Skip to main content
Erschienen in: Cardiovascular Diabetology 1/2018

Open Access 01.12.2018 | Review

The role of dipeptidylpeptidase-4 inhibitors in management of cardiovascular disease in diabetes; focus on linagliptin

verfasst von: Annayya R. Aroor, Camila Manrique-Acevedo, Vincent G. DeMarco

Erschienen in: Cardiovascular Diabetology | Ausgabe 1/2018

Abstract

Multiple population based analyses have demonstrated a high incidence of cardiovascular disease (CVD) and cardiovascular (CV) mortality in subjects with T2DM that reduces life expectancy by as much as 15 years. Importantly, the CV system is particularly sensitive to the metabolic and immune derangements present in obese pre-diabetic and diabetic individuals; consequently, CV dysfunction is often the initial CV derangement to occur and promotes the progression to end organ/tissue damage in T2DM. Specifically, diabetic CVD can manifest as microvascular complications, such as nephropathy, retinopathy, and neuropathy, as well as, macrovascular impairments, including ischemic heart disease, peripheral vascular disease, and cerebrovascular disease. Despite some progress in prevention and treatment of CVD, mainly via blood pressure and dyslipidemia control strategies, the impact of metabolic disease on CV outcomes is still a major challenge and persists in proportion to the epidemics of obesity and diabetes. There is abundant pre-clinical and clinical evidence implicating the DPP-4-incretin axis in CVD. In this regard, linagliptin is a unique DPP-4 inhibitor with both CV and renal safety profiles. Moreover, it exerts beneficial CV effects beyond glycemic control and beyond class effects. Linagliptin is protective for both macrovascular and microvascular complications of diabetes in preclinical models, as well as clinical models. Given the role of endothelial-immune cell interactions as one of the key events in the initiation and progression of CVD, linagliptin modulates these cell–cell interactions by affecting two important pathways involving stimulation of NO signaling and potent inhibition of a key immunoregulatory molecule.
Abkürzungen
DPP-4
dipeptidylpeptidase-4
GLP-1
glucagon-like peptide-1
GIP
glucose-dependent insulinotropic peptide
SU
sulfonylureas
TZD
thiazolidinedione
SDF-1α
stromal-cell-derived factor-1α
SGLT2
sodium-glucose co-transporter 2
ARB
angiotensin type 2 receptor blocker
ACE
angiotensin converting enzyme
TRAF3IP2
TRAF3 interacting protein2
MIP-1α
macrophage inflammatory protein-1α
Tregs
T regulatory cells
SAVOR-TIMI
saxagliptin assessment of vascular outcomes recorded in patients with diabetes mellitus (SAVOR)–thrombolysis in myocardial infarction (TIMI)
TECOS
trial evaluating cardiovascular outcomes with sitagliptin
EXAMINE
examination of cardiovascular outcomes with alogliptin versus standard of care

Background

Glycemic control, CVD and DPP-4 inhibitors

Overwhelming evidence indicates that CVD risk increases along with increases in glycated hemoglobin (HbA1c). For example, data from the Norfolk study indicated a linear relationship between HbA1c concentrations and CVD and mortality [1]. This analysis revealed that for every percentage point increase in HbA1c above 7%, the relative risk of CVD increases by 20–30% [1]. Surprisingly, this relationship also extended to those below the threshold for controlled T2DM, i.e., HbA1c between 5 and 6.9%. Nonetheless, there is conflicting evidence to support an intensive glucose-lowering regimen in T2DM to reduce major adverse CV events and deaths [2, 3]. Moreover, some conventional diabetes therapies, although effective at glucose control, may actually increase the risk of CVD events, increase hypoglycemic episodes and result in weight gain [46]. Newer anti-hyperglycemic drugs, such as DPP-4 inhibitors, GLP-1 agonists and SGLT2 inhibitors, are well tolerated and effective and are increasingly prescribed. These drugs may exert beneficial CV effects beyond glycemic control [79], thereby making them attractive strategies as either stand-alone or add-on therapy to conventional glucose lowering medications, such as metformin, sulfonylureas, thiazolidinediones and insulin. The emphasis on their CV safety is becoming an emerging issue. In this regard, large clinical trials have shown either neutral or beneficial effects for DPP-4 inhibitors [10]. Recently, the CV protective effects of different DPP-4 inhibitors have been reviewed [7, 1114]. It is noteworthy that the DPP-4 inhibitor, linagliptin, has unique kinetics, chemical nature and potent direct effects on the vasculature [15]. In this review we will update recent advances in our understanding of the cellular and molecular mechanisms of CV protection of DPP-4 inhibitors with a focus on linagliptin.

Cardiovascular protection as a treatment goal in treatment of type 2 diabetes mellitus (T2DM)

Prior to 2008, the US Food and Drug Administration (FDA) approval process for new diabetes therapies was based largely on whether a drug was effective at improving HbA1c and its general safety profile. Following reports that certain antihyperglycemic agents increased CV events [16, 17], the FDA issued new guidelines in 2008 requiring that drug developers perform comprehensive assessment of CV safety on any new diabetes drug to ensure that these therapies do not increase the risk of CV events. Results from three large randomized trials performed over the last 5 years to address the safety and efficacy of DPP-4 inhibitors in T2DM patients at high risk for CV events have been reported [1824]. The general consensus from these three trials is that DPP-4 inhibitors, relative to placebo, do not reduce or increase the risk of the primary composite endpoints of CV death, myocardial infarction, stroke or hospitalization for unstable angina (4 point MACE in TECOS only) when added to standard of care diabetes therapy. Nevertheless, some concerns have been raised for the CV safety profile of saxagliptin which led to slightly more hospitalizations for heart failure (HHF) (3.5 vs 2.8% versus placebo; hazard ratio, 1.27; 95% CI 1.07–1.51; p = 0.007) in the SAVOR-TIMI trial [18] and alogliptin which showed a non-statistically significant risk for HHF in subjects with pre-existing HF in the EXAMINE trial [25]. These adverse events were not detected in the TECOS trial that examined the CV profile of sitagliptin [20]. Whether the disparity between these clinical trials regarding HHF is related to the individual properties of the DPP-4 inhibitors, the subgroups of patients enrolled in the studies, differences in inclusion criteria or other aspects of clinical trial design remains to be determined. However, a recent survey of data from Medicare beneficiaries, older than 65 years of which 55% had baseline CVD, did not demonstrate increased risk of stroke, myocardial infarction or heart failure when comparing DPP-4 inhibitors with sulfonylureas (SU) or thiazolidinediones (TZDs) [26]. Similarly, an analysis of an insurance database in a Korean population reported no increased risk of HHF in DPP-4 inhibitor users (sitagliptin, linagliptin, vildagliptin and saxagliptin) when compared with SU [27]. It is notable that among the DPP-4 inhibitors examined in the Korean study, patients treated with sitagliptin or linagliptin were at lower risk for HF compared to SU therapy. Moreover, risk for MI in patients with pre-existing CVD and stroke were lower in patients treated with DPP-4 compared to SU. Furthermore, a Taiwanese case control study found that DPP-4 use was related to decreased risk of death after an acute myocardial infarction [28]. It is also noteworthy that small clinical studies (< 50 patients) examining the CV effects of DPP-4 inhibitors, including linagliptin, have shown protective effects, including decreases in aortic PWV, improved microvascular function and lower heart failure risk compared to SU [29, 30]. Most of the large trials investigating the CV safety profile of DPP-4 inhibitors (SAVOR-TIMI, TECOS, EXAMINE, and CARMELINA) were designed to compare the DPP-4 inhibitor to placebo; however the ongoing CAROLINA trial was designed to compare the CV effects of linagliptin to glimeperide as active comparator and will soon (second half of 2018) provide valuable information regarding the impact of DPP-4 inhibition in diabetic CV outcomes [27, 31, 32]. Lastly, a very recent systematic review and network meta-analysis, designed to evaluate the effects of long-term CV safety of DPP-4 inhibitors (and GLP-1 agonists), showed lower risk of MI compared to SU-based therapies when these drugs are administered for more than 1 year [33].

CV protection by linagliptin

Both pre-clinical and clinical studies have shown beneficial effects of linagliptin on CV dysfunction associated with obesity and diabetes (Fig. 1). These benefits include improvement in diastolic dysfunction [34, 35], atherosclerosis [36], coronary artery disease (CAD) [37], myocardial infarction [38], hypertension and stroke [3941], arterial stiffness [29, 42, 43], endothelial dysfunction [30, 37, 4451], and immune and inflammatory response [35, 52], all of which are depicted in Fig. 1 and reviewed below.

Heart failure and diastolic dysfunction

Accumulating evidence indicates that increased circulating DPP-4 activity is associated with poorer CV outcomes in experimental and clinical heart failure models [53]. Further, emerging evidence from preclinical and clinical studies support that DPP-4 inhibitors ameliorate the development and progression of heart failure [2730, 53]. In this regard, diastolic dysfunction (DD) is one of the early manifestations of CVD in insulin resistant conditions, such as obesity and T2DM and can be identified clinically by echocardiographic findings [5457]. Moreover, DD is an independent predictor of future CV events, progression to systolic HF and CV mortality and emerging evidence indicates that DD can antedate T2DM and predicts progression of T2DM [58]. Importantly, certain groups are at increased risk of developing DD, including obese children and adolescents [5961]. Moreover, obese and diabetic premenopausal women are also at heightened risk for CVD when compared to men [54, 55, 6266]. Significantly, preclinical data with DPP-4 inhibitors have shown promise to improve DD in both males and females [34, 67, 68]. Pre-clinical studies have demonstrated CV protective effects of DPP-4 inhibition in models of genetic and dietary induced obesity, as well as pressure overload [34, 6769]. We previously tested whether linagliptin reduces pathophysiologic abnormalities in diastolic and vascular endothelial dysfunction in two translationally relevant rodent models of obesity and insulin resistance, the Zucker Obese (ZO) rat [34] and the WD-fed mouse [35, 42]. In one study, male ZO rats were treated for 2 months with linagliptin [34], beginning at 2 months of age when they already display insulin-resistance, DD and mild hypertension [70]. Linagliptin-treated rats exhibited significant improvement in impaired LV diastolic function, as well as endothelial function of gastrocnemius feed arteries, and, somewhat surprisingly, this was associated with a reduction in BP [34]. We extended our investigation of the cardioprotective effects of linagliptin using a dietary murine model of over-nutrition in which 4 week old female mice were fed a high fat-high fructose diet for 4 months (WD-western diet) [35]. Unlike ZO rats that become hypertensive at an early age, 4 months of WD feeding does not induce hypertension in young female mice on a C57Bl/6J background. Our results show that linagliptin exerts robust cardioprotective effects, including the suppression of WD-induced DD, myocardial oxidative stress and inflammation [35]. These promising preclinical findings in translationally relevant models suggest that linagliptin may prevent the onset of DD in insulin resistant states caused by overnutrition, as well as improve DD in the setting of established insulin resistance, obesity and T2DM when there is a pre-existing cardiac relaxation abnormality.

Atherosclerosis, coronary artery disease (CAD) and myocardial infarction

The incidence of atherosclerosis and coronary artery disease (CAD) in patients with T2DM is greatly increased compared to individuals without diabetes [71]. Moreover, in the presence of CAD, T2DM subjects have worse clinical outcomes when compared with patients without diabetes [10]. Atherosclerosis accounts for half of all deaths in western countries and is increasing globally [14, 7274]. DPP-4 inhibitor therapy has been shown to reduce the risk for atherosclerosis and CAD through both glycemic control and direct effects on the atherosclerotic process, including atherosclerosis or plaque stability, and this topic has been addressed in recent reviews [11, 14]. Results have been mixed with respect to the role of DPP-4 inhibition on improvement in cardiac function and remodeling in experimental models of myocardial infarction [38, 53, 7580]. In this regard, linagliptin has been shown to significantly reduce infarct size and fibrosis after ischemia/reperfusion (I/R) injury in a rat model [38] in association with a significant increase in plasma GLP-1 levels [38]. These salutary effects were not accompanied by improved cardiac function. Despite these mixed results in preclinical settings, it has been reported that DPP-4 inhibitors improve DD or long term survival in T2DM patients after acute myocardial infarction [28, 33, 81]. Moreover, this improvement occurred in both sexes showing CV protection regardless of sex [28].

Hypertension and stroke

Hypertension is twice as prevalent in individuals with T2DM compared to non-diabetic individuals [82]. Blood pressure (BP) responses to DPP-4 inhibitor therapy in humans are either neutral [83, 84] or modestly reduced [8587]. In addition, linagliptin tended to further improve BP in a rat model of renovascular hypertension when administered along with the angiotensin receptor blocker (ARB), telmisartan [41], thereby suggesting that a combination of ARB/DPP-4i could be an additional option for the management of hypertension in T2DM patients.
Recent studies show a lower incidence of non-fatal stroke events in patients treated with linagliptin compared to glimepiride, thereby accounting for significantly fewer major CVD events for linagliptin treated patients compared to those receiving glimepiride [39]. The possibility that incretin enhancer therapy could be neuroprotective is likely given that GLP-1 receptors are expressed in neurons from rodents and humans [88, 89] and that native GLP-1 and GLP-1 analogs readily cross the blood brain barrier [90, 91]. Previous studies demonstrate that exendin-4, a GLP-1 receptor (GLP-1r) agonist, abrogates the severity of stroke in diabetic and non-diabetic rodent models [9294]. Linagliptin has also been tested for its efficacy in reducing complications from stroke utilizing middle-aged non-diabetic and diabetic mice subjected to middle cerebral artery occlusion [40]. In addition to reducing plasma DPP-4 activity, linagliptin increased plasma GLP-1 levels. This was associated with a significant increase in the number of surviving cortical neurons, despite no reduction in brain infarct size, in both non-diabetic and obese diabetic mice. GLP-1 mediated modulation of matrix metalloproteinases appears to be one of the important mechanisms contributing to vasculoprotective effects of linagliptin [93, 9597]. There is also evidence indicating that linagliptin can restore impaired cerebrovascular structure and function [98100]. Linagliptin has also been shown to ameliorate impaired cognitive function and brain atrophy induced by transient cerebral ischemia in diabetic db/db mice [101]. Thus, the cerebro-protective effects of DPP-4 inhibition may be another consideration for treatment of T2DM patients at risk for development of cerebrovascular disease or cognopathy.

Arterial stiffness, endothelial dysfunction and CVD

Arterial stiffness is an independent risk factor for CVD, including hypertension, heart failure with preserved ejection fraction, chronic kidney disease and stroke [102, 103]. Arterial stiffness is more prevalent in older individuals [104] and occurs naturally with aging as a consequence of fragmentation and degradation of elastin in the wall of the aorta and its replacement with much stiffer collagen fibers [105]. Nonetheless, arterial stiffness can develop in younger individuals in the setting of insulin resistance, obesity and T2DM and evidence indicates that obese, insulin resistant and diabetic women are more prone to develop vascular stiffness than men [106, 107]. The cellular and molecular mechanisms underlying vascular stiffening comprises endothelial stiffness/dysfunction, increased vascular tone, remodeling of extracellular matrix and dysfunction of adventitial and perivascular adipose tissue [108110]. Recent reports from our laboratory indicate that therapies targeting vascular stiffness could potentially improve CV outcomes in insulin resistance models [108, 111, 112]. We also reported that administration of linagliptin prevented the development of WD-induced aortic stiffness by an NO-dependent mechanism [42]. Moreover, recent clinical studies reported that linagliptin decreased aortic PWV in subjects with T2DM [29, 43]. These clinical results are consistent with our preclinical study of prediabetic mice fed an obesogenic diet in which linagliptin prevented arterial stiffening and vascular remodeling [42].
The vascular endothelium serves as interface between blood and surrounding tissue components and regulates normal vascular functions including control of vascular tone, extracellular matrix remodeling, coagulation, leukocyte trafficking and permeability, and immune and inflammatory responses [108, 113, 114]. Endothelial dysfunction is caused by both insulin resistance and hyperglycemia in diabetes mellitus and is associated with both development of macrovascular and microvascular complications of T2DM [115, 116]. In addition to protection of macrovascular complications of T2DM by DPP-4 inhibitors, including linagliptin [29, 43, 84, 117], emerging evidence from clinical studies conducted on small numbers of patients with T2DM reported improvement in microvascular function by linagliptin [30, 4951]. Endothelial dysfunction is usually indicated by decreased bioavailable NO in response to acetylcholine/insulin mediated vascular relaxation [34, 111] or impaired flow mediated vasodilation [29, 43, 84, 117], is strongly associated with insulin resistance and hyperglycemia in diabetes mellitus. In this regard, linagliptin has potent nitric oxide enhancing effects on vascular function [34, 118, 119].

Renoprotection

Development of kidney disease is one of the major sequelae of T2DM with approximately 50% of diabetic individuals progressing to chronic kidney disease (CKD) [120, 121]. Moreover, CKD is associated with development of CVD including arterial stiffness, hypertension and cardiac dysfunction [122]. Compared to other tissues, the kidneys express the highest level of DPP-4 and it is likely that the presence of DPP-4 in the glomerular endothelium and proximal renal tubules contributes importantly to sodium retention, tubular injury and glomerular injury. We and others have shown renoprotective effects of DPP-4 inhibitors, including linagliptin, in preclinical studies [48, 123128]. Similarly, the potential beneficial effects of DPP-4 inhibitors, including linagliptin, in preventing and treating progression of kidney disease in patients with T2DM is supported by retrospective analyses of clinical trials [49, 129]. The ongoing Cardiovascular and Renal Microvascular Outcome study with Linagliptin in patients with T2DM (CARMELINA), which is powered to evaluate kidney outcomes and renoprotective effects of this inhibitor, should begin to fill in a gap in our knowledge regarding the efficacy of DPP-4 inhibitors in T2DM patients with CKD with our without CVD [130].
With regard to treating diabetic nephropathy, the pharmacokinetics and pharmacodynamics of linagliptin make it an especially attractive drug for several reasons. First, unlike other DPP-4 inhibitors that are excreted largely in urine, linagliptin is mainly eliminated by a biliary route [131] and therefore does not require dose adjustment in patients with kidney disease [132, 133]. Additionally, compared to other DPP-4 inhibitors, linagliptin can penetrate deeply into renal tissue and therefore has the largest volume of distribution, and has the highest binding affinity for DPP-4 protein that is richly present in kidney [134137]. Microvascular dysfunction is one of the major factors contributing to progression of diabetic nephropathy and DPP-4 inhibitors have been shown to exert microvascular protection in pre-clinical studies. Importantly, data from prospective clinical trials is beginning to emerge [44, 45, 138]. In this regard, linagliptin-mediated CV effects can occur both in response to better glycemic control, as well as by mechanisms independent of glycemic control in animal models of pre-diabetes [35, 42, 4448]. Emerging evidence from clinical studies conducted on small numbers of patients with T2DM reported improvement in microvascular function by linagliptin [30, 4951]. Impairment of NO signaling is one of the pathways that contributes to CVD and linagliptin has potent nitric oxide enhancing effects on vascular function [34, 118, 119]. The nephroprotective effects of DPP-4 inhibitors, including linagliptin, and their potential underlying mechanisms have been the focus of several very extensive and recent reviews [121, 129, 139].

Cellular and molecular mechanisms

GLP-1-dependent and -independent effects

The beneficial effects of DPP-4 inhibitors on the CV system (Fig. 1) may occur through glycemic control, as well as mechanisms beyond glycemic control. Linagliptin mediated increase in GLP-1 levels through its classical effect to inhibit DPP-4 activity may partly account for improvement in vascular function and associated improvement in cardiac function (Fig. 2a–c). However, GLP-1-independent mechanisms beyond glycemic control, may also significantly account CV protection by linagliptin [11, 12, 14]. These mechanisms mainly include linagliptin suppression of inappropriate RAAS activation and maladaptive immune and inflammatory response [140] (Fig. 2c, d). The improvement in nitric oxide signaling contributes significantly to suppression of inflammation and improvement of insulin signaling and vascular function, thereby inhibiting the development of atherosclerotic vascular disease [141]. Recent studies also show modulatory effects of linagliptin on TRAF3IP2 signaling (Fig. 2e) and klotho/FGF23 signaling (Fig. 2f) [35, 42].
Pre-clinical studies demonstrate cardioprotective effects of GLP-1 agonists and favor the view that inhibition of DPP-4 resulting in increased levels of GLP-1 is one of the major pathways for CV protection by DPP-4 inhibitors [11, 12]. The cardioprotection by GLP-1 includes improvement in coronary blood flow [142, 143], decreases in cardiomyocyte apoptosis [144], and reduction in infarct size [97, 145]. GLP-1 signaling improves CV function by modulating various signaling pathways, including NO/cGMP, PKA and Akt [11, 146, 147]. A recent study showed GLP-1 mediated suppression of platelet activation thereby demonstrating one more mechanism for the anti-atherosclerotic effects of DPP-4 inhibitors [148].
In addition to GLP-1 mediated effects, DPP-4 inhibitors may have CV protection through GLP-1-independent mechanisms, including inhibition of degradation of other DPP-4 substrates such as gastric inhibitory peptide (GIP) and SDF-1α [12, 14, 31]. Moreover, direct effects of DPP-4, independent of its substrate effects, have also been reported, which in turn may account for CV effects of DPP-4 inhibitors [11, 12, 14].

