mouse Models. Cells. 2022;11(4). Rubel D, Boulanger J, Craciun F, Xu EY, Zhang Y, Phillips L, et al. Anti-microRNA-21 therapy on top of ACE inhibition delays renal failure in Alport Syndrome Mouse Models. Cells. 2022;11(4).
259.
Zurück zum Zitat Foinquinos A, Batkai S, Genschel C, Viereck J, Rump S, Gyongyosi M, et al. Preclinical development of a miR-132 inhibitor for heart failure treatment. Nat Commun. 2020;11(1):633.PubMedPubMedCentralCrossRef Foinquinos A, Batkai S, Genschel C, Viereck J, Rump S, Gyongyosi M, et al. Preclinical development of a miR-132 inhibitor for heart failure treatment. Nat Commun. 2020;11(1):633.PubMedPubMedCentralCrossRef
260.
Zurück zum Zitat Taubel J, Hauke W, Rump S, Viereck J, Batkai S, Poetzsch J, et al. Novel antisense therapy targeting microRNA-132 in patients with heart failure: results of a first-in-human phase 1b randomized, double-blind, placebo-controlled study. Eur Heart J. 2021;42(2):178–88.PubMedCrossRef Taubel J, Hauke W, Rump S, Viereck J, Batkai S, Poetzsch J, et al. Novel antisense therapy targeting microRNA-132 in patients with heart failure: results of a first-in-human phase 1b randomized, double-blind, placebo-controlled study. Eur Heart J. 2021;42(2):178–88.PubMedCrossRef
261.
Zurück zum Zitat Trajkovski M, Hausser J, Soutschek J, Bhat B, Akin A, Zavolan M, et al. MicroRNAs 103 and 107 regulate insulin sensitivity. Nature. 2011;474(7353):649–53.PubMedCrossRef Trajkovski M, Hausser J, Soutschek J, Bhat B, Akin A, Zavolan M, et al. MicroRNAs 103 and 107 regulate insulin sensitivity. Nature. 2011;474(7353):649–53.PubMedCrossRef
262.
Zurück zum Zitat American Diabetes A. 9. Pharmacologic Approaches to Glycemic Treatment: Standards of Medical Care in Diabetes—2020. Diabetes Care. 2019;43(Supplement_1):S98-S110. American Diabetes A. 9. Pharmacologic Approaches to Glycemic Treatment: Standards of Medical Care in Diabetes—2020. Diabetes Care. 2019;43(Supplement_1):S98-S110.
263.
Zurück zum Zitat Bharath LP, Agrawal M, McCambridge G, Nicholas DA, Hasturk H, Liu J, et al. Metformin enhances autophagy and normalizes mitochondrial function to Alleviate Aging-Associated inflammation. Cell metabolism. 2020;32(1):44–55.e6.PubMedPubMedCentralCrossRef Bharath LP, Agrawal M, McCambridge G, Nicholas DA, Hasturk H, Liu J, et al. Metformin enhances autophagy and normalizes mitochondrial function to Alleviate Aging-Associated inflammation. Cell metabolism. 2020;32(1):44–55.e6.PubMedPubMedCentralCrossRef
264.
Zurück zum Zitat Petrie JR, Chaturvedi N, Ford I, Brouwers MCGJ, Greenlaw N, Tillin T, et al. Cardiovascular and metabolic effects of metformin in patients with type 1 diabetes (REMOVAL): a double-blind, randomised, placebo-controlled trial. Lancet Diabetes Endocrinol. 2017;5(8):597–609.PubMedPubMedCentralCrossRef Petrie JR, Chaturvedi N, Ford I, Brouwers MCGJ, Greenlaw N, Tillin T, et al. Cardiovascular and metabolic effects of metformin in patients with type 1 diabetes (REMOVAL): a double-blind, randomised, placebo-controlled trial. Lancet Diabetes Endocrinol. 2017;5(8):597–609.PubMedPubMedCentralCrossRef
265.
Zurück zum Zitat Zhao D, Yang J, Yang L. Insights for oxidative stress and mTOR Signaling in Myocardial Ischemia/Reperfusion Injury under Diabetes. Oxidative medicine and cellular longevity. 2017;2017:6437467-.PubMedPubMedCentralCrossRef Zhao D, Yang J, Yang L. Insights for oxidative stress and mTOR Signaling in Myocardial Ischemia/Reperfusion Injury under Diabetes. Oxidative medicine and cellular longevity. 2017;2017:6437467-.PubMedPubMedCentralCrossRef
266.
Zurück zum Zitat Noren Hooten N, Martin-Montalvo A, Dluzen DF, Zhang Y, Bernier M, Zonderman AB, et al. Metformin-mediated increase in DICER1 regulates microRNA expression and cellular senescence. Aging cell. 2016;15(3):572–81.PubMedPubMedCentralCrossRef Noren Hooten N, Martin-Montalvo A, Dluzen DF, Zhang Y, Bernier M, Zonderman AB, et al. Metformin-mediated increase in DICER1 regulates microRNA expression and cellular senescence. Aging cell. 2016;15(3):572–81.PubMedPubMedCentralCrossRef
267.
Zurück zum Zitat Mateescu B, Batista L, Cardon M, Gruosso T, de Feraudy Y, Mariani O, et al. miR-141 and miR-200a act on ovarian tumorigenesis by controlling oxidative stress response. Nature Medicine. 2011;17(12):1627–35.PubMedCrossRef Mateescu B, Batista L, Cardon M, Gruosso T, de Feraudy Y, Mariani O, et al. miR-141 and miR-200a act on ovarian tumorigenesis by controlling oxidative stress response. Nature Medicine. 2011;17(12):1627–35.PubMedCrossRef
268.
Zurück zum Zitat Magenta A, Cencioni C, Fasanaro P, Zaccagnini G, Greco S, Sarra-Ferraris G, et al. miR-200c is upregulated by oxidative stress and induces endothelial cell apoptosis and senescence via ZEB1 inhibition. Cell Death Differ. 2011;18(10):1628–39.PubMedPubMedCentralCrossRef Magenta A, Cencioni C, Fasanaro P, Zaccagnini G, Greco S, Sarra-Ferraris G, et al. miR-200c is upregulated by oxidative stress and induces endothelial cell apoptosis and senescence via ZEB1 inhibition. Cell Death Differ. 2011;18(10):1628–39.PubMedPubMedCentralCrossRef
269.
