Abstract
Inflammatory bowel disease (IBD), consisting of ulcerative colitis and Crohn’s disease, mainly affects the gastrointestinal tract but is also known to have extraintestinal manifestations because of long-standing systemic inflammation. Several national cohort studies have found that IBD is an independent risk factor for the development of cardiovascular disorders. However, the molecular mechanisms by which IBD impairs the cardiovascular system are not fully understood. Although the gut-heart axis is attracting more attention in recent years, our knowledge of the organ-to-organ communication between the gut and the heart remains limited. In patients with IBD, upregulated inflammatory factors, altered microRNAs and lipid profiles, as well as dysbiotic gut microbiota, may induce adverse cardiac remodeling. In addition, patients with IBD have a three- to four times higher risk of developing thrombosis than people without IBD, and it is believed that the increased risk of thrombosis is largely due to increased procoagulant factors, platelet count/activity, and fibrinogen concentration, in addition to decreased anticoagulant factors. The predisposing factors for atherosclerosis are present in IBD and the possible mechanisms may involve oxidative stress system, overexpression of matrix metalloproteinases, and changes in vascular smooth muscle phenotype. This review focuses mainly on 1) the prevalence of cardiovascular diseases associated with IBD, 2) the potential pathogenic mechanisms of cardiovascular diseases in patients with IBD, and 3) adverse effects of IBD drugs on the cardiovascular system. Also, we introduce here a new paradigm for the gut-heart axis that includes exosomal microRNA and the gut microbiota as a cause for cardiac remodeling and fibrosis.
Keywords: cardiovascular disease, Crohn’s disease, gut microbiota, heart-gut axis, ulcerative colitis
INTRODUCTION
Systemic inflammation has been long regarded as a critical contributor to the onset and progression of cardiovascular diseases. Chronic inflammatory disorders, such as rheumatoid arthritis (RA), systemic lupus erythematosus, and psoriasis are associated with higher risks of atherosclerosis and arterial thromboembolic disease, including myocardial infarction (MI) and stroke (1–3). Inflammatory bowel diseases (IBD), including Crohn’s disease (CD) and ulcerative colitis (UC), are characterized by chronic inflammation of the gastrointestinal (GI) tract. According to the Centers for Disease Control and Prevention of the United States, an estimated 1.3% of US adults were diagnosed with IBD in 2015, and the prevalence is increasing. Although primarily a GI disorder, IBD affects other organs as well. Extraintestinal manifestations (EIMs) are common in both CD and UC, occurring in 25–40% of patients with IBD (4). As observed in other chronic inflammatory diseases, patients with IBD present a higher risk of developing cardiovascular diseases (CVD) (Fig. 1) (5–12). However, the gut-heart axis is a new field of research and the molecular mechanisms by which IBD induces CVD are poorly understood. There are several recent reviews of the pathogenesis of intestinal inflammation in IBD (13–16), therefore, the aim of this review is to introduce the concept of IBD-related heart and vascular diseases and particularly the potential causative mechanisms, including inflammatory cytokines such as tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, and IL-6, microRNAs (miRNAs), soluble suppression of tumorigenesis-2 (sST2), angiotensin (ANG) II, matrix metalloproteinases (MMPs), and lipopolysaccharide (LPS) (17–19). New concepts involving gut-derived microRNAs and brain-derived neurotrophic factor (BDNF) are considered. We will discuss the adverse effects of common IBD medications on the cardiovascular system and conclude with a brief description of major knowledge gaps and potential future studies.
Figure 1.
Overview of cardiovascular diseases in patients with inflammatory bowel disease (IBD). Increased proinflammatory cytokines such as tumor necrosis factor-α (TNFα), interleukin (IL)-6, and IL-1β dysregulate circulating microRNAs (miRNAs) including miR-155 and contribute to dysbiosis. Together, these factors induce molecular remodeling in the heart and vasculature, resulting in cardiovascular diseases. BDNF, brain-derived neurotrophic factor; VTE, venous thromboembolism.
INCREASED RISKS OF CVD IN IBD
Heart Diseases in Patients with IBD
Heart failure.
Patients with IBD have higher risks for heart failure (HF), pericarditis, myocarditis, and arrhythmias. A Danish nationwide cohort study found that IBD is associated with an increased risk of hospitalization for HF (8) and a worse prognosis after MI in those with IBD, in particular, in relation with flares of GI inflammation (20). A population-based cohort study from Mayo Clinic in Minnesota found that the relative risk of acute MI was significantly increased in patients with CD or UC (discussed under Vascular Diseases in Patients with IBD) (14). They reported that an increased relative risk of HF was noted among patients with UC, but not CD (11).
Pericarditis.
Pericarditis is an extraintestinal complication of IBD (21). A large population-based study by Bernstein et al. (22) revealed that patients with both CD (0.19% vs. 0.06% control) and UC (0.23% vs. 0.07% control) had a significantly greater likelihood of having pericarditis than population controls, with a similar increased risk for women and men. Although several case studies reported pericarditis in patients with IBD (23, 24), the actual prevalence of the condition is quite low.
Myocarditis.
Myocarditis had a total risk of 4.6 per 100,000 years in patients with IBD based on a 16-year Danish nationwide cohort study of 15,572 patients with IBD, which is significantly higher than the general population. The incidence rate ratio for myocarditis was 8.3 for CD and 2.6 for UC when compared with the general population (25, 26). As for pediatric patients with IBD, 0.9% of the patients were found to have myocarditis with the occurrence increasing with age, according to a Finnish registry study (27). The overall incidence of myocarditis was 1.95/100,000 person-years. There are multiple case reports on patients with IBD with myocarditis (28–30). Mesalamine, a therapy for UC, may also induce myocarditis (31–33).
Arrhythmia.
Arrhythmia is also a frequent manifestation of cardiac involvement in patients with IBD (34). Nationwide cohort studies in Denmark and South Korea found that IBD significantly increased the risk of atrial fibrillation (AF), particularly during active flare-ups, which represents the most common sustained cardiac arrhythmia among patients with IBD (35, 36). Pattanshetty et al. (37) found that the prevalence of AF in patients with IBD from Ohio was higher across all age groups compared with the general population and that the patients with IBD with AF were much younger compared with the controls. Yorulmaz et al. (38) reported that prolonged QT dispersion was significantly more frequent in patients with stricturing and penetrating CD when compared with control subjects. Pediatric patients with IBD in remission also have significantly longer P-wave dispersion, QT dispersion, and corrected QT interval dispersion compared with age- and sex-matched controls, suggesting that pediatric patients with IBD may carry potential risks for serious atrial and ventricular arrhythmias over time even during remission (39).
Vascular Diseases in Patients with IBD
Coronary heart disease.
A 2011 study of Haapamaki et al. (40), comprising 2,831 patients with IBD from Finland, found that coronary heart disease occurred significantly more frequently in patients with IBD than controls (P = 0.004). Biondi et al. (6) noted that patients with IBD presented a 6.45-fold higher risk of carotid atherosclerotic plaque. The aforementioned Danish nationwide cohort study also found that the disease activity in IBD is associated with increased risks of MI, stroke, and cardiovascular death (7). A systematic review and meta-analysis suggested that IBD is associated with a modest increase in the risk of cardiovascular morbidity, including cerebrovascular accidents and ischemic heart disease (IHD), particularly in women (9), and this was confirmed by subsequent meta-analyses (10, 41). Another retrospective cohort study revealed that although IHD prevalence was higher in patients with IBD (n = 19,163), most diagnoses predated the diagnosis of IBD (12), indicating that IHD may occur before or after the diagnosis of IBD. This idea was supported by studies showing that extraintestinal manifestations such as MI may occur in up to 24% of patients with IBD before the onset of intestinal symptoms (42). Recently, a large study with 29,090,220 patients from 26 nations found that the prevalence of MI was significantly higher in patients with UC or CD versus patients with non-IBD, and that younger patients have the highest relative risk (43). A Korean nationwide study in 2019 concluded that the risk of MI is higher in patients with CD, particularly patients aged <40 yr, than in the general population, but only female patients with UC had an increased risk of MI (44). More recently, a retrospective cohort study of 1,435 patients with IBD from 12 IBD centers and 1,588 non-IBD controls was conducted in China and the risk of IHD was found to be significantly higher in patients with IBD than in non-IBD controls, and it peaked between the ages of 18 and 35 yr (45).
Although most observations support an association of IBD with coronary heart disease, Osterman et al. (46) reported that patients with UC had a significantly increased risk of first-time acute MI compared with patients from general practice in unadjusted, but not adjusted, analysis; patients with CD did not have an increased risk of MI in either unadjusted or adjusted analyses. Using a nationwide inpatient database, Barnes et al. (47) found that hospitalizations for acute MI were decreased among patients with IBD. Taken together, we believe there is a higher risk of coronary heart disease in patients with IBD, especially in women, and there is conflicting, but no consistent, increased risk of coronary disease in one form of IBD over the other.
Venous thromboembolism.
Deep vein thrombosis, pulmonary embolism, cerebral venous sinus thrombosis, as well as retinal, hepatic, portal, and mesenteric vein thromboses, carry substantial morbidity and mortality with even higher mortality rates in patients with IBD (48, 49). The prevalence of venous thromboembolism (VTE) in patients with IBD ranges from 1 to 7%, which is at least three times higher than the general population (10). A prospective study that included 13,756 patients with IBD and 71,672 matched controls from the United Kingdom found that patients with IBD had a higher risk of VTE than did controls (P < 0.0001; absolute risk 2.6 per 1,000 per person-years); and the increase in risk was much more prominent (P < 0.0001; 9.0 per 1,000 person-years) at the time of an IBD flare (50). Using the US Nationwide Inpatient Sample between 2000 and 2018, Faye et al. (51) found that the VTE cases in hospitalized individuals with IBD significantly increased from 192 to 295 cases per 10,000 hospitalizations (P < 0.001); the difference remained significant when stratified by UC and CD as well as by deep vein thrombosis and pulmonary embolism. A Japanese cohort study found that the VTE incidence rate in IBD was 1.03% (102.5 per 100,000 IBD person-years) in Japan (52). Similarly, the incidence of VTE was 0.9% in the hospitalized cohort (115 per 100,000 IBD person-years) in an East Asian study and the incidence was comparable between UC and CD (53). A multicenter study followed 8,549 Chinese patients with IBD for 12,373 person-years showed that the incidence rate of VTE was 371.8 per 100,000 person-years (54). In comparison, three population-wide estimates of VTE rates in South Korea, Taiwan, and Hong Kong reported annual incidences of 13.8, 15.9, and 19.9 per 100,000 person-years, respectively, in the general Asian population (55).
Arterial thromboembolism.
It is important to evaluate the risks of arterial thromboembolism (ATE) because they are associated with higher morbidity and mortality and occur after a longer duration of disease (56, 57). Although it is less pronounced than VTE, the risk of ATE, including ischemic stroke, focal white matter ischemia, cardiac ischemia, peripheral vascular disease, and mesenteric ischemia, is increased in IBD (49, 56). A study included 17,487 patients with IBD and 69,948 controls showed that patients with IBD had a markedly increased risk of acute mesenteric ischemia. Subgroup analysis revealed that women > 40 yr of age with IBD were at increased risk of MI, whereas those <40 yr exhibited a twofold higher risk for stroke (58). Another study found that while the risk for IHD was significantly increased for all IBD in both males and females, only CD was associated with increased risk for cerebrovascular disease (57). For undifferentiated ATE only females and those aged 0 to 39 yr and 40 to 59 yr had significantly increased risks. Lin et al. analyzed a total of 11,067 patients with IBD and 43,765 controls for the risk of peripheral arterial disease (PAD) by using Cox proportional hazards regression models and found that the risk of developing PAD was 1.29-fold in patients with IBD compared with the control cohort, after age, sex, and comorbidities were adjusted. They also found that those patients with IBD who required two or more hospitalizations per year were nearly 27.5-fold more likely to have PAD compared with the controls (59).
POTENTIAL PATHOGENIC MECHANISMS OF CVD IN IBD
Pathophysiological Mechanisms Leading to IBD
It is known that the etiology of IBD is multifactorial and its pathophysiology involves complex genetic, environmental, epithelial, microbial, and immune factors (60). Here, we only briefly summarize the major risk factors contributing to IBD and a detailed description of IBD pathophysiology is beyond the scope of this review on cardiovascular manifestations.
Host genetics.
Multiple studies suggested a heritable risk for IBD. Genome-wide association studies and other analyses have identified more than 240 risk variants that affect intracellular pathways recognizing microbial products such as NOD2; the autophagy pathway facilitating the recycling of intracellular organelles and removal of intracellular microorganisms; genes regulating epithelial barrier function such as ECM1; and pathways modulating innate and adaptive immunity (13, 60).
Environmental factors.
Despite the exact mechanism is still unknown, accumulating evidence suggests that environmental risk factors play a role in developing both CD and UC. Early-life exposures, lifestyle and hygiene, vaccinations, exposure to drugs, surgeries, psychological factors, dietary intake and nutrients, and gastrointestinal pathogens may increase the risk of developing IBD (61).
Mucosal immunity.
The mucosal immune system represents the largest component of the immune system, and it is a consensus that immunological abnormalities play an important role in IBD pathogenesis (60). Dysregulation of the innate immune system as well as the adaptive immune system is attributed to many characteristics of chronic inflammatory processes in IBD reveals. Studies have found that B cells, effector T cells, regulatory T cells, and memory T cells are critical in maintaining gut mucosal homeostasis and health and may contribute to IBD development.
Gut microbiota.
Intestinal microbiota is considered the major driver of IBD because both commensal and pathogenic microorganisms determine the consequence of an infection (13). Although it is difficult to demonstrate a definitive cause-effect relationship between intestinal microbiota and IBD, it is believed that microbial factors play important roles in the pathophysiology of IBD through impacting the immune systems and affecting host metabolism.
Potential Mechanisms of Cardiac Remodeling
Inflammatory cytokines may mediate cardiac impairment in IBD.
The cytokine responses characterizing IBD are the crucial pathophysiological elements governing the initiation, evolution, and, ultimately, the resolution of the inflammation (62). Particularly, the imbalance between proinflammatory and anti-inflammatory cytokines in IBD often causes disease perpetuation and tissue damage. Well-known proinflammatory cytokines, including TNF-α, IL-1β, and IL-6 that are all increased in IBD, have been linked to diminished cardiac function (Fig. 2).
Figure 2.

Potential mechanisms of heart diseases in patients with inflammatory bowel diseases (IBDs). 1) Increased soluble suppression of tumorigenesis-2 (ST2) (sST2) in inflammatory bowel disease (IBD) blocks interleukin (IL)-33/ST2 signaling, stopping the protection effect of IL-33. 2) Inflammatory cytokines, such as tumor necrosis factor-α (TNFα) and IL-6, and angiotensin II (ANG II) activate cardiac fibroblasts, leading to proliferation/migration of fibroblasts, matrix degradation, extracellular matrix (ECM) deposition, recruitment of inflammatory cells, and generation of myofibroblasts. GM-CSF, granulocyte-macrophage colony-stimulating factor; MMP, matrix metalloproteinase.
TNF-α-mediated adverse cardiac remodeling may involve cardiomyocytes, macrophages, (myo)fibroblasts, and the extracellular matrix (ECM). In cardiomyocytes, TNF-α exerts negative inotropic actions by perturbing calcium homeostasis (63) and may trigger an apoptotic response by activating intrinsic cell death pathways (64). In macrophages, TNF-α could stimulate the synthesis of other proinflammatory cytokines with proapoptotic, negative inotropic, and matrix-degrading properties, and may upregulate the expression of inducible nitric oxide synthase (iNOS) (65). In fibroblasts, TNF-α may disrupt the balance between MMPs and their inhibitors, leading to abnormal ECM degradation (66, 67). In the microvasculature, TNF-α increases permeability through modulation of endothelial cyclooxygenase-2 (68) and induces expression of endothelial adhesion molecules, such as intercellular adhesion molecule 1 and vascular cell adhesion molecule 1, thus enhancing adhesive interactions between circulating leukocytes and the endothelial lining (69). Neutrophils or proinflammatory monocytes may be trapped in the cardiac microcirculation, thus contributing to tissue injury and cardiac dysfunction. A multicenter longitudinal study found that anti-TNF-α therapy effectively reduced inflammation and reduced cardiovascular risk in patients with IBD (70). Anti-TNF-α therapy also seems to prevent cardiovascular events in most patients with RA. In patients with RA with advanced heart damage, however, anti-TNF-α agents would interfere with the beneficial preconditioning effects of TNF-α, introducing a “rheumatological dilemma” in clinical practice (71).
