Skip to main content

Common mechanisms underlying diabetic vascular complications: focus on the interaction of metabolic disorders, immuno-inflammation, and endothelial dysfunction

Abstract

Diabetic vascular complications (DVCs), including macro- and micro- angiopathy, account for a high percentage of mortality in patients with diabetes mellitus (DM). Endothelial dysfunction is the initial and role step for the pathogenesis of DVCs. Hyperglycemia and lipid metabolism disorders contribute to endothelial dysfunction via direct injury of metabolism products, crosstalk between immunity and inflammation, as well as related interaction network. Although physiological and phenotypic differences support their specified changes in different targeted organs, there are still several common mechanisms underlying DVCs. Also, inhibitors of these common mechanisms may decrease the incidence of DVCs effectively. Thus, this review may provide new insights into the possible measures for the secondary prevention of DM. And we discussed the current limitations of those present preventive measures in DVCs research.

Video Abstract

Introduction

With the enhancement of our lifestyle, the global epidemic of metabolic diseases as represented by DM and its complications seriously threatens human life and health [1]. According to International Diabetes Federation (IDF 2021, version 10th), it is estimated that 537 million of the adult population were affected [2]. The high incidence of DM imposes huge personal and societal healthcare burdens. Global health spending on adult DM has grown from US$ 232 billion in 2007 to US$ 966 billion in 2021, representing a 316% increase over 15 years [2]. As for World Health Organization (WHO) statistics from 2005 to 2015, the economic burden of DM on our country was 557.7 billion US dollars, of which 80% was spent on diabetic complications [3].

DVCs, including microangiopathy (diabetic kidney disease (DKD), diabetic retinopathy (DR), diabetic cardiomyopathy (DCM), etc.) and macroangiopathy (DM-related coronary heart disease (CHD), etc.), play a leading role in morbidity and death among patients with DM [4,5,6]. Adults (20–79 years old) who died from DM or its complications equate to 12.2% of all-cause deaths in 2021 [2].

In recent years, the three-level system for prevention and control of DM has been proposed, in which secondary prevention (prevention of complications) and tertiary prevention (treatment of complications, delaying their death and disability) are both focused on the stage of complications [7]. Therefore, the early prevention and treatment of diabetic complications, especially DVCs, occupy a concernful position. At present, the intervention of diabetic microangiopathy is mostly at the level of tertiary prevention, such as ACEI/ARB drugs and SGLT2 inhibitors for DKD [8], calcium dobesilate combined with laser photocoagulation and anti-vascular endothelial growth factor (VEGF) agent for DR [9, 10], anti-heart failure therapy for DCM [11]. However, no clear evidence-based proof confirms the preventive ability of these therapeutic measures. Only general practical guidelines that control blood glucose, blood pressure, and blood lipids are recommended for DM secondary prevention, while more effective targeted measures are absent.

Known as one of basic pathological features of DVCs, vascular endothelial dysfunction is ordinarily caused by common upstream mechanisms of metabolic disorders and immuno-inflammation [12,13,14]. In this article, we reviewed the interaction of metabolic disorders, immuno-inflammation and endothelial dysfunction. By summarizing the common core mechanisms underlying DVCs, we hope to grasp opportunities to find multi-targeted approaches for the prevention and treatment of DVCs.

Endothelial dysfunction and DVCs

Endothelial cells (ECs) constitute a single-layer cell barrier covering the surface of the vascular lumen [14]. They bear the brunt of various stimuli from DVCs, highlighting their importance on vascular morphology, function, and metabolism homeostasis regulation [15].

There are two important hallmarks closely coordinated with each other to provoke endothelial dysfunction in DVCs: one is hyperglycemia induced by insufficient insulin secretion or insulin resistance (IR), and another is chronic, low-grade inflammation induced by proinflammatory immune cells and released cytokine [16]. It is characterized by reduced production or bioavailability of nitric oxide (NO), increased oxidative stress, increased vascular endothelial growth factor (VEGF), vasomotor dysfunction, and impaired endothelial recovery [17,18,19].

Macrovascular disease depends on vascular smooth muscle cells (VSMCs) to regulate vasomotor response while tiny blood vessels and capillaries are less affected by vascular tension for a lack of VSMCs [17]. So vascular endothelial homeostasis is more particularly significant to diabetic microangiopathy. For microvessels, the resistance to systemic blood flow in the body's blood vessels increased with high blood pressure. Small artery remodeling results in a decrease in blood flow reserve function, leading to abnormal tissue perfusion, and even preclinical or clinical cardiovascular diseases [17, 20]. Endothelial barriers (including blood–brain, blood-retina, and glomerular barrier) are destroyed for cellular connexin downregulation and integrin upregulation, thereby leading to vascular leakage and abnormal permeability [21, 22]. For diabetic macroangiopathy, endothelial dysfunction leads to vascular wall remodelling. Subsequently vascular becomes stiffer and less compliant, and even atherosclerosis (AS) forms [20, 23]. Thus, ECs are potential therapeutic targets for the prevention and treatment of DVCs, whether they are macrovascular or microvascular diseases.

Regulatory mechanisms of vascular endothelial homeostasis in DVCs

The maintenance of vascular endothelial homeostasis in DVCs, a complex regulatory process involving multiple steps, includes cell barrier and damage repair, vascular and blood flow regulation, redox, metabolic regulation and so on (Fig. 1).

Fig. 1
figure 1

Four main regulatory mechanisms of vascular endothelial homeostasis. Mechanisms required for endothelial homeostasis regulation include cell barrier and repair homeostasis, vascular and blood flow regulation homeostasis, redox homeostasis and metabolism homeostasis; cell barrier and repair involves cell-to-cell junction, communications and relative pathways; vascular and blood flow regulation homeostasis involves blood flow sensor and control, as well as angiogenesis; redox homeostasis involves intracellular and extracellular reduction–oxidation reaction associated with inflammation; metabolism homeostasis involves mitochondria balance, energy conversion, metabolite and ion exchange. eNOS: endothelial nitric oxide synthase; FFA: free fatty acid; NO: nitric oxide; NOX: NADPH oxidase; NRP1: neuropilin-1; OS: oscillatory shear; PGE2: prostaglandin E2; PKC: protein kinase C; PS: Pulsatile shear; ROS: reactive oxygen species; YAP: Yes-Associated Protein; VEGF: vascular endothelial growth factor; VEGFR2: VEGF receptor 2

Cell barrier and damage repair homeostasis

The normal cell barrier ensures complete structure and well function of vascular endothelium, which is the basis of vascular endothelial homeostasis [24]. Inner cell-to-cell connections include tight junction (TJ), adherens junction (AJ), and gap junction (GJ) [25]. Normally arranged ECs rely on tight junctions to maintain paracellular permeability and polarity, forming an inner membrane barrier [25]. TJ regulation-related signal pathways involved in DVCs include the protein kinase C (PKC), RhoA, MAPK, Src, PI3k/Akt, NF-κB and so on [24, 26]. AJ controls cell contact inhibition and permeability of inflammatory cytokine and solutes. Uncontrolled DM and DVCs cause inactivation and reduction in the expression of TJ protein, and related regulative signal pathways include Wnt/β-Catenin, PI3k/Akt, Ras, Rac, RhoA and so on [25, 27]. The endothelial glycocalyx (EG), which is associated with GJ, acts as a special barrier with glycosaminoglycans attached to prevent DVCs [28]. EG maintains structural and functional integrity of gap connexin proteins, ensuring interendothelial molecular transport [29]. More importantly, EG can reduce platelet and leukocyte adhesion, inflammatory stimulation, and oxidative stress damage to vascular ECs, underscoring the importance of EG preservation to avoid disease initiation and progression [30, 31].