Immune and inflammatory mechanisms

The role of maladaptive innate and adaptive immune and inflammatory responses contributing to CV stiffening and fibrosis is evidenced by changes in the polarization status of T lymphocytes and macrophages [110]. Macrophage polarization with predominant M1 pro-inflammatory response in visceral adipose tissue and perivascular adipose tissue results in increased pro-inflammatory cytokines in plasma and in the vascular wall [149154]. In addition to macrophages, T cell activation and dysregulation of T-cell polarization can also contribute to CV dysfunction. Moreover, T helper (Th) 1 cells not only induce a pro-inflammatory response, but also promote infiltration of M1 macrophages into adipose and CV tissues [150]. Th17 cells are another subset of CD4+ cells that secrete IL-17 which promotes CV injury in obesity, diabetes and hypertension [155]. CD4+ CD25+ T regulatory cells (Tregs) are a subpopulation of T-cells [156] that mediate anti-inflammatory effects by suppressing pro-inflammatory T-cell responses and promoting M2 macrophage polarization. IL-10 secreted by Tregs inhibits NADPH oxidase mediated oxidative stress thereby contributing to suppression of CV inflammation, improvement of cardiac function and lowering of blood pressure [157, 158].
DPP-4, also known as CD26, is a T-cell surface marker that is widely expressed in immune cells [159] and cleaves numerous chemokines and peptide hormones regulating the immune system, including CCL5, CXCL12, CCL22 and MIP-1α [52, 160162]. Therefore, DPP-4 inhibitor therapy may be beneficial in suppressing maladaptive innate and adaptive immunity [163] by regulating T cell activation and macrophage polarization in adipose and CV tissue [156, 164167]. In addition, DPP-4 expressed in dendritic cell/macrophages contributes to potentiating inflammation of adipose tissue in obesity [168]. DPP-4 is also characterized as an adipokine and regulates insulin sensitivity in adipose tissue and other insulin sensitive tissues and organs [169, 170]. DPP-4 is also expressed in F4/80+ M1 macrophages [52]. Recent studies show that linagliptin not only reduces migration of M1 polarized macrophages, but also induces M2 dominant macrophage phenotype within white adipose tissue as well as liver that resulted in suppression of inflammation and insulin resistance. Moreover, it decreased the expression of macrophage inflammatory protein-1α (MIP-1α) which is a chemokine, as well as, a DPP-4 substrate. In this regard, linagliptin was not effective in suppressing M1 polarization and insulin resistance in MIP-1α knock down mice suggesting that MIP-1α is a potential mediator contributing to immunoprotective effects of linagliptin [52].
In addition to the direct effect of DPP-4 on immune and inflammatory response, DPP-4 also suppresses RAAS-mediated immune responses [35, 156, 171]. Inappropriate activation of RAAS modulates activation of T-lymphocytes and macrophages [150, 151, 156, 172], thereby contributing to CV dysfunction in obesity and T2DM [150, 173]. In this regard, recent studies showed attenuation of Ang-2 induced cardiac fibrosis by alteration of AT1/AT2 receptor expression and ACE activity in rat hearts [174]. We recently reported increased myocardial expression of AT1r and MR in WD fed female mice; linagliptin suppressed elevated RAAS receptor expression with concomitant suppression of cardiac fibrosis and immune and inflammatory response [35].

Nitric oxide signaling

Nitric oxide is vital to CV homeostasis because it is a key regulator of, among other things, vascular function and remodeling and immune and inflammatory responses [175]. Nitric oxide regulates vascular flow, tone, monocyte activation and platelet aggregation, thereby modulating blood pressure, cardiac function, thrombosis, and atherosclerosis [175, 176]. Impairment of NO signaling is associated with most CVDs and is the hallmark of obesity and T2DM [175177]. Therefore, strategies that modulate NO signaling by way of enhancing endogenous NO signaling or its downstream signaling intermediates, or through delivery of NO precursors, are likely to have salutary effects in the CV tissue. In this regard, recent studies suggest that DPP-4 inhibitors exert CV protection by increasing bioavailability of NO in the vasculature [34, 42, 118, 178]. This occurs by both GLP-1-dependent and -independent mechanisms. Compared to other DPP-4 inhibitors, linagliptin exerts more potent vasodilatory effects in aortic rings and these effects are mediated by activation of the eNOS/Akt, NO/cGMP pathway [118, 178]. Consistent with these mechanistic studies performed ex vivo, previous reports demonstrate that long term administration of linagliptin improves vascular function and NO signaling in both genetic and dietary models of obesity, in both the presence and absence of BP change [34, 42]. Thus, the preclinical evidence suggests that the NO enhancing effects of linagliptin could translate into improved CV outcomes in patients with T2DM.

FGF23/klotho signaling

Klotho is an anti-aging protein that has received much attention for its role as an aging suppression gene [179]. Klotho is expressed in high amounts in kidney and lesser amounts in parathyroid cells, adipocytes, brain and vascular endothelial cells [180182]. The cardioprotective effects of klotho have been recently reported [183]. Among the downstream targets of klotho, Sirt1 and AMPK are considered to be CV protective molecules [184]. Importantly, aging is associated with a decrease in circulating klotho levels [185]. Emerging evidence indicates klotho deficiency may be a major contributor to not only, age-related aortic stiffening, but also obesity-associated aortic stiffness. In this regard, mice with a genetic deficiency in the klotho gene develop premature aortic stiffness that is associated with increased collagen deposition and reduced elastin in the medial layer of the aorta [186]. Moreover, klotho deficient mice had elevated circulating aldosterone concentrations and mineralocorticoid receptor blockade prevented the aortic stiffening and remodeling associated with klotho deficiency [186]. Moreover, klotho levels are decreased by Ang-2 [187] and one of the downstream effects of klotho is regulation of NO signaling (Fig. 2c, f) [188].
Emerging evidence suggests a new mechanism to explain the vasculo-protective effects of linagliptin (and perhaps other DPP-4 inhibitors) that involves modulation of aging pathways. We recently reported that long term consumption of a WD induces aortic stiffness in female mice and this was prevented with linagliptin [42]. We also determined that WD induced a deficiency in klotho protein expression in the aorta that was prevented with linagliptin administration [42]. To our knowledge, this was the first study suggesting that the vasculo-protective effects of linagliptin involve modulation of klotho signaling. In addition, a more recent study showed that amelioration of progression of premature aging in klotho knock out mice by linagliptin and these salutary effects were associated with increased bioavailable NO in the cerebral vasculature [188]. Angiotensin II decreases klotho levels and linagliptin improves Ang II signaling, including downregulation of AT1R [35]. Therefore, the klotho-mediated beneficial effects of linagliptin may be accounted for by both modulation of Ang II signaling upstream of klotho (Fig. 2c) and NO signaling downstream of klotho (Fig. 2f).

TRAF3IP2 (TRAF3 interacting protein2)

TRAF3IP2 is a key regulator of the immune and inflammatory response and exerts multiple effects to promote CV stiffening, inflammation, and fibrosis that contribute to cardiac dysfunction and vascular inflammation. TRAF3IP2 signaling is a convergent point in regulation of a pro-fibrotic response. The upstream regulators of TRAF3IP2 include oxidative stress, RAAS activation and cytokines, including IL-17, whereas the downstream targets are transcriptional factors, such as NF-κB and AP-1 and cell signaling pathways such as p38-MAPK and the crosslinking enzyme, lysyl oxidase [189] (Fig. 2e). Induction of TRAF3IP2 in cardiac fibroblasts in response to either Ang-2 [190] or aldosterone [191] promotes a fibrotic response. We recently showed upregulation of TRAF3IP2 in heart tissue from WD-fed female mice and linagliptin administration significantly suppressed this induction. We further demonstrated that the transcription factors and kinase signaling pathway regulated by TRAF3IP2 are also upregulated by WD feeding and this was associated with maladaptive cardiac immune and inflammatory response, as well as fibrosis [35]. Supporting these in vivo observations, our in vitro studies using isolated cardiac fibroblasts also demonstrated that linagliptin inhibits aldosterone-induced TRAF3IP2 expression, oxidative stress, inflammatory cytokine expression, and cardiac fibroblast activation and migration. Collectively, these findings suggest that one of the mechanisms by which linagliptin suppresses maladaptive immune and inflammatory response in CV tissue is through modulation of immune regulatory molecules, such as TRAF3IP2.

SDF-1 (stromal cell-derived factor-1)/CXCR4 signaling

The tissue repair process in response to CV injury is regulated by multiple factors including recruitment of progenitor/stem cells [192, 193]. Diabetes is characterized by a deficiency or dysfunction in circulating progenitor/stem cells which predicts future CV events, poor macro- and microvascular outcomes and death [194196]. In this regard, SDF-1α is a CXC chemokine and ligand for the CXCR4 receptor, in addition to being a substrate of DPP-4. SDF-1α is a potent chemoattractant for various stem cells involved in tissue repair and regeneration, including among others, endothelial progenitor cells, endogenous cardiac stem cells, bone marrow stem cells, mesenchymal stem cells, and T-lymphocytes [192]. Tissue injury can induce local endothelial cells to secrete SDF-1α which mediates adherence of circulating stem cells to the endothelium. In the setting of diabetes, SDF-1α may be rapidly degraded by local and circulating DPP-4 activity which may limit mobilization and attachment of stem cells to injured tissue. As there is no effective therapy to treat CV injury and fibrosis associated with T2DM, DPP-4 inhibitor-mediated targeting of SDF-1α, to prevent its degradation and enhance stem cell mobilization from bone marrow and recruitment to peripheral tissue, may be an attractive strategy to enhance CV tissue repair [35, 38, 197199]. It should be noted that a recent review article has proffered the notion that the potentiation of SDF-1α by DPP-4 inhibitor therapy may exacerbate rather than resolve tissue inflammation and fibrosis, thus neutralizing the benefits of potentiating GLP-1 signaling [200]. This idea has sparked debate [201]. In this regard, DPP-4 inhibitors have not been shown to have CV adverse effects for most of these inhibitors and the adverse effects of saxagliptin may be related to a subset of patients with pre-existing cardiac disease [20, 26, 27, 201]. Moreover, linagliptin has been shown to increase circulating SDF-1α, as well as putative vascular regenerative and anti-inflammatory cells in patients with T2DM, independently of its effects on glycemia [201, 202]. Other studies provide compelling evidence that linagliptin-induced increases in local and circulating levels of SDF-1α are associated with tissue repair. For example, linagliptin and the novel DPP-4 inhibitor, BI 14361 have been shown to significantly reduce infarct size and myocardial fibrosis in an experimental model of myocardial ischemia–reperfusion injury in Wistar rats [38]. Improvement in tissue remodeling in treated rats was associated with accumulation of cells positive for SDF-1α, CXCR-4 and CD34 within and around the infarcted area. This study supports the notion that SDF-1α is upregulated during myocardial ischemia and contributes to mobilization of bone marrow derived stem cells to the ischemic heart. In another study, we reported that linagliptin treatment increased glomerular and kidney tubular expression of SDF-1α, as well as, circulating SDF-1α levels and ameliorated kidney injury in rat model of diabetic nephropathy [48]. Importantly, SDF-1α has been shown to cause eNOS activation in endothelial cells and preserve microvascular integrity [193] and this is depicted in Fig. 2c.

Linagliptin combination with an SGLT2 inhibitor

The first line therapy for management of glycemia for T2DM throughout the world is metformin [203]; however, over time, the effectiveness of metformin monotherapy in achieving target HbA1c diminishes in a majority of patients [204]. This lack of adequate glycemic control necessitates the use of combinations of one or more additional anti-hyperglycemia agents to achieve target HbA1c. However, interest in the need for combination therapy as an initial treatment strategy is also increasing [205207]. Recent studies indicate that DPP-4 inhibitors, including linagliptin, could potentially complement the effects of other CV protective agents such as metformin [208], angiotensin type 2 receptor blockers (ARB) [41, 209], statins, and SGLT2 inhibitors [210212]. Importantly, there is little evidence of undesirable drug interactions between linagliptin and metformin, ARBs, statins and SGLT2 inhibitors [5, 213]. The increased risk of angioedema with combination DPP-4 and ACE inhibitor therapy is one potential limitation [214], however, this issue may be managed by consideration of replacing an ACE inhibitor with an ARB [128, 215].
Recent studies have shown that combination therapy of DPP-4 and SGLT2 inhibitors, either as an initial combination or stepwise addition, results in improvement in glycemic control [216221]. In contrast to other DPP-4 inhibitors, linagliptin does not require dose adjustment in patients with renal insufficiency given that it is not excreted by the kidneys [15]. Indeed, the evidence so far indicates that once daily single pill combination of linagliptin and the SGLT-2 inhibitor, empagliflozin, results in clinically meaningful and sustained reductions in HbA1c, fasting glucose, body weight and blood pressure [220, 222]. Other combinations of SGLT2 and DPP-4 inhibitors seem similarly promising [220]. The underlying mechanisms for these beneficial effects appears to be the convergence of complementary signaling pathways and physiological effects (e.g., reduced glucotoxicity, weight loss and BP reduction for SGLT2 inhibitor and reduced glucose-dependent glucagon secretion and anti-inflammatory effects for DPP-4 inhibitor) for beneficial effects on CV health or suppression of microvascular complications [220, 223].

Conclusions

Linagliptin is protective for both macrovascular and microvascular complications of diabetes in preclinical models, as well as clinical models. Linagliptin exerts beneficial CV effects through glycemic control, as well as effects beyond glycemic control and beyond class effects. Linagliptin modulation of endothelial and immune cell responses appear to be key mechanisms for ameliorating the progression of CVD.

Authors’ contributions

AA, CM and VGD reviewed the literature and wrote the paper. All authors read and approved the final manuscript.

Acknowledgements

Studio support with figure development was provided by Envision Scientific Solutions and supported financially by Boehringer Ingelheim.

Competing interests

VGD is currently funded by Boehringer Ingelheim.

Availability of data and materials

Not applicable.
Not applicable.
Not applicable-no human subjects or tissue.

Funding

This work was supported by unrestricted funding from Boehringer Ingelheim (VGD).