Experimental evidence suggests an important role for IL-1 signaling in the pathogenesis of cardiac dysfunction and adverse remodeling associated with HF (72). IL-1β could induce cardiovascular remodeling by activating nuclear factor κ B (NF-κB) and inducing MMPs (73, 74). Graff and Gozal found that intravenous injection of serial doses of recombinant IL-1β induced monophasic increases in ventilation, heart rate, and blood pressure in adults (75). Mice with genetic disruption of IL-1 signaling due to loss of IL1R1, the signaling receptor for IL-1, had attenuated adverse remodeling after MI, exhibiting suppressed inflammatory responses (76). IL-1 Trap, a dimeric fusion protein consisting of the ligand-binding domains of the extracellular portions of the IL-1 receptor components linked to the Fc portion of human immunoglobulin G1, also ameliorated cardiac remodeling and reduced cardiomyocyte apoptosis after experimental acute MI in mice (77). However, some studies suggested that endogenous activation of subinflammatory levels of IL-1β may also play an adaptive role following pressure overload mediating growth factor-induced compensatory hypertrophy (78).
IL-6 induces the transcription factor STAT-3 via trans-signaling and activation of IL-6/STAT3 signaling pathway in IBD is well documented. Inhibition of IL-6/JAK/STAT3 pathway is proven effective treatments of IBD (79, 80). The role of IL-6/STAT3 signaling in cardiovascular system has also been extensively investigated (81, 82). Cardiomyocyte-specific overexpression of STAT3 in mice led to spontaneous concentric cardiac hypertrophy, demonstrating an important role for STAT3 in cardiomyocyte growth (83). IL-6-induced hypertrophic growth of cardiomyocytes was abolished by a negative regulator of gp130/JAK/STAT3 signaling, SOCS3, suggesting that IL-6-mediated activation of STAT3 promotes cardiac hypertrophy (84). However, there is evidence that p42/p44-MAPK and PI3K are sufficient for IL-6/gp130 to induce hypertrophy as well as survival in cardiomyocytes without JAK/STAT activation (85). Of note, IL-6 can activate NF-κB (86), albeit IL-6 is also a proven target of NF-κB (87, 88), forming a positive feedback loop.
Interferon-γ (IFN-γ) released by activated T-lymphocytes is also a major cytokine in patients with IBD and IFN-γ induces production of C-reactive protein (CRP) in human monocyte-derived macrophages and dendritic cells. Of note, macrophages are critically involved in plaque formation. IFN-γ and macrophages also play important roles in the development of oxidative stress for antimicrobial and antitumoral defense within the cell-mediated immune response (89). In fact, IFN-γ is considered the most important trigger for reactive oxygen species (ROS) formation and release whereas oxidative stress is a key player in the atherogenesis and progression of cardiovascular disease. In addition, IFN-γ can stimulate indoleamine-2,3-dioxygenase, an enzyme degrading tryptophan to kynurenine. Recent studies demonstrated that IFN-γ also exerts potent vessel-directed pathologic breakdown of the vascular barrier through disruption of the adherens junction protein VE-cadherin (90). It is well known that IFN-γ activates the JAK-STAT pathways and transcriptional activation in the cardiovascular system (91). Furthermore, IFN-γ can modulate large-conductance, calcium-activated potassium channels through PKG and PKA-related pathways in human cardiac fibroblasts (92).
Granulocyte-macrophage colony-stimulating factor (GM-CSF) is well recognized as a proliferative agent for hematopoietic cells and exerts a priming function on neutrophils (93). In GM-CSF-induced bone marrow differentiation, myeloperoxidase (MPO) activity and oncogene expression are upregulated (94). GM-CSF functions as a potent chemoattractant in vitro and can recruit neutrophils from the microvasculature and induce extravascular migration in vivo, suggesting that GM-CSF may contribute significantly to recruitment during intestinal inflammation. In patients with CD, serum levels of CM-CSF antibody are significantly higher when compared with patients with UC or control subjects and high GM-CSF antibody titers have been considered as a risk marker for aggressive CD behavior and complications including surgery (95). Paradoxically, recombinant CSFs have emerged as a potential tool for the treatment of IBD, showing some promise in the treatment of patients with active CD with recombinant human GM-CSF (96). In the heart, Blyszczuk et al. (97) found that GM-CSF promotes inflammatory dendritic cell formation but does not contribute to disease progression in experimental autoimmune myocarditis, a model of postinflammatory dilated cardiomyopathy. Conversely, during the early phase of MI, GM-CSF was found to facilitate infarct expansion in association with increased monocyte recruitment and inappropriate collagen synthesis in the infarcted region (98). Further study by Anzai et al. (99) identified GM-CSF as both a key contributor to the pathogenesis of MI and a potential therapeutic target. Thus, the role of GM-CSF seems to be controversial in both IBD and heart disease.
Angiotensin II induces cardiac fibrosis.
Higher levels of angiotensin (ANG) I and II were detected in the colonic mucosal biopsies of patients with CD compared with healthy controls, and ANG II levels have been shown to correlate with the degree of macroscopic inflammation as well (100–102). The renin-angiotensin-aldosterone system, of which ANG II seems to be the predominant effector, promotes several physiological and pathological functions, including the development of cardiac fibrosis (Fig. 2). The ANG II type 1 receptor (AT1R) mediates several effects of ANG II in (myo)fibroblasts, including cell proliferation, migration, and induction of ECM protein synthesis. After cardiac injury, the levels of ANG II are quickly elevated, inducing proliferation of cardiac fibroblasts and stimulating the expression of collagen (103, 104). In addition, ANG II activation of AT1R induces the expression TGF-β1, which in turn induces both hypertrophy and fibrosis in the heart (105).
ST2/IL-33 signaling influences cardiac remodeling.
The cardiovascular mechanisms associated with maladaptive remodeling are complex and not yet fully understood. The mechano-sensitive signaling pathways are the main systems that have been investigated. These systems are closely associated with cardiac fibrosis since they are involved from the beginning to the late stages of remodeling responses. The first biomechanical system induced during cardiac stretching is ST2/IL-33 and is composed of three mediators belonging to the IL-1 family: two isoforms of ST2, i.e., soluble ST2 (sST2) and transmembrane ST2L, with opposing biological activities, and the third component of this system, IL-33 that drives activity after binding to the ST2 moieties (106). The ST2/IL-33 system is cardioprotective during heart stretch responses when IL-33 is released into the extracellular space where it associates with ST2L, promoting cell survival, and blocking profibrotic intracellular signaling. When sST2 is released into the extracellular space, however, the ST2/IL-33 system becomes harmful to the heart: sST2 acts as a decoy receptor of IL-33 to silence ST2L/IL33 signaling and can sequester most IL-33, stopping its cardioprotection through ST2L (107).
Although research is currently ongoing to identify sources and molecular mechanisms involved in sST2 production, there is evidence that total ST2 proteins, as well as IL-33 levels, were highly expressed in patients with active UC (108). sST2 was the predominant form expressed in IBD and its expression strongly correlates with ST2 levels in the serum. Furthermore, ST2 levels are correlated with disease severity and serum inflammatory cytokines and serum ST2 levels may differentiate active from inactive UC (109). More recently, Boga et al. (110) found that serum ST2 levels were significantly increased not only in active UC versus inactive UC but also in active CD compared with inactive CD. Proinflammatory pathways in UC and CD driven by cytokines produced during the inflammatory processes might be responsible for increased sST2 levels in patients with active IBD. In support of this, Kumar et al. (111) observed that fibroblasts exposed to the proinflammatory cytokines IL-1 and TNF-α expressed sST2 rather than ST2L. Abnormal sST2 levels in the cardiac extracellular space have become a major event in the principal cardiovascular disorders involving biomechanical stretch responses (112, 113).
Dysregulated microRNAs may impair heart function in IBD.
MicroRNAs (miRNAs) are small RNA molecules containing 18–25 nucleotides. Because miRNAs normally assist in fine tuning the complexity of gene expression, the alterations in their expression often play crucial roles in the development of many human diseases. It has been shown that miRNAs have a significant role in regulating the pathogenesis of myocardial remodeling by modulating cardiac hypertrophy, cardiomyocytes injury, cardiac fibrosis, angiogenesis, and inflammatory response through multiple mechanisms.
Using a rat model of chronic colitis induced by dextran sodium sulfate (DSS), our group found that chronic colitis significantly altered the miRNA profile in the adult rat and this was accompanied by a decreased ejection fraction, increased left ventricular mass, elevated B-type natriuretic protein in the plasma, and decreased brain-derived neurotrophic factor (BDNF) in the myocardium (114). Among the 56 dysregulated cardiac miRNAs, miR-155 was confirmed to suppress BDNF, a neurotrophin mediating wide-ranging effects on the cardiovascular system. We also found that recombinant IL-1β increased miR-155 in cardiac myoblast (H9c2) cells and in the rat heart in vivo, suggesting that chronic colitis impairs heart function through an IL-1β/miR-155/BDNF signaling axis. A pathogenic function of miR-155 in the heart was also supported by a previous study showing that intraperitoneal injection of LPS increased cardiac miR-155 and adversely induced heart dysfunction in mice and that pharmacological inhibition of miR-155 using antagomiR improved heart function (115). To determine if any circulating miRNAs contribute to cardiac remodeling in patients with IBD, we isolated plasma exosomes from patients with UC and control subjects and profiled miRNAs using next-generation sequencing. We identified 20 differentially expressed miRNAs, including miR-29b that was markedly upregulated in patients with UC (116). Furthermore, we demonstrated that exosomal miR-29b of gut origin suppressed cardiac BDNF just as miR-155 did, suggesting that exosomes derived from the inflamed intestine mediate the gut-heart cross talk and play a role in IBD-induced cardiac impairment.
Dysbiotic gut microbiota contributes to heart dysfunction.
It is believed that dysbiotic gut microbiome plays a role in HF and the “gut hypothesis,” which postulates that bowel wall edema and an impaired intestinal barrier during HF causes microbiome translocation into the host circulation, endotoxemia, and consequently a heightened systemic inflammatory state, has prevailed over the years (117). In the gut of patients with IBD, salutary bacteria, such as Bifidobacterium adolescentis and Faecalibacterium prausnitzii, become depleted, whereas virulent bacteria, such as Proteobacteria, proliferate (118). Using fecal microbiota from wild-type C57BL/6J mice, we performed fecal microbiota transplantation (FMT) on both normal C57BL/6J and IL-10 knockout mice with constitutive gut inflammation. Echocardiography revealed a significant decrease in left ventricle ejection fraction in the IL-10−/− mice compared with that in the C57BL/6J mice, and the decrease was significantly abrogated by FMT (119). Our findings suggest that healthy donor FMT will mitigate colitis-induced cardiac impairment, highlighting the importance of healthy gut microbiota to heart function. Further studies are warranted to validate the findings and elucidate the underlying mechanisms.
Potential Pathogenesis of Arrhythmia in IBD
Atrial electromechanical delay (EMD) has been considered as an early marker of AF. Efe et al. found that patients with active CD or UC had significantly prolonged left and right intra-atrial EMDs and interatrial EMD compared with patients on remission and healthy controls. The left and right intra-atrial EMDs and interatrial EMD were also higher in patients with IBD on remission compared with healthy controls, suggesting that chronic inflammation elsewhere may cause chronic structural and electrophysiological changes in the atrial tissue resulting in slow conduction (120). These findings were confirmed by Nar et al. (121) in patients with UC.
The acute phase reactant CRP and the cytokine IL-6 have been implicated in AF development (122). Increased circulating CRP may stay in atrial tissue and thereby induce myocarditis (37), as well as electrical changes in the atrium (120, 123), whereas IL-6 is strongly correlated with increased left atrium size (123) by activating MMP2 (124). In addition, P wave dispersion, which is accepted as a risk factor for the development of AF, was found to be high in patients with UC or CD (125). Furthermore, patients with UC were found to have significantly lower autonomic functions, especially parasympathetic function, which might trigger arrhythmia (126). It should be noted that patients with IBD treated with ciprofloxacin or azathioprine may develop arrhythmia (127, 128).
Vaze et al. (129) reported that circulating miR-106b, 26a-5p, 484, 20a-5p are associated with prevalent AF. Both miR-106b and miR-20a can regulate autophagy induced by leucine deprivation in C2C12 cells via targeting ULK1, suggesting that these miRNAs may affect myocardial remodeling by inhibiting autophagy in cardiomyocytes, which in turn leads to the development of AF (130). It is noteworthy that miR106b levels were significantly increased in the intestinal epithelia of patients with active CD compared with controls (131). miR-26a, an anti-inflammatory miRNA that suppresses IL-6 production in myeloid cells, was also increased in patients with both CD and UC (132). These findings suggest that dysregulated miR-106b and miR-26a may contribute to AF in patients with IBD.
Potential Mechanisms of Vascular Diseases in IBD
Altered lipid profile contributes to coronary artery disease.
Dyslipidemia is an established risk factor for coronary heart disease. A large cohort study found that low total cholesterol and high triglyceride levels are more frequent in patients with IBD (in particular, CD) compared with healthy controls (133). Multiple studies confirmed that lipid profile and lipid-related metabolites were significantly altered in patients with IBD, more specifically in CD (134–136). Epidemiologic studies provided evidence of a strong association between triglycerides and the development of coronary heart disease (137). It is also well documented that innate immune cells, including macrophages and dendritic cells, can sense certain lipid species, such as saturated fatty acids and oxidized low-density lipoprotein, and produce proinflammatory cytokines and chemokines detrimental to the heart (138, 139).
Reactive oxygen species induce endothelial dysfunction.
The activated immune cells that reach the mucosa in IBD stimulate the production of large quantities of ROS that are potentially harmful, such as superoxide and nitric oxide (NO), and the release of MPO producing hypochlorous acid that has strong oxidizing activity. Furthermore, endothelial dysfunction in IBD has been reported (140, 141). Endothelium-derived factors, such as NO and endothelin-1, may play a role in the development of arterial stiffness (142, 143) (Fig. 3). Inflammatory reactions lead to release of free radicals from leukocytes and activated macrophages (144).
Figure 3.

Pathogenic mechanisms of vascular diseases in inflammatory bowel diseases (IBDs). 1) Coagulant factors V, VII, VIII, X, VWF, and fibrinogen, as well as platelet count and activity, are increased while anticoagulation-related protein S and antithrombin are decreased in IBD. This leads to increased risks of venous thromboembolism. 2) Increased reactive oxygen species (ROS) and lipopolysaccharide (LPS) cause endothelial dysfunction, leading to vascular hyperplasia and foam cell formation. Matrix metalloproteinases (MMPs) and vascular calcification also contribute to arterial stiffness. GM-CSF, Granulocyte-macrophage colony-stimulating factor; HOCl, hypochlorous acid; MPO, myeloperoxidase; NO, nitric oxide; VWF, von Willebrand factor.
The most abundant free radical in human tissues is the superoxide anion, which is primarily produced in an enzymatic reaction catalyzed by xanthine oxidase (XO) in the GI tract (145, 146). Superoxide anion is then converted to H2O2. In neutrophils, H2O2 is utilized by MPO to produce the hypochlorite ion. In patients with IBD, circulating XO binds to vascular endothelial cells and produces site-specific oxidative injury of the artery. Although superoxide anion is a highly reactive, highly unstable, and very short-lived form of ROS that only acts near the place of its origin, H2O2 can freely diffuse across cell membranes and oxidize molecules located further, e.g., membrane lipids in arteries (146, 147).
Clinical studies reported the presence of high levels of nitrite/nitrate in the plasma, urine, and the lumen of the colon of patients with IBD, suggesting a deleterious role of NO (148, 149). It is known that proinflammatory cytokines are involved in the upregulation of iNOS expression in IBD, which generates high levels of NO (150). Excessive NO could be detrimental by creating reactive nitric oxygen species, such as peroxynitrite anion and nitroxyl anion. Gunnett et al. (151) reported that iNOS induces vascular dysfunction by activation of soluble guanylate cyclase and limiting availability of tetrahydrobiopterin. A study by Nagareddy et al. (152) found that induction of iNOS in cardiovascular tissues contributed to the depressed pressor responses to vasoactive agents and potentially endothelial dysfunction. It is widely accepted, however, that eNOS-produced NO reduces arterial stiffness. More research needs to be done to determine if and how NO generated from iNOS contributes to arterial stiffness in IBD.
The injured intestinal mucosal barrier in IBD allows microbial LPS and other endotoxins into circulation. LPS can induce the expression of proinflammatory cytokines, which in turn cause endothelial dysfunction and macrophage foam cell formation, and eventually lead to atherosclerosis (153). LPS has many detrimental effects, including oxidation of low-density lipoproteins toxic to endothelial cells and activation of macrophages that enhance atherosclerosis (154). Endothelial dysfunction may also be associated with hyperplasia of vascular smooth muscle cells and increased synthesis of collagen, resulting in structural arterial stiffening (155).
MMPs contribute to arterial stiffening.