Zurück zum Zitat Mensà E, Guescini M, Giuliani A, Bacalini MG, Ramini D, Corleone G, et al. Small extracellular vesicles deliver miR-21 and miR-217 as pro-senescence effectors to endothelial cells. J Extracell Vesicles. 2020;9(1):1725285-.PubMedPubMedCentralCrossRef Mensà E, Guescini M, Giuliani A, Bacalini MG, Ramini D, Corleone G, et al. Small extracellular vesicles deliver miR-21 and miR-217 as pro-senescence effectors to endothelial cells. J Extracell Vesicles. 2020;9(1):1725285-.PubMedPubMedCentralCrossRef
270.
Zurück zum Zitat Gu X, Wang XQ, Lin MJ, Liang H, Fan SY, Wang L, et al. Molecular interplay between microRNA-130a and PTEN in palmitic acid-mediated impaired function of endothelial progenitor cells: Effects of metformin. Int J Mol Med. 2019;43(5):2187–98.PubMed Gu X, Wang XQ, Lin MJ, Liang H, Fan SY, Wang L, et al. Molecular interplay between microRNA-130a and PTEN in palmitic acid-mediated impaired function of endothelial progenitor cells: Effects of metformin. Int J Mol Med. 2019;43(5):2187–98.PubMed
271.
Zurück zum Zitat Arunachalam G, Lakshmanan AP, Samuel SM, Triggle CR, Ding H. Molecular interplay between microRNA-34a and Sirtuin1 in hyperglycemia-mediated impaired angiogenesis in endothelial cells: Effects of Metformin. Journal of Pharmacology and Experimental Therapeutics. 2016;356(2):314.PubMedCrossRef Arunachalam G, Lakshmanan AP, Samuel SM, Triggle CR, Ding H. Molecular interplay between microRNA-34a and Sirtuin1 in hyperglycemia-mediated impaired angiogenesis in endothelial cells: Effects of Metformin. Journal of Pharmacology and Experimental Therapeutics. 2016;356(2):314.PubMedCrossRef
273.
Zurück zum Zitat Li W, Jin S, Hao J, Shi Y, Li W, Jiang L. Metformin attenuates ischemia/reperfusion-induced apoptosis of cardiac cells by downregulation of p53/microRNA-34a via activation of SIRT1. Can J Physiol Pharmacol. 2021;99(9):875–84.PubMedCrossRef Li W, Jin S, Hao J, Shi Y, Li W, Jiang L. Metformin attenuates ischemia/reperfusion-induced apoptosis of cardiac cells by downregulation of p53/microRNA-34a via activation of SIRT1. Can J Physiol Pharmacol. 2021;99(9):875–84.PubMedCrossRef
274.
Zurück zum Zitat Pizzo P, Scapin C, Vitadello M, Florean C, Gorza L. Grp94 acts as a mediator of curcumin-induced antioxidant defence in myogenic cells. J Cell Mol Med. 2010;14(4):970–81.PubMedPubMedCentralCrossRef Pizzo P, Scapin C, Vitadello M, Florean C, Gorza L. Grp94 acts as a mediator of curcumin-induced antioxidant defence in myogenic cells. J Cell Mol Med. 2010;14(4):970–81.PubMedPubMedCentralCrossRef
275.
Zurück zum Zitat Liu H, Bowes RC, van de Water B, Sillence C, Nagelkerke JF, Stevens JL. Endoplasmic reticulum chaperones GRP78 and calreticulin prevent oxidative stress, Ca2 + disturbances, and cell death in renal epithelial Cells*. Journal of Biological Chemistry. 1997;272(35):21751–9.PubMedCrossRef Liu H, Bowes RC, van de Water B, Sillence C, Nagelkerke JF, Stevens JL. Endoplasmic reticulum chaperones GRP78 and calreticulin prevent oxidative stress, Ca2 + disturbances, and cell death in renal epithelial Cells*. Journal of Biological Chemistry. 1997;272(35):21751–9.PubMedCrossRef
276.
Zurück zum Zitat Wang J, Cao B, Han D, Sun M, Feng J. Long non-coding RNA H19 induces cerebral ischemia reperfusion Injury via activation of Autophagy. Aging Dis. 2017;8(1):71–84.PubMedPubMedCentralCrossRef Wang J, Cao B, Han D, Sun M, Feng J. Long non-coding RNA H19 induces cerebral ischemia reperfusion Injury via activation of Autophagy. Aging Dis. 2017;8(1):71–84.PubMedPubMedCentralCrossRef
277.
Zurück zum Zitat Zeng J, Zhu L, Liu J, Zhu T, Xie Z, Sun X, et al. Metformin Protects against Oxidative Stress Injury Induced by Ischemia/Reperfusion via Regulation of the lncRNA-H19/miR-148a-3p/Rock2 Axis. Oxidative medicine and cellular longevity. 2019;2019:8768327-. Zeng J, Zhu L, Liu J, Zhu T, Xie Z, Sun X, et al. Metformin Protects against Oxidative Stress Injury Induced by Ischemia/Reperfusion via Regulation of the lncRNA-H19/miR-148a-3p/Rock2 Axis. Oxidative medicine and cellular longevity. 2019;2019:8768327-.
278.
Zurück zum Zitat Luo L, Chen J, Su D, Chen M, Luo B, Pi R, et al. L-F001, a multifunction ROCK inhibitor prevents 6-OHDA Induced Cell Death through activating Akt/GSK-3beta and Nrf2/HO-1 signaling pathway in PC12 cells and attenuates MPTP-Induced dopamine Neuron Toxicity in mice. Neurochemical Research. 2017;42(2):615–24.PubMedCrossRef Luo L, Chen J, Su D, Chen M, Luo B, Pi R, et al. L-F001, a multifunction ROCK inhibitor prevents 6-OHDA Induced Cell Death through activating Akt/GSK-3beta and Nrf2/HO-1 signaling pathway in PC12 cells and attenuates MPTP-Induced dopamine Neuron Toxicity in mice. Neurochemical Research. 2017;42(2):615–24.PubMedCrossRef
279.