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Khaw KT, Wareham N, Bingham S, Luben R, Welch A, Day N. Association of hemoglobin A1c with cardiovascular disease and mortality in adults: the European prospective investigation into cancer in Norfolk. Ann Intern Med. 2004;141(6):413–20.PubMedCrossRef Khaw KT, Wareham N, Bingham S, Luben R, Welch A, Day N. Association of hemoglobin A1c with cardiovascular disease and mortality in adults: the European prospective investigation into cancer in Norfolk. Ann Intern Med. 2004;141(6):413–20.PubMedCrossRef
2.
Zurück zum Zitat Mannucci E, Dicembrini I, Lauria A, Pozzilli P. Is glucose control important for prevention of cardiovascular disease in diabetes? Diabetes Care. 2013;36(Suppl 2):S259–63.PubMedPubMedCentralCrossRef Mannucci E, Dicembrini I, Lauria A, Pozzilli P. Is glucose control important for prevention of cardiovascular disease in diabetes? Diabetes Care. 2013;36(Suppl 2):S259–63.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Boussageon R, Bejan-Angoulvant T, Saadatian-Elahi M, Lafont S, Bergeonneau C, Kassai B, Erpeldinger S, Wright JM, Gueyffier F, Cornu C. Effect of intensive glucose lowering treatment on all cause mortality, cardiovascular death, and microvascular events in type 2 diabetes: meta-analysis of randomised controlled trials. BMJ. 2011;343:d4169.PubMedPubMedCentralCrossRef Boussageon R, Bejan-Angoulvant T, Saadatian-Elahi M, Lafont S, Bergeonneau C, Kassai B, Erpeldinger S, Wright JM, Gueyffier F, Cornu C. Effect of intensive glucose lowering treatment on all cause mortality, cardiovascular death, and microvascular events in type 2 diabetes: meta-analysis of randomised controlled trials. BMJ. 2011;343:d4169.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Upadhyay J, Polyzos SA, Perakakis N, Thakkar B, Paschou SA, Katsiki N, Underwood P, Park KH, Seufert J, Kang ES, et al. Pharmacotherapy of type 2 diabetes: an update. Metabolism. 2018;78:13–42.PubMedCrossRef Upadhyay J, Polyzos SA, Perakakis N, Thakkar B, Paschou SA, Katsiki N, Underwood P, Park KH, Seufert J, Kang ES, et al. Pharmacotherapy of type 2 diabetes: an update. Metabolism. 2018;78:13–42.PubMedCrossRef
5.
6.
Zurück zum Zitat UK Prospective Diabetes Study. (UKPDS) Group: Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34). UK Prospective Diabetes Study (UKPDS) Group. Lancet. 1998;352(9131):854–65.CrossRef UK Prospective Diabetes Study. (UKPDS) Group: Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34). UK Prospective Diabetes Study (UKPDS) Group. Lancet. 1998;352(9131):854–65.CrossRef
7.
Zurück zum Zitat Chin HJ, Nam JH, Lee EK, Shin JY. Comparative safety for cardiovascular outcomes of DPP-4 inhibitors versus glimepiride in patients with type 2 diabetes: a retrospective cohort study. Medicine (Baltimore). 2017;96(25):e7213.CrossRef Chin HJ, Nam JH, Lee EK, Shin JY. Comparative safety for cardiovascular outcomes of DPP-4 inhibitors versus glimepiride in patients with type 2 diabetes: a retrospective cohort study. Medicine (Baltimore). 2017;96(25):e7213.CrossRef
8.
Zurück zum Zitat Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck MA, Nissen SE, Pocock S, Poulter NR, Ravn LS, et al. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2016;375(4):311–22.PubMedPubMedCentralCrossRef Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck MA, Nissen SE, Pocock S, Poulter NR, Ravn LS, et al. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2016;375(4):311–22.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Zinman B, Lachin JM, Inzucchi SE. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2016;374(11):1094.PubMedCrossRef Zinman B, Lachin JM, Inzucchi SE. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2016;374(11):1094.PubMedCrossRef
10.
Zurück zum Zitat Lehrke M, Leiter LA, Hehnke U, Thiemann S, Bhandari A, Meinicke T, Johansen OE. Safety and efficacy of linagliptin in patients with type 2 diabetes mellitus and coronary artery disease: analysis of pooled events from 19 clinical trials. J Diabetes Complications. 2016;30(7):1378–84.PubMedCrossRef Lehrke M, Leiter LA, Hehnke U, Thiemann S, Bhandari A, Meinicke T, Johansen OE. Safety and efficacy of linagliptin in patients with type 2 diabetes mellitus and coronary artery disease: analysis of pooled events from 19 clinical trials. J Diabetes Complications. 2016;30(7):1378–84.PubMedCrossRef
11.
Zurück zum Zitat Aroor AR, Sowers JR, Jia G, DeMarco VG. Pleiotropic effects of the dipeptidylpeptidase-4 inhibitors on the cardiovascular system. Am J Physiol Heart Circ Physiol. 2014;15:H477–92.CrossRef Aroor AR, Sowers JR, Jia G, DeMarco VG. Pleiotropic effects of the dipeptidylpeptidase-4 inhibitors on the cardiovascular system. Am J Physiol Heart Circ Physiol. 2014;15:H477–92.CrossRef
12.
Zurück zum Zitat Nauck MA, Meier JJ, Cavender MA, Abd El Aziz M, Drucker DJ. Cardiovascular actions and clinical outcomes with glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors. Circulation. 2017;136(9):849–70.PubMedCrossRef Nauck MA, Meier JJ, Cavender MA, Abd El Aziz M, Drucker DJ. Cardiovascular actions and clinical outcomes with glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors. Circulation. 2017;136(9):849–70.PubMedCrossRef
13.
Zurück zum Zitat Zhong J, Maiseyeu A, Davis SN, Rajagopalan S. DPP4 in cardiometabolic disease: recent insights from the laboratory and clinical trials of DPP4 inhibition. Circ Res. 2015;116(8):1491–504.PubMedPubMedCentralCrossRef Zhong J, Maiseyeu A, Davis SN, Rajagopalan S. DPP4 in cardiometabolic disease: recent insights from the laboratory and clinical trials of DPP4 inhibition. Circ Res. 2015;116(8):1491–504.PubMedPubMedCentralCrossRef
14.
15.
Zurück zum Zitat Koibuchi N, Hasegawa Y, Katayama T, Toyama K, Uekawa K, Sueta D, Kusaka H, Ma M, Nakagawa T, Lin B, et al. DPP-4 inhibitor linagliptin ameliorates cardiovascular injury in salt-sensitive hypertensive rats independently of blood glucose and blood pressure. Cardiovasc Diabetol. 2014;13:157.PubMedPubMedCentralCrossRef Koibuchi N, Hasegawa Y, Katayama T, Toyama K, Uekawa K, Sueta D, Kusaka H, Ma M, Nakagawa T, Lin B, et al. DPP-4 inhibitor linagliptin ameliorates cardiovascular injury in salt-sensitive hypertensive rats independently of blood glucose and blood pressure. Cardiovasc Diabetol. 2014;13:157.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Nissen SE, Wolski K, Topol EJ. Effect of muraglitazar on death and major adverse cardiovascular events in patients with type 2 diabetes mellitus. JAMA. 2005;294(20):2581–6.PubMedCrossRef Nissen SE, Wolski K, Topol EJ. Effect of muraglitazar on death and major adverse cardiovascular events in patients with type 2 diabetes mellitus. JAMA. 2005;294(20):2581–6.PubMedCrossRef
17.
Zurück zum Zitat Nissen SE, Wolski K. Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes. N Engl J Med. 2007;356(24):2457–71.PubMedCrossRef Nissen SE, Wolski K. Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes. N Engl J Med. 2007;356(24):2457–71.PubMedCrossRef
18.
Zurück zum Zitat Scirica BM, Bhatt DL, Braunwald E, Steg PG, Davidson J, Hirshberg B, Ohman P, Frederich R, Wiviott SD, Hoffman EB, et al. Saxagliptin and cardiovascular outcomes in patients with type 2 diabetes mellitus. N Engl J Med. 2013;369(14):1317–26.PubMedCrossRef Scirica BM, Bhatt DL, Braunwald E, Steg PG, Davidson J, Hirshberg B, Ohman P, Frederich R, Wiviott SD, Hoffman EB, et al. Saxagliptin and cardiovascular outcomes in patients with type 2 diabetes mellitus. N Engl J Med. 2013;369(14):1317–26.PubMedCrossRef
19.
Zurück zum Zitat Zannad F, Cannon CP, Cushman WC, Bakris GL, Menon V, Perez AT, Fleck PR, Mehta CR, Kupfer S, Wilson C, et al. Heart failure and mortality outcomes in patients with type 2 diabetes taking alogliptin versus placebo in EXAMINE: a multicentre, randomised, double-blind trial. Lancet. 2015;385(9982):2067–76.PubMedCrossRef Zannad F, Cannon CP, Cushman WC, Bakris GL, Menon V, Perez AT, Fleck PR, Mehta CR, Kupfer S, Wilson C, et al. Heart failure and mortality outcomes in patients with type 2 diabetes taking alogliptin versus placebo in EXAMINE: a multicentre, randomised, double-blind trial. Lancet. 2015;385(9982):2067–76.PubMedCrossRef
20.
Zurück zum Zitat Green JB, Bethel MA, Armstrong PW, Buse JB, Engel SS, Garg J, Josse R, Kaufman KD, Koglin J, Korn S, et al. Effect of sitagliptin on cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2015;373(3):232–42.PubMedCrossRef Green JB, Bethel MA, Armstrong PW, Buse JB, Engel SS, Garg J, Josse R, Kaufman KD, Koglin J, Korn S, et al. Effect of sitagliptin on cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2015;373(3):232–42.PubMedCrossRef
21.
Zurück zum Zitat Paneni F. DPP-4 inhibitors, heart failure and type 2 diabetes: all eyes on safety. Cardiovasc Diagn Ther. 2015;5(6):471–8.PubMedPubMedCentral Paneni F. DPP-4 inhibitors, heart failure and type 2 diabetes: all eyes on safety. Cardiovasc Diagn Ther. 2015;5(6):471–8.PubMedPubMedCentral
22.
Zurück zum Zitat Standl E, Schnell O. DPP-4 inhibitors and risk of heart failure EXAMINEd. Lancet. 2015;385(9982):2022–4.PubMedCrossRef Standl E, Schnell O. DPP-4 inhibitors and risk of heart failure EXAMINEd. Lancet. 2015;385(9982):2022–4.PubMedCrossRef
23.
Zurück zum Zitat Udell JA, Cavender MA, Bhatt DL, Chatterjee S, Farkouh ME, Scirica BM. Glucose-lowering drugs or strategies and cardiovascular outcomes in patients with or at risk for type 2 diabetes: a meta-analysis of randomised controlled trials. Lancet Diabetes Endocrinol. 2015;3(5):356–66.PubMedCrossRef Udell JA, Cavender MA, Bhatt DL, Chatterjee S, Farkouh ME, Scirica BM. Glucose-lowering drugs or strategies and cardiovascular outcomes in patients with or at risk for type 2 diabetes: a meta-analysis of randomised controlled trials. Lancet Diabetes Endocrinol. 2015;3(5):356–66.PubMedCrossRef
24.
Zurück zum Zitat McGuire DK, Van de Werf F, Armstrong PW, Standl E, Koglin J, Green JB, Bethel MA, Cornel JH, Lopes RD, Halvorsen S, et al. Association between sitagliptin use and heart failure hospitalization and related outcomes in type 2 diabetes mellitus: secondary analysis of a randomized clinical trial. JAMA Cardiol. 2016;1(2):126–35.PubMedCrossRef McGuire DK, Van de Werf F, Armstrong PW, Standl E, Koglin J, Green JB, Bethel MA, Cornel JH, Lopes RD, Halvorsen S, et al. Association between sitagliptin use and heart failure hospitalization and related outcomes in type 2 diabetes mellitus: secondary analysis of a randomized clinical trial. JAMA Cardiol. 2016;1(2):126–35.PubMedCrossRef
25.
Zurück zum Zitat White WB, Pratley R, Fleck P, Munsaka M, Hisada M, Wilson C, Menon V. Cardiovascular safety of the dipetidyl peptidase-4 inhibitor alogliptin in type 2 diabetes mellitus. Diabetes Obes Metab. 2013;15(7):668–73.PubMedCrossRef White WB, Pratley R, Fleck P, Munsaka M, Hisada M, Wilson C, Menon V. Cardiovascular safety of the dipetidyl peptidase-4 inhibitor alogliptin in type 2 diabetes mellitus. Diabetes Obes Metab. 2013;15(7):668–73.PubMedCrossRef
26.
Zurück zum Zitat Gokhale M, Buse JB, Jonsson Funk M, Lund J, Pate V, Simpson RJ, Sturmer T. No increased risk of cardiovascular events in older adults initiating dipeptidyl peptidase-4 inhibitors vs therapeutic alternatives. Diabetes Obes Metab. 2017;19(7):970–8.PubMedCrossRefPubMedCentral Gokhale M, Buse JB, Jonsson Funk M, Lund J, Pate V, Simpson RJ, Sturmer T. No increased risk of cardiovascular events in older adults initiating dipeptidyl peptidase-4 inhibitors vs therapeutic alternatives. Diabetes Obes Metab. 2017;19(7):970–8.PubMedCrossRefPubMedCentral
27.
Zurück zum Zitat Kim YG, Yoon D, Park S, Han SJ, Kim DJ, Lee KW, Park RW, Kim HJ. Dipeptidyl peptidase-4 inhibitors and risk of heart failure in patients with type 2 diabetes mellitus: a population-based cohort study. Circ Heart Fail. 2017;10(9):e003957.PubMedCrossRef Kim YG, Yoon D, Park S, Han SJ, Kim DJ, Lee KW, Park RW, Kim HJ. Dipeptidyl peptidase-4 inhibitors and risk of heart failure in patients with type 2 diabetes mellitus: a population-based cohort study. Circ Heart Fail. 2017;10(9):e003957.PubMedCrossRef
28.
Zurück zum Zitat Wang MT, Lin SC, Tang PL, Hung WT, Cheng CC, Yang JS, Chang HT, Liu CP, Mar GY, Huang WC. The impact of DPP-4 inhibitors on long-term survival among diabetic patients after first acute myocardial infarction. Cardiovasc Diabetol. 2017;16(1):89.PubMedPubMedCentralCrossRef Wang MT, Lin SC, Tang PL, Hung WT, Cheng CC, Yang JS, Chang HT, Liu CP, Mar GY, Huang WC. The impact of DPP-4 inhibitors on long-term survival among diabetic patients after first acute myocardial infarction. Cardiovasc Diabetol. 2017;16(1):89.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat de Boer SA, Heerspink HJL, Juarez Orozco LE, van Roon AM, Kamphuisen PW, Smit AJ, Slart R, Lefrandt JD, Mulder DJ. Effect of linagliptin on pulse wave velocity in early type 2 diabetes: a randomized, double-blind, controlled 26-week trial (RELEASE). Diabetes Obes Metab. 2017;19(8):1147–54.PubMedCrossRef de Boer SA, Heerspink HJL, Juarez Orozco LE, van Roon AM, Kamphuisen PW, Smit AJ, Slart R, Lefrandt JD, Mulder DJ. Effect of linagliptin on pulse wave velocity in early type 2 diabetes: a randomized, double-blind, controlled 26-week trial (RELEASE). Diabetes Obes Metab. 2017;19(8):1147–54.PubMedCrossRef
30.
Zurück zum Zitat Jax T, Stirban A, Terjung A, Esmaeili H, Berk A, Thiemann S, Chilton R, von Eynatten M, Marx N. A randomised, active- and placebo-controlled, three-period crossover trial to investigate short-term effects of the dipeptidyl peptidase-4 inhibitor linagliptin on macro- and microvascular endothelial function in type 2 diabetes. Cardiovasc Diabetol. 2017;16(1):13.PubMedPubMedCentralCrossRef Jax T, Stirban A, Terjung A, Esmaeili H, Berk A, Thiemann S, Chilton R, von Eynatten M, Marx N. A randomised, active- and placebo-controlled, three-period crossover trial to investigate short-term effects of the dipeptidyl peptidase-4 inhibitor linagliptin on macro- and microvascular endothelial function in type 2 diabetes. Cardiovasc Diabetol. 2017;16(1):13.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Fiorentino TV, Sesti G. Lessons learned from cardiovascular outcome clinical trials with dipeptidyl peptidase 4 (DPP-4) inhibitors. Endocrine. 2016;53(2):373–80.PubMedCrossRef Fiorentino TV, Sesti G. Lessons learned from cardiovascular outcome clinical trials with dipeptidyl peptidase 4 (DPP-4) inhibitors. Endocrine. 2016;53(2):373–80.PubMedCrossRef
32.
Zurück zum Zitat Rosenstock J, Marx N, Neubacher D, Seck T, Patel S, Woerle HJ, Johansen OE. Cardiovascular safety of linagliptin in type 2 diabetes: a comprehensive patient-level pooled analysis of prospectively adjudicated cardiovascular events. Cardiovasc Diabetol. 2015;14:57.PubMedPubMedCentralCrossRef Rosenstock J, Marx N, Neubacher D, Seck T, Patel S, Woerle HJ, Johansen OE. Cardiovascular safety of linagliptin in type 2 diabetes: a comprehensive patient-level pooled analysis of prospectively adjudicated cardiovascular events. Cardiovasc Diabetol. 2015;14:57.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Chou CY, Chang YT, Yang JL, Wang JY, Lee TE, Wang RY, Hung CC. Effect of long-term incretin-based therapies on ischemic heart diseases in patients with type 2 diabetes mellitus: a network meta-analysis. Sci Rep. 2017;7(1):15795.PubMedPubMedCentralCrossRef Chou CY, Chang YT, Yang JL, Wang JY, Lee TE, Wang RY, Hung CC. Effect of long-term incretin-based therapies on ischemic heart diseases in patients with type 2 diabetes mellitus: a network meta-analysis. Sci Rep. 2017;7(1):15795.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Aroor AR, Sowers JR, Bender SB, Nistala R, Garro M, Mugerfeld I, Hayden MR, Johnson MS, Salam M, Whaley-Connell A, et al. Dipeptidylpeptidase inhibition is associated with improvement in blood pressure and diastolic function in insulin resistant male Zucker obese rats. Endocrinology. 2013;154(7):2501–13.PubMedPubMedCentralCrossRef Aroor AR, Sowers JR, Bender SB, Nistala R, Garro M, Mugerfeld I, Hayden MR, Johnson MS, Salam M, Whaley-Connell A, et al. Dipeptidylpeptidase inhibition is associated with improvement in blood pressure and diastolic function in insulin resistant male Zucker obese rats. Endocrinology. 2013;154(7):2501–13.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Aroor AR, Habibi J, Kandikattu HK, Garro-Kacher M, Barron B, Chen D, Hayden MR, Whaley-Connell A, Bender SB, Klein T, et al. Dipeptidyl peptidase-4 (DPP-4) inhibition with linagliptin reduces western diet-induced myocardial TRAF3IP2 expression, inflammation and fibrosis in female mice. Cardiovasc Diabetol. 2017;16(1):61.PubMedPubMedCentralCrossRef Aroor AR, Habibi J, Kandikattu HK, Garro-Kacher M, Barron B, Chen D, Hayden MR, Whaley-Connell A, Bender SB, Klein T, et al. Dipeptidyl peptidase-4 (DPP-4) inhibition with linagliptin reduces western diet-induced myocardial TRAF3IP2 expression, inflammation and fibrosis in female mice. Cardiovasc Diabetol. 2017;16(1):61.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Salim HM, Fukuda D, Higashikuni Y, Tanaka K, Hirata Y, Yagi S, Soeki T, Shimabukuro M, Sata M. Dipeptidyl peptidase-4 inhibitor, linagliptin, ameliorates endothelial dysfunction and atherogenesis in normoglycemic apolipoprotein-E deficient mice. Vasc Pharmacol. 2016;79:16–23.CrossRef Salim HM, Fukuda D, Higashikuni Y, Tanaka K, Hirata Y, Yagi S, Soeki T, Shimabukuro M, Sata M. Dipeptidyl peptidase-4 inhibitor, linagliptin, ameliorates endothelial dysfunction and atherogenesis in normoglycemic apolipoprotein-E deficient mice. Vasc Pharmacol. 2016;79:16–23.CrossRef