MMPs are zinc-dependent endopeptidases that cleave several ECM proteins and as such modulate outcome of various physiological and pathological processes including MI, atherosclerosis, and congestive HF. Several MMPs are overexpressed in IBD (156–158). Specifically, MMP9 upregulation in animal models of colitis and human IBD is unequivocal and plasma MMP9 has been considered a marker of disease activity in IBD (159, 160). Collagen and elastin are two important components of the ECM in the vessel wall. Because of their collagenolytic and elastinolytic activities, MMPs degrade the ECM and hence create uncoiled, less effective collagen and broken and frayed elastin molecules, stiffening vessel walls (161). In addition to structural ECM components, MMP substrates also include a multitude of ligand and receptor substrates such as cytokines, chemokines, growth factors, and adhesion molecules that alter cellular migration, adhesion, and activation. MMPs, therefore, exert a strong influence on cardiac remodeling through multiple mechanisms. Under inflammatory conditions, MMPs are activated in different cell types, including macrophages, (myo)fibroblasts, endothelial cells, and smooth muscle cells (162, 163).
Calcification of vascular smooth muscle cells.
A recent study showed that more than one-third of patients with IBD in Greece had moderate to severe abdominal aortic calcium deposition, a marker of early atherosclerosis (164). Vascular calcification is multifactorial, and phosphate plays a key role in the process. Smooth muscle cells take up extracellular phosphate through a neutral sodium-phosphate cotransporter and this phosphate uptake leads to serial cellular changes, which ultimately result in bioapatite formation (165–167). TNF-α has been shown to induce alkaline phosphatase expression and calcification in vascular smooth muscle cells (168, 169). IL-1β and IL-6 also induced osteogenic activity in vascular cells (170). Thus, increased expression of these proinflammatory cytokines is likely causative to vascular smooth muscle calcification in patients with IBD (Fig. 3).
Pathogenesis of hypercoagulability as a cause of VTE and ATE in IBD is multifactorial.
Accumulating evidence suggests that it is not one particular mechanism that leads to hypercoagulability in IBD but rather a complex interplay of systems (171). On the molecular level, the upregulation of the inflammatory and coagulation systems create an enhanced thrombotic state that involves the coagulation cascade, natural coagulation inhibitors, fibrinolytic system, endothelium, immune system, and platelets (172). More specifically, this increased risk of VTE has been attributed to higher levels of inflammatory cytokines, acute phase reactants, procoagulants, and lower levels of anticoagulants (173). It has been shown that coagulation factors V, VII, VIII, X, XI, XII, von Willebrand factor, and fibrinogen, as well as production of fibrin and thrombin formation are all elevated during an IBD flare (174). Patients with IBD also have increased platelet counts (thrombocytosis) and increased platelet activity during active disease states (175). In addition, studies have suggested that patients with IBD have lower levels of protein S and antithrombin during active disease states, which are important drivers of anticoagulation (Fig. 3) (176). Alterations in coagulation and platelet function and susceptibility to thrombosis have also been demonstrated in animal models of IBD. Animal studies have shown that inflammatory cytokines, including TNF-α, IL-1β, and IL-6, played important roles as mediators of the platelet abnormalities and augmented thrombus development (177).
Homocysteine plays a role in VTE in IBD.
Hyperhomocysteinemia is likely another risk factor for VTE in patients with IBD. Homocysteine is a nonprotein-forming sulfur amino acid derived from S-adenosylmethionine and it can be remethylated to methionine and transsulfurated to cystathionine (178). Homocysteine can function as a procoagulation factor because it causes alterations in the vessel wall, increasing tissue factor and factor V expression, decreasing thrombomodulin and t-plasminogen activator expression, and attenuating protein C activation (179). Both plasma and mucosal homocysteine levels were significantly higher in patients with IBD compared with healthy controls and correlated with disease activity (180). Elevated plasma homocysteine levels may promote the development of arterial and venous thrombosis.
Involvement of the Central and Peripheral Nervous System in IBD-Associated CVD
It is well known that IBD causes EIM in the central and peripheral nervous system, which could contribute to pathogenesis of CVD. The incidence of neurological disorders in patients with IBD ranges between 0.25% and 47.5% (181). Accumulating evidence suggests that the central nervous system interacts dynamically via the vagus nerve with the intestinal immune system to modulate inflammation through humoral and neural pathways, using a mechanism also referred to as the intestinal cholinergic anti-inflammatory pathway. Lindgren et al. (182) reported vagal dysfunction in patients with UC. Patients with UC in clinical remission were also found to have significantly lower parasympathetic function when compared with those with CD and healthy controls (126). Coruzzi et al. (183) found that all indexes related to cardiac vagal control were significantly lower in patients with quiescent UC versus controls or patients with CD. Patients with CD usually have normal peripheral motor nerve function, but almost half of the patients show signs of autonomic neuropathy, suggesting autonomic nerve dysfunction as a feature of CD (184). It is well known that elevated TNF-α in IBD activates neurons in sensory circumventricular organs including the subfornical organ (SFO) (185). The SFO projects to the hypothalamic paraventricular nucleus, which has projections to the preganglionic sympathetic neurons in the intermediolateral cell column of the spinal cord to increase sympathetic nerve activity. Increased sympathetic nerve activity is detrimental to the heart and would enhance the progression of the disease.
It has been recognized that the heart consists of neuronal and nonneuronal cholinergic system (186). There is evidence that acetylcholine of both neuronal and nonneuronal origin acts in the heart through muscarinic and nicotinic receptors (187). The action of acetylcholine in the heart is terminated rapidly by cholinesterases (ChE), acetylcholinesterase, and butyrylcholinesterase. Recently, a larger case-control study by Shao et al. (188) found a negative association between serum ChE levels and the Crohn’s disease activity index (CDAI) score of patients with CD and the simple clinical colitis activity index score of patients with UC. Patients with both CD and UC presented substantially lower serum ChE levels when compared with healthy controls. In addition, patients with CD displayed significantly lower serum ChE levels than patients with UC.
Alterations in the sympathetic system and parasympathetic nervous system, also known as the cholinergic system, have been clearly documented in patients with CVD and IBD (189). However, the interplay between the sympathetic and parasympathetic system is complex and poorly understood. Recent studies suggest that the increased cholinergic tone in the heart by cholinesterase inhibitors has a positive effect on some cardiovascular disorders such as heart failure, suggesting that the cholinesterase inhibitors could be beneficial in managing certain cardiovascular disorders including IBD-induced CVD (190).
Role of Obesity, Diabetes, and Metabolic Complications in CVD Development of IBD
The production of proinflammatory cytokines by obesity through adipose tissue can have a dramatic impact on the initiation, clinical course, and management of IBD. Because of overlapping alterations in immune and inflammatory pathways, concomitant diabetes may be an important comorbidity in patients with IBD. Studies have shown that diabetes is a predictor of surgery in patients with CD (191) and a risk factor for postoperative surgical site infections in patients with fulminant UC (192). Metabolic syndrome is also a comorbid condition of IBD (193). In fact, metabolic syndrome and IBD share some pathophysiological characteristics, such as inflammation, insufficient immune response, and dysregulation of adipose tissue. Both metabolic syndrome and IBD are defined by chronic inflammation due to shared immune pathways. It is reasonable to anticipate that obesity, diabetes, and metabolic complications increase the chance of CVD in patients with IBD. However, there is very limited data available regarding the cardiovascular disease outcomes associated with patients with IBD with concomitant obesity, diabetes, and metabolic complications.
ADVERSE EFFECTS OF CURRENT IBD DRUGS ON THE CARDIOVASCULAR SYSTEM
Higher risk of cardiovascular diseases, particularly thrombotic events, in patients with IBD has garnered attention from patients and gastroenterologists alike. Recent consensus meetings directed toward thromboembolism in IBD have provided recommendations to improve the quality of care of patients and to prevent potentially life-threatening complications of thrombosis (194, 195). Although it is imperative that caregivers continue to promote prevention of CVD in patients with IBD, it is important to understand that medicines used in IBD may have salutary or detrimental effects on the cardiovascular system. Thus, in this review, we will briefly discuss the benefits and side effects of therapeutic drugs currently used in patients with IBD. The effects of IBD medications on the cardiovascular system, as well as the impact of CVD medications on the GI tract, are summarized in Table 1.
Table 1.
Effects of IBD drugs on the cardiovascular system and CVD drugs on the GI tract
| Medication | CV Effects | GI Effects |
|---|---|---|
| IBD medications | ||
| 5-Aminosalicylates | Myopericarditis (196) | Induction and maintenance of remission in IBD (197, 198) |
| Arrythmia (199) | ||
| Corticosteroid | HF (200) Hypertension Hyperlipidemia |
Induction of IBD remission |
| Azathioprine | Arrythmia (201) | Induction and maintenance of remission |
| Anti-TNF | HF (202) Decreased ATE (203) |
Induction and maintenance of remission (204) |
| Anti-integrins (Vedolizumab) | Cerebral hemorrhage (205) | Induction and maintenance of remission |
| Anti-IL12/IL23 (Ustekinumab) | Dyslipidemia (206) | Induction and maintenance of remission |
| Tofacitinib | VTE (199) Induce herpes zoster (207) |
Induction and maintenance of remission (208–210) |
| CVD medications | ||
| ACEI/ARB | Lower BP and treat HF | Improve disease outcomes in patients with IBD (211) |
| Aspirin | Anti-platelet and prevention for patients with high risk of CVD | Risk of GI bleeding Possibly increase the risk of developing CD (212) |
| Heparin | Reduce risk of VTE | Alleviate IBD inflammation (213) |
| Ridogrel | Anti-platelet | Reduce mucosal thromboxane B2 (214) |
| Statins | Lower cholesterol | Decrease risk of new onset IBD (215) Relieve inflammation in CD patients (216) |
ACEI, angiotensin-converting enzyme inhibitors; ARB, angiotensin receptor blocker; BP, blood pressure; CD, Crohn’s disease; CV, cardiovascular; CVD, cardiovascular diseases; GI, gastrointestinal; HF, heart failure; IBD, inflammatory bowel disease; IL, interleukin; TNF, tumor necrosis factor; VTE, venous thromboembolism.
Direct Effect of IBD Drugs on the Cardiovascular System
TNF-α inhibitors.
Anti-TNF agents are effective in treating patients with IBD and they are the most commonly used biologics for IBD in the United Kingdom (204). A nationwide French cohort study suggested that anti-TNF treatment is associated with a decreased risk of acute arterial events in patients with IBD (203). However, some guidelines suggested that anti-TNF agents should be used with caution in patients with heart failure (71, 217). The in vivo evidence on the deleterious effects of TNF-α in the failing myocardium, and early clinical studies suggesting attenuated dysfunction in small groups of patients receiving TNF-α antagonists (218) fueled large clinical trials to investigate the effectiveness of TNF-α blockade with soluble TNFR (etanercept) in patients with heart failure with reduced ejection fraction (HFrEF). Unfortunately, the results were disappointing and the trials were halted prematurely because of the lack of clinical benefit (219). Combined trial data also showed no effect of etanercept on the primary end point of death or heart failure hospitalization in patients with HfrEF (220). A phase II anti-TNF-α trial examined the effects of infliximab, a chimeric monoclonal anti-TNF-α antibody, in patients with HfrEF, and found that TNF-α antagonism using infliximab had adverse effects, increasing all-cause mortality and heart failure hospitalizations in comparison with conventional treatment (221).
JAK-STAT inhibitors.
Tofacitinib is a first-generation pan JAK inhibitor primarily targeting JAK1 and JAK3 and the first oral small molecule approved for use in UC in the United States in 2012 (222). All tofacitinib studies demonstrated clinical improvement, which confirm its effectiveness for induction and maintenance in UC (208). Safety analyses concluded that tofacitinib treatment in patients with UC was associated with dose-dependent risk of herpes zoster, but the safety profile of tofacitinib in the UC clinical development program was manageable (207). In three phase 3, randomized, double-blind, placebo-controlled trials of tofacitinib therapy in adult patients with UC, adverse cardiovascular events occurred in 5 of 1,120 patients who received tofacitinib and in none who received placebo; tofacitinib was also associated with increased lipid levels compared with placebo. One patient in the 10-mg tofacitinib group in the induction 1 trial died from aortic dissection (209, 210). Of note, nearly all the involved patients had multiple risk factors that may have contributed to the development of these complications.
5-Aminosalicylic acid derivatives.
5-Aminosalicylic acid (5-ASA) derivatives work by blocking the activity of cyclooxygenase and lipoxygenase, thereby reducing the production of both limbs of arachidonic acid metabolism. Mesalazine, or mesalamine, is currently the core treatment of mild-to-moderate UC (223). It is also a less effective option for patients with CD preferring to avoid steroids (197, 198). Studies reported that pericarditis occurs as a complication of drug therapy by 5-ASA derivatives such as sulfasalazine, mesalamine, and balsalazide (196, 224). A mechanistic investigation revealed that mesalazine induced a dose- and time-dependent increase in mitochondrial ROS production, succinate dehydrogenase inhibition, mitochondrial enlargement, and cytochrome C release in rat heart mitochondria, suggesting that cardiotoxic effects of mesalazine are most likely associated with mitochondrial dysfunction and ROS formation (225).
Impact of IBD Drugs on Liver and Kidneys
TNF-α inhibitors.
TNF-α inhibitors are generally safe with a pretty low risk of causing severe drug-induced liver injury (DILI) (226). Alanine transaminase (ALT) levels are commonly elevated in patients with IBD treated with anti-TNF but, even in case of moderate or severe ALT increases, resolution seems to be the usual outcome (227). Liver injury is self-limiting in most patients and drug withdrawal is usually unnecessary. There are no standard guidelines for the management of anti-TNF-related liver injury. Patients with elevated ALT can continue the anti-TNF treatment because severe liver injury is rare and alternative medical options are limited in case of severe active IBD.
Drug-induced nephrotoxicity is rare in patients with IBD and there is paucity of data available (226). A possible mechanism underlying the development of renal complications during anti-TNF treatment could be interaction of anti-TNF antibodies with TNF in glomerular visceral epithelial cells. It has also been suggested that infliximab/TNF binding on the lymphocyte plasma membranes may induce apoptosis (228).
JAK-STAT inhibitors.
Currently, most safety data of JAK inhibitors are from patients with rheumatoid arthritis. Studies revealed elevated ALT or aspartate aminotransferase (AST) in patients exposed to tofacitinib, baricitinib, or upadacitinib compared with placebo, with most cases of elevated ALT/AST being asymptomatic and transient (229, 230). Elevated serum creatinine level is the most common adverse effect of tofacitinib (231). More studies, particularly long-term studies, are required to further illustrate the safety of JAK-STAT inhibitors.
5-ASA derivatives.
It has been reported that the incidence of elevated liver enzymes was in the range of 0–4% in patients treated with either low- or high dose of mesalazine (232). Drug-induced liver injury is the likely diagnosis if ALT and AST are commonly elevated to the range of 500–1,000 U/L. In multiple randomized controlled trials, however, no patient had to be withdrawn from the studies albeit case reports have indicated that mesalazine may cause a granulomatous hepatitis on biopsy (233, 234). Although sulfasalazine has been associated with cholestatic disease, granulomatous hepatitis, and acute liver failure, the incidence of DILI caused by sulfasalazine is low. A study including 4.7 million sulfasalazine prescriptions found only 3.1 cases of hepatitis per million prescriptions among patients with IBD (235). Mechanistically, transcriptomic studies in mice showed that sulfasalazine is involved in liver injury through multiple pathways, including redox processes, cytochrome p450 pathway, glutathione metabolism, epidermal growth factor receptor pathway, and the cytochrome p450 2C55 pathway (236, 237).
In 1990, the UK Medicines Safety Committee issued an alert on nephrotoxic reactions to mesalamine. Clinical trials suggested an annual risk of 0.26%, and questionnaire data estimated 1 case per 4,000 patient-years (238). Renal toxicity was reported in patients treated with the combined form of sulfasalazine (5-ASA bound to sulfapyridine) or the coated form of 5-ASA (mesalazine, olsalazine) (239). A few cases of renal toxicity have been reported with sulfasalazine in the form of an idiosyncratic. Mesalazine, on the other hand, has been reported to be responsible for a significant number of patients developing nephritis during its administration (240). Renal impairment may occur in up to 1 in 100 patients treated with 5-ASA, but clinically significant damage would occur in only 1 in 500 patients (241). Thus, measurements of serum creatinine before and during treatment have been advocated.
Management of CVD in patients with IBD is a significant challenge. Fortunately, traditional treatment for CVD, such as aspirin, statins, and propranolol are thought to be generally safe and useful. The use of angiotensin receptor blockers and angiotensin-converting enzyme inhibitors was also associated with improved disease outcomes in patients with IBD (242).
LIMITATIONS AND FUTURE STUDIES
Mounting evidence suggests that IBD is associated with increased risk for CVD especially in young adults and women, including heart failure, arrhythmias, and thrombosis. However, mechanistic studies are limited, and we just begin to understand how the heart communicates with the gut in normal and disease states. The potential mechanisms for increased risks of cardiovascular events in patients with IBD may involve elevated levels of inflammatory cytokines, ROS, MMPs, homocysteine and angiotensin, endothelial dysfunction, dyslipidemia, hypercoagulability, dysregulated miRNAs, dysbiotic gut microbiota, and drug toxicity. Multiple knowledge gaps remain and need to be addressed. More preclinical studies in animal models are warranted, which will help better understand the molecular mechanisms behind the development of CVD, particularly progressive remodeling of the cardiovascular system in response to long-term inflammation and thrombosis. Such research will also aid the development of new drugs targeting the gut-heart cross talk and VTE/ATE. Future studies on the role of dysbiosis in gut-heart cross talk will not only help understand how microbiota imbalance influences the cardiovascular system in IBD, but also identify specific microbial strains(s) responsible for the pathogenesis of CVD. Future research should pay more attention to how changes in gut microbial metabolites impact the cardiovascular system in patients with IBD. Molecular mechanisms by which MMPs of gut origin induce cardiac remodeling and fibrosis should be explored.