Zurück zum Zitat Zhang Q, Xiao X, Zheng J, Li M. A glucagon-like peptide-1 analog, liraglutide, ameliorates endothelial dysfunction through miRNAs to inhibit apoptosis in rats. PeerJ. 2019;7:e6567.PubMedPubMedCentralCrossRef Zhang Q, Xiao X, Zheng J, Li M. A glucagon-like peptide-1 analog, liraglutide, ameliorates endothelial dysfunction through miRNAs to inhibit apoptosis in rats. PeerJ. 2019;7:e6567.PubMedPubMedCentralCrossRef
280.
Zurück zum Zitat Giglio RV, Nikolic D, Volti GL, Stoian AP, Banerjee Y, Magan-Fernandez A, et al. Liraglutide increases serum levels of MicroRNA-27b, -130a and – 210 in patients with type 2 diabetes Mellitus: a Novel Epigenetic Effect. Metabolites. 2020;10(10). Giglio RV, Nikolic D, Volti GL, Stoian AP, Banerjee Y, Magan-Fernandez A, et al. Liraglutide increases serum levels of MicroRNA-27b, -130a and – 210 in patients with type 2 diabetes Mellitus: a Novel Epigenetic Effect. Metabolites. 2020;10(10).
281.
Zurück zum Zitat Stretti L, Zippo D, Coats AJS, Anker MS, von Haehling S, Metra M, et al. A year in heart failure: an update of recent findings. ESC Heart Fail. 2021;8(6):4370–93.PubMedPubMedCentralCrossRef Stretti L, Zippo D, Coats AJS, Anker MS, von Haehling S, Metra M, et al. A year in heart failure: an update of recent findings. ESC Heart Fail. 2021;8(6):4370–93.PubMedPubMedCentralCrossRef
282.
Zurück zum Zitat Mone P, Lombardi A, Kansakar U, Varzideh F, Jankauskas SS, Pansini A, et al. Empagliflozin improves the MicroRNA signature of endothelial dysfunction in patients with heart failure with preserved ejection fraction and diabetes. J Pharmacol Exp Ther. 2023;384(1):116–22.PubMedCrossRef Mone P, Lombardi A, Kansakar U, Varzideh F, Jankauskas SS, Pansini A, et al. Empagliflozin improves the MicroRNA signature of endothelial dysfunction in patients with heart failure with preserved ejection fraction and diabetes. J Pharmacol Exp Ther. 2023;384(1):116–22.PubMedCrossRef
283.
Zurück zum Zitat Smith Jb Fau - Araki H, Araki H Fau - Lefer AM, Lefer AM. Thromboxane A2, prostacyclin and aspirin: effects on vascular tone and platelet aggregation. (0009-7322 (Print)). Smith Jb Fau - Araki H, Araki H Fau - Lefer AM, Lefer AM. Thromboxane A2, prostacyclin and aspirin: effects on vascular tone and platelet aggregation. (0009-7322 (Print)).
284.
Zurück zum Zitat Raber I, McCarthy CP, Vaduganathan M, Bhatt DL, Wood DA, Cleland JGF, et al. The rise and fall of aspirin in the primary prevention of cardiovascular disease. The Lancet. 2019;393(10186):2155–67.CrossRef Raber I, McCarthy CP, Vaduganathan M, Bhatt DL, Wood DA, Cleland JGF, et al. The rise and fall of aspirin in the primary prevention of cardiovascular disease. The Lancet. 2019;393(10186):2155–67.CrossRef
285.
Zurück zum Zitat Kalgutkar AS, Kozak KR, Crews BC, Hochgesang GP, Marnett LJ. Covalent modification of Cyclooxygenase-2 (COX-2) by 2-Acetoxyphenyl Alkyl Sulfides, a New Class of selective COX-2 inactivators. Journal of Medicinal Chemistry. 1998;41(24):4800–18.PubMedCrossRef Kalgutkar AS, Kozak KR, Crews BC, Hochgesang GP, Marnett LJ. Covalent modification of Cyclooxygenase-2 (COX-2) by 2-Acetoxyphenyl Alkyl Sulfides, a New Class of selective COX-2 inactivators. Journal of Medicinal Chemistry. 1998;41(24):4800–18.PubMedCrossRef
286.
Zurück zum Zitat Pearson TA, Blair SN, Daniels SR, Eckel RH, Fair JM, Fortmann SP, et al. AHA Guidelines for primary Prevention of Cardiovascular Disease and Stroke: 2002 update. Circulation. 2002;106(3):388–91.PubMedCrossRef Pearson TA, Blair SN, Daniels SR, Eckel RH, Fair JM, Fortmann SP, et al. AHA Guidelines for primary Prevention of Cardiovascular Disease and Stroke: 2002 update. Circulation. 2002;106(3):388–91.PubMedCrossRef
288.
Zurück zum Zitat Du G, Lin Q, Wang J. A brief review on the mechanisms of aspirin resistance. International Journal of Cardiology. 2016;220:21–6.PubMedCrossRef Du G, Lin Q, Wang J. A brief review on the mechanisms of aspirin resistance. International Journal of Cardiology. 2016;220:21–6.PubMedCrossRef
289.
Zurück zum Zitat Zufferey A, Ibberson M, Reny J-L, Nolli S, Schvartz D, Docquier M, et al. New molecular insights into modulation of platelet reactivity in aspirin-treated patients using a network-based approach. Human Genetics. 2016;135(4):403–14.PubMedCrossRef Zufferey A, Ibberson M, Reny J-L, Nolli S, Schvartz D, Docquier M, et al. New molecular insights into modulation of platelet reactivity in aspirin-treated patients using a network-based approach. Human Genetics. 2016;135(4):403–14.PubMedCrossRef
290.
Zurück zum Zitat Kok MGM, Mandolini C, Moerland PD, de Ronde MWJ, Sondermeijer BM, Halliani A, et al. Low miR-19b-1-5p expression in isolated platelets after aspirin use is related to aspirin insensitivity. International Journal of Cardiology. 2016;203:262–3.PubMedCrossRef Kok MGM, Mandolini C, Moerland PD, de Ronde MWJ, Sondermeijer BM, Halliani A, et al. Low miR-19b-1-5p expression in isolated platelets after aspirin use is related to aspirin insensitivity. International Journal of Cardiology. 2016;203:262–3.PubMedCrossRef
291.