Apart from cell-to-cell junctions, maintaining barrier function needs updates and repairs of vascular ECs themselves. The sphingolipid signal pathway and related mediators, such as sphingosine 1-phosphate (S1P) and its G-protein coupled receptor 1 (S1PR1), perform barrier protective function to ECs and restore cell communication [29, 32]. Activation of S1PR1 prompts redistribution of connexins (VE-cadherin, β- Catenin, and α-Catenin, etc.) to the intercellular contact region, thereby tightening ECs barrier [33]. Concerning ECs repair, activation of transcription factors EGR1 and STAT3 is needed for transcribing sphingosine kinase 1 and sphingolipid transporter 2 to increase the generation and efflux of S1P [34, 35]. Then, the transition of S1PR1+ ECs amplifies and they activate endothelial regenerative programs to mediate vascular endothelial repair in DM [35, 36]. In addition, the role of immune cells and circulating angiogenic cells (CACs) are even more crucial to the sustainable complement for ECs [37, 38]. As one of the necessary regulatory factors maintaining the chemotaxis of CACs, endothelial nitric oxide synthase (eNOS) activates MMP-2 and MMP-9 to promote bone marrow differentiation and release CACs [30, 32, 38]. CACs position the site of endothelial injury and produce cytokines (VEGF, SDF-1, etc.) to promote angiogenesis and ECs repair in DM [38, 39].

Vascular and blood flow regulation homeostasis

Mechanical stress is a key factor underlying the pathophysiology of DVCs. Compared to straight vascular segments exposed to laminar flow, other vascular sites exposed to disturbed flow patterns appear more susceptible to endothelial dysfunction and atherosclerotic plaque formation [40]. Pulsatile shear (PS) stress in straight segments increases the expression of antioxidant and anti-inflammatory (flow-mediated genes (e.g. YAP-TAZ)) genes to keep a vascular protective phenotype, whereas this is not the case for oscillatory shear (OS) stress at the vessel curvatures, branchpoints, and bifurcations [41]. The signal transition mediated by various endothelial mechanosensors (e.g., caveolae and lipid rafts, membrane proteins, primary cilia, glycocalyx, cellular junction, and adhesion molecules) is responsible to regulate vascular homeostasis and adaptative ability [42]. Take the YAP protein (one mechanosensor) as an example, it interacts with STAT3 to regulate angiogenesis [43]. YAP/TAZ pathway, constitutively activated by hyperglycemia, has been proven to be associated with proinflammatory gene expression in endothelial cells as well as disturbed flow-induced diabetic vascular inflammation and renal damage [44, 45].

Under acute stress conditions (e.g., trauma, infection, fever), a sensitive mechanotransduction sensing mechanism regulates blood flow changes to match tissue supply–demand balance through negative feedback [46]. Meanwhile, ECs are regulated by sympathetic nerves to synthesize, release and balance endogenous regulators (e.g., NO, PGs, ET-1, and endothelium-derived hyperpolarizing factors) of vascular tone change [47, 48]. Vascular ECs can also secrete platelet-activating factors, and regulate factors related to the coagulation-fibrinolytic system to control blood flow [49].

Under long-term chronic injury, ECs extracellular matrix turnover interacts with smooth muscle cells that control intimal proliferation, thereby affecting vascular structure and function [46]. Numerous regulated signal pathways and targeted molecules (VEGF, FOXO1, and several miRNAs) get involved [50,51,52,53]. VEGF, a key factor in angiogenesis and normal vascular function maintenance, binds to its main receptor VEGFR-2 to regulate ECs proliferation and migration [54]. Neuropilin-1 (NRP-1) acts as a co-receptor to increase their binding ability [55]. Another regulatory factor FOXO1 inhibits the MYC signal pathway to decrease over-proliferated ECs and overgrown neovascular, maintaining a quiescent phase [56]. A variety of miRNAs, including miR-126, help to leverage the homeostasis maintenance of the neovascular and lymphatic network [57].

Redox homeostasis

Oxidative stress mentioned above is an important link to mediate inflammatory response, which leads to vascular ECs damage through direct cytotoxic effects. The balance between reactive oxygen species (ROS) accumulation and nitric oxide (NO) consumption in hyperglycemia ultimately contributes to normal vascular endothelial function [58]. On one hand, the overproduction of ROS can be a central mediator of injury to cell constituents, including lipids, nucleic acids, and proteins [59]. The accumulated ROS subsequently induces signaling cascade of MAPK and activate NFκB, which results in expression of pro-inflammatory cytokines, chemokines and receptors [60, 61]. Lipids peroxidation, the denaturation of nuclear or mitochondrial DNA, as well as inactivated proteins after oxidative modification, together bring about substantial changes in membrane permeability and elasticity [59, 62]. On the other hand, NO (also called an endothelium-derived relaxing factor), is one of the most important vasodilator factors. It continuously maintains the dynamic balance of vascular endothelium by activating ERK1/2 and p44/p42 mitogen-activated protein kinases and causing tyrosine phosphorylation of focal adhesion kinase (FAK) [63]. Except for antihypertensive and antiplatelet effects, NO also inhibits LDL oxidation and leukocyte adhesion, thereby reducing the risk of endothelial thrombosis in patients with arteriosclerosis or diabetic CHD [64]. NO is synthesized from L-arginine under the catalysis of eNOS in vascular ECs [65]. However, some studies have shown that eNOS is mainly expressed in large vascular ECs, rarely in arterioles, and not in capillary ECs [30, 66]. Based on this structural expression, eNOS may not be involved in diabetic microangiopathy, but it is not clear at present.

Metabolic homeostasis

Mitochondria are best known for cellular energy conversion and metabolic homeostasis. The balance of mitochondrial fusion and division, as well as normal autophagy, ensure the functional retention of mitochondria and timely clearance of damaged mitochondria, forming a highly dynamic regulatory mechanism against injury from DVCs [67, 68]. The OPA1 gene mediates mitochondrial fusion to protect ECs viability and reduce oxidative stress [68]. Normal release of ROS and stabilization of mitochondrial membrane potential reduce cell damage and aging, further preventing the vicious circle of mitochondrial damage caused by excessive peroxide in DVCs [67]. As for autophagy, VEGF equalizes autophagic flux through an AMPK/mTOR-dependent mechanism to prevent DVCs progression [69, 70].

Vascular ECs may also maintain their metabolic homeostasis by dynamically regulating ionic metabolite exchange. Iron homeostasis maintenance act as a vascular protector by reducing lipid peroxidation and excessive iron death [71, 72]. Zinc ions are required for eNOS dimerization and subsequent production of NO, while NO leads to the rapid mobilization of endothelial Zinc storage to mediate vascular cell protection and vasodilation [73]. Besides, other regulators Na+/H+ exchanger regulator-2 (NHERF-2) and second messenger Ca2+ also exist in the regulative process of endothelial homeostasis [74, 75].