Perspectives and Significance
Studies have identified highly expressed inflammatory factors, altered microRNAs and lipid profiles, and gut microbiota imbalance as risk factors of cardiac impairment. In addition, increased procoagulant factors, platelet count/activity, and fibrinogen concentration as well as decreased anticoagulant factors are thought to play important roles in the development of VTE. Furthermore, atherosclerosis is likely a consequence of oxidative stress, overexpression of matrix metalloproteinases, and changes in vascular smooth muscle phenotype. These studies provided valuable insights into the molecular mechanisms by which IBD predisposes patients to CVD. We must acknowledge, however, that IBD is a complex gastrointestinal tract disorder affecting many if not all organs in the body. There is also heterogeneity in the inflammatory burden among the IBD population, making it challenging to evaluate risk factors and identify pathogenic mechanisms for CVD in such patients. Further preclinical studies with animal models and clinical studies on patients are needed to decipher how the cardiovascular system as well as other organs respond to the disease signals emanating from the inflamed intestine and to unravel the complex mechanisms of CVD development. An important goal would be to identify patients with IBD with long standing or severe disease who are at most risk for the development of CVD. Research into mechanisms should uncover blood-borne biomarkers that might be useful for surveillance. Clinical epidemiologic studies might then indicate which patients with IBD should be treated with drugs discovered by this mechanistic research in both preclinical models and in patients with disease. It is hoped that research in IBD CVD will also inform research in other severe inflammatory diseases, such as rheumatoid arthritis, where the risk of developing CVD is also great or even greater. The clinical management of IBD has been forced to increasingly adopt a multidisciplinary team approach including gastroenterology, cardiology, and hematology. Clinicians are now aware of the detrimental effects of chronic inflammation on the cardiovascular system, and VTE prophylaxis is strongly considered for hospitalized patients with IBD and sometimes even extended postdischarge.
GRANTS
This work was supported in part by the National Institutes of Health Grant R01 HL152683 (to Q. Li).
DISCLOSURES
No conflicts of interest, financial or otherwise, are declared by the authors.
AUTHOR CONTRIBUTIONS
Q.L. conceived and designed research; Y.X. prepared figures; Y.X. and Q.L. drafted manuscript; D.W.P., X.L., and Q.L. edited and revised manuscript; Y.X., D.W.P., X.L., and Q.L. approved final version of manuscript.
REFERENCES
- 1. Zanatta E, Colombo C, D'Amico G, d'Humieres T, Dal Lin C, Tona F. Inflammation and coronary microvascular dysfunction in autoimmune rheumatic diseases. Int J Mol Sci 20: 5563, 2019. doi: 10.3390/ijms20225563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Frieri M, Stampfl H. Systemic lupus erythematosus and atherosclerosis: review of the literature. Autoimmun Rev 15: 16–21, 2016. doi: 10.1016/j.autrev.2015.08.007. [DOI] [PubMed] [Google Scholar]
- 3. Siddiqi HK, Ridker PM. Psoriasis and atherosclerosis. Circ Res 123: 1183–1184, 2018. doi: 10.1161/CIRCRESAHA.118.314073. [DOI] [PubMed] [Google Scholar]
- 4. Levine JS, Burakoff R. Extraintestinal manifestations of inflammatory bowel disease. Gastroenterol Hepatol 7: 235–241, 2011. [PMC free article] [PubMed] [Google Scholar]
- 5. Wolf D, Ley K. Immunity and inflammation in atherosclerosis. Circ Res 124: 315–327, 2019. doi: 10.1161/CIRCRESAHA.118.313591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Biondi RB, Salmazo PS, Bazan SGZ, Hueb JC, de Paiva SAR, Sassaki LY. Cardiovascular risk in individuals with inflammatory bowel disease. Clin Exp Gastroenterol 13: 107–113, 2020. doi: 10.2147/CEG.S243478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Kristensen SL, Ahlehoff O, Lindhardsen J, Erichsen R, Jensen GV, Torp-Pedersen C, Nielsen OH, Gislason GH, Hansen PR. Disease activity in inflammatory bowel disease is associated with increased risk of myocardial infarction, stroke and cardiovascular death—a Danish nationwide cohort study. PLoS One 8: e56944, 2013. [Erratum in PLoS One 8: 2013]. doi: 10.1371/journal.pone.0056944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Kristensen SL, Ahlehoff O, Lindhardsen J, Erichsen R, Lamberts M, Khalid U, Nielsen OH, Torp-Pedersen C, Gislason GH, Hansen PR. Inflammatory bowel disease is associated with an increased risk of hospitalization for heart failure: a Danish Nationwide Cohort study. Circ Heart Fail 7: 717–722, 2014. doi: 10.1161/CIRCHEARTFAILURE.114.001152. [DOI] [PubMed] [Google Scholar]
- 9. Singh S, Singh H, Loftus EV Jr, Pardi DS. Risk of cerebrovascular accidents and ischemic heart disease in patients with inflammatory bowel disease: a systematic review and meta-analysis. Clin Gastroenterol Hepatol 12: 382–393.e1: quiz e22, 2014. doi: 10.1016/j.cgh.2013.08.023. [DOI] [PubMed] [Google Scholar]
- 10. Sun HH, Tian F. Inflammatory bowel disease and cardiovascular disease incidence and mortality: a meta-analysis. Eur J Prev Cardiol 25: 1623–1631, 2018. doi: 10.1177/2047487318792952. [DOI] [PubMed] [Google Scholar]
- 11. Aniwan S, Pardi DS, Tremaine WJ, Loftus EV Jr.. Increased risk of acute myocardial infarction and heart failure in patients with inflammatory bowel diseases. Clin Gastroenterol Hepatol 16: 1607–1615.e1, 2018. doi: 10.1016/j.cgh.2018.04.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Close H, Mason JM, Wilson DW, Hungin AP, Jones R, Rubin G. Risk of ischaemic heart disease in patients with inflammatory bowel disease: cohort study using the General Practice Research Database. PLoS One 10: e0139745, 2015. doi: 10.1371/journal.pone.0139745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Guan Q. A comprehensive review and update on the pathogenesis of inflammatory bowel disease. J Immunol Res 2019: 7247238, 2019. doi: 10.1155/2019/7247238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Dipasquale V, Romano C. Genes vs environment in inflammatory bowel disease: an update. Expert Rev Clin Immunol 18: 1005–1013, 2022. doi: 10.1080/1744666X.2022.2108407. [DOI] [PubMed] [Google Scholar]
- 15. Tavakoli P, Vollmer-Conna U, Hadzi-Pavlovic D, Grimm MC. A review of inflammatory bowel disease: a model of microbial, immune and neuropsychological integration. Public Health Rev 42: 1603990, 2021. doi: 10.3389/phrs.2021.1603990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Kofla-Dłubacz A, Pytrus T, Akutko K, Sputa-Grzegrzółka P, Piotrowska A, Dzięgiel P. Etiology of IBD—is it still a mystery? Int J Mol Sci 23: 12445, 2022. doi: 10.3390/ijms232012445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Neurath MF. Cytokines in inflammatory bowel disease. Nat Rev Immunol 14: 329–342, 2014. doi: 10.1038/nri3661. [DOI] [PubMed] [Google Scholar]
- 18. Marafini I, Sedda S, Dinallo V, Monteleone G. Inflammatory cytokines: from discoveries to therapies in IBD. Expert Opin Biol Ther 19: 1207–1217, 2019. doi: 10.1080/14712598.2019.1652267. [DOI] [PubMed] [Google Scholar]
- 19. Violi F, Cammisotto V, Bartimoccia S, Pignatelli P, Carnevale R, Nocella C. Gut-derived low-grade endotoxaemia, atherothrombosis and cardiovascular disease. Nat Rev Cardiol 20: 24–37, 2023. doi: 10.1038/s41569-022-00737-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Kristensen SL, Ahlehoff O, Lindhardsen J, Erichsen R, Lamberts M, Khalid U, Nielsen OH, Torp-Pedersen C, Gislason GH, Hansen PR. Prognosis after first-time myocardial infarction in patients with inflammatory bowel disease according to disease activity: nationwide cohort study. Circ Cardiovasc Qual Outcomes 7: 857–862, 2014. doi: 10.1161/CIRCOUTCOMES.114.000918. [DOI] [PubMed] [Google Scholar]
- 21. Antonopoulos DA, Huse SM, Morrison HG, Schmidt TM, Sogin ML, Young VB. Reproducible community dynamics of the gastrointestinal microbiota following antibiotic perturbation. Infect Immun 77: 2367–2375, 2009. doi: 10.1128/IAI.01520-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Bernstein CN, Wajda A, Blanchard JF. The clustering of other chronic inflammatory diseases in inflammatory bowel disease: a population-based study. Gastroenterology 129: 827–836, 2005. doi: 10.1053/j.gastro.2005.06.021. [DOI] [PubMed] [Google Scholar]
- 23. Das BB, Dodson M, Guzman A. Recurrent pericarditis in an adolescent with Crohn’s colitis. Ann Pediatr Cardiol 13: 256–259, 2020. doi: 10.4103/apc.APC_16_20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Mukhopadhyay D, Nasr K, Grossman BJ, Kirsner JB. Pericarditis associated with inflammatory bowel disease. JAMA 211: 1540–1542, 1970. doi: 10.1001/jama.1970.03170090056015. [DOI] [PubMed] [Google Scholar]
- 25. Sørensen HT, Fonager KM. Myocarditis and inflammatory bowel disease. A 16-year Danish nationwide cohort study. Dan Med Bull 44: 442–444, 1997. [PubMed] [Google Scholar]
- 26. Cesa K, Cunningham C, Harris T, Sunseri W. A review of extraintestinal manifestations & medication-induced myocarditis and pericarditis in pediatric inflammatory bowel disease. Cureus 14: e26366, 2022. doi: 10.7759/cureus.26366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Arola A, Pikkarainen E, Sipilä JO, Pykäri J, Rautava P, Kytö V. Occurrence and features of childhood myocarditis: a nationwide study in Finland. J Am Heart Assoc 6: e005306, 2017. doi: 10.1161/JAHA.116.005306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Wang YY, Shi W, Wang J, Li Y, Tian Z, Jiao Y. Myocarditis as an extraintestinal manifestation of ulcerative colitis: a case report and review of the literature. World J Clin Cases 10: 9750–9759, 2022. doi: 10.12998/wjcc.v10.i27.9750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Caio G, Lungaro L, Caputo F, Muccinelli M, Marcello MC, Zoli E, Volta U, De Giorgio R, Zoli G. Recurrent myocarditis in a patient with active ulcerative colitis: a case report and review of the literature. BMJ Open Gastroenterol 8: e000587, 2021. doi: 10.1136/bmjgast-2020-000587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Piazza I, Burti C, Assolari A, Greco S, Benetti A, Cosentini R, Ferrero P. Acute myocarditis as first presentation of severe ulcerative colitis in a young man. Multidisciplinary management and long-term follow-up. J Cardiol Cases 26: 46–50, 2022. doi: 10.1016/j.jccase.2022.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Ibrahim A, Khalil C, Megaly M, Tawadros M, Mosleh W, Corbelli J. Mesalamine-induced myocarditis in a young athlete: can he run again? Cureus 11: e3978, 2019. doi: 10.7759/cureus.3978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Shergill S. Mesalazine-induced myopericarditis: a case report. Eur Heart J Case Rep 5: ytaa508, 2021. doi: 10.1093/ehjcr/ytaa508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Baker WL, Saulsberry WJ, Elliott K, Parker MW. Cardiac MRI-confirmed mesalamine-induced myocarditis. BMJ Case Rep 2015: bcr2015210689, 2015. doi: 10.1136/bcr-2015-210689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Gawałko M, Balsam P, Lodziński P, Grabowski M, Krzowski B, Opolski G, Kosiuk J. Cardiac arrhythmias in autoimmune diseases. Circ J 84: 685–694, 2020. doi: 10.1253/circj.CJ-19-0705. [DOI] [PubMed] [Google Scholar]
- 35. Kristensen SL, Lindhardsen J, Ahlehoff O, Erichsen R, Lamberts M, Khalid U, Torp-Pedersen C, Nielsen OH, Gislason GH, Hansen PR. Increased risk of atrial fibrillation and stroke during active stages of inflammatory bowel disease: a nationwide study. Europace 16: 477–484, 2014. doi: 10.1093/europace/eut312. [DOI] [PubMed] [Google Scholar]
- 36. Choi Y-J, Choi E-K, Han K-D, Park J, Moon I, Lee E, Choe W-S, Lee S-R, Cha M-J, Lim W-H, Oh S. Increased risk of atrial fibrillation in patients with inflammatory bowel disease: a nationwide population-based study. World J Gastroenterol 25: 2788–2798, 2019. doi: 10.3748/wjg.v25.i22.2788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Pattanshetty DJ, Anna K, Gajulapalli RD, Sappati-Biyyani RR. Inflammatory bowel “Cardiac” disease: Point prevalence of atrial fibrillation in inflammatory bowel disease population. Saudi J Gastroenterol 21: 325–329, 2015. doi: 10.4103/1319-3767.166208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Yorulmaz E, Sezgin A, Yorulmaz H, Adali G, Ciftci H. Prolonged QT dispersion in inflammatory bowel disease. World J Gastroenterol 19: 65–71, 2013. doi: 10.3748/wjg.v19.i1.65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Bornaun HA, Yılmaz N, Kutluk G, Dedeoğlu R, Öztarhan K, Keskindemirci G, Tulunoğlu A, Şap F. Prolonged P-wave and QT dispersion in children with inflammatory bowel disease in remission. Biomed Res Int 2017: 6960810, 2017. doi: 10.1155/2017/6960810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Haapamaki J, Roine RP, Turunen U, Farkkila MA, Arkkila PE. Increased risk for coronary heart disease, asthma, and connective tissue diseases in inflammatory bowel disease. J Crohns Colitis 5: 41–47, 2011. doi: 10.1016/j.crohns.2010.09.008. [DOI] [PubMed] [Google Scholar]
- 41. Feng W, Chen G, Cai D, Zhao S, Cheng J, Shen H. Inflammatory bowel disease and risk of ischemic heart disease: an updated meta-analysis of cohort studies. J Am Heart Assoc 6: e005892, 2017. doi: 10.1161/JAHA.117.005892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Rogler G, Singh A, Kavanaugh A, Rubin DT. Extraintestinal manifestations of inflammatory bowel disease: current concepts, treatment, and implications for disease management. Gastroenterology 161: 1118–1132, 2021. doi: 10.1053/j.gastro.2021.07.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Panhwar MS, Mansoor E, Al-Kindi SG, Sinh P, Katz J, Oliveira GH, Cooper GS, Ginwalla M. Risk of myocardial infarction in inflammatory bowel disease: a population-based national study. Inflamm Bowel Dis 25: 1080–1087, 2019. doi: 10.1093/ibd/izy354. [DOI] [PubMed] [Google Scholar]
- 44. Choi YJ, Lee DH, Shin DW, Han K-D, Yoon H, Shin CM, Park YS, Kim N. Patients with inflammatory bowel disease have an increased risk of myocardial infarction: a nationwide study. Aliment Pharmacol Ther 50: 769–779, 2019. doi: 10.1111/apt.15446. [DOI] [PubMed] [Google Scholar]