Zurück zum Zitat Singh S, de Ronde MWJ, Creemers EE, Van der Made I, Meijering R, Chan MY, et al. Low miR-19b-1-5p expression is related to aspirin resistance and major adverse cardio- cerebrovascular events in patients with Acute Coronary Syndrome. J Am Heart Assoc. 2021;10(2):e017120.PubMedPubMedCentralCrossRef Singh S, de Ronde MWJ, Creemers EE, Van der Made I, Meijering R, Chan MY, et al. Low miR-19b-1-5p expression is related to aspirin resistance and major adverse cardio- cerebrovascular events in patients with Acute Coronary Syndrome. J Am Heart Assoc. 2021;10(2):e017120.PubMedPubMedCentralCrossRef
292.
Zurück zum Zitat Paseban M, Marjaneh RM, Banach M, Riahi MM, Bo S, Sahebkar A. Modulation of microRNAs by aspirin in cardiovascular disease. Trends Cardiovasc Med. 2020;30(5):249–54.PubMedCrossRef Paseban M, Marjaneh RM, Banach M, Riahi MM, Bo S, Sahebkar A. Modulation of microRNAs by aspirin in cardiovascular disease. Trends Cardiovasc Med. 2020;30(5):249–54.PubMedCrossRef
293.
Zurück zum Zitat Vernstrom L, Funck KL, Grove EL, Laugesen E, Baier JM, Hvas AM, et al. Antiplatelet effect of aspirin during 24 h in patients with type 2 diabetes without cardiovascular disease. Thromb Res. 2018;161:1–6.PubMedCrossRef Vernstrom L, Funck KL, Grove EL, Laugesen E, Baier JM, Hvas AM, et al. Antiplatelet effect of aspirin during 24 h in patients with type 2 diabetes without cardiovascular disease. Thromb Res. 2018;161:1–6.PubMedCrossRef
294.
Zurück zum Zitat Bhatt DL, Marso SP, Hirsch AT, Ringleb PA, Hacke W, Topol EJ. Amplified benefit of clopidogrel versus aspirin in patients with diabetes mellitus. Am J Cardiol. 2002;90(6):625–8.PubMedCrossRef Bhatt DL, Marso SP, Hirsch AT, Ringleb PA, Hacke W, Topol EJ. Amplified benefit of clopidogrel versus aspirin in patients with diabetes mellitus. Am J Cardiol. 2002;90(6):625–8.PubMedCrossRef
295.
Zurück zum Zitat Parker WA, Storey RF. Long-term antiplatelet therapy following myocardial infarction: implications of PEGASUS-TIMI 54. Heart. 2016;102(10):783–9.PubMedCrossRef Parker WA, Storey RF. Long-term antiplatelet therapy following myocardial infarction: implications of PEGASUS-TIMI 54. Heart. 2016;102(10):783–9.PubMedCrossRef
296.
Zurück zum Zitat Parker WAE, Schulte C, Barwari T, Phoenix F, Pearson SM, Mayr M, et al. Aspirin, clopidogrel and prasugrel monotherapy in patients with type 2 diabetes mellitus: a double-blind randomised controlled trial of the effects on thrombotic markers and microRNA levels. Cardiovasc Diabetol. 2020;19(1):3.PubMedPubMedCentralCrossRef Parker WAE, Schulte C, Barwari T, Phoenix F, Pearson SM, Mayr M, et al. Aspirin, clopidogrel and prasugrel monotherapy in patients with type 2 diabetes mellitus: a double-blind randomised controlled trial of the effects on thrombotic markers and microRNA levels. Cardiovasc Diabetol. 2020;19(1):3.PubMedPubMedCentralCrossRef
297.
Zurück zum Zitat Willeit P, Zampetaki A, Dudek K, Kaudewitz D, King A, Kirkby NS, et al. Circulating microRNAs as novel biomarkers for platelet activation. Circ Res. 2013;112(4):595–600.PubMedCrossRef Willeit P, Zampetaki A, Dudek K, Kaudewitz D, King A, Kirkby NS, et al. Circulating microRNAs as novel biomarkers for platelet activation. Circ Res. 2013;112(4):595–600.PubMedCrossRef
298.
Zurück zum Zitat Zampetaki A, Willeit P, Tilling L, Drozdov I, Prokopi M, Renard JM, et al. Prospective study on circulating MicroRNAs and risk of myocardial infarction. J Am Coll Cardiol. 2012;60(4):290–9.PubMedCrossRef Zampetaki A, Willeit P, Tilling L, Drozdov I, Prokopi M, Renard JM, et al. Prospective study on circulating MicroRNAs and risk of myocardial infarction. J Am Coll Cardiol. 2012;60(4):290–9.PubMedCrossRef
300.
Zurück zum Zitat Ritter O, Kararigas G. Sex-Biased Vulnerability of the Heart to COVID-19. Mayo Clinic Proceedings. 2020;95(11):2332-5. Ritter O, Kararigas G. Sex-Biased Vulnerability of the Heart to COVID-19. Mayo Clinic Proceedings. 2020;95(11):2332-5.
302.
Zurück zum Zitat Nielsen SS, Vibholm LK, Monrad I, Olesen R, Frattari GS, Pahus MH, et al. SARS-CoV-2 elicits robust adaptive immune responses regardless of disease severity. EBioMedicine. 2021;68:103410.PubMedPubMedCentralCrossRef Nielsen SS, Vibholm LK, Monrad I, Olesen R, Frattari GS, Pahus MH, et al. SARS-CoV-2 elicits robust adaptive immune responses regardless of disease severity. EBioMedicine. 2021;68:103410.PubMedPubMedCentralCrossRef
303.
Zurück zum Zitat Augustine R, S A, Nayeem A, Salam SA, Augustine P, Dan P, et al. Increased complications of COVID-19 in people with cardiovascular disease: role of the renin-angiotensin-aldosterone system (RAAS) dysregulation. Chem Biol Interact. 2022;351:109738.PubMedCrossRef Augustine R, S A, Nayeem A, Salam SA, Augustine P, Dan P, et al. Increased complications of COVID-19 in people with cardiovascular disease: role of the renin-angiotensin-aldosterone system (RAAS) dysregulation. Chem Biol Interact. 2022;351:109738.PubMedCrossRef
304.
Zurück zum Zitat Liu F, Han K, Blair R, Kenst K, Qin Z, Upcin B, et al. SARS-CoV-2 infects endothelial cells in vivo and in Vitro. Front Cell Infect Microbiol. 2021;11:701278.PubMedPubMedCentralCrossRef Liu F, Han K, Blair R, Kenst K, Qin Z, Upcin B, et al. SARS-CoV-2 infects endothelial cells in vivo and in Vitro. Front Cell Infect Microbiol. 2021;11:701278.PubMedPubMedCentralCrossRef
305.