Common mechanisms of DVCs

Effects of metabolic disorders on ECs

Glucose metabolism disorder

Glucose metabolism disorder acts as the initiating step and important risk factor of DVCs [76]. The continuous high glucose status and accumulation of intermediate metabolites increase mitochondrial membrane potential and ROS production. Four classical pathways (hyperactivated polyol pathway and hexosamine pathway replacing normal glucose metabolism pathway, PKC activation, accelerated production and accumulation of advanced glycation end products (AGEs), reviewed by Ighodaro [77]) exacerbate the damage to vascular endothelial function [78]. Among them, the most important AGEs are produced by non-enzymatic glycosylation reaction of proteins, lipids, DNA, and other substances in the human body with reducing sugars [79]. Excessive AGEs bind to their receptors (RAGE) in ECs. On one hand, TLR-4 heterodimerization activates the NF-κB pathway to upregulate the expression of adhesion molecules. Along with inflammatory cells and cytokines migrating and infiltrating local tissues, oxidative stress is prone to increase the permeability of blood vessel wall [80]. On the other hand, AGEs-RAGE leads to VEGF overexpression and abnormal hyperplasia of neovascularization [79, 81]. Overlapped multi-effects not only directly lead to damage of vascular ECs (such as peroxidation of biofilm lipids, destruction of cell structure and increased apoptosis), but also accelerates oxidative stress to prompt further damage to ECs [82].

Lipid metabolism disorders

Obesity-induced insulin resistance (IR) is reported as the common soil and intermediate connecting mechanism for glucolipid metabolism disorders [83]. Firstly, IR inhibits eNOS activity to reduce NO concentration through the PI3KA-MAPK pathway, which severely impairs endothelium-dependent vasodilation and increases its permeability [84]. In addition, IR stimulates the proliferation of VSMCs in turn and the excessive release of free fatty acid (FFA) in adipose tissue to aggravate oxidative stress and PKC activation, eventually forming a vicious circle [85]. Suppressed endothelial fatty acid oxidation (FAO) increases intracellular calcium oscillation. Those negative effects reduce NADPH levels and increase the ratio of NADP+/NADPH, resulting in vascular leakage, weakened endothelial antioxidant stress capacity and aggravation of endothelial activation caused by lipopolysaccharide (LPS) [86, 87]. Under the stimulation of hyperinsulinemia, the synthesis of lipoproteins in the liver increases. AGEs participate in modifying lipoproteins as accelerators of lipid metabolism disorders. Ultimately, AGEs increase TG-rich lipoproteins and LDL and decrease HDL [88].

The binding affinity between sdLDL and LDL receptors decreases, which hinders sdLDL clearance pathway [89]. Moreover, due to its small particles, it is easy to adhere to the subendothelial layer and combine with proteoglycan. Combined lipoproteins are more prone to oxidative modification. Stimulated macrophages in circulation and tissues phagocytize the modified LDL and turn them into foam cells, which eventually accelerates the development of DVCs under the pressure of immuno-inflammation [86, 89, 90].

Gut microbiome metabolites

The metabolites of intestinal flora, such as short-chain fatty acids (SCFA), bile acids (BA), branched-chain amino acids (BCAA), methylamines and gaseous transmitters affect host metabolism [91]. Intestinal flora disorders may help inhibit the reverse cholesterol transport mechanism (RCT). Excess production of LPS and foam cells then inhibits cholesterol efflux [92]. Other products indoxyl sulfate and trimethylamine-N-oxide (TMAO), lead to the deterioration of DVCs [93]. Indoxyl sulfate is produced in the liver from the metabolism of indole. Indole, known as a protein-imported uremic toxin, is converted from dietary tryptophan by bacterial tryptase in the colon. TMAO is the ramification of trimethylamine derived from the intestinal microbiota oxidated by flavin-containing monooxy geniuses and acts as the promoter of AS [92, 93].

Inflammatory metabolite-arachidonic acid derivatives

Arachidonic acid can be metabolized by three pathways: COX, LOX, and CYP4. The leukotriene, thromboxanes TXA2 and TXB2, and EET produced during arachidonic acid metabolism are involved in vascular-related diseases [94]. In the pro-inflammatory environment, PI3K/AKT signal pathway directly regulates the activity of NF-κB. Subsequently, NF-κB upregulates the inducible expression of genes including cyclooxygenase-2 (COX-2) and promotes caspase-3 activity that triggers apoptosis of ECs. COX-2 catalyzes arachidonic acid and produces numerous prostaglandins (PGs) [95]. PGI2 and TXA2 are different kinds of PGs produced by vascular ECs and platelets, respectively. PGI2 induces vasodilation and inhibits platelet aggregation, while TXA2 induces vasoconstriction. The imbalance between them is a key factor to disrupt endothelial homeostasis [96, 97]. Activation of thromboxane/endoperoxide receptors can also aggravate endothelial dysfunction [95]. Moreover, COX-2 activity interacts with oxidative stress. COX2 stimulates the production of non-phagocytic cell NADPH oxidase 2 (NOX2), and activates COX2 reversely, resulting in a continuous vicious cycle of endothelial dysfunction [98].

Effect of immuno-inflammatory interaction on ECs

Under the regulation of various cells and their secreted cytokines, TLR2/4-NF-κB, p38/MAPK, IL-6/STAT3 and other key transcriptional regulatory pathways get involved to constitute the immuno-inflammation interaction mechanism in DVCs (Fig. 2). Innate immunity is enhanced in patients with DVCs, yet adaptive immunity is relatively less studied.

Fig. 2
figure 2

The interaction mechanism of metabolic disorders, immuno-inflammation and endothelial dysfunction in DVCs. The metabolic disorder process, characterized by hyperglycemia and IR, involves glucose, lipid, and gut microbe metabolism. The immuno-inflammation process is activated by immunocytes, proinflammatory cytokines, and NETs. Then induced inflammation and redox injury cause damage to ECs and vessels. AGEs: advanced glycation end products; CXCL: chemokine (C-X-C motif) ligand; FFA: free fatty acid; IR: insulin resistance; LPS: lipopolysaccharide; Mφ: macrophages; MCP-1: monocyte chemoattractant protein 1; MDSCs: marrow-derived myeloid cells; NETs: neutrophil extracellular traps; NO: nitric oxide; NOX: NADPH oxidase; PGI-2: prostaglandin I-2; PKC: protein kinase C; RAGE: receptor of AGEs; SCFA: short-chain fatty acids; Th: T helper; TLR: Toll-like receptors; TMAO: trimethylamine-N-oxide; Treg: T regulatory cells; TxA2: thromboxane A2; VEGF: vascular endothelial growth factor

Innate immune activation

Monocyte-macrophage system

As an important component of intrinsic immunity, the monocyte-macrophage system plays a key role in the chronic low-level inflammatory process [99]. Under the stimulus of metabolic disorders, Mφ polarized from anti-inflammatory M2-like macrophages (CD68+/Arg-1+) to pro-inflammatory M1-like macrophages (CD68+iNOS+) [100, 101]. M1-like macrophages secrete numerous proinflammatory cytokines (such as TNF-a, IL-1β, IL-6), chemokines (MCP-1/CCL-2) and inducible nitric oxide synthase (iNOS) [102]. Those cells, cytokines, chemokines and essential substances in tissue form an inflammatory M1-like immune microenvironment, maintaining a long-term inflammatory stress state of ECs [102, 103]. In parallel, metabolic disorders products FFA and LPS bind to membrane protein receptors of ECs and monocytes, leading to increased secretion of vascular cell adhesion molecules (intercellular adhesion molecule 1 (ICAM-1), etc.) and E-selectin [104]. Under the regulation of the AMPK-STAT3 axis, chemokines MCP-1 and CCR2 recruit circulating monocytes and M1-like differentiated macrophages to adhere to ECs in the vessel wall, accelerating metabolic disorders [103,104,105]. Infiltrating monocyte-macrophages transfer from the ECs gap to the tubular endothelial layer by rolling [104, 105]. Excess oxidized LDL can be phagocytosed and ingested by M1-like macrophages and VSMCs, which are the ultimate source of foam cell forms and AS plaques [106, 107]. Macrophages produce excessive metalloproteinases, which can dissolve and even rupture plaques [106, 108].