- 45. Fang L, Gao H, Gao X, Wu W, Miao Y, Zhang H, Guleng B, Zhang H, Wang Y, Li M, Yang H, Gao X, Liang J, Cao Q, Shen J, Ran Z, Wu K, Qian J, Chen M, Liu Z. Risks of cardiovascular events in patients with inflammatory bowel disease in China: a retrospective multicenter cohort study. Inflamm Bowel Dis 28 Suppl 2: S52–S58, 2022. doi: 10.1093/ibd/izab326. [DOI] [PubMed] [Google Scholar]
- 46. Osterman MT, Yang YX, Brensinger C, Forde KA, Lichtenstein GR, Lewis JD. No increased risk of myocardial infarction among patients with ulcerative colitis or Crohn’s disease. Clin Gastroenterol Hepatol 9: 875–880, 2011. doi: 10.1016/j.cgh.2011.06.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Barnes EL, Beery RM, Schulman AR, McCarthy EP, Korzenik JR, Winter RW. Hospitalizations for acute myocardial infarction are decreased among patients with inflammatory bowel disease using a nationwide inpatient database. Inflamm Bowel Dis 22: 2229–2237, 2016. doi: 10.1097/MIB.0000000000000899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Nguyen GC, Sam J. Rising prevalence of venous thromboembolism and its impact on mortality among hospitalized inflammatory bowel disease patients. Am J Gastroenterol 103: 2272–2280, 2008. doi: 10.1111/j.1572-0241.2008.02052.x. [DOI] [PubMed] [Google Scholar]
- 49. Stadnicki A, Stadnicka I. Venous and arterial thromboembolism in patients with inflammatory bowel diseases. World J Gastroenterol 27: 6757–6774, 2021. doi: 10.3748/wjg.v27.i40.6757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Grainge MJ, West J, Card TR. Venous thromboembolism during active disease and remission in inflammatory bowel disease: a cohort study. Lancet 375: 657–663, 2010. doi: 10.1016/S0140-6736(09)61963-2. [DOI] [PubMed] [Google Scholar]
- 51. Faye AS, Lee KE, Dodson J, Chodosh J, Hudesman D, Remzi F, Wright JD, Friedman AM, Shaukat A, Wen T. Increasing rates of venous thromboembolism among hospitalised patients with inflammatory bowel disease: a nationwide analysis. Aliment Pharmacol Ther 56: 1157–1167, 2022. doi: 10.1111/apt.17162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Ando K, Fujiya M, Watanabe K, Hiraoka S, Shiga H, Tanaka S, Iijima H, Mizushima T, Kobayashi T, Nagahori M, Ikeuchi H, Kato S, Torisu T, Kobayashi K, Higashiyama M, Fukui T, Kagaya T, Esaki M, Yanai S, Abukawa D, Naganuma M, Motoya S, Saruta M, Bamba S, Sasaki M, Uchiyama K, Fukuda K, Suzuki H, Nakase H, Shimizu T. A nationwide survey concerning the mortality and risk of progressing severity due to arterial and venous thromboembolism in inflammatory bowel disease in Japan. J Gastroenterol 56: 1062–1079, 2021. doi: 10.1007/s00535-021-01829-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Weng M-T, Park SH, Matsuoka K, Tung C-C, Lee JY, Chang C-H, Yang S-K, Watanabe M, Wong J-M, Wei S-C. Incidence and risk factor analysis of thromboembolic events in East Asian patients with inflammatory bowel disease, a multinational collaborative study. Inflamm Bowel Dis 24: 1791–1800, 2018. doi: 10.1093/ibd/izy058. [DOI] [PubMed] [Google Scholar]
- 54. Liu J, Gao X, Chen Y, Mei Q, Zhu L, Qian J, Hu P, Cao Q. Incidence and risk factors for venous thrombosis among patients with inflammatory bowel disease in China: a multicenter retrospective study. Intest Res 19: 313–322, 2021. doi: 10.5217/ir.2020.00017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Lee LH, Gallus A, Jindal R, Wang C, Wu CC. Incidence of venous thromboembolism in Asian populations: a systematic review. Thromb Haemost 117: 2243–2260, 2017. doi: 10.1160/TH17-02-0134. [DOI] [PubMed] [Google Scholar]
- 56. Kohoutova D, Moravkova P, Kruzliak P, Bures J. Thromboembolic complications in inflammatory bowel disease. J Thromb Thrombolysis 39: 489–498, 2015. doi: 10.1007/s11239-014-1129-7. [DOI] [PubMed] [Google Scholar]
- 57. Gala D, Newsome T, Roberson N, Lee SM, Thekkanal M, Shah M, Kumar V, Bandaru P, Gayam V. Thromboembolic events in patients with inflammatory bowel disease: a comprehensive overview. Diseases (Basel, Switzerland). 10: 73, 2022. doi: 10.3390/diseases10040073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Ha C, Magowan S, Accortt NA, Chen J, Stone CD. Risk of arterial thrombotic events in inflammatory bowel disease. Am J Gastroenterol 104: 1445–1451, 2009. doi: 10.1038/ajg.2009.81. [DOI] [PubMed] [Google Scholar]
- 59. Lin TY, Chen YG, Lin CL, Huang WS, Kao CH. Inflammatory bowel disease increases the risk of peripheral arterial disease: a nationwide cohort study. Medicine (Baltimore) 94: e2381, 2015. doi: 10.1097/MD.0000000000002381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Chang JT. Pathophysiology of inflammatory bowel diseases. N Engl J Med 383: 2652–2664, 2020. doi: 10.1056/NEJMra2002697. [DOI] [PubMed] [Google Scholar]
- 61. Singh N, Bernstein CN. Environmental risk factors for inflammatory bowel disease. United European Gastroenterol J 10: 1047–1053, 2022. doi: 10.1002/ueg2.12319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Strober W, Fuss IJ. Proinflammatory cytokines in the pathogenesis of inflammatory bowel diseases. Gastroenterology 140: 1756–1767, 2011. doi: 10.1053/j.gastro.2011.02.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Yokoyama T, Vaca L, Rossen RD, Durante W, Hazarika P, Mann DL. Cellular basis for the negative inotropic effects of tumor necrosis factor-α in the adult mammalian heart. J Clin Invest 92: 2303–2312, 1993. doi: 10.1172/JCI116834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Haudek SB, Taffet GE, Schneider MD, Mann DL. TNF provokes cardiomyocyte apoptosis and cardiac remodeling through activation of multiple cell death pathways. J Clin Invest 117: 2692–2701, 2007. doi: 10.1172/JCI29134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Sanders DB, Larson DF, Hunter K, Gorman M, Yang B. Comparison of tumor necrosis factor-α effect on the expression of iNOS in macrophage and cardiac myocytes. Perfusion 16: 67–74, 2001. doi: 10.1177/026765910101600110. [DOI] [PubMed] [Google Scholar]
- 66. Sivasubramanian N, Coker ML, Kurrelmeyer KM, MacLellan WR, DeMayo FJ, Spinale FG, Mann DL. Left ventricular remodeling in transgenic mice with cardiac restricted overexpression of tumor necrosis factor. Circulation 104: 826–831, 2001. doi: 10.1161/hc3401.093154. [DOI] [PubMed] [Google Scholar]
- 67. Li YY, Feng YQ, Kadokami T, McTiernan CF, Draviam R, Watkins SC, Feldman AM. Myocardial extracellular matrix remodeling in transgenic mice overexpressing tumor necrosis factor α can be modulated by anti-tumor necrosis factor α therapy. Proc Natl Acad Sci USA 97: 12746–12751, 2000. doi: 10.1073/pnas.97.23.12746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Mark KS, Trickler WJ, Miller DW. Tumor necrosis factor-α induces cyclooxygenase-2 expression and prostaglandin release in brain microvessel endothelial cells. J Pharmacol Exp Ther 297: 1051–1058, 2001. [PubMed] [Google Scholar]
- 69. Mattila P, Majuri ML, Mattila PS, Renkonen R. TNF α-induced expression of endothelial adhesion molecules, ICAM-1 and VCAM-1, is linked to protein kinase C activation. Scand J Immunol 36: 159–165, 1992. doi: 10.1111/j.1365-3083.1992.tb03087.x. [DOI] [PubMed] [Google Scholar]
- 70. Zanoli L, Ozturk K, Cappello M, Inserra G, Geraci G, Tuttolomondo A, Torres D, Pinto A, Duminuco A, Riguccio G, Aykan MB, Mulé G, Cottone S, Perna AF, Laurent S, Fatuzzo P, Castellino P, Boutouyrie P. Inflammation and aortic pulse wave velocity: a multicenter longitudinal study in patients with inflammatory bowel disease. J Am Heart Assoc 8: e010942, 2019. doi: 10.1161/JAHA.118.010942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Cacciapaglia F, Navarini L, Menna P, Salvatorelli E, Minotti G, Afeltra A. Cardiovascular safety of anti-TNF-α therapies: facts and unsettled issues. Autoimmun Rev 10: 631–635, 2011. doi: 10.1016/j.autrev.2011.04.014. [DOI] [PubMed] [Google Scholar]
- 72. Bujak M, Frangogiannis NG. The role of IL-1 in the pathogenesis of heart disease. Arch Immunol Ther Exp (Warsz) 57: 165–176, 2009. doi: 10.1007/s00005-009-0024-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Xie Z, Singh M, Singh K. Differential regulation of matrix metalloproteinase-2 and -9 expression and activity in adult rat cardiac fibroblasts in response to interleukin-1β. J Biol Chem 279: 39513–39519, 2004. doi: 10.1074/jbc.M405844200. [DOI] [PubMed] [Google Scholar]
- 74. Zitta K, Brandt B, Wuensch A, Meybohm P, Bein B, Steinfath M, Scholz J, Albrecht M. Interleukin-1β regulates cell proliferation and activity of extracellular matrix remodelling enzymes in cultured primary pig heart cells. Biochem Biophys Res Commun 399: 542–547, 2010. doi: 10.1016/j.bbrc.2010.07.106. [DOI] [PubMed] [Google Scholar]
- 75. Graff GR, Gozal D. Cardiorespiratory responses to interleukin-1β in adult rats: role of nitric oxide, eicosanoids and glucocorticoids. Arch Physiol Biochem 107: 97–112, 1999. doi: 10.1076/apab.107.2.97.4344. [DOI] [PubMed] [Google Scholar]
- 76. Bujak M, Dobaczewski M, Chatila K, Mendoza LH, Li N, Reddy A, Frangogiannis NG. Interleukin-1 receptor type I signaling critically regulates infarct healing and cardiac remodeling. Am J Pathol 173: 57–67, 2008. doi: 10.2353/ajpath.2008.070974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Van Tassell BW, Varma A, Salloum FN, Das A, Seropian IM, Toldo S, Smithson L, Hoke NN, Chau VQ, Robati R, Abbate A. Interleukin-1 trap attenuates cardiac remodeling after experimental acute myocardial infarction in mice. J Cardiovasc Pharmacol 55: 117–122, 2010. doi: 10.1097/FJC.0b013e3181c87e53. [DOI] [PubMed] [Google Scholar]
- 78. Honsho S, Nishikawa S, Amano K, Zen K, Adachi Y, Kishita E, Matsui A, Katsume A, Yamaguchi S, Nishikawa K, Isoda K, Riches DWH, Matoba S, Okigaki M, Matsubara H. Pressure-mediated hypertrophy and mechanical stretch induces IL-1 release and subsequent IGF-1 generation to maintain compensative hypertrophy by affecting Akt and JNK pathways. Circ Res 105: 1149–1158, 2009. doi: 10.1161/CIRCRESAHA.109.208199. [DOI] [PubMed] [Google Scholar]
- 79. Schreiber S, Aden K, Bernardes JP, Conrad C, Tran F, Höper H, Volk V, Mishra N, Blase JI, Nikolaus S, Bethge J, Kühbacher T, Röcken C, Chen M, Cottingham I, Petri N, Rasmussen BB, Lokau J, Lenk L, Garbers C, Feuerhake F, Rose-John S, Waetzig GH, Rosenstiel P. Therapeutic interleukin-6 trans-signaling inhibition by olamkicept (sgp130Fc) in patients with active inflammatory bowel disease. Gastroenterology 160: 2354–2366.e11, 2021. doi: 10.1053/j.gastro.2021.02.062. [DOI] [PubMed] [Google Scholar]
- 80. Spalinger MR, Sayoc-Becerra A, Ordookhanian C, Canale V, Santos AN, King SJ, Krishnan M, Nair MG, Scharl M, McCole DF. The JAK inhibitor tofacitinib rescues intestinal barrier defects caused by disrupted epithelial-macrophage interactions. J Crohns Colitis 15: 471–484, 2021. doi: 10.1093/ecco-jcc/jjaa182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Boengler K, Hilfiker-Kleiner D, Drexler H, Heusch G, Schulz R. The myocardial JAK/STAT pathway: from protection to failure. Pharmacol Ther 120: 172–185, 2008. doi: 10.1016/j.pharmthera.2008.08.002. [DOI] [PubMed] [Google Scholar]
- 82. Kurdi M, Zgheib C, Booz GW. Recent developments on the crosstalk between STAT3 and inflammation in heart function and disease. Front Immunol 9: 3029, 2018. doi: 10.3389/fimmu.2018.03029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Haghikia A, Stapel B, Hoch M, Hilfiker-Kleiner D. STAT3 and cardiac remodeling. Heart Fail Rev 16: 35–47, 2011. doi: 10.1007/s10741-010-9170-x. [DOI] [PubMed] [Google Scholar]
- 84. Fischer P, Hilfiker-Kleiner D. Role of gp130-mediated signalling pathways in the heart and its impact on potential therapeutic aspects. Br J Pharmacol 153 Suppl 1: S414–S427, 2008. doi: 10.1038/bjp.2008.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Fahmi A, Smart N, Punn A, Jabr R, Marber M, Heads R. p42/p44-MAPK and PI3K are sufficient for IL-6 family cytokines/gp130 to signal to hypertrophy and survival in cardiomyocytes in the absence of JAK/STAT activation. Cell Signal 25: 898–909, 2013. doi: 10.1016/j.cellsig.2012.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Wang L, Walia B, Evans J, Gewirtz AT, Merlin D, Sitaraman SV. IL-6 induces NF-κB activation in the intestinal epithelia. J Immunol (Baltimore, MD: 1950) 171: 3194–3201, 2003. doi: 10.4049/jimmunol.171.6.3194. [DOI] [PubMed] [Google Scholar]
- 87. Craig R, Larkin A, Mingo AM, Thuerauf DJ, Andrews C, McDonough PM, Glembotski CC. p38 MAPK and NF-κB collaborate to induce interleukin-6 gene expression and release. Evidence for a cytoprotective autocrine signaling pathway in a cardiac myocyte model system. J Biol Chem 275: 23814–23824, 2000. doi: 10.1074/jbc.M909695199. [DOI] [PubMed] [Google Scholar]
- 88. Matsui H, Ihara Y, Fujio Y, Kunisada K, Akira S, Kishimoto T, Yamauchi-Takihara K. Induction of interleukin (IL)-6 by hypoxia is mediated by nuclear factor (NF)-κB and NF-IL6 in cardiac myocytes. Cardiovasc Res 42: 104–112, 1999. doi: 10.1016/s0008-6363(98)00285-5. [DOI] [PubMed] [Google Scholar]
- 89. Schroecksnadel K, Frick B, Winkler C, Fuchs D. Crucial role of interferon-γ and stimulated macrophages in cardiovascular disease. Curr Vasc Pharmacol 4: 205–213, 2006. doi: 10.2174/157016106777698379. [DOI] [PubMed] [Google Scholar]
- 90. Langer V, Vivi E, Regensburger D, Winkler TH, Waldner MJ, Rath T, Schmid B, Skottke L, Lee S, Jeon NL, Wohlfahrt T, Kramer V, Tripal P, Schumann M, Kersting S, Handtrack C, Geppert CI, Suchowski K, Adams RH, Becker C, Ramming A, Naschberger E, Britzen-Laurent N, Stürzl M. IFN-γ drives inflammatory bowel disease pathogenesis through VE-cadherin-directed vascular barrier disruption. J Clin Invest 129: 4691–4707, 2019. doi: 10.1172/JCI124884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Darnell JE Jr, Kerr IM, Stark GR. Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science 264: 1415–1421, 1994. doi: 10.1126/science.8197455. [DOI] [PubMed] [Google Scholar]
- 92. Mitrokhin MV, Kalsin V, Kamkina O, Babkina I, Zotov A, Troitskiy VA, Mladenov MI, Kamkin GA. Participation of PKG and PKA-related pathways in the IFN-γ induced modulation of the BK(Ca) channel activity in human cardiac fibroblasts. J Pharmacol Sci 141: 25–31, 2019. doi: 10.1016/j.jphs.2019.08.006. [DOI] [PubMed] [Google Scholar]