Zurück zum Zitat Odilov A, Volkov A, Abdullaev A, Gasanova T, Lipina T, Babichenko I. COVID-19: Multiorgan dissemination of SARS-CoV-2 is driven by pulmonary factors. Viruses. 2021;14(1). Odilov A, Volkov A, Abdullaev A, Gasanova T, Lipina T, Babichenko I. COVID-19: Multiorgan dissemination of SARS-CoV-2 is driven by pulmonary factors. Viruses. 2021;14(1).
306.
Zurück zum Zitat Giustino G, Pinney SP, Lala A, Reddy VY, Johnston-Cox HA, Mechanick JI, et al. Coronavirus and Cardiovascular Disease, Myocardial Injury, and Arrhythmia: JACC Focus Seminar. J Am Coll Cardiol. 2020;76(17):2011–23.PubMedPubMedCentralCrossRef Giustino G, Pinney SP, Lala A, Reddy VY, Johnston-Cox HA, Mechanick JI, et al. Coronavirus and Cardiovascular Disease, Myocardial Injury, and Arrhythmia: JACC Focus Seminar. J Am Coll Cardiol. 2020;76(17):2011–23.PubMedPubMedCentralCrossRef
307.
Zurück zum Zitat Lindner D, Fitzek A, Brauninger H, Aleshcheva G, Edler C, Meissner K, et al. Association of Cardiac infection with SARS-CoV-2 in confirmed COVID-19 autopsy cases. JAMA Cardiol. 2020;5(11):1281–5.PubMedCrossRef Lindner D, Fitzek A, Brauninger H, Aleshcheva G, Edler C, Meissner K, et al. Association of Cardiac infection with SARS-CoV-2 in confirmed COVID-19 autopsy cases. JAMA Cardiol. 2020;5(11):1281–5.PubMedCrossRef
308.
Zurück zum Zitat Caramaschi S, Kapp ME, Miller SE, Eisenberg R, Johnson J, Epperly G, et al. Histopathological findings and clinicopathologic correlation in COVID-19: a systematic review. Mod Pathol. 2021;34(9):1614–33.PubMedPubMedCentralCrossRef Caramaschi S, Kapp ME, Miller SE, Eisenberg R, Johnson J, Epperly G, et al. Histopathological findings and clinicopathologic correlation in COVID-19: a systematic review. Mod Pathol. 2021;34(9):1614–33.PubMedPubMedCentralCrossRef
309.
Zurück zum Zitat Puelles VG, Lutgehetmann M, Lindenmeyer MT, Sperhake JP, Wong MN, Allweiss L, et al. Multiorgan and renal tropism of SARS-CoV-2. N Engl J Med. 2020;383(6):590–2.PubMedCrossRef Puelles VG, Lutgehetmann M, Lindenmeyer MT, Sperhake JP, Wong MN, Allweiss L, et al. Multiorgan and renal tropism of SARS-CoV-2. N Engl J Med. 2020;383(6):590–2.PubMedCrossRef
310.
Zurück zum Zitat Patel VB, Bodiga S, Basu R, Das SK, Wang W, Wang Z, et al. Loss of angiotensin-converting enzyme-2 exacerbates diabetic cardiovascular complications and leads to systolic and vascular dysfunction: a critical role of the angiotensin II/AT1 receptor axis. Circ Res. 2012;110(10):1322–35.PubMedPubMedCentralCrossRef Patel VB, Bodiga S, Basu R, Das SK, Wang W, Wang Z, et al. Loss of angiotensin-converting enzyme-2 exacerbates diabetic cardiovascular complications and leads to systolic and vascular dysfunction: a critical role of the angiotensin II/AT1 receptor axis. Circ Res. 2012;110(10):1322–35.PubMedPubMedCentralCrossRef
311.
Zurück zum Zitat Jackson CB, Farzan M, Chen B, Choe H. Mechanisms of SARS-CoV-2 entry into cells. Nat Rev Mol Cell Biol. 2022;23(1):3–20.PubMedCrossRef Jackson CB, Farzan M, Chen B, Choe H. Mechanisms of SARS-CoV-2 entry into cells. Nat Rev Mol Cell Biol. 2022;23(1):3–20.PubMedCrossRef
312.
Zurück zum Zitat Bugert CL, Kwiat V, Valera IC, Bugert JJ, Parvatiyar MS. Cardiovascular Injury due to SARS-CoV-2. Curr Clin Microbiol Rep. 2021:1–11. Bugert CL, Kwiat V, Valera IC, Bugert JJ, Parvatiyar MS. Cardiovascular Injury due to SARS-CoV-2. Curr Clin Microbiol Rep. 2021:1–11.
313.
Zurück zum Zitat Mahamat-Saleh Y, Fiolet T, Rebeaud ME, Mulot M, Guihur A, El Fatouhi D, et al. Diabetes, hypertension, body mass index, smoking and COVID-19-related mortality: a systematic review and meta-analysis of observational studies. BMJ Open. 2021;11(10):e052777.PubMedCrossRef Mahamat-Saleh Y, Fiolet T, Rebeaud ME, Mulot M, Guihur A, El Fatouhi D, et al. Diabetes, hypertension, body mass index, smoking and COVID-19-related mortality: a systematic review and meta-analysis of observational studies. BMJ Open. 2021;11(10):e052777.PubMedCrossRef
314.
Zurück zum Zitat Viswanathan V, Puvvula A, Jamthikar AD, Saba L, Johri AM, Kotsis V, et al. Bidirectional link between diabetes mellitus and coronavirus disease 2019 leading to cardiovascular disease: a narrative review. World J Diabetes. 2021;12(3):215–37.PubMedPubMedCentralCrossRef Viswanathan V, Puvvula A, Jamthikar AD, Saba L, Johri AM, Kotsis V, et al. Bidirectional link between diabetes mellitus and coronavirus disease 2019 leading to cardiovascular disease: a narrative review. World J Diabetes. 2021;12(3):215–37.PubMedPubMedCentralCrossRef
316.