37.
Zurück zum Zitat Koyama T, Tanaka A, Yoshida H, Oyama JI, Toyoda S, Sakuma M, Inoue T, Otsuka Y, Node K. Comparison of the effects of linagliptin and voglibose on endothelial function in patients with type 2 diabetes and coronary artery disease: a prospective, randomized, pilot study (EFFORT). Heart Vessels. 2018. https://doi.org/10.1007/s00380-018-1136-2.PubMedCrossRef Koyama T, Tanaka A, Yoshida H, Oyama JI, Toyoda S, Sakuma M, Inoue T, Otsuka Y, Node K. Comparison of the effects of linagliptin and voglibose on endothelial function in patients with type 2 diabetes and coronary artery disease: a prospective, randomized, pilot study (EFFORT). Heart Vessels. 2018. https://​doi.​org/​10.​1007/​s00380-018-1136-2.PubMedCrossRef
38.
Zurück zum Zitat Hocher B, Sharkovska Y, Mark M, Klein T, Pfab T. The novel DPP-4 inhibitors linagliptin and BI 14361 reduce infarct size after myocardial ischemia/reperfusion in rats. Int J Cardiol. 2013;167(1):87–93.PubMedCrossRef Hocher B, Sharkovska Y, Mark M, Klein T, Pfab T. The novel DPP-4 inhibitors linagliptin and BI 14361 reduce infarct size after myocardial ischemia/reperfusion in rats. Int J Cardiol. 2013;167(1):87–93.PubMedCrossRef
39.
Zurück zum Zitat Gallwitz B, Rosenstock J, Rauch T, Bhattacharya S, Patel S, von Eynatten M, Dugi KA, Woerle HJ. 2-year efficacy and safety of linagliptin compared with glimepiride in patients with type 2 diabetes inadequately controlled on metformin: a randomised, double-blind, non-inferiority trial. Lancet. 2012;380(9840):475–83.PubMedCrossRef Gallwitz B, Rosenstock J, Rauch T, Bhattacharya S, Patel S, von Eynatten M, Dugi KA, Woerle HJ. 2-year efficacy and safety of linagliptin compared with glimepiride in patients with type 2 diabetes inadequately controlled on metformin: a randomised, double-blind, non-inferiority trial. Lancet. 2012;380(9840):475–83.PubMedCrossRef
40.
Zurück zum Zitat Darsalia V, Ortsater H, Olverling A, Darlof E, Wolbert P, Nystrom T, Klein T, Sjoholm A, Patrone C. The DPP-4 inhibitor linagliptin counteracts stroke in the normal and diabetic mouse brain: a comparison with glimepiride. Diabetes. 2012;62:1289–96.PubMedCrossRef Darsalia V, Ortsater H, Olverling A, Darlof E, Wolbert P, Nystrom T, Klein T, Sjoholm A, Patrone C. The DPP-4 inhibitor linagliptin counteracts stroke in the normal and diabetic mouse brain: a comparison with glimepiride. Diabetes. 2012;62:1289–96.PubMedCrossRef
41.
Zurück zum Zitat Chaykovska L, Alter ML, von Websky K, Hohmann M, Tsuprykov O, Reichetzeder C, Kutil B, Kraft R, Klein T, Hocher B. Effects of telmisartan and linagliptin when used in combination on blood pressure and oxidative stress in rats with 2-kidney-1-clip hypertension. J Hypertens. 2013;31(11):2290–9.PubMedCrossRef Chaykovska L, Alter ML, von Websky K, Hohmann M, Tsuprykov O, Reichetzeder C, Kutil B, Kraft R, Klein T, Hocher B. Effects of telmisartan and linagliptin when used in combination on blood pressure and oxidative stress in rats with 2-kidney-1-clip hypertension. J Hypertens. 2013;31(11):2290–9.PubMedCrossRef
42.
Zurück zum Zitat Manrique C, Habibi J, Aroor AR, Sowers JR, Jia G, Hayden MR, Garro M, Martinez-Lemus LA, Ramirez-Perez FI, Klein T, et al. Dipeptidyl peptidase-4 inhibition with linagliptin prevents western diet-induced vascular abnormalities in female mice. Cardiovasc Diabetol. 2016;15(1):94.PubMedPubMedCentralCrossRef Manrique C, Habibi J, Aroor AR, Sowers JR, Jia G, Hayden MR, Garro M, Martinez-Lemus LA, Ramirez-Perez FI, Klein T, et al. Dipeptidyl peptidase-4 inhibition with linagliptin prevents western diet-induced vascular abnormalities in female mice. Cardiovasc Diabetol. 2016;15(1):94.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Shigiyama F, Kumashiro N, Miyagi M, Iga R, Kobayashi Y, Kanda E, Uchino H, Hirose T. Linagliptin improves endothelial function in patients with type 2 diabetes: a randomized study of linagliptin effectiveness on endothelial function. J Diabetes Investig. 2017;8(3):330–40.PubMedCrossRef Shigiyama F, Kumashiro N, Miyagi M, Iga R, Kobayashi Y, Kanda E, Uchino H, Hirose T. Linagliptin improves endothelial function in patients with type 2 diabetes: a randomized study of linagliptin effectiveness on endothelial function. J Diabetes Investig. 2017;8(3):330–40.PubMedCrossRef
44.
Zurück zum Zitat Kang YM, Jung CH. Effects of incretin-based therapies on diabetic microvascular complications. Endocrinol Metab (Seoul). 2017;32(3):316–25.CrossRef Kang YM, Jung CH. Effects of incretin-based therapies on diabetic microvascular complications. Endocrinol Metab (Seoul). 2017;32(3):316–25.CrossRef
45.
Zurück zum Zitat Avogaro A, Fadini GP. The effects of dipeptidyl peptidase-4 inhibition on microvascular diabetes complications. Diabetes Care. 2014;37(10):2884–94.PubMedCrossRef Avogaro A, Fadini GP. The effects of dipeptidyl peptidase-4 inhibition on microvascular diabetes complications. Diabetes Care. 2014;37(10):2884–94.PubMedCrossRef
46.
Zurück zum Zitat Gallwitz B. Emerging DPP-4 inhibitors: focus on linagliptin for type 2 diabetes. Diabetes Metabol Syndr Obesity. 2013;6:1–9. Gallwitz B. Emerging DPP-4 inhibitors: focus on linagliptin for type 2 diabetes. Diabetes Metabol Syndr Obesity. 2013;6:1–9.
47.
Zurück zum Zitat Dietrich N, Kolibabka M, Busch S, Bugert P, Kaiser U, Lin J, Fleming T, Morcos M, Klein T, Schlotterer A, et al. The DPP4 inhibitor linagliptin protects from experimental diabetic retinopathy. PLoS ONE. 2016;11(12):e0167853.PubMedPubMedCentralCrossRef Dietrich N, Kolibabka M, Busch S, Bugert P, Kaiser U, Lin J, Fleming T, Morcos M, Klein T, Schlotterer A, et al. The DPP4 inhibitor linagliptin protects from experimental diabetic retinopathy. PLoS ONE. 2016;11(12):e0167853.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Nistala R, Habibi J, Aroor A, Sowers JR, Hayden MR, Meuth A, Knight W, Hancock T, Klein T, DeMarco VG, et al. DPP4 inhibition attenuates filtration barrier injury and oxidant stress in the Zucker obese rat. Obesity (Silver Spring). 2014;22(10):2172–9.CrossRef Nistala R, Habibi J, Aroor A, Sowers JR, Hayden MR, Meuth A, Knight W, Hancock T, Klein T, DeMarco VG, et al. DPP4 inhibition attenuates filtration barrier injury and oxidant stress in the Zucker obese rat. Obesity (Silver Spring). 2014;22(10):2172–9.CrossRef
49.
Zurück zum Zitat Groop PH, Cooper ME, Perkovic V, Emser A, Woerle HJ, von Eynatten M. Linagliptin lowers albuminuria on top of recommended standard treatment in patients with type 2 diabetes and renal dysfunction. Diabetes Care. 2013;36(11):3460–8.PubMedPubMedCentralCrossRef Groop PH, Cooper ME, Perkovic V, Emser A, Woerle HJ, von Eynatten M. Linagliptin lowers albuminuria on top of recommended standard treatment in patients with type 2 diabetes and renal dysfunction. Diabetes Care. 2013;36(11):3460–8.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Baltzis D, Dushay JR, Loader J, Wu J, Greenman RL, Roustit M, Veves A. Effect of linagliptin on vascular function: a randomized. Placebo-controlled Study. J Clin Endocrinol Metab. 2016;101(11):4205–13.PubMedPubMedCentralCrossRef Baltzis D, Dushay JR, Loader J, Wu J, Greenman RL, Roustit M, Veves A. Effect of linagliptin on vascular function: a randomized. Placebo-controlled Study. J Clin Endocrinol Metab. 2016;101(11):4205–13.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Ott C, Kistner I, Keller M, Friedrich S, Willam C, Bramlage P, Schmieder RE. Effects of linagliptin on renal endothelial function in patients with type 2 diabetes: a randomised clinical trial. Diabetologia. 2016;59(12):2579–87.PubMedCrossRef Ott C, Kistner I, Keller M, Friedrich S, Willam C, Bramlage P, Schmieder RE. Effects of linagliptin on renal endothelial function in patients with type 2 diabetes: a randomised clinical trial. Diabetologia. 2016;59(12):2579–87.PubMedCrossRef
52.
Zurück zum Zitat Zhuge F, Ni Y, Nagashimada M, Nagata N, Xu L, Mukaida N, Kaneko S, Ota T. DPP-4 inhibition by linagliptin attenuates obesity-related inflammation and insulin resistance by regulating M1/M2 macrophage polarization. Diabetes. 2016;65:2966–79.PubMedCrossRef Zhuge F, Ni Y, Nagashimada M, Nagata N, Xu L, Mukaida N, Kaneko S, Ota T. DPP-4 inhibition by linagliptin attenuates obesity-related inflammation and insulin resistance by regulating M1/M2 macrophage polarization. Diabetes. 2016;65:2966–79.PubMedCrossRef
53.
Zurück zum Zitat Dos Santos L, Salles TA, Arruda-Junior DF, Campos LC, Pereira AC, Barreto AL, Antonio EL, Mansur AJ, Tucci PJ, Krieger JE, et al. Circulating dipeptidyl peptidase iv activity correlates with cardiac dysfunction in human and experimental heart failure. Circ Heart Fail. 2013;6:1029–38.PubMedCrossRef Dos Santos L, Salles TA, Arruda-Junior DF, Campos LC, Pereira AC, Barreto AL, Antonio EL, Mansur AJ, Tucci PJ, Krieger JE, et al. Circulating dipeptidyl peptidase iv activity correlates with cardiac dysfunction in human and experimental heart failure. Circ Heart Fail. 2013;6:1029–38.PubMedCrossRef
54.
Zurück zum Zitat Peterson LR, Waggoner AD, Schechtman KB, Meyer T, Gropler RJ, Barzilai B, Davila-Roman VG. Alterations in left ventricular structure and function in young healthy obese women: assessment by echocardiography and tissue Doppler imaging. J Am Coll Cardiol. 2004;43(8):1399–404.PubMedCrossRef Peterson LR, Waggoner AD, Schechtman KB, Meyer T, Gropler RJ, Barzilai B, Davila-Roman VG. Alterations in left ventricular structure and function in young healthy obese women: assessment by echocardiography and tissue Doppler imaging. J Am Coll Cardiol. 2004;43(8):1399–404.PubMedCrossRef
55.
Zurück zum Zitat Kenchaiah S, Evans JC, Levy D, Wilson PW, Benjamin EJ, Larson MG, Kannel WB, Vasan RS. Obesity and the risk of heart failure. N Engl J Med. 2002;347(5):305–13.PubMedCrossRef Kenchaiah S, Evans JC, Levy D, Wilson PW, Benjamin EJ, Larson MG, Kannel WB, Vasan RS. Obesity and the risk of heart failure. N Engl J Med. 2002;347(5):305–13.PubMedCrossRef
56.
Zurück zum Zitat Santos JL, Salemi VM, Picard MH, Mady C, Coelho OR. Subclinical regional left ventricular dysfunction in obese patients with and without hypertension or hypertrophy. Obesity (Silver Spring). 2011;19(6):1296–303.CrossRef Santos JL, Salemi VM, Picard MH, Mady C, Coelho OR. Subclinical regional left ventricular dysfunction in obese patients with and without hypertension or hypertrophy. Obesity (Silver Spring). 2011;19(6):1296–303.CrossRef
57.
Zurück zum Zitat Van Putte-Katier N, Rooman RP, Haas L, Verhulst SL, Desager KN, Ramet J, Suys BE. Early cardiac abnormalities in obese children: importance of obesity per se versus associated cardiovascular risk factors. Pediatr Res. 2008;64(2):205–9.PubMedCrossRef Van Putte-Katier N, Rooman RP, Haas L, Verhulst SL, Desager KN, Ramet J, Suys BE. Early cardiac abnormalities in obese children: importance of obesity per se versus associated cardiovascular risk factors. Pediatr Res. 2008;64(2):205–9.PubMedCrossRef
58.
Zurück zum Zitat Park J, Kim JS, Kim SH, Kim S, Lim SY, Lim HE, Cho GY, Sung KC, Kim JY, Baik I, et al. Subclinical left ventricular diastolic dysfunction and incident type 2 diabetes risk: the Korean Genome and Epidemiology Study. Cardiovasc Diabetol. 2017;16(1):36.PubMedPubMedCentralCrossRef Park J, Kim JS, Kim SH, Kim S, Lim SY, Lim HE, Cho GY, Sung KC, Kim JY, Baik I, et al. Subclinical left ventricular diastolic dysfunction and incident type 2 diabetes risk: the Korean Genome and Epidemiology Study. Cardiovasc Diabetol. 2017;16(1):36.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Ogden CL, Carroll MD, Kit BK, Flegal KM. Prevalence of obesity and trends in body mass index among US children and adolescents, 1999-2010. JAMA. 2012;307(5):483–90.PubMedCrossRefPubMedCentral Ogden CL, Carroll MD, Kit BK, Flegal KM. Prevalence of obesity and trends in body mass index among US children and adolescents, 1999-2010. JAMA. 2012;307(5):483–90.PubMedCrossRefPubMedCentral
60.
Zurück zum Zitat Sharpe JA, Naylor LH, Jones TW, Davis EA, O’Driscoll G, Ramsay JM, Green DJ. Impact of obesity on diastolic function in subjects < or = 16 years of age. Am J Cardiol. 2006;98(5):691–3.PubMedCrossRef Sharpe JA, Naylor LH, Jones TW, Davis EA, O’Driscoll G, Ramsay JM, Green DJ. Impact of obesity on diastolic function in subjects < or = 16 years of age. Am J Cardiol. 2006;98(5):691–3.PubMedCrossRef
61.
Zurück zum Zitat Batalli-Kepuska A, Bajraktari G, Zejnullahu M, Azemi M, Shala M, Batalli A, Ibrahimi P, Jashari F, Henein MY. Abnormal systolic and diastolic myocardial function in obese asymptomatic adolescents. Int J Cardiol. 2013;168(3):2347–51.PubMedCrossRef Batalli-Kepuska A, Bajraktari G, Zejnullahu M, Azemi M, Shala M, Batalli A, Ibrahimi P, Jashari F, Henein MY. Abnormal systolic and diastolic myocardial function in obese asymptomatic adolescents. Int J Cardiol. 2013;168(3):2347–51.PubMedCrossRef
62.
Zurück zum Zitat Peterson LR, Saeed IM, McGill JB, Herrero P, Schechtman KB, Gunawardena R, Recklein CL, Coggan AR, Demoss AJ, Dence CS, et al. Sex and type 2 diabetes: obesity-independent effects on left ventricular substrate metabolism and relaxation in humans. Obesity (Silver Spring). 2012;20(4):802–10.CrossRef Peterson LR, Saeed IM, McGill JB, Herrero P, Schechtman KB, Gunawardena R, Recklein CL, Coggan AR, Demoss AJ, Dence CS, et al. Sex and type 2 diabetes: obesity-independent effects on left ventricular substrate metabolism and relaxation in humans. Obesity (Silver Spring). 2012;20(4):802–10.CrossRef
63.
Zurück zum Zitat De Simone G, Devereux RB, Chinali M, Roman MJ, Barac A, Panza JA, Lee ET, Howard BV. Sex differences in obesity-related changes in left ventricular morphology: the strong heart study. J Hypertens. 2011;29(7):1431–8.PubMedPubMedCentralCrossRef De Simone G, Devereux RB, Chinali M, Roman MJ, Barac A, Panza JA, Lee ET, Howard BV. Sex differences in obesity-related changes in left ventricular morphology: the strong heart study. J Hypertens. 2011;29(7):1431–8.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Rutter MK, Parise H, Benjamin EJ, Levy D, Larson MG, Meigs JB, Nesto RW, Wilson PW, Vasan RS. Impact of glucose intolerance and insulin resistance on cardiac structure and function: sex-related differences in the Framingham heart study. Circulation. 2003;107(3):448–54.PubMedCrossRef Rutter MK, Parise H, Benjamin EJ, Levy D, Larson MG, Meigs JB, Nesto RW, Wilson PW, Vasan RS. Impact of glucose intolerance and insulin resistance on cardiac structure and function: sex-related differences in the Framingham heart study. Circulation. 2003;107(3):448–54.PubMedCrossRef
65.
Zurück zum Zitat From AM, Scott CG, Chen HH. The development of heart failure in patients with diabetes mellitus and pre-clinical diastolic dysfunction a population-based study. J Am Coll Cardiol. 2010;55(4):300–5.PubMedCrossRef From AM, Scott CG, Chen HH. The development of heart failure in patients with diabetes mellitus and pre-clinical diastolic dysfunction a population-based study. J Am Coll Cardiol. 2010;55(4):300–5.PubMedCrossRef
66.
Zurück zum Zitat Gregg EW, Zhuo X, Cheng YJ, Albright AL, Narayan KM, Thompson TJ. Trends in lifetime risk and years of life lost due to diabetes in the USA, 1985–2011: a modelling study. Lancet Diabetes Endocrinol. 2014;2(11):867–74.PubMedCrossRef Gregg EW, Zhuo X, Cheng YJ, Albright AL, Narayan KM, Thompson TJ. Trends in lifetime risk and years of life lost due to diabetes in the USA, 1985–2011: a modelling study. Lancet Diabetes Endocrinol. 2014;2(11):867–74.PubMedCrossRef
67.
Zurück zum Zitat Bostick B, Habibi J, Ma L, Aroor A, Rehmer N, Hayden MR, Sowers JR. Dipeptidyl peptidase inhibition prevents diastolic dysfunction and reduces myocardial fibrosis in a mouse model of Western diet induced obesity. Metabolism. 2014;63(8):1000–11.PubMedPubMedCentralCrossRef Bostick B, Habibi J, Ma L, Aroor A, Rehmer N, Hayden MR, Sowers JR. Dipeptidyl peptidase inhibition prevents diastolic dysfunction and reduces myocardial fibrosis in a mouse model of Western diet induced obesity. Metabolism. 2014;63(8):1000–11.PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Brown SM, Smith CE, Meuth AI, Khan M, Aroor AR, Cleeton HM, Meininger GA, Sowers JR, DeMarco VG, Chandrasekar B, et al. Dipeptidyl peptidase-4 inhibition with saxagliptin ameliorates angiotensin ii-induced cardiac diastolic dysfunction in male mice. Endocrinology. 2017;158(10):3592–604.PubMedCrossRefPubMedCentral Brown SM, Smith CE, Meuth AI, Khan M, Aroor AR, Cleeton HM, Meininger GA, Sowers JR, DeMarco VG, Chandrasekar B, et al. Dipeptidyl peptidase-4 inhibition with saxagliptin ameliorates angiotensin ii-induced cardiac diastolic dysfunction in male mice. Endocrinology. 2017;158(10):3592–604.PubMedCrossRefPubMedCentral
69.
Zurück zum Zitat Hiemstra JA, Lee DI, Chakir K, Gutierrez-Aguilar M, Marshall KD, Zgoda PJ, Cruz Rivera N, Dozier DG, Ferguson BS, Heublein DM, et al. Saxagliptin and tadalafil differentially alter cyclic guanosine monophosphate (cGMP) signaling and left ventricular function in aortic-banded mini-swine. J Am Heart Assoc. 2016;5(4):e003277.PubMedPubMedCentralCrossRef Hiemstra JA, Lee DI, Chakir K, Gutierrez-Aguilar M, Marshall KD, Zgoda PJ, Cruz Rivera N, Dozier DG, Ferguson BS, Heublein DM, et al. Saxagliptin and tadalafil differentially alter cyclic guanosine monophosphate (cGMP) signaling and left ventricular function in aortic-banded mini-swine. J Am Heart Assoc. 2016;5(4):e003277.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Zhou X, Ma L, Habibi J, Whaley-Connel AT, Hayden MR, Tilmon RD, Brown AN, DeMarco VG, Sowers JR. Nebivolol improves diastolic dysfunction and myocardial tissue remodeling through reductions in oxidative stress in the Zucker Obese rat. Hypertension. 2010;55(4):880–8.PubMedCrossRef Zhou X, Ma L, Habibi J, Whaley-Connel AT, Hayden MR, Tilmon RD, Brown AN, DeMarco VG, Sowers JR. Nebivolol improves diastolic dysfunction and myocardial tissue remodeling through reductions in oxidative stress in the Zucker Obese rat. Hypertension. 2010;55(4):880–8.PubMedCrossRef