- 93. Khajah M, Millen B, Cara DC, Waterhouse C, McCafferty DM. Granulocyte-macrophage colony-stimulating factor (GM-CSF): a chemoattractive agent for murine leukocytes in vivo. J Leukoc Biol 89: 945–953, 2011. doi: 10.1189/jlb.0809546. [DOI] [PubMed] [Google Scholar]
- 94. Jaffe BD, Sabath DE, Johnson GD, Moscinski LC, Johnson KR, Rovera G, Nauseef WM, Prystowsky MB. Myeloperoxidase and oncogene expression in GM-CSF induced bone marrow differentiation. Oncogene 2: 167–174, 1988. [PubMed] [Google Scholar]
- 95. Gathungu G, Kim M-O, Ferguson JP, Sharma Y, Zhang W, Ng SME, Bonkowski E, Ning K, Simms LA, Croft AR, Stempak JM, Walker N, Huang N, Xiao Y, Silverberg MS, Trapnell B, Cho JH, Radford-Smith GL, Denson LA. Granulocyte-macrophage colony-stimulating factor autoantibodies: a marker of aggressive Crohn’s disease. Inflamm Bowel Dis 19: 1671–1680, 2013. doi: 10.1097/MIB.0b013e318281f506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Goldstein JI, Kominsky DJ, Jacobson N, Bowers B, Regalia K, Austin GL, Yousefi M, Falta MT, Fontenot AP, Gerich ME, Golden-Mason L, Colgan SP. Defective leukocyte GM-CSF receptor (CD116) expression and function in inflammatory bowel disease. Gastroenterology 141: 208–216, 2011. doi: 10.1053/j.gastro.2011.03.060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Blyszczuk P, Behnke S, Lüscher TF, Eriksson U, Kania G. GM-CSF promotes inflammatory dendritic cell formation but does not contribute to disease progression in experimental autoimmune myocarditis. Biochim Biophys Acta 1833: 934–944, 2013. doi: 10.1016/j.bbamcr.2012.10.008. [DOI] [PubMed] [Google Scholar]
- 98. Maekawa Y, Anzai T, Yoshikawa T, Sugano Y, Mahara K, Kohno T, Takahashi T, Ogawa S. Effect of granulocyte-macrophage colony-stimulating factor inducer on left ventricular remodeling after acute myocardial infarction. J Am Coll Cardiol 44: 1510–1520, 2004. doi: 10.1016/j.jacc.2004.05.083. [DOI] [PubMed] [Google Scholar]
- 99. Anzai A, Choi JL, He S, Fenn AM, Nairz M, Rattik S, McAlpine CS, Mindur JE, Chan CT, Iwamoto Y, Tricot B, Wojtkiewicz GR, Weissleder R, Libby P, Nahrendorf M, Stone JR, Becher B, Swirski FK. The infarcted myocardium solicits GM-CSF for the detrimental oversupply of inflammatory leukocytes. J Exp Med 214: 3293–3310, 2017. doi: 10.1084/jem.20170689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Devani M, Cugno M, Vecchi M, Ferrero S, Di Berardino F, Avesani EC, de Franchis R, Colman RW. Kallikrein-kinin system activation in Crohn's disease: differences in intestinal and systemic markers. Am J Gastroenterol 97: 2026–2032, 2002. doi: 10.1111/j.1572-0241.2002.05919.x. [DOI] [PubMed] [Google Scholar]
- 101. Devani M, Vecchi M, Ferrero S, Avesani EC, Arizzi C, Chao L, Colman RW, Cugno M. Kallikrein-kinin system in inflammatory bowel diseases: intestinal involvement and correlation with the degree of tissue inflammation. Dig Liver Dis 37: 665–673, 2005. doi: 10.1016/j.dld.2005.01.021. [DOI] [PubMed] [Google Scholar]
- 102. Stadnicki A, Mazurek U, Gonciarz M, Plewka D, Nowaczyk G, Orchel J, Pastucha E, Plewka A, Wilczok T, Colman RW. Immunolocalization and expression of kallistatin and tissue kallikrein in human inflammatory bowel disease. Dig Dis Sci 48: 615–623, 2003. doi: 10.1023/a:1022569623350. [DOI] [PubMed] [Google Scholar]
- 103. Siddesha JM, Valente AJ, Sakamuri SSVP, Yoshida T, Gardner JD, Somanna N, Takahashi C, Noda M, Chandrasekar B. Angiotensin II stimulates cardiac fibroblast migration via the differential regulation of matrixins and RECK. J Mol Cell Cardiol 65: 9–18, 2013. doi: 10.1016/j.yjmcc.2013.09.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Gao X, He X, Luo B, Peng L, Lin J, Zuo Z. Angiotensin II increases collagen I expression via transforming growth factor-β1 and extracellular signal-regulated kinase in cardiac fibroblasts. Eur J Pharmacol 606: 115–120, 2009. doi: 10.1016/j.ejphar.2008.12.049. [DOI] [PubMed] [Google Scholar]
- 105. Schultz JEJ, Witt SA, Glascock BJ, Nieman ML, Reiser PJ, Nix SL, Kimball TR, Doetschman T. TGF-β1 mediates the hypertrophic cardiomyocyte growth induced by angiotensin II. J Clin Invest 109: 787–796, 2002. doi: 10.1172/JCI0214190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Ip C, Luk KS, Yuen VLC, Chiang L, Chan CK, Ho K, Gong M, Lee TTL, Leung KSK, Roever L, Bazoukis G, Lampropoulos K, Li KHC, Tse G, Liu T; International Health Informatics Study (IHIS) Network. Soluble suppression of tumorigenicity 2 (sST2) for predicting disease severity or mortality outcomes in cardiovascular diseases: a systematic review and meta-analysis. Int J Cardiol Heart Vasc 37: 100887, 2021. doi: 10.1016/j.ijcha.2021.100887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Sanada S, Hakuno D, Higgins LJ, Schreiter ER, McKenzie AN, Lee RT. IL-33 and ST2 comprise a critical biomechanically induced and cardioprotective signaling system. J Clin Invest 117: 1538–1549, 2007. doi: 10.1172/JCI30634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Beltrán CJ, Núñez LE, Díaz-Jiménez D, Farfan N, Candia E, Heine C, López F, González MJ, Quera R, Hermoso MA. Characterization of the novel ST2/IL-33 system in patients with inflammatory bowel disease. Inflamm Bowel Dis 16: 1097–1107, 2010. doi: 10.1002/ibd.21175. [DOI] [PubMed] [Google Scholar]
- 109. Díaz-Jiménez D, Núñez LE, Beltrán CJ, Candia E, Suazo C, Alvarez-Lobos M, González M-J, Hermoso MA, Quera R. Soluble ST2: a new and promising activity marker in ulcerative colitis. World J Gastroenterol 17: 2181–2190, 2011. doi: 10.3748/wjg.v17.i17.2181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Boga S, Alkim H, Koksal AR, Ozagari AA, Bayram M, Tekin Neijmann S, Sen I, Alkim C. Serum ST2 in inflammatory bowel disease: a potential biomarker for disease activity. J Investig Med 64: 1016–1024, 2016. doi: 10.1136/jim-2016-000062. [DOI] [PubMed] [Google Scholar]
- 111. Kumar S, Tzimas MN, Griswold DE, Young PR. Expression of ST2, an interleukin-1 receptor homologue, is induced by proinflammatory stimuli. Biochem Biophys Res Commun 235: 474–478, 1997. doi: 10.1006/bbrc.1997.6810. [DOI] [PubMed] [Google Scholar]
- 112. Kim H-L, Lee JP, Wong N, Lim W-H, Seo J-B, Zo J-H, Kim M-A, Kim S-H. Prognostic value of serum soluble ST2 in stable coronary artery disease: a prospective observational study. Sci Rep 11: 15203, 2021. doi: 10.1038/s41598-021-94714-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Willems S, Hoefer I, Pasterkamp G. The role of the Interleukin 1 receptor-like 1 (ST2) and Interleukin-33 pathway in cardiovascular disease and cardiovascular risk assessment. Minerva Med 103: 513–524, 2012. [PubMed] [Google Scholar]
- 114. Tang Y, Kline KT, Zhong XS, Xiao Y, Lian H, Peng J, Liu X, Powell DW, Tang G, Li Q. Chronic colitis upregulates microRNAs suppressing brain-derived neurotrophic factor in the adult heart. PLoS One 16: e0257280, 2021. doi: 10.1371/journal.pone.0257280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Wang H, Bei Y, Huang P, Zhou Q, Shi J, Sun Q, Zhong J, Li X, Kong X, Xiao J. Inhibition of miR-155 protects against LPS-induced cardiac dysfunction and apoptosis in mice. Mol Ther Nucleic Acids 5: e374, 2016. doi: 10.1038/mtna.2016.80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Lian H, Zhong XS, Xiao Y, Sun Z, Shen Y, Zhao K, Ma X, Li Y, Niu Q, Liu M, Powell DW, Liu C, Li Q. Exosomal miR-29b of gut origin in patients with ulcerative colitis suppresses heart brain-derived neurotrophic factor. Front Mol Biosci 9: 759689, 2022. doi: 10.3389/fmolb.2022.759689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Cui X, Ye L, Li J, Jin L, Wang W, Li S, Bao M, Wu S, Li L, Geng B, Zhou X, Zhang J, Cai J. Metagenomic and metabolomic analyses unveil dysbiosis of gut microbiota in chronic heart failure patients. Sci Rep 8: 635, 2018. doi: 10.1038/s41598-017-18756-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Chu Y, Jiang MZ, Xu B, Wang WJ, Chen D, Li XW, Zhang YJ, Liang J. Specific changes of enteric mycobiota and virome in inflammatory bowel disease. J Dig Dis 19: 2–7, 2018. doi: 10.1111/1751-2980.12570. [DOI] [PubMed] [Google Scholar]
- 119. Xiao Y, Zhong XS, Liu X, Li Q. Therapeutic evaluation of fecal microbiota transplantation in an interleukin 10-deficient mouse model. J Vis Exp 182: e63350, 2022. doi: 10.3791/63350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Efe TH, Cimen T, Ertem AG, Coskun Y, Bilgin M, Sahan HF, Pamukcu HE, Yayla C, Sunman H, Yuksel I, Yeter E. Atrial electromechanical properties in inflammatory bowel disease. Echocardiography 33: 1309–1316, 2016. doi: 10.1111/echo.13261. [DOI] [PubMed] [Google Scholar]
- 121. Nar G, Ergul B, Aksan G, Inci S. Assessment of atrial electromechanical delay and left atrial mechanical functions in patients with ulcerative colitis. Echocardiography 33: 970–976, 2016. doi: 10.1111/echo.13213. [DOI] [PubMed] [Google Scholar]
- 122. Mazlam MZ, Hodgson HJ. Interrelations between interleukin-6, interleukin-1 β, plasma C-reactive protein values, and in vitro C-reactive protein generation in patients with inflammatory bowel disease. Gut 35: 77–83, 1994. doi: 10.1136/gut.35.1.77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Psychari SN, Apostolou TS, Sinos L, Hamodraka E, Liakos G, Kremastinos DT. Relation of elevated C-reactive protein and interleukin-6 levels to left atrial size and duration of episodes in patients with atrial fibrillation. Am J Cardiol 95: 764–767, 2005. doi: 10.1016/j.amjcard.2004.11.032. [DOI] [PubMed] [Google Scholar]
- 124. Luckett LR, Gallucci RM. Interleukin-6 (IL-6) modulates migration and matrix metalloproteinase function in dermal fibroblasts from IL-6KO mice. Br J Dermatol 156: 1163–1171, 2007. doi: 10.1111/j.1365-2133.2007.07867.x. [DOI] [PubMed] [Google Scholar]
- 125. Dogan Y, Soylu A, Eren GA, Poturoglu S, Dolapcioglu C, Sonmez K, Duman H, Sevindir I. Evaluation of QT and P wave dispersion and mean platelet volume among inflammatory bowel disease patients. Int J Med Sci 8: 540–546, 2011. doi: 10.7150/ijms.8.540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Sharma P, Makharia GK, Ahuja V, Dwivedi SN, Deepak KK. Autonomic dysfunctions in patients with inflammatory bowel disease in clinical remission. Dig Dis Sci 54: 853–861, 2009. doi: 10.1007/s10620-008-0424-6. [DOI] [PubMed] [Google Scholar]
- 127. Knorr JP, Moshfeghi M, Sokoloski MC. Ciprofloxacin-induced Q-T interval prolongation. Am J Health Syst Pharm 65: 547–551, 2008. doi: 10.2146/ajhp070081. [DOI] [PubMed] [Google Scholar]
- 128. Riccioni G, Bucciarelli V, Di Ilio E, Scotti L, Aceto A, D'Orazio N, Martini F, Bucciarelli T. Recurrent atrial fibrillation in a patient with ulcerative colitis treated with azathioprine: case report and review of the literature. Int J Immunopathol Pharmacol 24: 247–249, 2011. doi: 10.1177/039463201102400131. [DOI] [PubMed] [Google Scholar]
- 129. Vaze A, Tran K-V, Tanriverdi K, Sardana M, Lessard D, Donahue JK, Barton B, Aurigemma G, Lubitz SA, Lin H, Nasr GH, Mandapati A, Benjamin EJ, Vasan RS, Freedman JE, McManus DD. Relations between plasma microRNAs, echocardiographic markers of atrial remodeling, and atrial fibrillation: data from the Framingham Offspring study. PLoS One 15: e0236960, 2020. doi: 10.1371/journal.pone.0236960. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Wu H, Wang F, Hu S, Yin C, Li X, Zhao S, Wang J, Yan X. MiR-20a and miR-106b negatively regulate autophagy induced by leucine deprivation via suppression of ULK1 expression in C2C12 myoblasts. Cell Signal 24: 2179–2186, 2012. doi: 10.1016/j.cellsig.2012.07.001. [DOI] [PubMed] [Google Scholar]
- 131. Lu C, Chen J, Xu H-G, Zhou X, He Q, Li Y-L, Jiang G, Shan Y, Xue B, Zhao R-X, Wang Y, Werle KD, Cui R, Liang J, Xu Z-X. MIR106B and MIR93 prevent removal of bacteria from epithelial cells by disrupting ATG16L1-mediated autophagy. Gastroenterology 146: 188–199, 2014. doi: 10.1053/j.gastro.2013.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Fasseu M, Tréton X, Guichard C, Pedruzzi E, Cazals-Hatem D, Richard C, Aparicio T, Daniel F, Soulé J-C, Moreau R, Bouhnik Y, Laburthe M, Groyer A, Ogier-Denis E. Identification of restricted subsets of mature microRNA abnormally expressed in inactive colonic mucosa of patients with inflammatory bowel disease. PLoS One 5: e13160, 2010. doi: 10.1371/journal.pone.0013160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Koutroumpakis E, Ramos-Rivers C, Regueiro M, Hashash JG, Barrie A, Swoger J, Baidoo L, Schwartz M, Dunn MA, Koutroubakis IE, Binion DG. Association between long-term lipid profiles and disease severity in a large cohort of patients with inflammatory bowel disease. Dig Dis Sci 61: 865–871, 2016. doi: 10.1007/s10620-015-3932-1. [DOI] [PubMed] [Google Scholar]
- 134. Scoville EA, Allaman MM, Brown CT, Motley AK, Horst SN, Williams CS, Koyama T, Zhao Z, Adams DW, Beaulieu DB, Schwartz DA, Wilson KT, Coburn LA. Alterations in lipid, amino acid, and energy metabolism distinguish Crohn's disease from ulcerative colitis and control subjects by serum metabolomic profiling. Metabolomics 14: 17, 2018. doi: 10.1007/s11306-017-1311-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Murgia A, Hinz C, Liggi S, Denes J, Hall Z, West J, Santoru ML, Piras C, Manis C, Usai P, Atzori L, Griffin JL, Caboni P. Italian cohort of patients affected by inflammatory bowel disease is characterised by variation in glycerophospholipid, free fatty acids and amino acid levels. Metabolomics 14: 140, 2018. doi: 10.1007/s11306-018-1439-4. [DOI] [PubMed] [Google Scholar]
- 136. Lai Y, Xue J, Liu C-W, Gao B, Chi L, Tu P, Lu K, Ru H. Serum metabolomics identifies altered bioenergetics, signaling cascades in parallel with exposome markers in Crohn's disease. Molecules 24: 449, 2019. doi: 10.3390/molecules24030449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Morrison A, Hokanson JE. The independent relationship between triglycerides and coronary heart disease. Vasc Health Risk Manag 5: 89–95, 2009. [PMC free article] [PubMed] [Google Scholar]
- 138. Ryu H, Kim J, Kim D, Lee JE, Chung Y. Cellular and molecular links between autoimmunity and lipid metabolism. Mol Cells 42: 747–754, 2019. doi: 10.14348/molcells.2019.0196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Cinoku II, Mavragani CP, Moutsopoulos HM. Atherosclerosis: beyond the lipid storage hypothesis. The role of autoimmunity. Eur J Clin Invest 50: e13195, 2020. doi: 10.1111/eci.13195. [DOI] [PubMed] [Google Scholar]