Granulocytes, MDSCs and NETs

The granulocyte system mainly includes neutrophils, eosinophils, and basophils. As a component of innate immunity, the granulocyte system often participates in immuno-inflammation by releasing proinflammatory mediators and cytokines [109]. Neutrophils are the most abundant inflammation-related immune cells in circulation and play a wide role in the inflammatory process [110]. Mechanisms of neutrophils that respond to immuno-inflammation in ECs mainly include the production of ROS, the secretion of inflammatory cytokines and peroxidase MPO, as well as the formation of NETs [111, 112]. With the spontaneous migration of granulocytes to targeted organs, the expression of adhesion molecules CD11b and ICAM-1 are also up-regulated [113, 114].

A special granulocyte system called proangiogenic granulocytes (PAGs) is composed of 20% neutrophils and 80% eosinophils [115]. PAGs are measured with CD49d+Ly6G+ adhesion marker on their surface and rich in VEGFR1 and CXCR4 molecules [116]. PAGs support the angiogenesis of ECs in vitro but PAG level is inversely proportional to blood glucose level [115]. The eosinophil count in DKD always rises, along with wider glomerular basement membrane (GBM) and positively correlated count with male albumin excretion rate [117, 118]. Eosinophils are rarely studied in other DVCs, and basophils are even rarely reported.

A large number of polymorphonuclear marrow-derived myeloid cells (PMN-MDSCs) are generated by bone marrow and spleen [112]. Under high glucose and chronic inflammatory conditions, IL-6/STAT3 pathway is activated. After recruitment by inflammatory chemokines (CXCL1, CXCL2, CXCL5, and S100A8/A9 (Calprotectin)), and activation by proinflammatory (IL-6, VEGF, IFN-γ, IL-1β, HMGB1), PMN-MDSCs transmit to inflammatory ECs to triggers neutrophil extracellular traps (NETs) [112, 119]. Up-regulated PMN-MDSCs express the components p47phox and gp91phox of NOX2. NOX2 not only accelerates the inflammatory response by excessive ROS production but also promotes the occurrence of NETs [102, 120]. Moreover, activated NLRP3 inflammasome also promotes NETosis in CHD and atherosclerosis [121]. That is an important mechanism of diabetic microangiopathy, which may be used as a potential therapeutic target.

The important role of other innate immune cells

Numerous previous studies have focused on the role of mast cells (MCs) in AS and CHD. It has been found that MCs have involved in vascular plaque formation and inflammatory infiltration to atherosclerotic vessel walls through cell cytoplasmic vacuolization, extracellular extrusion of granules and nuclear sequestration [122]. MCs can secrete various mediators to activate other inflammatory cells (such as lymphocytes and foam cells) and affect the metabolism and circulation of HDL and LDL [123, 124]. MCs activated by endogenous factors (ET-1, component C5R, etc.) can release proteases through granule exocytosis to degrade inflammatory markers and limit the progress of the inflammatory process [124].

DCs help to capture, process, and present antigens to antigen-presenting cells (APC). APC stimulates the activation and proliferation of antigen-specific T and B cells to initiate adaptive immunity [125, 126]. Activated dendritic cells (DCs) promote the secretion of proinflammatory mediators, including antibacterial mediators and chemokines, to help recruit more immune cells to infiltrate to local inflammatory vascular endothelium [125, 127]. In addition, DCs regulates the differentiation of T cells into different subsets [126].

Complement system

The systemic complement system is activated, and deposited serum membrane attack complex (MAC) results in cell damage to targeted organs [128,129,130]. In addition to inducing epithelial-mesenchymal transition and inflammatory cell infiltration, complement C3 and C5 also participate in transforming growth factor β (TGF-β) mediated endothelial EndMT [130, 131]. Complement regulator CD59, a membrane glycoprotein, can inhibit the polymerization of C9 to prevent the formation of MAC. CD59 is more susceptible to glucose-dependent nonenzymatic glycation-inactivation in DM. Autoantibodies against glycosylated proteins initiate complement activation again via classical pathways [132].

Specific immunity activation

Diverse immunosuppressive cells (like Treg cells) and secretory cytokines mainly composed the vascular endothelial immunosuppressive microenvironment of patients with DVCs [133, 134]. AGEs may act as a new epitope bound by the mannose-binding lectin (MBL) pathway to motivate adaptive immunity [135]. The balance of the circulating T lymphocyte subset is dynamically affected by hyperglycemia, with significantly increased CD4+ T cells and reduced CD8+ T cells [136]. Infiltrating CD8+ T cells in targeted tissues exerts negative regulatory effects on angiogenesis and endothelial function [137]. Moreover, microparticles released by T cells regulate the expression of eNOS and caveolin-1, accelerating endothelial dysfunction by NO and prostacyclin pathways [138].

T helper type 1 (Th1), Th2, Th17, regulatory T (Treg) cells, and cytotoxic T cells participate in the occurrence and development of DVCs, but the related molecular mechanism of endothelial dysfunction in various organs is not equal [139]. B cells contribute to the production of antibodies and cytokines, whereas few studies focus on the interconnection between them and inflammation in DVCs [140].

Immuno-inflammation related cytokines

Proinflammatory cytokines

The majority of proinflammatory cytokines infiltrated into local tissues originate from polarized M1-like macrophages or leukocytes [141, 142]. Toll-like receptors (TLRs) recognize damage-associated molecular patterns (DAMPs) released during cell stress and injury [143]. Combined TLR 2/4 in monocytes increases with NF-κB translocation to the nucleus and proinflammatory genes (encoding proinflammatory factors, like TNF-α, IL-6, IL-1β, IL-18) transcription. These changes result in NLRP3 inflammasome overexpression and NLRP3 activation-associated endothelial dysfunction [129, 144]. NLRP3 inflammasome also activates caspase-1 to increase the speed and severity of inflammation [145]. In this regard, TLR2/4 is positively correlated with transcription factor NF- κB expression and the severity of inflammation [146].

As one of the earliest detectable cytokines, TNF-α promotes the generation of ROS to aggravate intimal injury. The amount of TNF-α goes up with disease progression to form a vicious cycle. Its continuous secretion leads to the release of ICAM-1, which controls recruited macrophage infiltration [147]. Additionally, TNF-α is a key signaling molecule of the AMPK/NF-κB /NLRP3 pathway. Activation of NLRP3 inflammasome can trigger IL-1β and proIL-18 secretion to promote further inflammatory processes and oxidative stress [148]. Analogously, IL-6 interacts with TNF-α to enhance oxidative stress and reduce eNOS phosphorylation [149]. IL-6 receptor family activates JAK/STAT signal pathway to cooperatively regulate B cells differentiation, plasma cell genesis, and acute phase response [150, 151].