Zurück zum Zitat Abe T, Egbuche O, Igwe J, Jegede O, Wagle B, Olanipekun T, et al. Cardiovascular complications in COVID-19 patients with or without diabetes mellitus. Endocrinology, Diabetes & Metabolism. 2021;4(2):e00218.CrossRef Abe T, Egbuche O, Igwe J, Jegede O, Wagle B, Olanipekun T, et al. Cardiovascular complications in COVID-19 patients with or without diabetes mellitus. Endocrinology, Diabetes & Metabolism. 2021;4(2):e00218.CrossRef
317.
Zurück zum Zitat Spertus JA, Birmingham MC, Nassif M, Damaraju CV, Abbate A, Butler J, et al. The SGLT2 inhibitor canagliflozin in heart failure: the CHIEF-HF remote, patient-centered randomized trial. Nat Med. 2022;28(4):809–13.PubMedPubMedCentralCrossRef Spertus JA, Birmingham MC, Nassif M, Damaraju CV, Abbate A, Butler J, et al. The SGLT2 inhibitor canagliflozin in heart failure: the CHIEF-HF remote, patient-centered randomized trial. Nat Med. 2022;28(4):809–13.PubMedPubMedCentralCrossRef
318.
Zurück zum Zitat Ma CX, Ma XN, Guan CH, Li YD, Mauricio D, Fu SB. Cardiovascular disease in type 2 diabetes mellitus: progress toward personalized management. Cardiovasc Diabetol. 2022;21(1):74.PubMedPubMedCentralCrossRef Ma CX, Ma XN, Guan CH, Li YD, Mauricio D, Fu SB. Cardiovascular disease in type 2 diabetes mellitus: progress toward personalized management. Cardiovasc Diabetol. 2022;21(1):74.PubMedPubMedCentralCrossRef
319.
Zurück zum Zitat Lim S, Bae JH, Kwon HS, Nauck MA. COVID-19 and diabetes mellitus: from pathophysiology to clinical management. Nat Rev Endocrinol. 2021;17(1):11–30.PubMedCrossRef Lim S, Bae JH, Kwon HS, Nauck MA. COVID-19 and diabetes mellitus: from pathophysiology to clinical management. Nat Rev Endocrinol. 2021;17(1):11–30.PubMedCrossRef
320.
Zurück zum Zitat Garg A, Seeliger B, Derda AA, Xiao K, Gietz A, Scherf K, et al. Circulating cardiovascular microRNAs in critically ill COVID-19 patients. Eur J Heart Fail. 2021;23(3):468–75.PubMedCrossRef Garg A, Seeliger B, Derda AA, Xiao K, Gietz A, Scherf K, et al. Circulating cardiovascular microRNAs in critically ill COVID-19 patients. Eur J Heart Fail. 2021;23(3):468–75.PubMedCrossRef
321.
Zurück zum Zitat Schulte C, Barwari T, Joshi A, Theofilatos K, Zampetaki A, Barallobre-Barreiro J, et al. Comparative analysis of circulating noncoding RNAs versus protein biomarkers in the detection of myocardial Injury. Circ Res. 2019;125(3):328–40.PubMedPubMedCentralCrossRef Schulte C, Barwari T, Joshi A, Theofilatos K, Zampetaki A, Barallobre-Barreiro J, et al. Comparative analysis of circulating noncoding RNAs versus protein biomarkers in the detection of myocardial Injury. Circ Res. 2019;125(3):328–40.PubMedPubMedCentralCrossRef
322.
Zurück zum Zitat Gutmann C, Khamina K, Theofilatos K, Diendorfer AB, Burnap SA, Nabeebaccus A, et al. Association of cardiometabolic microRNAs with COVID-19 severity and mortality. Cardiovasc Res. 2022;118(2):461–74.PubMedCrossRef Gutmann C, Khamina K, Theofilatos K, Diendorfer AB, Burnap SA, Nabeebaccus A, et al. Association of cardiometabolic microRNAs with COVID-19 severity and mortality. Cardiovasc Res. 2022;118(2):461–74.PubMedCrossRef
323.
Zurück zum Zitat de Gonzalo-Calvo D, Benitez ID, Pinilla L, Carratala A, Moncusi-Moix A, Gort-Paniello C, et al. Circulating microRNA profiles predict the severity of COVID-19 in hospitalized patients. Transl Res. 2021;236:147–59.PubMedPubMedCentralCrossRef de Gonzalo-Calvo D, Benitez ID, Pinilla L, Carratala A, Moncusi-Moix A, Gort-Paniello C, et al. Circulating microRNA profiles predict the severity of COVID-19 in hospitalized patients. Transl Res. 2021;236:147–59.PubMedPubMedCentralCrossRef
324.
Zurück zum Zitat Lopez-Martinez C, Martin-Vicente P, Gomez de Ona J, Lopez-Alonso I, Gil-Pena H, Cuesta-Llavona E, et al. Transcriptomic clustering of critically ill COVID-19 patients. Eur Respir J. 2023;61(1). Lopez-Martinez C, Martin-Vicente P, Gomez de Ona J, Lopez-Alonso I, Gil-Pena H, Cuesta-Llavona E, et al. Transcriptomic clustering of critically ill COVID-19 patients. Eur Respir J. 2023;61(1).
325.
Zurück zum Zitat Made A, Greco S, Vausort M, Miliotis M, Schordan E, Baksi S, et al. Association of miR-144 levels in the peripheral blood with COVID-19 severity and mortality. Sci Rep. 2022;12(1):20048.PubMedPubMedCentralCrossRef Made A, Greco S, Vausort M, Miliotis M, Schordan E, Baksi S, et al. Association of miR-144 levels in the peripheral blood with COVID-19 severity and mortality. Sci Rep. 2022;12(1):20048.PubMedPubMedCentralCrossRef
326.
Zurück zum Zitat Garcia-Hidalgo MC, Gonzalez J, Benitez ID, Carmona P, Santisteve S, Perez-Pons M, et al. Identification of circulating microRNA profiles associated with pulmonary function and radiologic features in survivors of SARS-CoV-2-induced ARDS. Emerg Microbes Infect. 2022;11(1):1537–49.PubMedPubMedCentralCrossRef Garcia-Hidalgo MC, Gonzalez J, Benitez ID, Carmona P, Santisteve S, Perez-Pons M, et al. Identification of circulating microRNA profiles associated with pulmonary function and radiologic features in survivors of SARS-CoV-2-induced ARDS. Emerg Microbes Infect. 2022;11(1):1537–49.PubMedPubMedCentralCrossRef
327.