- 140. Roifman I, Sun YC, Fedwick JP, Panaccione R, Buret AG, Liu H, Rostom A, Anderson TJ, Beck PL. Evidence of endothelial dysfunction in patients with inflammatory bowel disease. Clin Gastroenterol Hepatol 7: 175–182, 2009. doi: 10.1016/j.cgh.2008.10.021. [DOI] [PubMed] [Google Scholar]
- 141. Horowitz S, Binion DG, Nelson VM, Kanaa Y, Javadi P, Lazarova Z, Andrekopoulos C, Kalyanaraman B, Otterson MF, Rafiee P. Increased arginase activity and endothelial dysfunction in human inflammatory bowel disease. Am J Physiol Gastrointest Liver Physiol 292: G1323–G1336, 2007. doi: 10.1152/ajpgi.00499.2006. [DOI] [PubMed] [Google Scholar]
- 142. McEniery CM, Qasem A, Schmitt M, Avolio AP, Cockcroft JR, Wilkinson IB. Endothelin-1 regulates arterial pulse wave velocity in vivo. J Am Coll Cardiol 42: 1975–1981, 2003. doi: 10.1016/j.jacc.2003.06.016. [DOI] [PubMed] [Google Scholar]
- 143. Wilkinson IB, Qasem A, McEniery CM, Webb DJ, Avolio AP, Cockcroft JR. Nitric oxide regulates local arterial distensibility in vivo. Circulation 105: 213–217, 2002. doi: 10.1161/hc0202.101970. [DOI] [PubMed] [Google Scholar]
- 144. Dudzińska E, Gryzinska M, Ognik K, Gil-Kulik P, Kocki J. Oxidative stress and effect of treatment on the oxidation product decomposition processes in IBD. Oxid Med Cell Longev 2018: 7918261, 2018. doi: 10.1155/2018/7918261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Dutta S, Rittinger K. Regulation of NOXO1 activity through reversible interactions with p22 and NOXA1. PLoS One 5: e10478, 2010. doi: 10.1371/journal.pone.0010478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Piechota-Polanczyk A, Fichna J. Review article: the role of oxidative stress in pathogenesis and treatment of inflammatory bowel diseases. Naunyn Schmiedebergs Arch Pharmacol 387: 605–620, 2014. doi: 10.1007/s00210-014-0985-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Lacolley P, Regnault V, Laurent S. Mechanisms of arterial stiffening: from mechanotransduction to epigenetics. Arterioscler Thromb Vasc Biol 40: 1055–1062, 2020. doi: 10.1161/ATVBAHA.119.313129. [DOI] [PubMed] [Google Scholar]
- 148. Rachmilewitz D, Eliakim R, Ackerman Z, Karmeli F. Direct determination of colonic nitric oxide level—a sensitive marker of disease activity in ulcerative colitis. Am J Gastroenterol 93: 409–412, 1998. doi: 10.1111/j.1572-0241.1998.00409.x. [DOI] [PubMed] [Google Scholar]
- 149. Cross RK, Wilson KT. Nitric oxide in inflammatory bowel disease. Inflamm Bowel Dis 9: 179–189, 2003. doi: 10.1097/00054725-200305000-00006. [DOI] [PubMed] [Google Scholar]
- 150. Soufli I, Toumi R, Rafa H, Touil-Boukoffa C. Overview of cytokines and nitric oxide involvement in immuno-pathogenesis of inflammatory bowel diseases. World J Gastrointest Pharmacol Ther 7: 353–360, 2016. doi: 10.4292/wjgpt.v7.i3.353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Gunnett CA, Lund DD, McDowell AK, Faraci FM, Heistad DD. Mechanisms of inducible nitric oxide synthase-mediated vascular dysfunction. Arterioscler Thromb Vasc Biol 25: 1617–1622, 2005. doi: 10.1161/01.ATV.0000172626.00296.ba. [DOI] [PubMed] [Google Scholar]
- 152. Nagareddy PR, Xia Z, McNeill JH, MacLeod KM. Increased expression of iNOS is associated with endothelial dysfunction and impaired pressor responsiveness in streptozotocin-induced diabetes. Am J Physiol Heart Circ Physiol 289: H2144–H2152, 2005. doi: 10.1152/ajpheart.00591.2005. [DOI] [PubMed] [Google Scholar]
- 153. Howell KW, Meng X, Fullerton DA, Jin C, Reece TB, Cleveland JC Jr.. Toll-like receptor 4 mediates oxidized LDL-induced macrophage differentiation to foam cells. J Surg Res 171: e27–e31, 2011. doi: 10.1016/j.jss.2011.06.033. [DOI] [PubMed] [Google Scholar]
- 154. Maziere C, Conte MA, Dantin F, Maziere J-C. Lipopolysaccharide enhances oxidative modification of low density lipoprotein by copper ions, endothelial and smooth muscle cells. Atherosclerosis 143: 75–80, 1999. doi: 10.1016/s0021-9150(98)00277-9. [DOI] [PubMed] [Google Scholar]
- 155. Li M, Qian M, Kyler K, Xu J. Endothelial-vascular smooth muscle cells interactions in atherosclerosis. Front Cardiovasc Med 5: 151, 2018. doi: 10.3389/fcvm.2018.00151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Shamseya AM, Hussein WM, Elnely DA, Adel F, Header DA. Serum matrix metalloproteinase-9 concentration as a marker of disease activity in patients with inflammatory bowel disease. Eur J Gastroenterol Hepatol 33: e803–e809, 2021. doi: 10.1097/MEG.0000000000002264. [DOI] [PubMed] [Google Scholar]
- 157. Fonseca-Camarillo G, Furuzawa-Carballeda J, Martínez-Benitez B, Barreto-Zuñiga R, Yamamoto-Furusho JK. Increased expression of extracellular matrix metalloproteinase inducer (EMMPRIN) and MMP10, MMP23 in inflammatory bowel disease: cross-sectional study. Scand J Immunol 93: e12962, 2021. doi: 10.1111/sji.12962. [DOI] [PubMed] [Google Scholar]
- 158. Mäkitalo L, Kolho KL, Karikoski R, Anthoni H, Saarialho-Kere U. Expression profiles of matrix metalloproteinases and their inhibitors in colonic inflammation related to pediatric inflammatory bowel disease. Scand J Gastroenterol 45: 862–871, 2010. doi: 10.3109/00365520903583863. [DOI] [PubMed] [Google Scholar]
- 159. Faubion WA, Fletcher JG, O'Byrne S, Feagan BG, de Villiers WJ, Salzberg B, Plevy S, Proctor DD, Valentine JF, Higgins PD, Harris JM, Diehl L, Wright L, Tew GW, Luca D, Basu K, Keir ME. EMerging BiomARKers in Inflammatory Bowel Disease (EMBARK) study identifies fecal calprotectin, serum MMP9, and serum IL-22 as a novel combination of biomarkers for Crohn's disease activity: role of cross-sectional imaging. Am J Gastroenterol 108: 1891–1900, 2013. [Erratum in Am J Gastroenterol 109: 780, 2014] doi: 10.1038/ajg.2013.354. [DOI] [PubMed] [Google Scholar]
- 160. Yablecovitch D, Kopylov U, Lahat A, Amitai MM, Klang E, Ben-Ami Shor D, Neuman S, Levhar N, Fudim E, Avidan B, Laish I, Selinger L, Zingboim-Orbach N, Picard O, Yavzori M, Eliakim R, Ben-Horin S. Serum MMP-9: a novel biomarker for prediction of clinical relapse in patients with quiescent Crohn’s disease, a post hoc analysis. Therap Adv Gastroenterol 12: 1756284819881590, 2019. doi: 10.1177/1756284819881590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Zieman SJ, Melenovsky V, Kass DA. Mechanisms, pathophysiology, and therapy of arterial stiffness. Arterioscler Thromb Vasc Biol 25: 932–943, 2005. doi: 10.1161/01.ATV.0000160548.78317.29. [DOI] [PubMed] [Google Scholar]
- 162. Haas TL. Endothelial cell regulation of matrix metalloproteinases. Can J Physiol Pharmacol 83: 1–7, 2005. doi: 10.1139/y04-120. [DOI] [PubMed] [Google Scholar]
- 163. Xiao Y, Lian H, Zhong XS, Krishnachaitanya SS, Cong Y, Dashwood RH, Savidge TC, Powell DW, Liu X, Li Q. Matrix metalloproteinase 7 contributes to intestinal barrier dysfunction by degrading tight junction protein Claudin-7. Front Immunol 13: 1020902, 2022. doi: 10.3389/fimmu.2022.1020902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Mantaka A, Galanakis N, Tsetis D, Koutroubakis IE. Abdominal aortic calcification in patients with inflammatory bowel disease: does anti-tumor necrosis factor α use protect from chronic inflammation-induced atherosclerosis? Intest Res 20: 495–505, 2022. doi: 10.5217/ir.2022.00017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Giachelli CM, Jono S, Shioi A, Nishizawa Y, Mori K, Morii H. Vascular calcification and inorganic phosphate. Am J Kidney Dis 38: S34–S37, 2001. doi: 10.1053/ajkd.2001.27394. [DOI] [PubMed] [Google Scholar]
- 166. Floege J, Ketteler M. Vascular calcification in patients with end-stage renal disease. Nephrol Dial Transplant 19 Suppl 5: V59–V66, 2004. doi: 10.1093/ndt/gfh1058. [DOI] [PubMed] [Google Scholar]
- 167. Travers JG, Kamal FA, Robbins J, Yutzey KE, Blaxall BC. Cardiac fibrosis: the fibroblast awakens. Circ Res 118: 1021–1040, 2016. doi: 10.1161/CIRCRESAHA.115.306565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Tintut Y, Patel J, Territo M, Saini T, Parhami F, Demer LL. Monocyte/macrophage regulation of vascular calcification in vitro. Circulation 105: 650–655, 2002. doi: 10.1161/hc0502.102969. [DOI] [PubMed] [Google Scholar]
- 169. Shioi A, Katagi M, Okuno Y, Mori K, Jono S, Koyama H, Nishizawa Y. Induction of bone-type alkaline phosphatase in human vascular smooth muscle cells: roles of tumor necrosis factor-α and oncostatin M derived from macrophages. Circ Res 91: 9–16, 2002. doi: 10.1161/01.res.0000026421.61398.f2. [DOI] [PubMed] [Google Scholar]
- 170. Parhami F, Basseri B, Hwang J, Tintut Y, Demer LL. High-density lipoprotein regulates calcification of vascular cells. Circ Res 91: 570–576, 2002. doi: 10.1161/01.res.0000036607.05037.da. [DOI] [PubMed] [Google Scholar]
- 171. Danese S, Sans M, Fiocchi C. Inflammatory bowel disease: the role of environmental factors. Autoimmun Rev 3: 394–400, 2004. doi: 10.1016/j.autrev.2004.03.002. [DOI] [PubMed] [Google Scholar]
- 172. Giannotta M, Tapete G, Emmi G, Silvestri E, Milla M. Thrombosis in inflammatory bowel diseases: what's the link? Thromb J 13: 14, 2015. doi: 10.1186/s12959-015-0044-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Papa A, Gerardi V, Marzo M, Felice C, Rapaccini GL, Gasbarrini A. Venous thromboembolism in patients with inflammatory bowel disease: focus on prevention and treatment. World J Gastroenterol 20: 3173–3179, 2014. doi: 10.3748/wjg.v20.i12.3173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Alkim H, Ayaz S, Alkim C, Ulker A, Sahin B. Continuous active state of coagulation system in patients with nonthrombotic inflammatory bowel disease. Clin Appl Thromb Hemost 17: 600–604, 2011. doi: 10.1177/1076029611405034. [DOI] [PubMed] [Google Scholar]
- 175. Collins CE, Cahill MR, Newland AC, Rampton DS. Platelets circulate in an activated state in inflammatory bowel disease. Gastroenterology 106: 840–845, 1994. doi: 10.1016/0016-5085(94)90741-2. [DOI] [PubMed] [Google Scholar]
- 176. Zezos P, Kouklakis G, Saibil F. Inflammatory bowel disease and thromboembolism. World J Gastroenterol 20: 13863–13878, 2014. doi: 10.3748/wjg.v20.i38.13863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Senchenkova E, Seifert H, Granger DN. Hypercoagulability and platelet abnormalities in inflammatory bowel disease. Semin Thromb Hemost 41: 582–589, 2015. doi: 10.1055/s-0035-1556590. [DOI] [PubMed] [Google Scholar]
- 178. Selhub J. Homocysteine metabolism. Annu Rev Nutr 19: 217–246, 1999. doi: 10.1146/annurev.nutr.19.1.217. [DOI] [PubMed] [Google Scholar]
- 179. Danese S, Papa A, Saibeni S, Repici A, Malesci A, Vecchi M. Inflammation and coagulation in inflammatory bowel disease: the clot thickens. Am J Gastroenterol 102: 174–186, 2007. doi: 10.1111/j.1572-0241.2006.00943.x. [DOI] [PubMed] [Google Scholar]
- 180. Oussalah A, Guéant JL, Peyrin-Biroulet L. Meta-analysis: hyperhomocysteinaemia in inflammatory bowel diseases. Aliment Pharmacol Ther 34: 1173–1184, 2011. doi: 10.1111/j.1365-2036.2011.04864.x. [DOI] [PubMed] [Google Scholar]
- 181. Ferro JM, Oliveira Santos M. Neurology of inflammatory bowel disease. J Neurol Sci 424: 117426, 2021. doi: 10.1016/j.jns.2021.117426. [DOI] [PubMed] [Google Scholar]
- 182. Lindgren S, Stewenius J, Sjölund K, Lilja B, Sundkvist G. Autonomic vagal nerve dysfunction in patients with ulcerative colitis. Scand J Gastroenterol 28: 638–642, 1993. doi: 10.3109/00365529309096103. [DOI] [PubMed] [Google Scholar]
- 183. Coruzzi P, Castiglioni P, Parati G, Brambilla V, Brambilla L, Gualerzi M, Cademartiri F, Franzè A, De Angelis G, Di Rienzo M, Di Mario F. Autonomic cardiovascular regulation in quiescent ulcerative colitis and Crohn’s disease. Eur J Clin Invest 37: 964–970, 2007. doi: 10.1111/j.1365-2362.2007.01887.x. [DOI] [PubMed] [Google Scholar]
- 184. Lindgren S, Lilja B, Rosén I, Sundkvist G. Disturbed autonomic nerve function in patients with Crohn’s disease. Scand J Gastroenterol 26: 361–366, 1991. doi: 10.3109/00365529108996495. [DOI] [PubMed] [Google Scholar]
- 185. Yao ST, McKinley MJ, May CN. Circumventing a broken heart: cytokines and the subfornical organ. Am J Physiol Heart Circ Physiol 313: H729–H731, 2017. doi: 10.1152/ajpheart.00480.2017. [DOI] [PubMed] [Google Scholar]
- 186. Saw EL, Kakinuma Y, Fronius M, Katare R. The non-neuronal cholinergic system in the heart: a comprehensive review. J Mol Cell Cardiol 125: 129–139, 2018. doi: 10.1016/j.yjmcc.2018.10.013. [DOI] [PubMed] [Google Scholar]
- 187. Kučera M, Hrabovská A. Cholinergic system of the heart. Ceska Slov Farm 64: 254–263, 2015. [PubMed] [Google Scholar]
- 188. Shao X, Yang L, Hu K, Shen R, Ye Q, Yuan X, Zhao Q, Shen J. Serum cholinesterases, a novel marker of clinical activity in inflammatory bowel disease: a retrospective case-control study. Mediators Inflamm 2020: 4694090, 2020. doi: 10.1155/2020/4694090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Dayer M, MacIver DH, Rosen SD. The central nervous system and heart failure. Future Cardiol 17: 363–381, 2021. doi: 10.2217/fca-2020-0059. [DOI] [PubMed] [Google Scholar]
- 190. Serafini MA, Paz AH, Nunes NS. Cholinergic immunomodulation in inflammatory bowel diseases. Brain Behav Immun Health 19: 100401, 2022. doi: 10.1016/j.bbih.2021.100401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191. Harper JW, Welch MP, Sinanan MN, Wahbeh GT, Lee SD. Co-morbid diabetes in patients with Crohn’s disease predicts a greater need for surgical intervention. Aliment Pharmacol Ther 35: 126–132, 2012. doi: 10.1111/j.1365-2036.2011.04915.x. [DOI] [PubMed] [Google Scholar]
- 192. Coakley BA, Divino CM. Identifying factors predictive of surgical-site infections after colectomy for fulminant ulcerative colitis. Am Surg 78: 481–484, 2012. doi: 10.1177/000313481207800442. [DOI] [PubMed] [Google Scholar]
- 193. Argollo M, Gilardi D, Peyrin-Biroulet C, Chabot JF, Peyrin-Biroulet L, Danese S. Comorbidities in inflammatory bowel disease: a call for action. Lancet Gastroenterol Hepatol 4: 643–654, 2019. doi: 10.1016/S2468-1253(19)30173-6. [DOI] [PubMed] [Google Scholar]