71.
Zurück zum Zitat Naito R, Kasai T. Coronary artery disease in type 2 diabetes mellitus: recent treatment strategies and future perspectives. World J Cardiol. 2015;7(3):119–24.PubMedPubMedCentralCrossRef Naito R, Kasai T. Coronary artery disease in type 2 diabetes mellitus: recent treatment strategies and future perspectives. World J Cardiol. 2015;7(3):119–24.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Herrington W, Lacey B, Sherliker P, Armitage J, Lewington S. Epidemiology of atherosclerosis and the potential to reduce the global burden of atherothrombotic disease. Circ Res. 2016;118(4):535–46.PubMedCrossRef Herrington W, Lacey B, Sherliker P, Armitage J, Lewington S. Epidemiology of atherosclerosis and the potential to reduce the global burden of atherothrombotic disease. Circ Res. 2016;118(4):535–46.PubMedCrossRef
73.
Zurück zum Zitat Raskob GE, Angchaisuksiri P, Blanco AN, Buller H, Gallus A, Hunt BJ, Hylek EM, Kakkar A, Konstantinides SV, McCumber M, et al. Thrombosis: a major contributor to global disease burden. Arterioscler Thromb Vasc Biol. 2014;34(11):2363–71.PubMedCrossRef Raskob GE, Angchaisuksiri P, Blanco AN, Buller H, Gallus A, Hunt BJ, Hylek EM, Kakkar A, Konstantinides SV, McCumber M, et al. Thrombosis: a major contributor to global disease burden. Arterioscler Thromb Vasc Biol. 2014;34(11):2363–71.PubMedCrossRef
74.
Zurück zum Zitat Palombo C, Kozakova M. Arterial stiffness, atherosclerosis and cardiovascular risk: pathophysiologic mechanisms and emerging clinical indications. Vascul Pharmacol. 2016;77:1–7.PubMedCrossRef Palombo C, Kozakova M. Arterial stiffness, atherosclerosis and cardiovascular risk: pathophysiologic mechanisms and emerging clinical indications. Vascul Pharmacol. 2016;77:1–7.PubMedCrossRef
75.
Zurück zum Zitat Poulsen MK, Henriksen JE, Dahl J, Johansen A, Gerke O, Vach W, Haghfelt T, Hoilund-Carlsen PF, Beck-Nielsen H, Moller JE. Left ventricular diastolic function in type 2 diabetes mellitus: prevalence and association with myocardial and vascular disease. Circ Cardiovasc Imaging. 2010;3(1):24–31.PubMedCrossRef Poulsen MK, Henriksen JE, Dahl J, Johansen A, Gerke O, Vach W, Haghfelt T, Hoilund-Carlsen PF, Beck-Nielsen H, Moller JE. Left ventricular diastolic function in type 2 diabetes mellitus: prevalence and association with myocardial and vascular disease. Circ Cardiovasc Imaging. 2010;3(1):24–31.PubMedCrossRef
76.
Zurück zum Zitat Connelly KA, Zhang Y, Advani A, Advani SL, Thai K, Yuen DA, Gilbert RE. DPP-4 inhibition attenuates cardiac dysfunction and adverse remodeling following myocardial infarction in rats with experimental diabetes. Cardiovasc Ther. 2013;31(5):259–67.PubMedCrossRef Connelly KA, Zhang Y, Advani A, Advani SL, Thai K, Yuen DA, Gilbert RE. DPP-4 inhibition attenuates cardiac dysfunction and adverse remodeling following myocardial infarction in rats with experimental diabetes. Cardiovasc Ther. 2013;31(5):259–67.PubMedCrossRef
77.
Zurück zum Zitat Sauve M, Ban K, Momen MA, Zhou YQ, Henkelman RM, Husain M, Drucker DJ. Genetic deletion or pharmacological inhibition of dipeptidyl peptidase-4 improves cardiovascular outcomes after myocardial infarction in mice. Diabetes. 2010;59(4):1063–73.PubMedPubMedCentralCrossRef Sauve M, Ban K, Momen MA, Zhou YQ, Henkelman RM, Husain M, Drucker DJ. Genetic deletion or pharmacological inhibition of dipeptidyl peptidase-4 improves cardiovascular outcomes after myocardial infarction in mice. Diabetes. 2010;59(4):1063–73.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Ye Y, Keyes KT, Zhang C, Perez-Polo JR, Lin Y, Birnbaum Y. The myocardial infarct size-limiting effect of sitagliptin is PKA-dependent, whereas the protective effect of pioglitazone is partially dependent on PKA. Am J Physiol Heart Circ Physiol. 2010;298(5):H1454–65.PubMedCrossRef Ye Y, Keyes KT, Zhang C, Perez-Polo JR, Lin Y, Birnbaum Y. The myocardial infarct size-limiting effect of sitagliptin is PKA-dependent, whereas the protective effect of pioglitazone is partially dependent on PKA. Am J Physiol Heart Circ Physiol. 2010;298(5):H1454–65.PubMedCrossRef
79.
Zurück zum Zitat Hausenloy DJ, Whittington HJ, Wynne AM, Begum SS, Theodorou L, Riksen N, Mocanu MM, Yellon DM. Dipeptidyl peptidase-4 inhibitors and GLP-1 reduce myocardial infarct size in a glucose-dependent manner. Cardiovasc Diabetol. 2013;12(1):154.PubMedPubMedCentralCrossRef Hausenloy DJ, Whittington HJ, Wynne AM, Begum SS, Theodorou L, Riksen N, Mocanu MM, Yellon DM. Dipeptidyl peptidase-4 inhibitors and GLP-1 reduce myocardial infarct size in a glucose-dependent manner. Cardiovasc Diabetol. 2013;12(1):154.PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Yin M, Sillje HH, Meissner M, van Gilst WH, de Boer RA. Early and late effects of the DPP-4 inhibitor vildagliptin in a rat model of post-myocardial infarction heart failure. Cardiovasc Diabetol. 2011;10:85.PubMedPubMedCentralCrossRef Yin M, Sillje HH, Meissner M, van Gilst WH, de Boer RA. Early and late effects of the DPP-4 inhibitor vildagliptin in a rat model of post-myocardial infarction heart failure. Cardiovasc Diabetol. 2011;10:85.PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Fujiwara T, Yoshida M, Nakamura T, Sakakura K, Wada H, Arao K, Katayama T, Funayama H, Sugawara Y, Mitsuhashi T, et al. Dipeptidyl peptidase-4 inhibitors are associated with improved left ventricular diastolic function after acute myocardial infarction in diabetic patients. Heart Vessels. 2015;30(5):696–701.PubMedCrossRef Fujiwara T, Yoshida M, Nakamura T, Sakakura K, Wada H, Arao K, Katayama T, Funayama H, Sugawara Y, Mitsuhashi T, et al. Dipeptidyl peptidase-4 inhibitors are associated with improved left ventricular diastolic function after acute myocardial infarction in diabetic patients. Heart Vessels. 2015;30(5):696–701.PubMedCrossRef
82.
Zurück zum Zitat Lago RM, Singh PP, Nesto RW. Diabetes and hypertension. Nat Clin Pract Endocrinol Metab. 2007;3(10):667.PubMedCrossRef Lago RM, Singh PP, Nesto RW. Diabetes and hypertension. Nat Clin Pract Endocrinol Metab. 2007;3(10):667.PubMedCrossRef
83.
Zurück zum Zitat von Eynatten M, Gong Y, Emser A, Woerle HJ. Efficacy and safety of linagliptin in type 2 diabetes subjects at high risk for renal and cardiovascular disease: a pooled analysis of six phase III clinical trials. Cardiovasc Diabetol. 2013;12:60.CrossRef von Eynatten M, Gong Y, Emser A, Woerle HJ. Efficacy and safety of linagliptin in type 2 diabetes subjects at high risk for renal and cardiovascular disease: a pooled analysis of six phase III clinical trials. Cardiovasc Diabetol. 2013;12:60.CrossRef
84.
Zurück zum Zitat Kubota Y, Miyamoto M, Takagi G, Ikeda T, Kirinoki-Ichikawa S, Tanaka K, Mizuno K. The dipeptidyl peptidase-4 inhibitor sitagliptin improves vascular endothelial function in type 2 diabetes. J Korean Med Sci. 2012;27(11):1364–70.PubMedPubMedCentralCrossRef Kubota Y, Miyamoto M, Takagi G, Ikeda T, Kirinoki-Ichikawa S, Tanaka K, Mizuno K. The dipeptidyl peptidase-4 inhibitor sitagliptin improves vascular endothelial function in type 2 diabetes. J Korean Med Sci. 2012;27(11):1364–70.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Mistry GC, Maes AL, Lasseter KC, Davies MJ, Gottesdiener KM, Wagner JA, Herman GA. Effect of sitagliptin, a dipeptidyl peptidase-4 inhibitor, on blood pressure in nondiabetic patients with mild to moderate hypertension. J Clin Pharmacol. 2008;48(5):592–8.PubMedCrossRef Mistry GC, Maes AL, Lasseter KC, Davies MJ, Gottesdiener KM, Wagner JA, Herman GA. Effect of sitagliptin, a dipeptidyl peptidase-4 inhibitor, on blood pressure in nondiabetic patients with mild to moderate hypertension. J Clin Pharmacol. 2008;48(5):592–8.PubMedCrossRef
86.
Zurück zum Zitat Ogawa S, Ishiki M, Nako K, Okamura M, Senda M, Mori T, Ito S. Sitagliptin, a dipeptidyl peptidase-4 inhibitor, decreases systolic blood pressure in Japanese hypertensive patients with type 2 diabetes. Tohoku J Exp Med. 2011;223(2):133–5.PubMedCrossRef Ogawa S, Ishiki M, Nako K, Okamura M, Senda M, Mori T, Ito S. Sitagliptin, a dipeptidyl peptidase-4 inhibitor, decreases systolic blood pressure in Japanese hypertensive patients with type 2 diabetes. Tohoku J Exp Med. 2011;223(2):133–5.PubMedCrossRef
87.
Zurück zum Zitat Cobble ME, Frederich R. Saxagliptin for the treatment of type 2 diabetes mellitus: assessing cardiovascular data. Cardiovasc Diabetol. 2012;11:6.PubMedPubMedCentralCrossRef Cobble ME, Frederich R. Saxagliptin for the treatment of type 2 diabetes mellitus: assessing cardiovascular data. Cardiovasc Diabetol. 2012;11:6.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Hamilton A, Holscher C. Receptors for the incretin glucagon-like peptide-1 are expressed on neurons in the central nervous system. Neuro Report. 2009;20(13):1161–6. Hamilton A, Holscher C. Receptors for the incretin glucagon-like peptide-1 are expressed on neurons in the central nervous system. Neuro Report. 2009;20(13):1161–6.
89.
Zurück zum Zitat Goke R, Larsen PJ, Mikkelsen JD, Sheikh SP. Distribution of GLP-1 binding sites in the rat brain: evidence that exendin-4 is a ligand of brain GLP-1 binding sites. Eur J Neurosci. 1995;7(11):2294–300.PubMedCrossRef Goke R, Larsen PJ, Mikkelsen JD, Sheikh SP. Distribution of GLP-1 binding sites in the rat brain: evidence that exendin-4 is a ligand of brain GLP-1 binding sites. Eur J Neurosci. 1995;7(11):2294–300.PubMedCrossRef
90.
Zurück zum Zitat Hunter K, Holscher C. Drugs developed to treat diabetes, liraglutide and lixisenatide, cross the blood brain barrier and enhance neurogenesis. BMC Neurosci. 2012;13:33.PubMedPubMedCentralCrossRef Hunter K, Holscher C. Drugs developed to treat diabetes, liraglutide and lixisenatide, cross the blood brain barrier and enhance neurogenesis. BMC Neurosci. 2012;13:33.PubMedPubMedCentralCrossRef
91.
Zurück zum Zitat Holst JJ, Burcelin R, Nathanson E. Neuroprotective properties of GLP-1: theoretical and practical applications. Curr Med Res Opin. 2011;27(3):547–58.PubMedCrossRef Holst JJ, Burcelin R, Nathanson E. Neuroprotective properties of GLP-1: theoretical and practical applications. Curr Med Res Opin. 2011;27(3):547–58.PubMedCrossRef
92.
Zurück zum Zitat Briyal S, Gulati K, Gulati A. Repeated administration of exendin-4 reduces focal cerebral ischemia-induced infarction in rats. Brain Res. 2012;1427:23–34.PubMedCrossRef Briyal S, Gulati K, Gulati A. Repeated administration of exendin-4 reduces focal cerebral ischemia-induced infarction in rats. Brain Res. 2012;1427:23–34.PubMedCrossRef
93.
Zurück zum Zitat Darsalia V, Mansouri S, Ortsater H, Olverling A, Nozadze N, Kappe C, Iverfeldt K, Tracy LM, Grankvist N, Sjoholm A, et al. Glucagon-like peptide-1 receptor activation reduces ischaemic brain damage following stroke in type 2 diabetic rats. Clin Sci (Lond). 2012;122(10):473–83.CrossRef Darsalia V, Mansouri S, Ortsater H, Olverling A, Nozadze N, Kappe C, Iverfeldt K, Tracy LM, Grankvist N, Sjoholm A, et al. Glucagon-like peptide-1 receptor activation reduces ischaemic brain damage following stroke in type 2 diabetic rats. Clin Sci (Lond). 2012;122(10):473–83.CrossRef
94.
Zurück zum Zitat Lee CH, Yan B, Yoo KY, Choi JH, Kwon SH, Her S, Sohn Y, Hwang IK, Cho JH, Kim YM, et al. Ischemia-induced changes in glucagon-like peptide-1 receptor and neuroprotective effect of its agonist, exendin-4, in experimental transient cerebral ischemia. J Neurosci Res. 2011;89(7):1103–13.PubMedCrossRef Lee CH, Yan B, Yoo KY, Choi JH, Kwon SH, Her S, Sohn Y, Hwang IK, Cho JH, Kim YM, et al. Ischemia-induced changes in glucagon-like peptide-1 receptor and neuroprotective effect of its agonist, exendin-4, in experimental transient cerebral ischemia. J Neurosci Res. 2011;89(7):1103–13.PubMedCrossRef
96.
Zurück zum Zitat Kaczmarek L. Mmp-9 inhibitors in the brain: can old bullets shoot new targets? Curr Pharm Des. 2013;19(6):1085–9.PubMedCrossRef Kaczmarek L. Mmp-9 inhibitors in the brain: can old bullets shoot new targets? Curr Pharm Des. 2013;19(6):1085–9.PubMedCrossRef
97.
Zurück zum Zitat Noyan-Ashraf MH, Momen MA, Ban K, Sadi AM, Zhou YQ, Riazi AM, Baggio LL, Henkelman RM, Husain M, Drucker DJ. GLP-1R agonist liraglutide activates cytoprotective pathways and improves outcomes after experimental myocardial infarction in mice. Diabetes. 2009;58(4):975–83.PubMedPubMedCentralCrossRef Noyan-Ashraf MH, Momen MA, Ban K, Sadi AM, Zhou YQ, Riazi AM, Baggio LL, Henkelman RM, Husain M, Drucker DJ. GLP-1R agonist liraglutide activates cytoprotective pathways and improves outcomes after experimental myocardial infarction in mice. Diabetes. 2009;58(4):975–83.PubMedPubMedCentralCrossRef
98.
Zurück zum Zitat Hardigan T, Abdul Y, Ergul A. Linagliptin reduces effects of ET-1 and TLR2-mediated cerebrovascular hyperreactivity in diabetes. Life Sci. 2016;159:90–6.PubMedPubMedCentralCrossRef Hardigan T, Abdul Y, Ergul A. Linagliptin reduces effects of ET-1 and TLR2-mediated cerebrovascular hyperreactivity in diabetes. Life Sci. 2016;159:90–6.PubMedPubMedCentralCrossRef
99.
Zurück zum Zitat Yasir A, Hardigan T, Ergul A. Diabetes-mediated middle cerebral artery remodeling is restored by linagliptin: interaction with the vascular smooth muscle cell endothelin system. Life Sci. 2016;159:76–82.PubMedPubMedCentralCrossRef Yasir A, Hardigan T, Ergul A. Diabetes-mediated middle cerebral artery remodeling is restored by linagliptin: interaction with the vascular smooth muscle cell endothelin system. Life Sci. 2016;159:76–82.PubMedPubMedCentralCrossRef
100.
Zurück zum Zitat Hardigan T, Yasir A, Abdelsaid M, Coucha M, El-Shaffey S, Li W, Johnson MH, Ergul A. Linagliptin treatment improves cerebrovascular function and remodeling and restores reduced cerebral perfusion in Type 2 diabetes. Am J Physiol Regul Integr Comp Physiol. 2016;311(3):R466–77.PubMedPubMedCentralCrossRef Hardigan T, Yasir A, Abdelsaid M, Coucha M, El-Shaffey S, Li W, Johnson MH, Ergul A. Linagliptin treatment improves cerebrovascular function and remodeling and restores reduced cerebral perfusion in Type 2 diabetes. Am J Physiol Regul Integr Comp Physiol. 2016;311(3):R466–77.PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Ma M, Hasegawa Y, Koibuchi N, Toyama K, Uekawa K, Nakagawa T, Lin B, Kim-Mitsuyama S. DPP-4 inhibition with linagliptin ameliorates cognitive impairment and brain atrophy induced by transient cerebral ischemia in type 2 diabetic mice. Cardiovasc Diabetol. 2015;14:54.PubMedPubMedCentralCrossRef Ma M, Hasegawa Y, Koibuchi N, Toyama K, Uekawa K, Nakagawa T, Lin B, Kim-Mitsuyama S. DPP-4 inhibition with linagliptin ameliorates cognitive impairment and brain atrophy induced by transient cerebral ischemia in type 2 diabetic mice. Cardiovasc Diabetol. 2015;14:54.PubMedPubMedCentralCrossRef
102.
Zurück zum Zitat Coutinho T. Arterial stiffness and its clinical implications in women. Can J Cardiol. 2014;30(7):756–64.PubMedCrossRef Coutinho T. Arterial stiffness and its clinical implications in women. Can J Cardiol. 2014;30(7):756–64.PubMedCrossRef
103.
Zurück zum Zitat Weisbrod RM, Shiang T, Al Sayah L, Fry JL, Bajpai S, Reinhart-King CA, Lob HE, Santhanam L, Mitchell G, Cohen RA, et al. Arterial stiffening precedes systolic hypertension in diet-induced obesity. Hypertension. 2013;62:1105–10.PubMedCrossRef Weisbrod RM, Shiang T, Al Sayah L, Fry JL, Bajpai S, Reinhart-King CA, Lob HE, Santhanam L, Mitchell G, Cohen RA, et al. Arterial stiffening precedes systolic hypertension in diet-induced obesity. Hypertension. 2013;62:1105–10.PubMedCrossRef
104.
Zurück zum Zitat Lakatta EG, Levy D. Arterial and cardiac aging: major shareholders in cardiovascular disease enterprises: part I: aging arteries: a “set up” for vascular disease. Circulation. 2003;107(1):139–46.PubMedCrossRef Lakatta EG, Levy D. Arterial and cardiac aging: major shareholders in cardiovascular disease enterprises: part I: aging arteries: a “set up” for vascular disease. Circulation. 2003;107(1):139–46.PubMedCrossRef
105.
Zurück zum Zitat Wagenseil JE, Mecham RP. Elastin in large artery stiffness and hypertension. J cardiovasc Trans Res. 2012;5(3):264–73.CrossRef Wagenseil JE, Mecham RP. Elastin in large artery stiffness and hypertension. J cardiovasc Trans Res. 2012;5(3):264–73.CrossRef
106.
Zurück zum Zitat Webb DR, Khunti K, Silverman R, Gray LJ, Srinivasan B, Lacy PS, Williams B, Davies MJ. Impact of metabolic indices on central artery stiffness: independent association of insulin resistance and glucose with aortic pulse wave velocity. Diabetologia. 2010;53(6):1190–8.PubMedCrossRef Webb DR, Khunti K, Silverman R, Gray LJ, Srinivasan B, Lacy PS, Williams B, Davies MJ. Impact of metabolic indices on central artery stiffness: independent association of insulin resistance and glucose with aortic pulse wave velocity. Diabetologia. 2010;53(6):1190–8.PubMedCrossRef
107.
Zurück zum Zitat Park JS, Nam JS, Cho MH, Yoo JS, Ahn CW, Jee SH, Lee HS, Cha BS, Kim KR, Lee HC. Insulin resistance independently influences arterial stiffness in normoglycemic normotensive postmenopausal women. Menopause. 2010;17(4):779–84.PubMed Park JS, Nam JS, Cho MH, Yoo JS, Ahn CW, Jee SH, Lee HS, Cha BS, Kim KR, Lee HC. Insulin resistance independently influences arterial stiffness in normoglycemic normotensive postmenopausal women. Menopause. 2010;17(4):779–84.PubMed