Zurück zum Zitat Meidert AS, Hermann S, Brandes F, Kirchner B, Buschmann D, Billaud JN, et al. Extracellular vesicle Associated miRNAs regulate signaling pathways involved in COVID-19 pneumonia and the progression to severe Acute Respiratory Corona Virus-2 syndrome. Front Immunol. 2021;12:784028.PubMedPubMedCentralCrossRef Meidert AS, Hermann S, Brandes F, Kirchner B, Buschmann D, Billaud JN, et al. Extracellular vesicle Associated miRNAs regulate signaling pathways involved in COVID-19 pneumonia and the progression to severe Acute Respiratory Corona Virus-2 syndrome. Front Immunol. 2021;12:784028.PubMedPubMedCentralCrossRef
328.
Zurück zum Zitat Martinez-Fleta P, Vera-Tome P, Jimenez-Fernandez M, Requena S, Roy-Vallejo E, Sanz-Garcia A, et al. A Differential signature of circulating miRNAs and cytokines between COVID-19 and Community-Acquired Pneumonia uncovers Novel Physiopathological Mechanisms of COVID-19. Front Immunol. 2021;12:815651.PubMedCrossRef Martinez-Fleta P, Vera-Tome P, Jimenez-Fernandez M, Requena S, Roy-Vallejo E, Sanz-Garcia A, et al. A Differential signature of circulating miRNAs and cytokines between COVID-19 and Community-Acquired Pneumonia uncovers Novel Physiopathological Mechanisms of COVID-19. Front Immunol. 2021;12:815651.PubMedCrossRef
329.
Zurück zum Zitat Wilson JC, Kealy D, James SR, Plowman T, Newling K, Jagger C, et al. Integrated miRNA/cytokine/chemokine profiling reveals severity-associated step changes and principal correlates of fatality in COVID-19. iScience. 2022;25(1):103672.PubMedCrossRef Wilson JC, Kealy D, James SR, Plowman T, Newling K, Jagger C, et al. Integrated miRNA/cytokine/chemokine profiling reveals severity-associated step changes and principal correlates of fatality in COVID-19. iScience. 2022;25(1):103672.PubMedCrossRef
330.
Zurück zum Zitat Houshmandfar S, Saeedi-Boroujeni A, Rashno M, Khodadadi A, Mahmoudian-Sani MR. miRNA-223 as a regulator of inflammation and NLRP3 inflammasome, the main fragments in the puzzle of immunopathogenesis of different inflammatory diseases and COVID-19. Naunyn Schmiedebergs Arch Pharmacol. 2021;394(11):2187–95.PubMedPubMedCentralCrossRef Houshmandfar S, Saeedi-Boroujeni A, Rashno M, Khodadadi A, Mahmoudian-Sani MR. miRNA-223 as a regulator of inflammation and NLRP3 inflammasome, the main fragments in the puzzle of immunopathogenesis of different inflammatory diseases and COVID-19. Naunyn Schmiedebergs Arch Pharmacol. 2021;394(11):2187–95.PubMedPubMedCentralCrossRef
331.
Zurück zum Zitat Wang Y, Zhu X, Jiang XM, Guo J, Fu Z, Zhou Z, et al. Decreased inhibition of exosomal miRNAs on SARS-CoV-2 replication underlies poor outcomes in elderly people and diabetic patients. Signal Transduct Target Ther. 2021;6(1):300.PubMedPubMedCentralCrossRef Wang Y, Zhu X, Jiang XM, Guo J, Fu Z, Zhou Z, et al. Decreased inhibition of exosomal miRNAs on SARS-CoV-2 replication underlies poor outcomes in elderly people and diabetic patients. Signal Transduct Target Ther. 2021;6(1):300.PubMedPubMedCentralCrossRef
332.
Zurück zum Zitat Falaleeva M, Welden JR, Duncan MJ, Stamm S. C/D-box snoRNAs form methylating and non-methylating ribonucleoprotein complexes: old dogs show new tricks. Bioessays. 2017;39(6). Falaleeva M, Welden JR, Duncan MJ, Stamm S. C/D-box snoRNAs form methylating and non-methylating ribonucleoprotein complexes: old dogs show new tricks. Bioessays. 2017;39(6).
333.
Zurück zum Zitat Noren Hooten N, Martin-Montalvo A, Dluzen DF, Zhang Y, Bernier M, Zonderman AB, et al. Metformin-mediated increase in DICER1 regulates microRNA expression and cellular senescence. Aging Cell. 2016;15(3):572–81.PubMedPubMedCentralCrossRef Noren Hooten N, Martin-Montalvo A, Dluzen DF, Zhang Y, Bernier M, Zonderman AB, et al. Metformin-mediated increase in DICER1 regulates microRNA expression and cellular senescence. Aging Cell. 2016;15(3):572–81.PubMedPubMedCentralCrossRef
334.
Zurück zum Zitat Cufi S, Vazquez-Martin A, Oliveras-Ferraros C, Quirantes R, Segura-Carretero A, Micol V, et al. Metformin lowers the threshold for stress-induced senescence: a role for the microRNA-200 family and miR-205. Cell Cycle. 2012;11(6):1235–46.PubMedCrossRef Cufi S, Vazquez-Martin A, Oliveras-Ferraros C, Quirantes R, Segura-Carretero A, Micol V, et al. Metformin lowers the threshold for stress-induced senescence: a role for the microRNA-200 family and miR-205. Cell Cycle. 2012;11(6):1235–46.PubMedCrossRef
335.
Zurück zum Zitat Giuliani A, Londin E, Ferracin M, Mensa E, Prattichizzo F, Ramini D, et al. Long-term exposure of human endothelial cells to metformin modulates miRNAs and isomiRs. Sci Rep. 2020;10(1):21782.PubMedPubMedCentralCrossRef Giuliani A, Londin E, Ferracin M, Mensa E, Prattichizzo F, Ramini D, et al. Long-term exposure of human endothelial cells to metformin modulates miRNAs and isomiRs. Sci Rep. 2020;10(1):21782.PubMedPubMedCentralCrossRef
336.