- 194. Olivera PA, Zuily S, Kotze PG, Regnault V, Al Awadhi S, Bossuyt P, Gearry RB, Ghosh S, Kobayashi T, Lacolley P, Louis E, Magro F, Ng SC, Papa A, Raine T, Teixeira FV, Rubin DT, Danese S, Peyrin-Biroulet L. International consensus on the prevention of venous and arterial thrombotic events in patients with inflammatory bowel disease. Nat Rev Gastroenterol Hepatol 18: 857–873, 2021. doi: 10.1038/s41575-021-00492-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Nguyen GC, Bernstein CN, Bitton A, Chan AK, Griffiths AM, Leontiadis GI, Geerts W, Bressler B, Butzner JD, Carrier M, Chande N, Marshall JK, Williams C, Kearon C. Consensus statements on the risk, prevention, and treatment of venous thromboembolism in inflammatory bowel disease: Canadian Association of Gastroenterology. Gastroenterology 146: 835–848.e6, 2014. doi: 10.1053/j.gastro.2014.01.042. [DOI] [PubMed] [Google Scholar]
- 196. Downton TD, Willcocks BJ, Boutlis CS. Acute myopericarditis in a patient recently started on mesalazine for Crohn disease. Intern Med J 49: 676–677, 2019. doi: 10.1111/imj.14273. [DOI] [PubMed] [Google Scholar]
- 197. Coward S, Kuenzig ME, Hazlewood G, Clement F, McBrien K, Holmes R, Panaccione R, Ghosh S, Seow CH, Rezaie A, Kaplan GG. Comparative effectiveness of mesalamine, sulfasalazine, corticosteroids, and budesonide for the induction of remission in Crohn’s disease: a bayesian network meta-analysis. Inflamm Bowel Dis 23: 461–472, 2017. [Erratum in Inflamm Bowel Dis 23: E25, 2017]. doi: 10.1097/MIB.0000000000001023. [DOI] [PubMed] [Google Scholar]
- 198. Tamura S, Ishida N, Miyazu T, Onoue S, Tani S, Yamade M, Hamaya Y, Iwaizumi M, Osawa S, Furuta T, Sugimoto K. Mesalazine granule formulation improves clinical data in Crohn's disease compared with tablet formulation. Sci Rep 10: 21353, 2020. doi: 10.1038/s41598-020-78603-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Asirvatham S, Sebastian C, Thadani U. Severe symptomatic sinus bradycardia associated with mesalamine use. Am J Gastroenterol 93: 470–471, 1998. [Erratum in Am J Gastroenterol 93: 1196, 1998]. doi: 10.1111/j.1572-0241.1998.468_2.x. [DOI] [PubMed] [Google Scholar]
- 200. Pujades-Rodriguez M, Morgan AW, Cubbon RM, Wu J. Dose-dependent oral glucocorticoid cardiovascular risks in people with immune-mediated inflammatory diseases: a population-based cohort study. PLoS Med 17: e1003432, 2020. doi: 10.1371/journal.pmed.1003432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Cassinotti A, Massari A, Ferrara E, Greco S, Bosani M, Ardizzone S, Bianchi Porro G. New onset of atrial fibrillation after introduction of azathioprine in ulcerative colitis: case report and review of the literature. Eur J Clin Pharmacol 63: 875–878, 2007. doi: 10.1007/s00228-007-0328-y. [DOI] [PubMed] [Google Scholar]
- 202. Sinagra E, Perricone G, Romano C, Cottone M. Heart failure and anti-tumor necrosis factor-α in systemic chronic inflammatory diseases. Eur J Intern Med 24: 385–392, 2013. doi: 10.1016/j.ejim.2012.12.015. [DOI] [PubMed] [Google Scholar]
- 203. Kirchgesner J, Nyboe Andersen N, Carrat F, Jess T, Beaugerie L; BERENICE study group. Risk of acute arterial events associated with treatment of inflammatory bowel diseases: nationwide French cohort study. Gut 69: 852–858, 2020. doi: 10.1136/gutjnl-2019-318932. [DOI] [PubMed] [Google Scholar]
- 204. Akbar A, Orchard T, Powell N, Selinger C, Tibbatts C. Influence of comorbidities on treatment considerations for first-line biologic prescribing in patients with inflammatory bowel disease in the UK. Frontline Gastroenterol 13: 490–496, 2022. doi: 10.1136/flgastro-2021-101995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205. Sinh P, Cross R. Cardiovascular risk assessment and impact of medications on cardiovascular disease in inflammatory bowel disease. Inflamm Bowel Dis 27: 1107–1115, 2021. doi: 10.1093/ibd/izaa258. [DOI] [PubMed] [Google Scholar]
- 206. Trakaki A, Wolf P, Weger W, Eichmann TO, Scharnagl H, Stadler JT, Salmhofer W, Knuplez E, Holzer M, Marsche G. Biological anti-psoriatic therapy profoundly affects high-density lipoprotein function. Biochim Biophys Acta Mol Cell Biol Lipids 1866: 158943, 2021. doi: 10.1016/j.bbalip.2021.158943. [DOI] [PubMed] [Google Scholar]
- 207. Sandborn WJ, Panés J, D'Haens GR, Sands BE, Su C, Moscariello M, Jones T, Pedersen R, Friedman GS, Lawendy N, Chan G. Safety of tofacitinib for treatment of ulcerative colitis, based on 4.4 years of data from global clinical trials. Clin Gastroenterol Hepatol 17: 1541–1550, 2019. doi: 10.1016/j.cgh.2018.11.035. [DOI] [PubMed] [Google Scholar]
- 208. Panés J, Gisbert JP. Efficacy of tofacitinib treatment in ulcerative colitis. Gastroenterol Hepatol 42: 403–412, 2019. doi: 10.1016/j.gastrohep.2019.03.002. [DOI] [PubMed] [Google Scholar]
- 209. Padda IS, Bhatt R, Parmar M. Tofacitinib. In: StatPearls. Treasure Island, FL: StatPearls Publishing, 2022. [Google Scholar]
- 210. Sandborn WJ, Su C, Sands BE, D'Haens GR, Vermeire S, Schreiber S, Danese S, Feagan BG, Reinisch W, Niezychowski W, Friedman G, Lawendy N, Yu D, Woodworth D, Mukherjee A, Zhang H, Healey P, Panés J; OCTAVE Induction 1, OCTAVE Induction 2, and OCTAVE Sustain Investigators. Tofacitinib as induction and maintenance therapy for ulcerative colitis. N Engl J Med 376: 1723–1736, 2017. doi: 10.1056/NEJMoa1606910. [DOI] [PubMed] [Google Scholar]
- 211. Jacobs JD, Wagner T, Gulotta G, Liao C, Li YC, Bissonnette M, Pekow J. Impact of angiotensin II signaling blockade on clinical outcomes in patients with inflammatory bowel disease. Dig Dis Sci 64: 1938–1944, 2019. doi: 10.1007/s10620-019-5474-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212. Chan SSM, Luben R, Bergmann MM, Boeing H, Olsen A, Tjonneland A, Overvad K, Kaaks R, Kennedy H, Khaw K-T, Riboli E, Hart AR. Aspirin in the aetiology of Crohn's disease and ulcerative colitis: a European prospective cohort study. Aliment Pharmacol Ther 34: 649–655, 2011. doi: 10.1111/j.1365-2036.2011.04784.x. [DOI] [PubMed] [Google Scholar]
- 213. Chande N, MacDonald JK, Wang JJ, McDonald JW. Unfractionated or low molecular weight heparin for induction of remission in ulcerative colitis: a Cochrane inflammatory bowel disease and functional bowel disorders systematic review of randomized trials. Inflamm Bowel Dis 17: 1979–1986, 2011. doi: 10.1002/ibd.21776. [DOI] [PubMed] [Google Scholar]
- 214. Auwerda JJ, Zijlstra FJ, Tak CJ, van den Ingh HF, Wilson JH, Ouwendijk RJ. Ridogrel enemas in distal ulcerative colitis. Eur J Gastroenterol Hepatol 13: 397–400, 2001. doi: 10.1097/00042737-200104000-00016. [DOI] [PubMed] [Google Scholar]
- 215. Ungaro R, Chang HL, Côté-Daigneault J, Mehandru S, Atreja A, Colombel JF. Statins associated with decreased risk of new onset inflammatory bowel disease. Am J Gastroenterol 111: 1416–1423, 2016. [Erratum in Am J Gastroenterol 111: 1370, 2016]. doi: 10.1038/ajg.2016.233. [DOI] [PubMed] [Google Scholar]
- 216. Grip O, Janciauskiene S, Bredberg A. Use of atorvastatin as an anti-inflammatory treatment in Crohn's disease. Br J Pharmacol 155: 1085–1092, 2008. doi: 10.1038/bjp.2008.369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217. Rutgeerts P, Van Assche G, Vermeire S. Optimizing anti-TNF treatment in inflammatory bowel disease. Gastroenterology 126: 1593–1610, 2004. doi: 10.1053/j.gastro.2004.02.070. [DOI] [PubMed] [Google Scholar]
- 218. Deswal A, Bozkurt B, Seta Y, Parilti-Eiswirth S, Hayes FA, Blosch C, Mann DL. Safety and efficacy of a soluble P75 tumor necrosis factor receptor (Enbrel, etanercept) in patients with advanced heart failure. Circulation 99: 3224–3226, 1999. doi: 10.1161/01.cir.99.25.3224. [DOI] [PubMed] [Google Scholar]
- 219. Adamo L, Rocha-Resende C, Prabhu SD, Mann D. Reappraising the role of inflammation in heart failure. Nat Rev Cardiol 17: 269–285, 2020. [Erratum in Nat Rev Cardiol 18: 735, 2021]. doi: 10.1038/s41569-019-0315-x. [DOI] [PubMed] [Google Scholar]
- 220. Mann DL, McMurray JJV, Packer M, Swedberg K, Borer JS, Colucci WS, Djian J, Drexler H, Feldman A, Kober L, Krum H, Liu P, Nieminen M, Tavazzi L, van Veldhuisen DJ, Waldenstrom A, Warren M, Westheim A, Zannad F, Fleming T. Targeted anticytokine therapy in patients with chronic heart failure: results of the Randomized Etanercept Worldwide Evaluation (RENEWAL). Circulation 109: 1594–1602, 2004. doi: 10.1161/01.CIR.0000124490.27666.B2. [DOI] [PubMed] [Google Scholar]
- 221. Chung ES, Packer M, Lo KH, Fasanmade AA, Willerson JTJC; Anti-TNF Therapy Against Congestive Heart Failure Investigators. Randomized, double-blind, placebo-controlled, pilot trial of infliximab, a chimeric monoclonal antibody to tumor necrosis factor-α, in patients with moderate-to-severe heart failure: results of the Anti-TNF Therapy Against Congestive Heart Failure (ATTACH) trial. Circulation 107: 3133–3140, 2003. doi: 10.1161/01.CIR.0000077913.60364.D2. [DOI] [PubMed] [Google Scholar]
- 222. López-Sanromán A, Esplugues JV, Domènech E. Pharmacology and safety of tofacitinib in ulcerative colitis. Gastroenterol Hepatol 44: 39–48, 2021. doi: 10.1016/j.gastrohep.2020.04.012. [DOI] [PubMed] [Google Scholar]
- 223. Magro F, Cordeiro G, Dias AM, Estevinho MM. Inflammatory bowel disease—non-biological treatment. Pharmacol Res 160: 105075, 2020. doi: 10.1016/j.phrs.2020.105075. [DOI] [PubMed] [Google Scholar]
- 224. Patel RS, Rohit Reddy S, Llukmani A, Hashim A, Haddad DR, Patel DS, Ahmad F, Gordon DK. Cardiovascular manifestations in inflammatory bowel disease: a systematic review of the pathogenesis and management of pericarditis. Cureus 13: e14010, 2021. doi: 10.7759/cureus.14010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225. Salimi A, Bahreini F, Jamali Z, Pourahmad J. Mesalazine induces oxidative stress and cytochrome c release in isolated rat heart mitochondria: an analysis of cardiotoxic effects. Int J Toxicol 9: 241–247, 2020. doi: 10.1177/1091581820918163. [DOI] [PubMed] [Google Scholar]
- 226. Oikonomou KA, Kapsoritakis AN, Stefanidis I, Potamianos SP. Drug-induced nephrotoxicity in inflammatory bowel disease. Nephron Clin Pract 119: c89–c94, 2011. doi: 10.1159/000326682. [DOI] [PubMed] [Google Scholar]
- 227. Rossi RE, Parisi I, Despott EJ, Burroughs AK, O'Beirne J, Conte D, Hamilton MI, Murray CD. Anti-tumour necrosis factor agent and liver injury: literature review, recommendations for management. World J Gastroenterol 20: 17352–17359, 2014. doi: 10.3748/wjg.v20.i46.17352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228. Charles PJ, Smeenk RJ, De Jong J, Feldmann M, Maini RN. Assessment of antibodies to double-stranded DNA induced in rheumatoid arthritis patients following treatment with infliximab, a monoclonal antibody to tumor necrosis factor α: findings in open-label and randomized placebo-controlled trials. Arthritis Rheum 43: 2383–2390, 2000. doi:. [DOI] [PubMed] [Google Scholar]
- 229. van Vollenhoven RF, Fleischmann R, Cohen S, Lee EB, García Meijide JA, Wagner S, Forejtova S, Zwillich SH, Gruben D, Koncz T, Wallenstein GV, Krishnaswami S, Bradley JD, Wilkinson B; ORAL Standard Investigators. Tofacitinib or adalimumab versus placebo in rheumatoid arthritis. N Engl J Med 367: 508–519, 2012. [Erratum in N Engl J Med 369: 293, 2013]. doi: 10.1056/NEJMoa1112072. [DOI] [PubMed] [Google Scholar]
- 230. Genovese MC, Kremer J, Zamani O, Ludivico C, Krogulec M, Xie L, Beattie SD, Koch AE, Cardillo TE, Rooney TP, Macias WL, de Bono S, Schlichting DE, Smolen JS. Baricitinib in patients with refractory rheumatoid arthritis. N Engl J Med 374: 1243–1252, 2016. doi: 10.1056/NEJMoa1507247. [DOI] [PubMed] [Google Scholar]
- 231. Morinobu A. JAK inhibitors for the treatment of rheumatoid arthritis. Immunol Med 43: 148–155, 2020. doi: 10.1080/25785826.2020.1770948. [DOI] [PubMed] [Google Scholar]
- 232. Sehgal P, Colombel JF, Aboubakr A, Narula N. Systematic review: safety of mesalazine in ulcerative colitis. Aliment Pharmacol Ther 47: 1597–1609, 2018. doi: 10.1111/apt.14688. [DOI] [PubMed] [Google Scholar]
- 233. Stelzer T, Kohler S, Marques Maggio E, Heuss LT. An unusual cause of febrile hepatitis. BMJ Case Rep 2015: bcr2014205857, 2015. doi: 10.1136/bcr-2014-205857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234. Braun M, Fraser GM, Kunin M, Salamon F, Tur-Kaspa R. Mesalamine-induced granulomatous hepatitis. Am J Gastroenterol 94: 1973–1974, 1999. doi: 10.1111/j.1572-0241.1999.01245.x. [DOI] [PubMed] [Google Scholar]
- 235. Ransford RA, Langman MJ. Sulphasalazine and mesalazine: serious adverse reactions re-evaluated on the basis of suspected adverse reaction reports to the Committee on Safety of Medicines. Gut 51: 536–539, 2002. doi: 10.1136/gut.51.4.536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236. Li S, Tan H-Y, Wang N, Zhang Z-J, Lao L, Wong C-W, Feng Y. The role of oxidative stress and antioxidants in liver diseases. Int J Mol Sci 16: 26087–26124, 2015. doi: 10.3390/ijms161125942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237. Li Y-C, Shen J-D, Lu S-F, Zhu L-L, Wang B-Y, Bai M, Xu E-P. Transcriptomic analysis reveals the mechanism of sulfasalazine-induced liver injury in mice. Toxicol Lett 321: 12–20, 2020. doi: 10.1016/j.toxlet.2019.12.011. [DOI] [PubMed] [Google Scholar]
- 238. Muller AF, Stevens PE, McIntyre AS, Ellison H, Logan RF. Experience of 5-aminosalicylate nephrotoxicity in the United Kingdom. Aliment Pharmacol Ther 21: 1217–1224, 2005. doi: 10.1111/j.1365-2036.2005.02462.x. [DOI] [PubMed] [Google Scholar]
- 239. de Jong DJ, Tielen J, Habraken CM, Wetzels JF, Naber AH. 5-Aminosalicylates and effects on renal function in patients with Crohn's disease. Inflamm Bowel Dis 11: 972–976, 2005. doi: 10.1097/01.mib.0000185402.65288.19. [DOI] [PubMed] [Google Scholar]
- 240. Corrigan G, Stevens PE. Review article: interstitial nephritis associated with the use of mesalazine in inflammatory bowel disease. Aliment Pharmacol Ther 14: 1–6, 2000. doi: 10.1046/j.1365-2036.2000.00683.x. [DOI] [PubMed] [Google Scholar]
- 241. Gisbert JP, González-Lama Y, Maté J. 5-Aminosalicylates and renal function in inflammatory bowel disease: a systematic review. Inflamm Bowel Dis 13: 629–638, 2007. doi: 10.1002/ibd.20099. [DOI] [PubMed] [Google Scholar]
- 242. Garg M, Royce SG, Tikellis C, Shallue C, Batu D, Velkoska E, Burrell LM, Patel SK, Beswick L, Jackson A, Britto K, Lukies M, Sluka P, Wardan H, Hirokawa Y, Tan CW, Faux M, Burgess AW, Hosking P, Monagle S, Thomas M, Gibson PR, Lubel J. Imbalance of the renin-angiotensin system may contribute to inflammation and fibrosis in IBD: a novel therapeutic target? Gut 69: 841–851, 2020. doi: 10.1136/gutjnl-2019-318512. [DOI] [PubMed] [Google Scholar]