108.
Zurück zum Zitat Aroor AA, DeMarco VG, Jia G, Sun Z, Nistala R, Meininger GA, Sowers JR. The role of tissue renin-angiotensin-aldosterone system in the development of endothelial dysfunction and arterial stiffness. Front Endocrinol. 2013;4:161.CrossRef Aroor AA, DeMarco VG, Jia G, Sun Z, Nistala R, Meininger GA, Sowers JR. The role of tissue renin-angiotensin-aldosterone system in the development of endothelial dysfunction and arterial stiffness. Front Endocrinol. 2013;4:161.CrossRef
109.
Zurück zum Zitat Leopold JA. Cellular and molecular mechanisms of arterial stiffness associated with obesity. Hypertension. 2013;62(6):1003–4.PubMedCrossRef Leopold JA. Cellular and molecular mechanisms of arterial stiffness associated with obesity. Hypertension. 2013;62(6):1003–4.PubMedCrossRef
110.
Zurück zum Zitat Aroor AR, Jia G, Sowers JR. Cellular mechanisms underlying obesity-induced arterial stiffness. Am J Physiol Regul Integr Comp Physiol. 2018;314:R387–98.PubMedCrossRef Aroor AR, Jia G, Sowers JR. Cellular mechanisms underlying obesity-induced arterial stiffness. Am J Physiol Regul Integr Comp Physiol. 2018;314:R387–98.PubMedCrossRef
111.
Zurück zum Zitat DeMarco VG, Habibi J, Jia G, Aroor AR, Ramirez-Perez FI, Martinez-Lemus LA, Bender SB, Garro M, Hayden MR, Sun Z, et al. Low-dose mineralocorticoid receptor blockade prevents western diet-induced arterial stiffening in female mice. Hypertension. 2015;66:99–107.PubMedCrossRef DeMarco VG, Habibi J, Jia G, Aroor AR, Ramirez-Perez FI, Martinez-Lemus LA, Bender SB, Garro M, Hayden MR, Sun Z, et al. Low-dose mineralocorticoid receptor blockade prevents western diet-induced arterial stiffening in female mice. Hypertension. 2015;66:99–107.PubMedCrossRef
112.
Zurück zum Zitat Aroor AR, Jia G, Habibi J, Sun Z, Ramirez-Perez FI, Brady B, Chen D, Martinez-Lemus LA, Manrique C, Nistala R, et al. Uric acid promotes vascular stiffness, maladaptive inflammatory responses and proteinuria in western diet fed mice. Metabolism. 2017;74:32–40.PubMedCrossRefPubMedCentral Aroor AR, Jia G, Habibi J, Sun Z, Ramirez-Perez FI, Brady B, Chen D, Martinez-Lemus LA, Manrique C, Nistala R, et al. Uric acid promotes vascular stiffness, maladaptive inflammatory responses and proteinuria in western diet fed mice. Metabolism. 2017;74:32–40.PubMedCrossRefPubMedCentral
113.
Zurück zum Zitat Potenza MA, Nacci C, De Salvia MA, Sgarra L, Collino M, Montagnani M. Targeting endothelial metaflammation to counteract diabesity cardiovascular risk: current and perspective therapeutic options. Pharmacol Res. 2017;120:226–41.PubMedCrossRef Potenza MA, Nacci C, De Salvia MA, Sgarra L, Collino M, Montagnani M. Targeting endothelial metaflammation to counteract diabesity cardiovascular risk: current and perspective therapeutic options. Pharmacol Res. 2017;120:226–41.PubMedCrossRef
114.
Zurück zum Zitat Roberts AC, Porter KE. Cellular and molecular mechanisms of endothelial dysfunction in diabetes. Diab Vasc Dis Res. 2013;10(6):472–82.PubMedCrossRef Roberts AC, Porter KE. Cellular and molecular mechanisms of endothelial dysfunction in diabetes. Diab Vasc Dis Res. 2013;10(6):472–82.PubMedCrossRef
115.
Zurück zum Zitat Jamwal S, Sharma S. Vascular endothelium dysfunction: a conservative target in metabolic disorders. Inflamm Res. 2018;67:395–401.CrossRef Jamwal S, Sharma S. Vascular endothelium dysfunction: a conservative target in metabolic disorders. Inflamm Res. 2018;67:395–401.CrossRef
116.
Zurück zum Zitat Hansen NW, Hansen AJ, Sams A. The endothelial border to health: mechanistic evidence of the hyperglycemic culprit of inflammatory disease acceleration. IUBMB Life. 2017;69(3):148–61.PubMedCrossRef Hansen NW, Hansen AJ, Sams A. The endothelial border to health: mechanistic evidence of the hyperglycemic culprit of inflammatory disease acceleration. IUBMB Life. 2017;69(3):148–61.PubMedCrossRef
117.
Zurück zum Zitat Dell’Oro R, Maloberti A, Nicoli F, Villa P, Gamba P, Bombelli M, Mancia G, Grassi G. Long-term saxagliptin treatment improves endothelial function but not pulse wave velocity and intima-media thickness in type 2 diabetic patients. High Blood Press Cardiovasc Prev. 2017;24(4):393–400.PubMedCrossRef Dell’Oro R, Maloberti A, Nicoli F, Villa P, Gamba P, Bombelli M, Mancia G, Grassi G. Long-term saxagliptin treatment improves endothelial function but not pulse wave velocity and intima-media thickness in type 2 diabetic patients. High Blood Press Cardiovasc Prev. 2017;24(4):393–400.PubMedCrossRef
118.
Zurück zum Zitat Kroller-Schon S, Knorr M, Hausding M, Oelze M, Schuff A, Schell R, Sudowe S, Scholz A, Daub S, Karbach S, et al. Glucose-independent improvement of vascular dysfunction in experimental sepsis by dipeptidyl-peptidase 4 inhibition. Cardiovasc Res. 2012;96(1):140–9.PubMedCrossRef Kroller-Schon S, Knorr M, Hausding M, Oelze M, Schuff A, Schell R, Sudowe S, Scholz A, Daub S, Karbach S, et al. Glucose-independent improvement of vascular dysfunction in experimental sepsis by dipeptidyl-peptidase 4 inhibition. Cardiovasc Res. 2012;96(1):140–9.PubMedCrossRef
119.
Zurück zum Zitat Takai S, Sakonjo H, Jin D. Significance of vascular dipeptidyl peptidase-4 inhibition on vascular protection in Zucker diabetic fatty rats. J Pharmacol Sci. 2014;125(4):386–93.PubMedCrossRef Takai S, Sakonjo H, Jin D. Significance of vascular dipeptidyl peptidase-4 inhibition on vascular protection in Zucker diabetic fatty rats. J Pharmacol Sci. 2014;125(4):386–93.PubMedCrossRef
120.
Zurück zum Zitat Thomas MC, Cooper ME, Zimmet P. Changing epidemiology of type 2 diabetes mellitus and associated chronic kidney disease. Nat Rev Nephrol. 2016;12(2):73–81.PubMedCrossRef Thomas MC, Cooper ME, Zimmet P. Changing epidemiology of type 2 diabetes mellitus and associated chronic kidney disease. Nat Rev Nephrol. 2016;12(2):73–81.PubMedCrossRef
121.
Zurück zum Zitat Kanasaki K. The role of renal dipeptidyl peptidase-4 in kidney disease: renal effects of dipeptidyl peptidase-4 inhibitors with a focus on linagliptin. Clin Sci (Lond). 2018;132(4):489–507.CrossRef Kanasaki K. The role of renal dipeptidyl peptidase-4 in kidney disease: renal effects of dipeptidyl peptidase-4 inhibitors with a focus on linagliptin. Clin Sci (Lond). 2018;132(4):489–507.CrossRef
122.
Zurück zum Zitat Aroor AR, Jia G, Sowers JR. Cellular mechanisms underlying obesity-induced arterial stiffness. Am J Physiol Regul Integr Comp Physiol. 2018;314(3):R387–98.PubMedCrossRef Aroor AR, Jia G, Sowers JR. Cellular mechanisms underlying obesity-induced arterial stiffness. Am J Physiol Regul Integr Comp Physiol. 2018;314(3):R387–98.PubMedCrossRef
123.
Zurück zum Zitat Liu WJ, Xie SH, Liu YN, Kim W, Jin HY, Park SK, Shao YM, Park TS. Dipeptidyl peptidase IV inhibitor attenuates kidney injury in streptozotocin-induced diabetic rats. J Pharmacol Exp Ther. 2012;340(2):248–55.PubMedCrossRef Liu WJ, Xie SH, Liu YN, Kim W, Jin HY, Park SK, Shao YM, Park TS. Dipeptidyl peptidase IV inhibitor attenuates kidney injury in streptozotocin-induced diabetic rats. J Pharmacol Exp Ther. 2012;340(2):248–55.PubMedCrossRef
124.
Zurück zum Zitat Mega C, de Lemos ET, Vala H, Fernandes R, Oliveira J, Mascarenhas-Melo F, Teixeira F, Reis F. Diabetic nephropathy amelioration by a low-dose sitagliptin in an animal model of type 2 diabetes (Zucker diabetic fatty rat). Exp Diabetes Res. 2011;2011:162092.PubMedPubMedCentralCrossRef Mega C, de Lemos ET, Vala H, Fernandes R, Oliveira J, Mascarenhas-Melo F, Teixeira F, Reis F. Diabetic nephropathy amelioration by a low-dose sitagliptin in an animal model of type 2 diabetes (Zucker diabetic fatty rat). Exp Diabetes Res. 2011;2011:162092.PubMedPubMedCentralCrossRef
125.
Zurück zum Zitat Eun Lee J, Kim JE, Lee MH, Song HK, Ghee JY, Kang YS, Min HS, Kim HW, Cha JJ, Han JY, et al. DA-1229, a dipeptidyl peptidase IV inhibitor, protects against renal injury by preventing podocyte damage in an animal model of progressive renal injury. Lab Invest. 2016;96(5):547–60.PubMedCrossRef Eun Lee J, Kim JE, Lee MH, Song HK, Ghee JY, Kang YS, Min HS, Kim HW, Cha JJ, Han JY, et al. DA-1229, a dipeptidyl peptidase IV inhibitor, protects against renal injury by preventing podocyte damage in an animal model of progressive renal injury. Lab Invest. 2016;96(5):547–60.PubMedCrossRef
126.
Zurück zum Zitat Sharkovska Y, Reichetzeder C, Alter M, Tsuprykov O, Bachmann S, Secher T, Klein T, Hocher B. Blood pressure and glucose independent renoprotective effects of dipeptidyl peptidase-4 inhibition in a mouse model of type-2 diabetic nephropathy. J Hypertens. 2014;32(11):2211–23 (discussion 2223).PubMedCrossRef Sharkovska Y, Reichetzeder C, Alter M, Tsuprykov O, Bachmann S, Secher T, Klein T, Hocher B. Blood pressure and glucose independent renoprotective effects of dipeptidyl peptidase-4 inhibition in a mouse model of type-2 diabetic nephropathy. J Hypertens. 2014;32(11):2211–23 (discussion 2223).PubMedCrossRef
127.
Zurück zum Zitat Kanasaki K, Shi S, Kanasaki M, He J, Nagai T, Nakamura Y, Ishigaki Y, Kitada M, Srivastava SP, Koya D. Linagliptin-mediated DPP-4 inhibition ameliorates kidney fibrosis in streptozotocin-induced diabetic mice by inhibiting endothelial-to-mesenchymal transition in a therapeutic regimen. Diabetes. 2014;63(6):2120–31.PubMedCrossRef Kanasaki K, Shi S, Kanasaki M, He J, Nagai T, Nakamura Y, Ishigaki Y, Kitada M, Srivastava SP, Koya D. Linagliptin-mediated DPP-4 inhibition ameliorates kidney fibrosis in streptozotocin-induced diabetic mice by inhibiting endothelial-to-mesenchymal transition in a therapeutic regimen. Diabetes. 2014;63(6):2120–31.PubMedCrossRef
128.
Zurück zum Zitat Tsuprykov O, Ando R, Reichetzeder C, von Websky K, Antonenko V, Sharkovska Y, Chaykovska L, Rahnenfuhrer J, Hasan AA, Tammen H, et al. The dipeptidyl peptidase inhibitor linagliptin and the angiotensin II receptor blocker telmisartan show renal benefit by different pathways in rats with 5/6 nephrectomy. Kidney Int. 2016;89(5):1049–61.PubMedCrossRef Tsuprykov O, Ando R, Reichetzeder C, von Websky K, Antonenko V, Sharkovska Y, Chaykovska L, Rahnenfuhrer J, Hasan AA, Tammen H, et al. The dipeptidyl peptidase inhibitor linagliptin and the angiotensin II receptor blocker telmisartan show renal benefit by different pathways in rats with 5/6 nephrectomy. Kidney Int. 2016;89(5):1049–61.PubMedCrossRef
129.
Zurück zum Zitat Coppolino G, Leporini C, Rivoli L, Ursini F, di Paola ED, Cernaro V, Arturi F, Bolignano D, Russo E, De Sarro G, et al. Exploring the effects of DPP-4 inhibitors on the kidney from the bench to clinical trials. Pharmacol Res. 2018;129:274–94.PubMedCrossRef Coppolino G, Leporini C, Rivoli L, Ursini F, di Paola ED, Cernaro V, Arturi F, Bolignano D, Russo E, De Sarro G, et al. Exploring the effects of DPP-4 inhibitors on the kidney from the bench to clinical trials. Pharmacol Res. 2018;129:274–94.PubMedCrossRef
130.
Zurück zum Zitat Rosenstock J, Perkovic V, Alexander JH, Cooper ME, Marx N, Pencina MJ, Toto RD, Wanner C, Zinman B, Baanstra D, et al. Rationale, design, and baseline characteristics of the CArdiovascular safety and Renal Microvascular outcomE study with LINAgliptin (CARMELINA((R))): a randomized, double-blind, placebo-controlled clinical trial in patients with type 2 diabetes and high cardio-renal risk. Cardiovasc Diabetol. 2018;17(1):39.PubMedPubMedCentralCrossRef Rosenstock J, Perkovic V, Alexander JH, Cooper ME, Marx N, Pencina MJ, Toto RD, Wanner C, Zinman B, Baanstra D, et al. Rationale, design, and baseline characteristics of the CArdiovascular safety and Renal Microvascular outcomE study with LINAgliptin (CARMELINA((R))): a randomized, double-blind, placebo-controlled clinical trial in patients with type 2 diabetes and high cardio-renal risk. Cardiovasc Diabetol. 2018;17(1):39.PubMedPubMedCentralCrossRef
131.
Zurück zum Zitat Blech S, Ludwig-Schwellinger E, Grafe-Mody EU, Withopf B, Wagner K. The metabolism and disposition of the oral dipeptidyl peptidase-4 inhibitor, linagliptin, in humans. Drug Metab Dispos. 2010;38(4):667–78.PubMedCrossRef Blech S, Ludwig-Schwellinger E, Grafe-Mody EU, Withopf B, Wagner K. The metabolism and disposition of the oral dipeptidyl peptidase-4 inhibitor, linagliptin, in humans. Drug Metab Dispos. 2010;38(4):667–78.PubMedCrossRef
132.
Zurück zum Zitat Golightly LK, Drayna CC, McDermott MT. Comparative clinical pharmacokinetics of dipeptidyl peptidase-4 inhibitors. Clin Pharmacokinet. 2012;51(8):501–14.PubMedCrossRef Golightly LK, Drayna CC, McDermott MT. Comparative clinical pharmacokinetics of dipeptidyl peptidase-4 inhibitors. Clin Pharmacokinet. 2012;51(8):501–14.PubMedCrossRef
133.
Zurück zum Zitat Graefe-Mody U, Friedrich C, Port A, Ring A, Retlich S, Heise T, Halabi A, Woerle HJ. Effect of renal impairment on the pharmacokinetics of the dipeptidyl peptidase-4 inhibitor linagliptin(*). Diabetes Obes Metab. 2011;13(10):939–46.PubMedCrossRef Graefe-Mody U, Friedrich C, Port A, Ring A, Retlich S, Heise T, Halabi A, Woerle HJ. Effect of renal impairment on the pharmacokinetics of the dipeptidyl peptidase-4 inhibitor linagliptin(*). Diabetes Obes Metab. 2011;13(10):939–46.PubMedCrossRef
134.
Zurück zum Zitat Fuchs H, Binder R, Greischel A. Tissue distribution of the novel DPP-4 inhibitor BI 1356 is dominated by saturable binding to its target in rats. Biopharm Drug Dispos. 2009;30(5):229–40.PubMedCrossRef Fuchs H, Binder R, Greischel A. Tissue distribution of the novel DPP-4 inhibitor BI 1356 is dominated by saturable binding to its target in rats. Biopharm Drug Dispos. 2009;30(5):229–40.PubMedCrossRef
135.
Zurück zum Zitat Greischel A, Binder R, Baierl J. The dipeptidyl peptidase-4 inhibitor linagliptin exhibits time- and dose-dependent localization in kidney, liver, and intestine after intravenous dosing: results from high resolution autoradiography in rats. Drug Metab Dispos. 2010;38(9):1443–8.PubMedCrossRef Greischel A, Binder R, Baierl J. The dipeptidyl peptidase-4 inhibitor linagliptin exhibits time- and dose-dependent localization in kidney, liver, and intestine after intravenous dosing: results from high resolution autoradiography in rats. Drug Metab Dispos. 2010;38(9):1443–8.PubMedCrossRef
136.
Zurück zum Zitat Schnapp G, Klein T, Hoevels Y, Bakker RA, Nar H. Comparative analysis of binding kinetics and thermodynamics of dipeptidyl peptidase-4 inhibitors and their relationship to structure. J Med Chem. 2016;59(16):7466–77.PubMedCrossRef Schnapp G, Klein T, Hoevels Y, Bakker RA, Nar H. Comparative analysis of binding kinetics and thermodynamics of dipeptidyl peptidase-4 inhibitors and their relationship to structure. J Med Chem. 2016;59(16):7466–77.PubMedCrossRef
137.
Zurück zum Zitat Davis TM. Dipeptidyl peptidase-4 inhibitors: pharmacokinetics, efficacy, tolerability and safety in renal impairment. Diabetes Obes Metab. 2014;16(10):891–9.PubMedCrossRef Davis TM. Dipeptidyl peptidase-4 inhibitors: pharmacokinetics, efficacy, tolerability and safety in renal impairment. Diabetes Obes Metab. 2014;16(10):891–9.PubMedCrossRef
138.
Zurück zum Zitat Fong DS, Aiello LP, Ferris FL 3rd, Klein R. Diabetic retinopathy. Diabetes Care. 2004;27(10):2540–53.PubMedCrossRef Fong DS, Aiello LP, Ferris FL 3rd, Klein R. Diabetic retinopathy. Diabetes Care. 2004;27(10):2540–53.PubMedCrossRef
139.
Zurück zum Zitat Nistala R, Savin V. Diabetes, hypertension, and chronic kidney disease progression: role of DPP4. Am J Physiol Renal Physiol. 2017;312(4):F661–70.PubMedCrossRef Nistala R, Savin V. Diabetes, hypertension, and chronic kidney disease progression: role of DPP4. Am J Physiol Renal Physiol. 2017;312(4):F661–70.PubMedCrossRef
140.
Zurück zum Zitat Aroor A, McKarns S, Nistala R, Demarco V, Gardner M, Garcia-Touza M, Whaley-Connell A, Sowers JR. DPP-4 inhibitors as therapeutic modulators of immune cell function and associated cardiovascular and renal insulin resistance in obesity and diabetes. Cardiorenal Med. 2013;3(1):48–56.PubMedPubMedCentralCrossRef Aroor A, McKarns S, Nistala R, Demarco V, Gardner M, Garcia-Touza M, Whaley-Connell A, Sowers JR. DPP-4 inhibitors as therapeutic modulators of immune cell function and associated cardiovascular and renal insulin resistance in obesity and diabetes. Cardiorenal Med. 2013;3(1):48–56.PubMedPubMedCentralCrossRef
142.
Zurück zum Zitat Sokos GG, Nikolaidis LA, Mankad S, Elahi D, Shannon RP. Glucagon-like peptide-1 infusion improves left ventricular ejection fraction and functional status in patients with chronic heart failure. J Card Fail. 2006;12(9):694–9.PubMedCrossRef Sokos GG, Nikolaidis LA, Mankad S, Elahi D, Shannon RP. Glucagon-like peptide-1 infusion improves left ventricular ejection fraction and functional status in patients with chronic heart failure. J Card Fail. 2006;12(9):694–9.PubMedCrossRef
143.
Zurück zum Zitat Zhao T, Parikh P, Bhashyam S, Bolukoglu H, Poornima I, Shen YT, Shannon RP. Direct effects of glucagon-like peptide-1 on myocardial contractility and glucose uptake in normal and postischemic isolated rat hearts. J Pharmacol Exp Ther. 2006;317(3):1106–13.PubMedCrossRef Zhao T, Parikh P, Bhashyam S, Bolukoglu H, Poornima I, Shen YT, Shannon RP. Direct effects of glucagon-like peptide-1 on myocardial contractility and glucose uptake in normal and postischemic isolated rat hearts. J Pharmacol Exp Ther. 2006;317(3):1106–13.PubMedCrossRef
144.