Zurück zum Zitat Arunachalam G, Lakshmanan AP, Samuel SM, Triggle CR, Ding H. Molecular interplay between microRNA-34a and Sirtuin1 in hyperglycemia-mediated impaired angiogenesis in endothelial cells: Effects of Metformin. J Pharmacol Exp Ther. 2016;356(2):314–23.PubMedCrossRef Arunachalam G, Lakshmanan AP, Samuel SM, Triggle CR, Ding H. Molecular interplay between microRNA-34a and Sirtuin1 in hyperglycemia-mediated impaired angiogenesis in endothelial cells: Effects of Metformin. J Pharmacol Exp Ther. 2016;356(2):314–23.PubMedCrossRef
337.
Zurück zum Zitat Zhang Y, Liu X, Zhang L, Li X, Zhou Z, Jiao L, et al. Metformin protects against H(2)O(2)-Induced Cardiomyocyte Injury by inhibiting the miR-1a-3p/GRP94 pathway. Mol Ther Nucleic Acids. 2018;13:189–97.PubMedPubMedCentralCrossRef Zhang Y, Liu X, Zhang L, Li X, Zhou Z, Jiao L, et al. Metformin protects against H(2)O(2)-Induced Cardiomyocyte Injury by inhibiting the miR-1a-3p/GRP94 pathway. Mol Ther Nucleic Acids. 2018;13:189–97.PubMedPubMedCentralCrossRef
338.
Zurück zum Zitat Zeng J, Zhu L, Liu J, Zhu T, Xie Z, Sun X, et al. Metformin protects against oxidative stress Injury Induced by Ischemia/Reperfusion via Regulation of the lncRNA-H19/miR-148a-3p/Rock2 Axis. Oxid Med Cell Longev. 2019;2019:8768327.PubMedPubMedCentralCrossRef Zeng J, Zhu L, Liu J, Zhu T, Xie Z, Sun X, et al. Metformin protects against oxidative stress Injury Induced by Ischemia/Reperfusion via Regulation of the lncRNA-H19/miR-148a-3p/Rock2 Axis. Oxid Med Cell Longev. 2019;2019:8768327.PubMedPubMedCentralCrossRef
339.
Zurück zum Zitat Guo X, Yu L, Chen M, Wu T, Peng X, Guo R, et al. miR-145 mediated the role of aspirin in resisting VSMCs proliferation and anti-inflammation through CD40. J Transl Med. 2016;14(1):211.PubMedPubMedCentralCrossRef Guo X, Yu L, Chen M, Wu T, Peng X, Guo R, et al. miR-145 mediated the role of aspirin in resisting VSMCs proliferation and anti-inflammation through CD40. J Transl Med. 2016;14(1):211.PubMedPubMedCentralCrossRef
340.
Zurück zum Zitat Zufferey A, Ibberson M, Reny JL, Nolli S, Schvartz D, Docquier M, et al. New molecular insights into modulation of platelet reactivity in aspirin-treated patients using a network-based approach. Hum Genet. 2016;135(4):403–14.PubMedCrossRef Zufferey A, Ibberson M, Reny JL, Nolli S, Schvartz D, Docquier M, et al. New molecular insights into modulation of platelet reactivity in aspirin-treated patients using a network-based approach. Hum Genet. 2016;135(4):403–14.PubMedCrossRef
341.
Zurück zum Zitat Kok MG, Mandolini C, Moerland PD, de Ronde MW, Sondermeijer BM, Halliani A, et al. Low miR-19b-1-5p expression in isolated platelets after aspirin use is related to aspirin insensitivity. Int J Cardiol. 2016;203:262–3.PubMedCrossRef Kok MG, Mandolini C, Moerland PD, de Ronde MW, Sondermeijer BM, Halliani A, et al. Low miR-19b-1-5p expression in isolated platelets after aspirin use is related to aspirin insensitivity. Int J Cardiol. 2016;203:262–3.PubMedCrossRef
Metadaten
Titel
Cardiovascular complications of diabetes: role of non-coding RNAs in the crosstalk between immune and cardiovascular systems
verfasst von
Gaia Spinetti
Martina Mutoli
Simona Greco
Federica Riccio
Soumaya Ben-Aicha
Franziska Kenneweg
Amela Jusic
David de Gonzalo-Calvo
Anne Yaël Nossent
Susana Novella
Georgios Kararigas
Thomas Thum
Costanza Emanueli
Yvan Devaux
Fabio Martelli
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
Cardiovascular Diabetology / Ausgabe 1/2023
Elektronische ISSN: 1475-2840
DOI
https://doi.org/10.1186/s12933-023-01842-3

Weitere Artikel der Ausgabe 1/2023

Cardiovascular Diabetology 1/2023 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Hodgkin Lymphom: BrECADD-Regime übertrifft die Erwartungen

05.06.2024 ASCO 2024 Kongressbericht

Das Kombinationsregime BrECADD mit Brentuximab vedotin ermöglichte in der Studie HD21 beim fortgeschrittenen klassischen Hodgkin-Lymphom eine unerwartet hohe progressionsfreie Überlebensrate von 94,3% nach vier Jahren. Gleichzeitig war das Regime besser tolerabel als der bisherige Standard eBEACOPP.

Antikörper-Drug-Konjugat verdoppelt PFS bei Multiplem Myelom

05.06.2024 ASCO 2024 Nachrichten

Zwei Phase-3-Studien deuten auf erhebliche Vorteile des Antikörper-Wirkstoff-Konjugats Belantamab-Mafodotin bei vorbehandelten Personen mit Multiplem Myelom: Im Vergleich mit einer Standard-Tripeltherapie wurde das progressionsfreie Überleben teilweise mehr als verdoppelt.

Neuer TKI gegen CML: Höhere Wirksamkeit, seltener Nebenwirkungen

05.06.2024 Chronische myeloische Leukämie Nachrichten

Der Tyrosinkinasehemmer (TKI) Asciminib ist älteren Vertretern dieser Gruppe bei CML offenbar überlegen: Personen mit frisch diagnostizierter CML entwickelten damit in einer Phase-3-Studie häufiger eine gut molekulare Response, aber seltener ernste Nebenwirkungen.

Hereditäres Angioödem: Tablette könnte Akuttherapie erleichtern

05.06.2024 Hereditäres Angioödem Nachrichten

Medikamente zur Bedarfstherapie bei hereditärem Angioödem sind bisher nur als Injektionen und Infusionen verfügbar. Der Arzneistoff Sebetralstat kann oral verabreicht werden und liefert vielversprechende Daten.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.