Chemokines

Chemokine MCP-1 and ICAM-1 can attract monocytes, macrophages, T cells, and DCs to inflammatory sites [139, 152]. MCP-1 promotes monocytes and macrophages activation and also upregulates the expression of adhesion molecules and proinflammatory cytokines [153, 154]. In addition, CXCL1, CXCL2, CXCL5, and calprotectin (S100A8/A9, calprotectin) are involved in neutrophil recruitment [155].

Other cytokines

VEGF is by far the strongest specific angiogenic factor and is also a signal communication bridge between islet β cells and ECs [66]. Under the condition of metabolic disorders, VEGF overexpression, mediated by various proinflammatory mediators, leads to increased vascular permeability, disordered angiogenesis, increased adhesion molecules, as well as broken barrier protection and function repair of ECs [156]. Multiple mechanisms related to VEGF include Wnt, PI3K/Akt/mTOR/eNOS and p38/MAPK [66, 156, 157]. TGF-β mainly participates in the process of ECs-related organ fibrosis. In kidney, retina, and heart fibrosis, disorders of glucose and lipid metabolism mediate EndMT in ECs through PKC β/TGF-β/PAI-1, TGF-β/SMAD, and a part of lncRNA signal pathways [158,159,160,161]. High glucose stimulation triggers ROS production and activates TGF-β/SAMD3 pathway. Thus, caused proinflammatory cytokines and chemokines upregulate fibroblast-stromal cell proteins by stimulating the AGE-RAGE axis [162].

The important role of oxidative stress in immuno-inflammation

Oxidative stress is associated with IR, B cell dysfunction, and damage to cell membrane integrity. Induced apoptosis, microvascular damage, and barrier break, ultimately lead to the progression of vascular complications [163, 164].

ROS in ECs is mainly generated by NOX2, mitochondrial respiratory chain, and eNOS uncoupling [165]. The systemic chronic inflammatory state induced by the disorders of glucose and lipid metabolism stimulates the overproduction of ROS from different sources. Endothelial NO inhibits NF-κB activation and upregulation of VCAM-1, E-selectin, and ICAM-2, thereby inhibiting leukocyte adhesion to vascular endothelium [166]. Total eNOS levels decreases, however, endothelial NO production is impaired and consumed by oxidative inactivation [64]. Imbalanced NO and ROS contribute to a vicious circle of oxidative stress. Pathways and molecules involved in this process include the polyol pathway, MAPK, NF-KB, forkhead box O (FOXO), Keap1-Nrf2/ARE, Nrf2/HO signal pathway and so on [90, 167,168,169]. Oxidative stress disturbs endothelial signal transduction and enhances vascular endothelial permeability and leukocyte adhesion through these above mechanisms, which ultimately exacerbates vascular endothelial dysfunction [167].

Interaction between metabolic disorder and immuno-inflammation

Increased levels of circulating inflammatory biomarkers in DM patients appear to predict the onset and progression of DVCs [170]. Long-term high glucose environment triggers innate immunity and homeostasis maintaining response once AGEs are recognized by pattern recognition receptors (PRRS). Induced abnormal function of mitochondria and endoplasmic reticulum promotes oxidative stress and endothelium structural damage. The process of oxidative stress is related to the elevated levels of proinflammatory cytokines and autoantibodies. AGEs/RAGE pathway activates downstream NF-κB signal pathway to stimulate blood vessels continuously.

Lipotoxicity leads to lipid metabolism products and visceral adipose tissue levels dynamic storage addition. These products induce M1 polarization of macrophages. LDL adhesion remains in the subendothelial layer of blood vessels and receives oxidative modification to oxidized low-density lipoprotein (ox-LDL), which can be engulfed by M1-like macrophages and transformed into foam cells. The expression of systemic inflammatory cytokines increases, to form a chronic low-level inflammatory state in the body, which runs through the process of DVCs.

Downstream effector mechanisms of DKD, DR and DHD

Different targeted organs consist of different cells. Inevitably, the main functional cells of kidney, retina, heart and coronary artery seem to be affected by endothelial dysfunction (Fig. 3).

Fig. 3
figure 3

The inner linkage of endothelial dysfunction and different DVCs. The complex relationship of metabolic disorders, immuno-inflammation and endothelial dysfunction (left part) and different downstream effector mechanisms of DKD, DR and DHD (right part) are shown. ECs: endothelial cells; RAAS: renin-angiotensin aldosterone system; RPE: retinal pigment epithelial

Glomerular ECs, podocytes, mesangial cells and DKD

As the most common diabetic microangiopathy, DKD developed with the cellular structure destruction and dysfunction of glomerular ECs, podocytes, and mesangial cells. VEGF secreted by ECs or podocytes binds to its receptors VEGFR, regulating the normal vascular permeability and angiogenic function [171]. Once ECs are damaged, the function of renal mesangial cells will be seriously affected along with plenty of podocytes lost. Besides, activated local renin-angiotensin aldosterone system (RAAS) results in the consequences of glomerular hyperperfusion, high pressure and high filtration. Other mechanisms include immune activation related to immune complex deposition, inflammatory response caused by the chemokines serum amyloid A (SAA) and regulated upon activation of normal T-cell expressed and secreted (RANTES) [172, 173], as well as the profibrotic cascade with multiple cytokines involved [174, 175].

Retinal vascular ECs, RPE cells, microglia and DR

The high metabolic demand and limited vascular supply to retinal cells make them extremely sensitive to metabolic disorders. The disorder of vascular structure is more significant in DR for the influence of VEGF overexpression [8]. Thrombus and haemorrhage coexist after the formation of microangioma [176]. Even fibrovascular membrane hyperplasia or traction retinal detachment occurs in the proliferative stage, accompanied by retinal neurodegeneration [177]. In fact, the retina is an immune privilege tissue. The blood-retinal barrier (BRB) is formed by a tight junction of retinal vascular ECs (inner BRB) and retinal pigment epithelial (RPE) cells (outer BRB) [188]. Unless BRB structural disruption in severe DR, circulating cells and molecules cannot freely enter the retinal parenchyma, much less specific autoantibodies or T cell infiltration [178]. Additionally, there is no lymphatic system in the retina so that it is difficult to recognize DAMPs.

Retinal microglia are known as the most important local immune cells in DR. Others include perivascular macrophages, persistent transparent cells and DCs. Hyperglycemia inhibits the activation of myeloid cells (microglia/macrophages) and T cells, but it can induce Tregs cells formation through CD200-CD200R or CX3CLL-CX3CRL pathway [179, 180]. RPE cells induce apoptosis of infiltrating immune cells through FAS ligand and TRAIL pathway. RPE cells also inhibit complement activation with the help of CD55, CD46 and CD59. Recently, it has been found that CD11b+ monocytes in the peripheral blood of DM mice were more active and expressed higher levels of the chemokine receptor CCR5 [181]. They preferentially stay in retinal microvessels and may be the risk factor for DR.