Zurück zum Zitat Ravassa S, Zudaire A, Carr RD, Diez J. Antiapoptotic effects of Glp-1 in murine Hl-1 cardiomyocytes. Am J Physiol Heart Circ Physiol. 2011;300:1361–72.CrossRef Ravassa S, Zudaire A, Carr RD, Diez J. Antiapoptotic effects of Glp-1 in murine Hl-1 cardiomyocytes. Am J Physiol Heart Circ Physiol. 2011;300:1361–72.CrossRef
145.
Zurück zum Zitat Bose AK, Mocanu MM, Carr RD, Yellon DM. Myocardial ischaemia-reperfusion injury is attenuated by intact glucagon like peptide-1 (GLP-1) in the in vitro rat heart and may involve the p70s6K pathway. Cardiovasc Drugs Ther. 2007;21(4):253–6.PubMedCrossRef Bose AK, Mocanu MM, Carr RD, Yellon DM. Myocardial ischaemia-reperfusion injury is attenuated by intact glucagon like peptide-1 (GLP-1) in the in vitro rat heart and may involve the p70s6K pathway. Cardiovasc Drugs Ther. 2007;21(4):253–6.PubMedCrossRef
146.
Zurück zum Zitat Okerson T, Chilton RJ. The cardiovascular effects of GLP-1 receptor agonists. Cardiovasc Ther. 2012;30(3):e146–55.PubMedCrossRef Okerson T, Chilton RJ. The cardiovascular effects of GLP-1 receptor agonists. Cardiovasc Ther. 2012;30(3):e146–55.PubMedCrossRef
147.
Zurück zum Zitat Drucker DJ. The cardiovascular biology of glucagon-like peptide-1. Cell Metab. 2016;24(1):15–30.PubMedCrossRef Drucker DJ. The cardiovascular biology of glucagon-like peptide-1. Cell Metab. 2016;24(1):15–30.PubMedCrossRef
148.
Zurück zum Zitat Jia G, Aroor AR, Sowers JR. Glucagon-like peptide 1 receptor activation and platelet function: beyond glycemic control. Diabetes. 2016;65(6):1487–9.PubMedPubMedCentralCrossRef Jia G, Aroor AR, Sowers JR. Glucagon-like peptide 1 receptor activation and platelet function: beyond glycemic control. Diabetes. 2016;65(6):1487–9.PubMedPubMedCentralCrossRef
149.
Zurück zum Zitat Ryan MJ. An update on immune system activation in the pathogenesis of hypertension. Hypertension. 2013;62(2):226–30.PubMedCrossRef Ryan MJ. An update on immune system activation in the pathogenesis of hypertension. Hypertension. 2013;62(2):226–30.PubMedCrossRef
150.
Zurück zum Zitat Schiffrin EL. Immune mechanisms in hypertension and vascular injury. Clin Sci (Lond). 2014;126(4):267–74.CrossRef Schiffrin EL. Immune mechanisms in hypertension and vascular injury. Clin Sci (Lond). 2014;126(4):267–74.CrossRef
152.
Zurück zum Zitat Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest. 2007;117(1):175–84.PubMedPubMedCentralCrossRef Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest. 2007;117(1):175–84.PubMedPubMedCentralCrossRef
154.
155.
Zurück zum Zitat Kalupahana NS, Moustaid-Moussa N, Claycombe KJ. Immunity as a link between obesity and insulin resistance. Mol Aspects Med. 2012;33(1):26–34.PubMedCrossRef Kalupahana NS, Moustaid-Moussa N, Claycombe KJ. Immunity as a link between obesity and insulin resistance. Mol Aspects Med. 2012;33(1):26–34.PubMedCrossRef
156.
Zurück zum Zitat Aroor AR, McKarns S, Demarco VG, Jia G, Sowers JR. Maladaptive immune and inflammatory pathways lead to cardiovascular insulin resistance. Metabolism. 2013;62:1543–52.PubMedCrossRef Aroor AR, McKarns S, Demarco VG, Jia G, Sowers JR. Maladaptive immune and inflammatory pathways lead to cardiovascular insulin resistance. Metabolism. 2013;62:1543–52.PubMedCrossRef
157.
Zurück zum Zitat Kassan M, Galan M, Partyka M, Trebak M, Matrougui K. Interleukin-10 released by CD4(+) CD25(+) natural regulatory T cells improves microvascular endothelial function through inhibition of NADPH oxidase activity in hypertensive mice. Arterioscler Thromb Vasc Biol. 2011;31(11):2534–42.PubMedPubMedCentralCrossRef Kassan M, Galan M, Partyka M, Trebak M, Matrougui K. Interleukin-10 released by CD4(+) CD25(+) natural regulatory T cells improves microvascular endothelial function through inhibition of NADPH oxidase activity in hypertensive mice. Arterioscler Thromb Vasc Biol. 2011;31(11):2534–42.PubMedPubMedCentralCrossRef
158.
Zurück zum Zitat Ohshima K, Mogi M, Jing F, Iwanami J, Tsukuda K, Min LJ, Higaki J, Horiuchi M. Roles of interleukin 17 in angiotensin II type 1 receptor-mediated insulin resistance. Hypertension. 2012;59(2):493–9.PubMedCrossRef Ohshima K, Mogi M, Jing F, Iwanami J, Tsukuda K, Min LJ, Higaki J, Horiuchi M. Roles of interleukin 17 in angiotensin II type 1 receptor-mediated insulin resistance. Hypertension. 2012;59(2):493–9.PubMedCrossRef
159.
Zurück zum Zitat Yaron A, Naider F. Proline-dependent structural and biological properties of peptides and proteins. Crit Rev Biochem Mol Biol. 1993;28(1):31–81.PubMedCrossRef Yaron A, Naider F. Proline-dependent structural and biological properties of peptides and proteins. Crit Rev Biochem Mol Biol. 1993;28(1):31–81.PubMedCrossRef
160.
Zurück zum Zitat Zhong J, Rao X, Rajagopalan S. An emerging role of dipeptidyl peptidase 4 (DPP4) beyond glucose control: potential implications in cardiovascular disease. Atherosclerosis. 2013;226(2):305–14.PubMedCrossRef Zhong J, Rao X, Rajagopalan S. An emerging role of dipeptidyl peptidase 4 (DPP4) beyond glucose control: potential implications in cardiovascular disease. Atherosclerosis. 2013;226(2):305–14.PubMedCrossRef
161.
Zurück zum Zitat Drucker DJ. Dipeptidyl peptidase-4 inhibition and the treatment of type 2 diabetes: preclinical biology and mechanisms of action. Diabetes Care. 2007;30(6):1335–43.PubMedCrossRef Drucker DJ. Dipeptidyl peptidase-4 inhibition and the treatment of type 2 diabetes: preclinical biology and mechanisms of action. Diabetes Care. 2007;30(6):1335–43.PubMedCrossRef
162.
Zurück zum Zitat Muskiet MH, Smits MM, Morsink LM, Diamant M. The gut-renal axis: do incretin-based agents confer renoprotection in diabetes? Nat Rev Nephrol. 2014;10(2):88–103.PubMedCrossRef Muskiet MH, Smits MM, Morsink LM, Diamant M. The gut-renal axis: do incretin-based agents confer renoprotection in diabetes? Nat Rev Nephrol. 2014;10(2):88–103.PubMedCrossRef
163.
Zurück zum Zitat Yazbeck R, Howarth GS, Abbott CA. Dipeptidyl peptidase inhibitors, an emerging drug class for inflammatory disease? Trends Pharmacol Sci. 2009;30(11):600–7.PubMedCrossRef Yazbeck R, Howarth GS, Abbott CA. Dipeptidyl peptidase inhibitors, an emerging drug class for inflammatory disease? Trends Pharmacol Sci. 2009;30(11):600–7.PubMedCrossRef
164.
Zurück zum Zitat Shirakawa J, Fujii H, Ohnuma K, Sato K, Ito Y, Kaji M, Sakamoto E, Koganei M, Sasaki H, Nagashima Y, et al. Diet-induced adipose tissue inflammation and liver steatosis are prevented by DPP-4 inhibition in diabetic mice. Diabetes. 2011;60(4):1246–57.PubMedPubMedCentralCrossRef Shirakawa J, Fujii H, Ohnuma K, Sato K, Ito Y, Kaji M, Sakamoto E, Koganei M, Sasaki H, Nagashima Y, et al. Diet-induced adipose tissue inflammation and liver steatosis are prevented by DPP-4 inhibition in diabetic mice. Diabetes. 2011;60(4):1246–57.PubMedPubMedCentralCrossRef
165.
Zurück zum Zitat Shah Z, Kampfrath T, Deiuliis JA, Zhong J, Pineda C, Ying Z, Xu X, Lu B, Moffatt-Bruce S, Durairaj R, et al. Long-term dipeptidyl-peptidase 4 inhibition reduces atherosclerosis and inflammation via effects on monocyte recruitment and chemotaxis. Circulation. 2011;124:2338–49.PubMedPubMedCentralCrossRef Shah Z, Kampfrath T, Deiuliis JA, Zhong J, Pineda C, Ying Z, Xu X, Lu B, Moffatt-Bruce S, Durairaj R, et al. Long-term dipeptidyl-peptidase 4 inhibition reduces atherosclerosis and inflammation via effects on monocyte recruitment and chemotaxis. Circulation. 2011;124:2338–49.PubMedPubMedCentralCrossRef
166.
Zurück zum Zitat Ervinna N, Mita T, Yasunari E, Azuma K, Tanaka R, Fujimura S, Sukmawati D, Nomiyama T, Kanazawa A, Kawamori R, et al. Anagliptin, a DPP-4 inhibitor, suppresses proliferation of vascular smooth muscles and monocyte inflammatory reaction and attenuates atherosclerosis in male apo E-deficient mice. Endocrinology. 2013;154(3):1260–70.PubMedCrossRef Ervinna N, Mita T, Yasunari E, Azuma K, Tanaka R, Fujimura S, Sukmawati D, Nomiyama T, Kanazawa A, Kawamori R, et al. Anagliptin, a DPP-4 inhibitor, suppresses proliferation of vascular smooth muscles and monocyte inflammatory reaction and attenuates atherosclerosis in male apo E-deficient mice. Endocrinology. 2013;154(3):1260–70.PubMedCrossRef
167.
Zurück zum Zitat Ta NN, Schuyler CA, Li Y, Lopes-Virella MF, Huang Y. DPP-4 (CD26) inhibitor alogliptin inhibits atherosclerosis in diabetic apolipoprotein E-deficient mice. J Cardiovasc Pharmacol. 2011;58(2):157–66.PubMedPubMedCentralCrossRef Ta NN, Schuyler CA, Li Y, Lopes-Virella MF, Huang Y. DPP-4 (CD26) inhibitor alogliptin inhibits atherosclerosis in diabetic apolipoprotein E-deficient mice. J Cardiovasc Pharmacol. 2011;58(2):157–66.PubMedPubMedCentralCrossRef
168.
Zurück zum Zitat Zhong J, Rao X, Deiuliis J, Braunstein Z, Narula V, Hazey J, Mikami D, Needleman B, Satoskar AR, Rajagopalan S. A potential role for dendritic cell/macrophage-expressing DPP4 in obesity-induced visceral inflammation. Diabetes. 2013;62(1):149–57.PubMedCrossRef Zhong J, Rao X, Deiuliis J, Braunstein Z, Narula V, Hazey J, Mikami D, Needleman B, Satoskar AR, Rajagopalan S. A potential role for dendritic cell/macrophage-expressing DPP4 in obesity-induced visceral inflammation. Diabetes. 2013;62(1):149–57.PubMedCrossRef
169.
Zurück zum Zitat Lamers D, Famulla S, Wronkowitz N, Hartwig S, Lehr S, Ouwens DM, Eckardt K, Kaufman JM, Ryden M, Muller S, et al. Dipeptidyl peptidase 4 is a novel adipokine potentially linking obesity to the metabolic syndrome. Diabetes. 2011;60(7):1917–25.PubMedPubMedCentralCrossRef Lamers D, Famulla S, Wronkowitz N, Hartwig S, Lehr S, Ouwens DM, Eckardt K, Kaufman JM, Ryden M, Muller S, et al. Dipeptidyl peptidase 4 is a novel adipokine potentially linking obesity to the metabolic syndrome. Diabetes. 2011;60(7):1917–25.PubMedPubMedCentralCrossRef
170.
Zurück zum Zitat Sell H, Bluher M, Kloting N, Schlich R, Willems M, Ruppe F, Knoefel WT, Dietrich A, Fielding BA, Arner P, et al. Adipose dipeptidyl peptidase-4 and obesity: correlation with insulin resistance and depot-specific release from adipose tissue in vivo and in vitro. Diabetes Care. 2013;36(12):4083–90.PubMedPubMedCentralCrossRef Sell H, Bluher M, Kloting N, Schlich R, Willems M, Ruppe F, Knoefel WT, Dietrich A, Fielding BA, Arner P, et al. Adipose dipeptidyl peptidase-4 and obesity: correlation with insulin resistance and depot-specific release from adipose tissue in vivo and in vitro. Diabetes Care. 2013;36(12):4083–90.PubMedPubMedCentralCrossRef
171.
Zurück zum Zitat Zhong J, Gong Q, Goud A, Srinivasamaharaj S, Rajagopalan S. Recent advances in dipeptidyl-peptidase-4 inhibition therapy: lessons from the bench and clinical trials. J Diabetes Res. 2015;2015:606031.PubMedPubMedCentralCrossRef Zhong J, Gong Q, Goud A, Srinivasamaharaj S, Rajagopalan S. Recent advances in dipeptidyl-peptidase-4 inhibition therapy: lessons from the bench and clinical trials. J Diabetes Res. 2015;2015:606031.PubMedPubMedCentralCrossRef
172.
Zurück zum Zitat Guzik TJ, Hoch NE, Brown KA, McCann LA, Rahman A, Dikalov S, Goronzy J, Weyand C, Harrison DG. Role of the T cell in the genesis of angiotensin II induced hypertension and vascular dysfunction. J Exp Med. 2007;204(10):2449–60.PubMedPubMedCentralCrossRef Guzik TJ, Hoch NE, Brown KA, McCann LA, Rahman A, Dikalov S, Goronzy J, Weyand C, Harrison DG. Role of the T cell in the genesis of angiotensin II induced hypertension and vascular dysfunction. J Exp Med. 2007;204(10):2449–60.PubMedPubMedCentralCrossRef
173.
Zurück zum Zitat Kasal DA, Barhoumi T, Li MW, Yamamoto N, Zdanovich E, Rehman A, Neves MF, Laurant P, Paradis P, Schiffrin EL. T regulatory lymphocytes prevent aldosterone-induced vascular injury. Hypertension. 2012;59(2):324–30.PubMedCrossRef Kasal DA, Barhoumi T, Li MW, Yamamoto N, Zdanovich E, Rehman A, Neves MF, Laurant P, Paradis P, Schiffrin EL. T regulatory lymphocytes prevent aldosterone-induced vascular injury. Hypertension. 2012;59(2):324–30.PubMedCrossRef
174.
Zurück zum Zitat Zhang LH, Pang XF, Bai F, Wang NP, Shah AI, McKallip RJ, Li XW, Wang X, Zhao ZQ. Preservation of glucagon-like peptide-1 level attenuates angiotensin ii-induced tissue fibrosis by altering AT1/AT 2 receptor expression and angiotensin-converting enzyme 2 activity in rat heart. Cardiovasc Drugs Ther. 2015;29(3):243–55.PubMedCrossRef Zhang LH, Pang XF, Bai F, Wang NP, Shah AI, McKallip RJ, Li XW, Wang X, Zhao ZQ. Preservation of glucagon-like peptide-1 level attenuates angiotensin ii-induced tissue fibrosis by altering AT1/AT 2 receptor expression and angiotensin-converting enzyme 2 activity in rat heart. Cardiovasc Drugs Ther. 2015;29(3):243–55.PubMedCrossRef
175.
Zurück zum Zitat Lundberg JO, Gladwin MT, Weitzberg E. Strategies to increase nitric oxide signalling in cardiovascular disease. Nat Rev Drug Discovery. 2015;14(9):623–41.PubMedCrossRef Lundberg JO, Gladwin MT, Weitzberg E. Strategies to increase nitric oxide signalling in cardiovascular disease. Nat Rev Drug Discovery. 2015;14(9):623–41.PubMedCrossRef
176.
Zurück zum Zitat Forte M, Conti V, Damato A, Ambrosio M, Puca AA, Sciarretta S, Frati G, Vecchione C, Carrizzo A. Targeting nitric oxide with natural derived compounds as a therapeutic strategy in vascular diseases. Oxid Med Cell Longev. 2016;2016:7364138.PubMedPubMedCentralCrossRef Forte M, Conti V, Damato A, Ambrosio M, Puca AA, Sciarretta S, Frati G, Vecchione C, Carrizzo A. Targeting nitric oxide with natural derived compounds as a therapeutic strategy in vascular diseases. Oxid Med Cell Longev. 2016;2016:7364138.PubMedPubMedCentralCrossRef
177.
Zurück zum Zitat Chen JY, Ye ZX, Wang XF, Chang J, Yang MW, Zhong HH, Hong FF, Yang SL. Nitric oxide bioavailability dysfunction involves in atherosclerosis. Biomed Pharmacother Biomed Pharmacother. 2017;97:423–8.PubMedCrossRef Chen JY, Ye ZX, Wang XF, Chang J, Yang MW, Zhong HH, Hong FF, Yang SL. Nitric oxide bioavailability dysfunction involves in atherosclerosis. Biomed Pharmacother Biomed Pharmacother. 2017;97:423–8.PubMedCrossRef
178.
Zurück zum Zitat Shah Z, Pineda C, Kampfrath T, Maiseyeu A, Ying Z, Racoma I, Deiuliis J, Xu X, Sun Q, Moffatt-Bruce S, et al. Acute DPP-4 inhibition modulates vascular tone through GLP-1 independent pathways. Vasc Pharmacol. 2011;55(1–3):2–9.CrossRef Shah Z, Pineda C, Kampfrath T, Maiseyeu A, Ying Z, Racoma I, Deiuliis J, Xu X, Sun Q, Moffatt-Bruce S, et al. Acute DPP-4 inhibition modulates vascular tone through GLP-1 independent pathways. Vasc Pharmacol. 2011;55(1–3):2–9.CrossRef
180.
Zurück zum Zitat Donate-Correa J, Mora-Fernandez C, Martinez-Sanz R, Muros-de-Fuentes M, Perez H, Meneses-Perez B, Cazana-Perez V, Navarro-Gonzalez JF. Expression of FGF23/KLOTHO system in human vascular tissue. Int J Cardiol. 2013;165(1):179–83.PubMedCrossRef Donate-Correa J, Mora-Fernandez C, Martinez-Sanz R, Muros-de-Fuentes M, Perez H, Meneses-Perez B, Cazana-Perez V, Navarro-Gonzalez JF. Expression of FGF23/KLOTHO system in human vascular tissue. Int J Cardiol. 2013;165(1):179–83.PubMedCrossRef
181.
Zurück zum Zitat Lim SC, Liu JJ, Subramaniam T, Sum CF. Elevated circulating alpha-klotho by angiotensin II receptor blocker losartan is associated with reduction of albuminuria in type 2 diabetic patients. J Renin Angiotensin Aldosterone Syst. 2014;15(4):487–90.PubMedCrossRef Lim SC, Liu JJ, Subramaniam T, Sum CF. Elevated circulating alpha-klotho by angiotensin II receptor blocker losartan is associated with reduction of albuminuria in type 2 diabetic patients. J Renin Angiotensin Aldosterone Syst. 2014;15(4):487–90.PubMedCrossRef
182.
Zurück zum Zitat Lim K, Groen A, Molostvov G, Lu T, Lilley KS, Snead D, James S, Wilkinson IB, Ting S, Hsiao LL, et al. Alpha-klotho expression in human tissues. J Clin Endocrinol Metab. 2015;100(10):E1308–18.PubMedPubMedCentralCrossRef Lim K, Groen A, Molostvov G, Lu T, Lilley KS, Snead D, James S, Wilkinson IB, Ting S, Hsiao LL, et al. Alpha-klotho expression in human tissues. J Clin Endocrinol Metab. 2015;100(10):E1308–18.PubMedPubMedCentralCrossRef
183.
Zurück zum Zitat Dalton GD, Xie J, An SW, Huang CL. New insights into the mechanism of action of soluble klotho. Front Endocrinol (Lausanne). 2017;8:323.CrossRef Dalton GD, Xie J, An SW, Huang CL. New insights into the mechanism of action of soluble klotho. Front Endocrinol (Lausanne). 2017;8:323.CrossRef
184.
Zurück zum Zitat Pierce GL. Recent advances: mechanisms and subclinical consequences of aortic stiffness. Hypertension. 2017;70:848–53.PubMedCrossRef Pierce GL. Recent advances: mechanisms and subclinical consequences of aortic stiffness. Hypertension. 2017;70:848–53.PubMedCrossRef
185.
Zurück zum Zitat Xiao NM, Zhang YM, Zheng Q, Gu J. Klotho is a serum factor related to human aging. Chin Med J (Engl). 2004;117(5):742–7. Xiao NM, Zhang YM, Zheng Q, Gu J. Klotho is a serum factor related to human aging. Chin Med J (Engl). 2004;117(5):742–7.
186.
Zurück zum Zitat Chen K, Zhou X, Sun Z. Haplodeficiency of klotho gene causes arterial stiffening via upregulation of scleraxis expression and induction of autophagy. Hypertension. 2015;66(5):1006–13.PubMedCrossRef Chen K, Zhou X, Sun Z. Haplodeficiency of klotho gene causes arterial stiffening via upregulation of scleraxis expression and induction of autophagy. Hypertension. 2015;66(5):1006–13.PubMedCrossRef
187.
Zurück zum Zitat de Borst MH, Vervloet MG, ter Wee PM, Navis G. Cross talk between the renin-angiotensin-aldosterone system and vitamin D-FGF-23-klotho in chronic kidney disease. J Am Soc Nephrol. 2011;22(9):1603–9.PubMedPubMedCentralCrossRef de Borst MH, Vervloet MG, ter Wee PM, Navis G. Cross talk between the renin-angiotensin-aldosterone system and vitamin D-FGF-23-klotho in chronic kidney disease. J Am Soc Nephrol. 2011;22(9):1603–9.PubMedPubMedCentralCrossRef
188.
Zurück zum Zitat Hasegawa Y, Hayashi K, Takemoto Y, Cheng C, Takane K, Lin B, Komohara Y, Kim-Mitsuyama S. DPP-4 inhibition with linagliptin ameliorates the progression of premature aging in klotho-/- mice. Cardiovasc Diabetol. 2017;16(1):154.PubMedPubMedCentralCrossRef Hasegawa Y, Hayashi K, Takemoto Y, Cheng C, Takane K, Lin B, Komohara Y, Kim-Mitsuyama S. DPP-4 inhibition with linagliptin ameliorates the progression of premature aging in klotho-/- mice. Cardiovasc Diabetol. 2017;16(1):154.PubMedPubMedCentralCrossRef
189.
Zurück zum Zitat Erickson JM, Valente AJ, Hemanthkumar K, Raikwar SP, DeMarco VG, Bender SB, Fay WP, Siebenlist U, Chandrasekar B: Targeting TRAF3IP2 by genetic and interventional approaches inhibits ischemia/reperfusion-induced myocardial injury. J Biol Chem. 2017;292:2345–58.CrossRef Erickson JM, Valente AJ, Hemanthkumar K, Raikwar SP, DeMarco VG, Bender SB, Fay WP, Siebenlist U, Chandrasekar B: Targeting TRAF3IP2 by genetic and interventional approaches inhibits ischemia/reperfusion-induced myocardial injury. J Biol Chem. 2017;292:2345–58.CrossRef
190.
Zurück zum Zitat Siddesha JM, Valente AJ, Sakamuri SS, 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. 2013;65:9–18.PubMedPubMedCentralCrossRef Siddesha JM, Valente AJ, Sakamuri SS, 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. 2013;65:9–18.PubMedPubMedCentralCrossRef
191.
Zurück zum Zitat Somanna NK, Yariswamy M, Garagliano JM, Siebenlist U, Mummidi S, Valente AJ, Chandrasekar B. Aldosterone-induced cardiomyocyte growth, and fibroblast migration and proliferation are mediated by TRAF3IP2. Cell Signal. 2015;27(10):1928–38.PubMedCrossRef Somanna NK, Yariswamy M, Garagliano JM, Siebenlist U, Mummidi S, Valente AJ, Chandrasekar B. Aldosterone-induced cardiomyocyte growth, and fibroblast migration and proliferation are mediated by TRAF3IP2. Cell Signal. 2015;27(10):1928–38.PubMedCrossRef
192.
Zurück zum Zitat Zhong J, Rajagopalan S. Dipeptidyl peptidase-4 regulation of SDF-1/CXCR4 axis: implications for cardiovascular disease. Front Immunol. 2015;6:477.PubMedPubMedCentralCrossRef Zhong J, Rajagopalan S. Dipeptidyl peptidase-4 regulation of SDF-1/CXCR4 axis: implications for cardiovascular disease. Front Immunol. 2015;6:477.PubMedPubMedCentralCrossRef
193.
Zurück zum Zitat Chen LH, Advani SL, Thai K, Kabir MG, Sood MM, Gibson IW, Yuen DA, Connelly KA, Marsden PA, Kelly DJ, et al. SDF-1/CXCR4 signaling preserves microvascular integrity and renal function in chronic kidney disease. PLoS ONE. 2014;9(3):e92227.PubMedPubMedCentralCrossRef Chen LH, Advani SL, Thai K, Kabir MG, Sood MM, Gibson IW, Yuen DA, Connelly KA, Marsden PA, Kelly DJ, et al. SDF-1/CXCR4 signaling preserves microvascular integrity and renal function in chronic kidney disease. PLoS ONE. 2014;9(3):e92227.PubMedPubMedCentralCrossRef
194.
Zurück zum Zitat Fadini GP, Losordo D, Dimmeler S. Critical reevaluation of endothelial progenitor cell phenotypes for therapeutic and diagnostic use. Circ Res. 2012;110(4):624–37.PubMedPubMedCentralCrossRef Fadini GP, Losordo D, Dimmeler S. Critical reevaluation of endothelial progenitor cell phenotypes for therapeutic and diagnostic use. Circ Res. 2012;110(4):624–37.PubMedPubMedCentralCrossRef
195.
Zurück zum Zitat Rigato M, Bittante C, Albiero M, Avogaro A, Fadini GP. Circulating progenitor cell count predicts microvascular outcomes in type 2 diabetic patients. J Clin Endocrinol Metab. 2015;100(7):2666–72.PubMedCrossRef Rigato M, Bittante C, Albiero M, Avogaro A, Fadini GP. Circulating progenitor cell count predicts microvascular outcomes in type 2 diabetic patients. J Clin Endocrinol Metab. 2015;100(7):2666–72.PubMedCrossRef
196.
Zurück zum Zitat Patel RS, Li Q, Ghasemzadeh N, Eapen DJ, Moss LD, Janjua AU, Manocha P, Kassem HA, Veledar E, Samady H, et al. Circulating CD34+ progenitor cells and risk of mortality in a population with coronary artery disease. Circ Res. 2015;116(2):289–97.PubMedCrossRef Patel RS, Li Q, Ghasemzadeh N, Eapen DJ, Moss LD, Janjua AU, Manocha P, Kassem HA, Veledar E, Samady H, et al. Circulating CD34+ progenitor cells and risk of mortality in a population with coronary artery disease. Circ Res. 2015;116(2):289–97.PubMedCrossRef
197.
Zurück zum Zitat Negro R, Greco EL, Greco G. Active stromal cell-derived factor 1alpha and endothelial progenitor cells are equally increased by alogliptin in good and poor diabetes control. Clin Med Insights Endocrinol Diabetes. 2017;10:1179551417743980.PubMedPubMedCentralCrossRef Negro R, Greco EL, Greco G. Active stromal cell-derived factor 1alpha and endothelial progenitor cells are equally increased by alogliptin in good and poor diabetes control. Clin Med Insights Endocrinol Diabetes. 2017;10:1179551417743980.PubMedPubMedCentralCrossRef
198.
Zurück zum Zitat Takahashi A, Asakura M, Ito S, Min KD, Shindo K, Yan Y, Liao Y, Yamazaki S, Sanada S, Asano Y, et al. Dipeptidyl-peptidase IV inhibition improves pathophysiology of heart failure and increases survival rate in pressure-overloaded mice. Am J Physiol Heart Circ Physiol. 2013;304(10):H1361–9.PubMedCrossRef Takahashi A, Asakura M, Ito S, Min KD, Shindo K, Yan Y, Liao Y, Yamazaki S, Sanada S, Asano Y, et al. Dipeptidyl-peptidase IV inhibition improves pathophysiology of heart failure and increases survival rate in pressure-overloaded mice. Am J Physiol Heart Circ Physiol. 2013;304(10):H1361–9.PubMedCrossRef
199.
Zurück zum Zitat Anderluh M, Kocic G, Tomovic K, Kocic R, Deljanin-Ilic M, Smelcerovic A. Cross-talk between the dipeptidyl peptidase-4 and stromal cell-derived factor-1 in stem cell homing and myocardial repair: potential impact of dipeptidyl peptidase-4 inhibitors. Pharmacol Ther. 2016;167:100–7.PubMedCrossRef Anderluh M, Kocic G, Tomovic K, Kocic R, Deljanin-Ilic M, Smelcerovic A. Cross-talk between the dipeptidyl peptidase-4 and stromal cell-derived factor-1 in stem cell homing and myocardial repair: potential impact of dipeptidyl peptidase-4 inhibitors. Pharmacol Ther. 2016;167:100–7.PubMedCrossRef
200.
Zurück zum Zitat Packer M. Have dipeptidyl peptidase-4 inhibitors ameliorated the vascular complications of type 2 diabetes in large-scale trials? The potential confounding effect of stem-cell chemokines. Cardiovasc Diabetol. 2018;17(1):9.PubMedPubMedCentralCrossRef Packer M. Have dipeptidyl peptidase-4 inhibitors ameliorated the vascular complications of type 2 diabetes in large-scale trials? The potential confounding effect of stem-cell chemokines. Cardiovasc Diabetol. 2018;17(1):9.PubMedPubMedCentralCrossRef
201.
Zurück zum Zitat Fadini GP, Avogaro A. How to interpret the role of SDF-1alpha on diabetic complications during therapy with DPP-4 inhibitors. Cardiovasc Diabetol. 2018;17(1):22.PubMedPubMedCentralCrossRef Fadini GP, Avogaro A. How to interpret the role of SDF-1alpha on diabetic complications during therapy with DPP-4 inhibitors. Cardiovasc Diabetol. 2018;17(1):22.PubMedPubMedCentralCrossRef
202.
Zurück zum Zitat Fadini GP, Bonora BM, Cappellari R, Menegazzo L, Vedovato M, Iori E, Marescotti MC, Albiero M, Avogaro A. Acute effects of linagliptin on progenitor cells, monocyte phenotypes, and soluble mediators in type 2 diabetes. J Clin Endocrinol Metab. 2016;101(2):748–56.PubMedCrossRef Fadini GP, Bonora BM, Cappellari R, Menegazzo L, Vedovato M, Iori E, Marescotti MC, Albiero M, Avogaro A. Acute effects of linagliptin on progenitor cells, monocyte phenotypes, and soluble mediators in type 2 diabetes. J Clin Endocrinol Metab. 2016;101(2):748–56.PubMedCrossRef
204.
Zurück zum Zitat Turner RC, Cull CA, Frighi V, Holman RR. Glycemic control with diet, sulfonylurea, metformin, or insulin in patients with type 2 diabetes mellitus: progressive requirement for multiple therapies (UKPDS 49). UK prospective diabetes study (UKPDS) group. JAMA. 1999;281(21):2005–12.PubMedCrossRef Turner RC, Cull CA, Frighi V, Holman RR. Glycemic control with diet, sulfonylurea, metformin, or insulin in patients with type 2 diabetes mellitus: progressive requirement for multiple therapies (UKPDS 49). UK prospective diabetes study (UKPDS) group. JAMA. 1999;281(21):2005–12.PubMedCrossRef
205.
Zurück zum Zitat Kim ES, Deeks ED. Empagliflozin/linagliptin: a review in type 2 diabetes. Drugs. 2015;75(13):1547–57.PubMedCrossRef Kim ES, Deeks ED. Empagliflozin/linagliptin: a review in type 2 diabetes. Drugs. 2015;75(13):1547–57.PubMedCrossRef
206.
Zurück zum Zitat Zinman B. Initial combination therapy for type 2 diabetes mellitus: is it ready for prime time? Am J Med. 2011;124(1 Suppl):S19–34.PubMedCrossRef Zinman B. Initial combination therapy for type 2 diabetes mellitus: is it ready for prime time? Am J Med. 2011;124(1 Suppl):S19–34.PubMedCrossRef
207.
Zurück zum Zitat Phung OJ, Sobieraj DM, Engel SS, Rajpathak SN. Early combination therapy for the treatment of type 2 diabetes mellitus: systematic review and meta-analysis. Diabetes Obes Metab. 2014;16(5):410–7.PubMedCrossRef Phung OJ, Sobieraj DM, Engel SS, Rajpathak SN. Early combination therapy for the treatment of type 2 diabetes mellitus: systematic review and meta-analysis. Diabetes Obes Metab. 2014;16(5):410–7.PubMedCrossRef
208.
Zurück zum Zitat Ma RC, Del Prato S, Gallwitz B, Shivane VK, Lewis-D’Agostino D, Bailes Z, Patel S, Lee J, von Eynatten M, Di Domenico M, et al. Oral glucose lowering with linagliptin and metformin compared with linagliptin alone as initial treatment in Asian patients with newly diagnosed type 2 diabetes and marked hyperglycemia: subgroup analysis of a randomized clinical trial. J Diabetes Investig. 2017. https://doi.org/10.1111/jdi.12746.CrossRefPubMedPubMedCentral Ma RC, Del Prato S, Gallwitz B, Shivane VK, Lewis-D’Agostino D, Bailes Z, Patel S, Lee J, von Eynatten M, Di Domenico M, et al. Oral glucose lowering with linagliptin and metformin compared with linagliptin alone as initial treatment in Asian patients with newly diagnosed type 2 diabetes and marked hyperglycemia: subgroup analysis of a randomized clinical trial. J Diabetes Investig. 2017. https://​doi.​org/​10.​1111/​jdi.​12746.CrossRefPubMedPubMedCentral
209.
Zurück zum Zitat Alter ML, Ott IM, von Websky K, Tsuprykov O, Sharkovska Y, Krause-Relle K, Raila J, Henze A, Klein T, Hocher B. DPP-4 inhibition on top of angiotensin receptor blockade offers a new therapeutic approach for diabetic nephropathy. Kidney Blood Press Res. 2012;36(1):119–30.PubMedCrossRef Alter ML, Ott IM, von Websky K, Tsuprykov O, Sharkovska Y, Krause-Relle K, Raila J, Henze A, Klein T, Hocher B. DPP-4 inhibition on top of angiotensin receptor blockade offers a new therapeutic approach for diabetic nephropathy. Kidney Blood Press Res. 2012;36(1):119–30.PubMedCrossRef
210.
Zurück zum Zitat Rizos CV, Filippatos TD, Elisaf MS. Pharmacokinetic drug evaluation of empagliflozin plus linagliptin for the treatment of type 2 diabetes. Expert Opin Drug Metab Toxicol. 2018;14(1):117–25.PubMedCrossRef Rizos CV, Filippatos TD, Elisaf MS. Pharmacokinetic drug evaluation of empagliflozin plus linagliptin for the treatment of type 2 diabetes. Expert Opin Drug Metab Toxicol. 2018;14(1):117–25.PubMedCrossRef
211.
Zurück zum Zitat Gallwitz B. A safety evaluation of empagliflozin plus linagliptin for treating type 2 diabetes. Expert Opin Drug Saf. 2017;16(12):1399–405.PubMedCrossRef Gallwitz B. A safety evaluation of empagliflozin plus linagliptin for treating type 2 diabetes. Expert Opin Drug Saf. 2017;16(12):1399–405.PubMedCrossRef
212.
Zurück zum Zitat Aronson R. Single-pill combination therapy for type 2 diabetes mellitus: linagliptin plus empagliflozin. Curr Med Res Opin. 2015;31(5):901–11.PubMedCrossRef Aronson R. Single-pill combination therapy for type 2 diabetes mellitus: linagliptin plus empagliflozin. Curr Med Res Opin. 2015;31(5):901–11.PubMedCrossRef
213.
Zurück zum Zitat Scheen AJ. Dipeptidylpeptidase-4 inhibitors (gliptins): focus on drug-drug interactions. Clin Pharmacokinet. 2010;49(9):573–88.PubMedCrossRef Scheen AJ. Dipeptidylpeptidase-4 inhibitors (gliptins): focus on drug-drug interactions. Clin Pharmacokinet. 2010;49(9):573–88.PubMedCrossRef
214.
Zurück zum Zitat Brown NJ, Byiers S, Carr D, Maldonado M, Warner BA. Dipeptidyl peptidase-IV inhibitor use associated with increased risk of ACE inhibitor-associated angioedema. Hypertension. 2009;54(3):516–23.PubMedCrossRef Brown NJ, Byiers S, Carr D, Maldonado M, Warner BA. Dipeptidyl peptidase-IV inhibitor use associated with increased risk of ACE inhibitor-associated angioedema. Hypertension. 2009;54(3):516–23.PubMedCrossRef
215.
Zurück zum Zitat Zhao S, Chan LK, Chen L, Cheng TW, Klein T, Leung PS. Combination of telmisartan and linagliptin preserves pancreatic islet cell function and morphology in db/db mice. Pancreas. 2016;45(4):584–92.PubMedCrossRef Zhao S, Chan LK, Chen L, Cheng TW, Klein T, Leung PS. Combination of telmisartan and linagliptin preserves pancreatic islet cell function and morphology in db/db mice. Pancreas. 2016;45(4):584–92.PubMedCrossRef
216.
Zurück zum Zitat Rosenstock J, Hansen L, Zee P, Li Y, Cook W, Hirshberg B, Iqbal N. dual add-on therapy in type 2 diabetes poorly controlled with metformin monotherapy: a randomized double-blind trial of saxagliptin plus dapagliflozin addition versus single addition of saxagliptin or dapagliflozin to metformin. Diabetes Care. 2015;38(3):376–83.PubMedCrossRef Rosenstock J, Hansen L, Zee P, Li Y, Cook W, Hirshberg B, Iqbal N. dual add-on therapy in type 2 diabetes poorly controlled with metformin monotherapy: a randomized double-blind trial of saxagliptin plus dapagliflozin addition versus single addition of saxagliptin or dapagliflozin to metformin. Diabetes Care. 2015;38(3):376–83.PubMedCrossRef
217.
Zurück zum Zitat Softeland E, Meier JJ, Vangen B, Toorawa R, Maldonado-Lutomirsky M, Broedl UC. Empagliflozin as add-on therapy in patients with type 2 diabetes inadequately controlled with linagliptin and metformin: a 24-week randomized, double-blind. Parallel-group trial. Diabetes Care. 2017;40(2):201–9.PubMedCrossRef Softeland E, Meier JJ, Vangen B, Toorawa R, Maldonado-Lutomirsky M, Broedl UC. Empagliflozin as add-on therapy in patients with type 2 diabetes inadequately controlled with linagliptin and metformin: a 24-week randomized, double-blind. Parallel-group trial. Diabetes Care. 2017;40(2):201–9.PubMedCrossRef
218.
Zurück zum Zitat DeFronzo RA, Lewin A, Patel S, Liu D, Kaste R, Woerle HJ, Broedl UC. Combination of empagliflozin and linagliptin as second-line therapy in subjects with type 2 diabetes inadequately controlled on metformin. Diabetes Care. 2015;38(3):384–93.PubMedCrossRef DeFronzo RA, Lewin A, Patel S, Liu D, Kaste R, Woerle HJ, Broedl UC. Combination of empagliflozin and linagliptin as second-line therapy in subjects with type 2 diabetes inadequately controlled on metformin. Diabetes Care. 2015;38(3):384–93.PubMedCrossRef
219.
Zurück zum Zitat Lewin A, DeFronzo RA, Patel S, Liu D, Kaste R, Woerle HJ, Broedl UC. Initial combination of empagliflozin and linagliptin in subjects with type 2 diabetes. Diabetes Care. 2015;38(3):394–402.PubMedCrossRef Lewin A, DeFronzo RA, Patel S, Liu D, Kaste R, Woerle HJ, Broedl UC. Initial combination of empagliflozin and linagliptin in subjects with type 2 diabetes. Diabetes Care. 2015;38(3):394–402.PubMedCrossRef
220.
Zurück zum Zitat Dey J. SGLT2 inhibitor/DPP-4 inhibitor combination therapy—complementary mechanisms of action for management of type 2 diabetes mellitus. Postgrad Med. 2017;129(4):409–20.PubMedCrossRef Dey J. SGLT2 inhibitor/DPP-4 inhibitor combination therapy—complementary mechanisms of action for management of type 2 diabetes mellitus. Postgrad Med. 2017;129(4):409–20.PubMedCrossRef
221.
Zurück zum Zitat Scheen AJ. DPP-4 inhibitor plus SGLT-2 inhibitor as combination therapy for type 2 diabetes: from rationale to clinical aspects. Expert Opin Drug Metab Toxicol. 2016;12(12):1407–17.PubMedCrossRef Scheen AJ. DPP-4 inhibitor plus SGLT-2 inhibitor as combination therapy for type 2 diabetes: from rationale to clinical aspects. Expert Opin Drug Metab Toxicol. 2016;12(12):1407–17.PubMedCrossRef
222.
Zurück zum Zitat Triplitt C, Solis-Herrera C, Cersosimo E, Abdul-Ghani M, Defronzo RA. Empagliflozin and linagliptin combination therapy for treatment of patients with type 2 diabetes mellitus. Expert Opin Pharmacother. 2015;16(18):2819–33.PubMedCrossRef Triplitt C, Solis-Herrera C, Cersosimo E, Abdul-Ghani M, Defronzo RA. Empagliflozin and linagliptin combination therapy for treatment of patients with type 2 diabetes mellitus. Expert Opin Pharmacother. 2015;16(18):2819–33.PubMedCrossRef
223.
Zurück zum Zitat Singh AK, Singh R. Sodium-glucose co-transporter-2 inhibitors and dipeptidyl peptidase-4 inhibitors combination therapy in type 2 diabetes: a systematic review of current evidence. Indian J Endocrinol Metab. 2016;20(2):245–53.PubMedPubMedCentralCrossRef Singh AK, Singh R. Sodium-glucose co-transporter-2 inhibitors and dipeptidyl peptidase-4 inhibitors combination therapy in type 2 diabetes: a systematic review of current evidence. Indian J Endocrinol Metab. 2016;20(2):245–53.PubMedPubMedCentralCrossRef
Metadaten
Titel
The role of dipeptidylpeptidase-4 inhibitors in management of cardiovascular disease in diabetes; focus on linagliptin
verfasst von
Annayya R. Aroor
Camila Manrique-Acevedo
Vincent G. DeMarco
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Cardiovascular Diabetology / Ausgabe 1/2018
Elektronische ISSN: 1475-2840
DOI
https://doi.org/10.1186/s12933-018-0704-1

Weitere Artikel der Ausgabe 1/2018

Cardiovascular Diabetology 1/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

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