Coronary artery and microvascular ECs, cardiomyocytes and DHD

Adverse outcomes of diabetic heart disease (DHD) include significant reduction of glucose supply and utilization, depletion of glucose transporter 4 (GLUT4) mediated PPARs pathway, pyruvate dehydrogenase activity inhibition by β-oxidated FFA, the burden of lipid metabolism in cardiomyocytes aggravated by ubiquitin ligase MG53, and cardiac systolic and diastolic dysfunction caused by myocardial lipotoxicity intermediates such as ceramide [182,183,184,185]. There are intimate relationships and intercellular dependence between ECs and cardiomyocytes. DM-related coronary heart disease is mainly associated with atherosclerosis caused by glucose and lipid metabolism disorders, whereas the targeted cells of diabetic cardiomyopathy are mainly cardiomyocytes. Cardiomyocytes are more sensitive to energy metabolism and ion changes, among which Ca2+ is the main ion causing myocardial contraction. Endoplasmic reticulum stress promotes the disorder of Ca2+ metabolism in the cardiomyocyte membrane and eventually causes cardiomyocyte death [186, 187].

Discussion

Vascular disease is a leading cause of death and disability for DM patients worldwide [188]. The burden of DM and its complications are unmet public health issues. Secondary prevention of DM, which means preventing diabetic complications and decreasing risks of major organ injury, is immediately needed. If so, a large quantity of DM patients might have the opportunity to escape from those bad consequences. However, the absence of suitable drugs for the prevention of major vascular events after DM leaves clinicians with dilemmas. For example, ACEI/ARB drugs could not prevent disease progression in the kidneys but increase the incidence of cardiovascular events [189, 190]. Aspirin and clopidogrel for primary and secondary prevention of CHD are recommended, while few proofs of they improve microvascular complications [191, 192].

Proofs of DVCs prevention

Potential preclinical interventions (Table 1) bring new light on realizing secondary prevention of DM. These potential medications were associated with a possible structural benefit in administration of different DVCs. With respect to diabetes-related CHD, combined secondary prevention practices of CHD (e.g., antiplatelet, lipid-lowering, blood pressure control and lifestyle intervention) and antidiabetic drugs without cardiovascular risk have been proposed [191]. The scope of the research on diabetes-related CHD gradually increases and present studies are carried out around therapies of novel glucose-lowering drugs, lipid regulators, antithrombotic drugs, and so on [193]. Eventually, these data should be confirmed with prospective, randomized, controlled clinical trials.

Table 1 Potential preventive intervention for DVCs from experimental data

A retrospective summary of registered clinical trials (WHO international clinical trials registry platform, https://trialsearch.who.int/) on the prevention of DVCs (Table 2) demonstrates that evidence of both safety and effectiveness is still absent. PRIORITY is a multicentre, prospective, observational randomised controlled trial initiated by Steno Diabetes Center [219]. This study illustrated that spironolactone could not prevent progression to microalbuminuria and diabetic kidney disease, which is contrary to what the previous experiment showed [195, 219]. Another exciting trial PRECIDENTD is currently in progress and is due to be completed in 2028. Up-to-now guidelines recommend the initiation of SGLT2 inhibitors or GLP1 receptor agonists with proven cardiovascular benefits in adult patients with T2DM [220]. PRECIDENTD trial will innovatively compare two drugs to prevent cardiovascular and diabetic kidney disease.

Table 2 Registered clinical trials on the prevention of diabetic vascular complications

Possibilities and challenges of parallel targets in DVCs

Treatment of metabolic disorders is based on lifestyle intervention and patient education [221]. By controlling blood glucose, blood pressure, blood lipids and other risk factors, it is expected that DM patients keep a healthy lifestyle and get effective follow-up care strategies. Loss of body weight and drugs assist with ideal metabolic control to protect the function of vital targeted organs [222]. SGLT2 inhibitors are high-profile glycaemic control drugs and associated with reductions in body mass and blood pressure, as well as with both kidney and cardiovascular protective effects [223]. Remarkably, metabolism regulation makes a meaningful impact on both primary and secondary prevention of DM.

Treatment on immunity and inflammation mechanisms includes extracts from Traditional Chinese Medicine (TCM, like rhein, hirudin, polysaccharides), SGLT2 inhibitors, ACEI/ARB agents and so on. TCM therapy has unique advantages for DVCs prevention, which still deserves further study [224]. Rhein, derived from the roots of Rheum L., protects islets β cells by inhibiting abnormal activation of the hexosamine biosynthesis pathway and reverse IR. Besides, Rhein antagonizes TGF-β and protects ECs function [225]. Hirudin, an acidic polypeptide secreted by the salivary glands of Hirudo medicinalis, inhibits HIF1α/VEGF and p38 MAPK/NF-κB pathways and activates NRF-2/HO-1 pathways to prevent DN and other vascular complications [226]. Polysaccharides from different Chinese herbs have significant anti-diabetic and anti-DVCs effects through various mechanisms with almost no side effects [227]. Similarly, SGLT2 inhibitors also modify inflammatory responses by various underlying mechanisms (e.g., oxidative stress, RAAS activation, and immune system function) [223]. Moreover, clinical investigators have been always trying to use ACEI/ARB drugs for secondary prevention of DM, but results are still controversial. ACEI/ARB drugs reduce the formation of angiotensin II or prevent angiotensin II from binding to angiotensin receptors to inhibit RAAS system [228]. They also help relieve local inflammation on the vessel wall to reduce urine protein and regulate blood pressure for patients with DVCs, yet losartan probably increases incidence of macroalbuminuria [229, 230]. Whether other ACEI/ARB use should be recommended for secondary prevention of DM need support from further research data.

Treatment of the endothelial injury ET-1 receptor inhibitors (avosentan, atrasentan, etc.) have been used to lower blood pressure and urine protein, but increase the risk of edema and heart failure [231,232,233]. Moreover, the effects of ET-1 receptor inhibitors for DKD and DHD lack clinical trial validation. Vasodilators can prevent DVCs and their progression by improving the microcirculation of the kidney, retina and myocardium [234]. Pancreatic kallidinogenase (PK), a vital vasodilator, increases capillary blood and tissue perfusion flow by dilating small pulsations and regulating blood rheology. It is reported that PK is beneficial to control the process of DKD, DR and DHD [235,236,237]. The mechanisms of PK may involve the protection of ECs, the influence of angiogenesis and permeability, as well as the reduction of tissue fibrosis [237,238,239,240].

Opportunities for new therapeutics and secondary prevention

In the future, an improved understanding of the integration and regulation of the crosstalk network between metabolic disorder, immuno-inflammation and endothelial dysfunction may provide novel and effective therapeutic targets for DVCs prevention. If effective interventions can be taken on those common mechanisms, multi-disciplinary comprehensive therapy of DM-related vascular disease may provide new ideas for further improved clinical efficacy. For now, though, this is still up for debate. Therefore, the combinatorial drug treatment on the strong interaction network or natural products like TCM prescriptions characterized by multi-component, multi-target, multi-path comprehensive prevention and treatment of diseases may be a preferable alternative.

Availability of data and materials

Not applicable.

Abbreviations

ACEI:

Angiotensin-converting enzyme inhibitor

AGEs:

Advanced glycation end products

AJ:

Adhesive junction

AMPK:

Adenosine 5′-monophosphate (AMP)-activated protein kinase

ARB:

Angiotensin receptor blocker

AS:

Atherosclerosis

BA:

Bile acids

BCAA:

Branched-chain amino acids

CACs:

Circulating angiogenic cells

CHD:

Coronary heart disease

COX-2:

Cyclooxygenase-2

DCM:

Diabetic cardiomyopathy

DCs:

Dendritic cells

DHD:

Diabetic heart disease

DKD:

Diabetic kidney disease

DM:

Diabetes mellitus

DR:

Diabetic retinopathy

DVCs:

Diabetic vascular complications

ECs:

Endothelial cells

EGC:

Endothelial glycocalyx

eNOS:

Endothelial nitric oxide synthase

ERK:

Extracellular regulated protein kinases

FAK:

Focal adhesion kinase

FFA:

Free fatty acid

FAO:

Fatty acid oxidation

GJ:

Gap junction

GLUT-4:

Glucose transporter 4

ICAM-1:

Intercellular cell adhesion molecule 1

IL:

Interleukin

iNOS:

Inducible nitric oxide synthase

IR:

Insulin resistance

LDL:

Low-density lipoprotein

LPS:

Lipopolysaccharide

MAPK:

Mitogen-activated protein kinase

MCP-1:

Monocyte chemoattractant protein 1

MCs:

Mast cells

MEF-2:

Myocyte Enhancer Factor 2

mTOR:

Mechanistic target of rapamycin

mtorc1:

MTOR complex 1

NO:

Nitric oxide

NOX2:

NADPH oxidase 2

NETs:

Neutrophil extracellular traps

NRP-1:

Neuropilin-1

OS:

Oscillatory shear

PAGs:

Proangiogenic granulocytes

PGs:

Prostaglandins

PKC:

Protein kinase C

PMN-MDSCs:

Polymorphonuclear marrow-derived myeloid cells

PS:

Pulsatile shear

RAAS:

Renin angiotensin aldosterone system

RhoA:

Ras Homolog Family Member A

ROS:

Reactive oxygen species

S1P:

Sphingosine 1-phosphate

S1PR1:

Sphingosine 1-phosphate receptor 1

SCFA:

Short-chain fatty acids

SGLT2:

Sodium-dependent glucose transporters 2

STAT3:

Signal transducer and activator of transcription 3

TAP:

Yes-Associated Protein

TGF-β:

Transforming growth factor β

TJ:

Tight junction

TMAO:

Trimethylamine-N-oxide

TNF-α:

Tumor necrosis factor α

VEGF:

Vascular endothelial growth factor

VEGFR2:

VEGF receptor 2

VSMCs:

Vascular smooth muscle cells

References

  1. Farooqui AA. High calorie diet and the human brain. Cham: Springer International Publishing; 2015 [cited 2022 Aug 16]. Available from: http://link.springer.com.

  2. International Diabetes Federation. IDF diabetes atlas 10th edition 2021[EB/OL]. (2021-12-13) [2022-08-16]. https://diabetesatlas.org/atlas/tenth-edition/.

  3. World Health Organization. Diabetes [EB/OL]. (2021-12-13) [2022-08-16]. https://www.who.int/news-room/fact-sheets/detail/diabetes.

  4. Lind M, Svensson A-M, Rosengren A. Glycemic control and excess mortality in type 1 diabetes. N Engl J Med. 2015;372:880–1.

    CAS  PubMed  Google Scholar 

  5. Tancredi M, Rosengren A, Svensson A-M, Kosiborod M, Pivodic A, Gudbjörnsdottir S, et al. Excess mortality among persons with type 2 diabetes. N Engl J Med. 2015;373:1720–32.

    Article  CAS  PubMed  Google Scholar 

  6. Beckman JA, Creager MA. Vascular complications of diabetes. Circ Res. 2016;118:1771–85.

    Article  CAS  PubMed  Google Scholar 

  7. Skyler JS. Primary and secondary prevention of type 1 diabetes. Diabet Med. 2013;30:161–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Yang J, Liu Z. Mechanistic pathogenesis of endothelial dysfunction in diabetic nephropathy and retinopathy. Front Endocrinol (Lausanne). 2022;13:816400.

    Article  PubMed  Google Scholar 

  9. Evans JR, Michelessi M, Virgili G. Laser photocoagulation for proliferative diabetic retinopathy. Cochrane Database Syst Rev. 2014;CD011234.

  10. Zhang X, Liu W, Wu S, Jin J, Li W, Wang N. Calcium dobesilate for diabetic retinopathy: a systematic review and meta-analysis. Sci China Life Sci. 2015;58:101–7.

    Article  CAS  PubMed  Google Scholar 

  11. Gilbert RE, Krum H. Heart failure in diabetes: effects of anti-hyperglycaemic drug therapy. Lancet. 2015;385:2107–17.

    Article  CAS  PubMed  Google Scholar 

  12. Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444:860–7.

    Article  CAS  PubMed  Google Scholar 

  13. Rohm TV, Meier DT, Olefsky JM, Donath MY. Inflammation in obesity, diabetes, and related disorders. Immunity. 2022;55:31–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Takeda Y, Matoba K, Sekiguchi K, Nagai Y, Yokota T, Utsunomiya K, et al. Endothelial dysfunction in diabetes. Biomedicines. 2020;8:E182.

    Article  Google Scholar 

  15. Ren B, Qin W, Wu F, Wang S, Pan C, Wang L, et al. Apigenin and naringenin regulate glucose and lipid metabolism, and ameliorate vascular dysfunction in type 2 diabetic rats. Eur J Pharmacol. 2016;773:13–23.

    Article  CAS  PubMed  Google Scholar 

  16. Samuel VT, Shulman GI. The pathogenesis of insulin resistance: integrating signaling pathways and substrate flux. J Clin Investig. 2016;126:12–22.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Shi Y, Vanhoutte PM. Macro- and microvascular endothelial dysfunction in diabetes. J Diabetes. 2017;9:434–49.

    Article  CAS  PubMed  Google Scholar 

  18. Matsumoto S, Shimabukuro M, Fukuda D, Soeki T, Yamakawa K, Masuzaki H, et al. Azilsartan, an angiotensin II type 1 receptor blocker, restores endothelial function by reducing vascular inflammation and by increasing the phosphorylation ratio Ser(1177)/Thr(497) of endothelial nitric oxide synthase in diabetic mice. Cardiovasc Diabetol. 2014;13:30.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Paneni F, Costantino S, Castello L, Battista R, Capretti G, Chiandotto S, et al. Targeting prolyl-isomerase Pin1 prevents mitochondrial oxidative stress and vascular dysfunction: insights in patients with diabetes. Eur Heart J. 2015;36:817–28.

    Article  CAS  PubMed  Google Scholar 

  20. Wimmer RA, Leopoldi A, Aichinger M, Wick N, Hantusch B, Novatchkova M, et al. Human blood vessel organoids as a model of diabetic vasculopathy. Nature. 2019;565:505–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Abu El-Asrar AM, Mohammad G, Nawaz MI, Abdelsaid M, Siddiquei MM, Alam K, et al. The chemokine platelet factor-4 variant (PF-4var)/CXCL4L1 inhibits diabetes-induced blood-retinal barrier breakdown. Invest Ophthalmol Vis Sci. 2015;56:1956–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Lee Y-J, Jung S-H, Kim S-H, Kim M-S, Lee S, Hwang J, et al. Essential role of transglutaminase 2 in vascular endothelial growth factor-induced vascular leakage in the retina of diabetic mice. Diabetes. 2016;65:2414–28.

    Article  CAS  PubMed  Google Scholar 

  23. Gimbrone MA, García-Cardeña G. Endothelial cell dysfunction and the pathobiology of atherosclerosis. Circ Res. 2016;118:620–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Rudraraju M, Narayanan SP, Somanath PR. Regulation of blood-retinal barrier cell-junctions in diabetic retinopathy. Pharmacol Res. 2020;161:105115.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Bazzoni G, Dejana E. Endothelial cell-to-cell junctions: molecular organization and role in vascular homeostasis. Physiol Rev. 2004;84:869–901.

    Article  CAS  PubMed  Google Scholar 

  26. Cong X, Kong W. Endothelial tight junctions and their regulatory signaling pathways in vascular homeostasis and disease. Cell Signal. 2020;66:109485.

    Article  CAS  PubMed  Google Scholar 

  27. Feng J, Liu Y, Singh AK, Ehsan A, Sellke N, Liang J, et al. Effects of diabetes and cardiopulmonary bypass on expression of Adherens junction proteins in human peripheral tissue. Surgery. 2017;161:823–9.

    Article  PubMed  Google Scholar 

  28. Dogné S, Flamion B, Caron N. Endothelial glycocalyx as a shield against diabetic vascular complications: involvement of hyaluronan and hyaluronidases. Arterioscler Thromb Vasc Biol. 2018;38:1427–39.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Mensah SA, Cheng MJ, Homayoni H, Plouffe BD, Coury AJ, Ebong EE. Regeneration of glycocalyx by heparan sulfate and sphingosine 1-phosphate restores inter-endothelial communication. PLoS ONE. 2017;12:e0186116.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Heiss C, Rodriguez-Mateos A, Kelm M. Central role of eNOS in the maintenance of endothelial homeostasis. Antioxid Redox Signal. 2015;22:1230–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Hiebert LM, Han J, Mandal AK. Glycosaminoglycans, hyperglycemia, and disease. Antioxid Redox Signal. 2014;21:1032–43.

    Article  CAS  PubMed  Google Scholar 

  32. Chakravarthy H, Navitskaya S, O’Reilly S, Gallimore J, Mize H, Beli E, et al. Role of acid sphingomyelinase in shifting the balance between proinflammatory and reparative bone marrow cells in diabetic retinopathy. Stem Cells. 2016;34:972–83.

    Article  CAS  PubMed  Google Scholar 

  33. Huwiler A, Pfeilschifter J. Recuperation of vascular homeostasis. Circ Res. 2021;129:237–9.

    Article  CAS  PubMed  Google Scholar 

  34. Proia RL, Hla T. Emerging biology of sphingosine-1-phosphate: its role in pathogenesis and therapy. J Clin Invest. 2015;125:1379–87.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Akhter MZ, Chandra Joshi J, Balaji Ragunathrao VA, Maienschein-Cline M, Proia RL, Malik AB, et al. Programming to S1PR1+ endothelial cells promotes restoration of vascular integrity. Circ Res. 2021;129:221–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Tong X, Lv P, Mathew AV, Liu D, Niu C, Wang Y, et al. The compensatory enrichment of sphingosine-1-phosphate harbored on glycated high-density lipoprotein restores endothelial protective function in type 2 diabetes mellitus. Cardiovasc Diabetol. 2014;13:82.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Kady N, Yan Y, Salazar T, Wang Q, Chakravarthy H, Huang C, et al. Increase in acid sphingomyelinase level in human retinal endothelial cells and CD34+ circulating angiogenic cells isolated from diabetic individuals is associated with dysfunctional retinal vasculature and vascular repair process in diabetes. J Clin Lipidol. 2017;11:694–703.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Zafar N, Krishnasamy SS, Shah J, Rai SN, Riggs DW, Bhatnagar A, et al. Circulating angiogenic stem cells in type 2 diabetes are associated with glycemic control and endothelial dysfunction. PLoS ONE. 2018;13:e0205851.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Kachamakova-Trojanowska N, Bukowska-Strakova K, Zukowska M, Dulak J, Jozkowicz A. The real face of endothelial progenitor cells—circulating angiogenic cells as endothelial prognostic marker? Pharmacol Rep. 2015;67:793–802.

    Article  CAS  PubMed  Google Scholar 

  40. Hahn C, Schwartz MA. Mechanotransduction in vascular physiology and atherogenesis. Nat Rev Mol Cell Biol. 2009;10:53–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. He M, Martin M, Marin T, Chen Z, Gongol B. Endothelial mechanobiology. APL Bioeng. 2020;4:010904.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Deng Q, Huo Y, Luo J. Endothelial mechanosensors: the gatekeepers of vascular homeostasis and adaptation under mechanical stress. Sci China Life Sci. 2014;57:755–62.

    Article  CAS  PubMed  Google Scholar 

  43. He J, Bao Q, Zhang Y, Liu M, Lv H, Liu Y, et al. Yes-Associated Protein promotes angiogenesis via signal transducer and activator of transcription 3 in endothelial cells. Circ Res. 2018;122:591–605.

    Article  CAS  PubMed  Google Scholar 

  44. Ortillon J, Le Bail J-C, Villard E, Léger B, Poirier B, Girardot C, et al. High glucose activates YAP signaling to promote vascular inflammation. Front Physiol. 2021;12:665994.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Wang L, Luo J-Y, Li B, Tian XY, Chen L-J, Huang Y, et al. Integrin-YAP/TAZ-JNK cascade mediates atheroprotective effect of unidirectional shear flow. Nature. 2016;540:579–82.

    Article  CAS  PubMed  Google Scholar 

  46. Han J, Zern BJ, Shuvaev VV, Davies PF, Muro S, Muzykantov V. Acute and chronic shear stress differently regulate endothelial internalization of nanocarriers targeted to platelet-endothelial cell adhesion molecule-1. ACS Nano. 2012;6:8824–36.

    Article  CAS  PubMed  Google Scholar 

  47. Peng Y-J, Nanduri J, Zhang X, Wang N, Raghuraman G, Seagard J, et al. Endothelin-1 mediates attenuated carotid baroreceptor activity by intermittent hypoxia. J Appl Physiol. 1985;2012(112):187–96.

    Google Scholar 

  48. Jacobsen TN, Converse RL, Victor RG. Contrasting effects of propranolol on sympathetic nerve activity and vascular resistance during orthostatic stress. Circulation. 1992;85:1072–6.

    Article  CAS  PubMed  Google Scholar 

  49. Machovich R. Blood coagulation-fibrinolytic system and endothelial cells. Acta Biochim Biophys Acad Sci Hung. 1985;20:135–53.

    CAS  PubMed  Google Scholar 

  50. Sohn SJ, Li D, Lee LK, Winoto A. Transcriptional regulation of tissue-specific genes by the ERK5 mitogen-activated protein kinase. Mol Cell Biol. 2005;25:8553–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Maiti D, Xu Z, Duh EJ. Vascular endothelial growth factor induces MEF2C and MEF2-dependent activity in endothelial cells. Invest Ophthalmol Vis Sci. 2008;49:3640–8.

    Article  PubMed  Google Scholar 

  52. Fledderus JO, Boon RA, Volger OL, Hurttila H, Ylä-Herttuala S, Pannekoek H, et al. KLF2 primes the antioxidant transcription factor Nrf2 for activation in endothelial cells. Arterioscler Thromb Vasc Biol. 2008;28:1339–46.

    Article  CAS  PubMed  Google Scholar 

  53. Lee D-Y, Lee C-I, Lin T-E, Lim SH, Zhou J, Tseng Y-C, et al. Role of histone deacetylases in transcription factor regulation and cell cycle modulation in endothelial cells in response to disturbed flow. Proc Natl Acad Sci U S A. 2012;109:1967–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Lazarus A, Keshet E. Vascular endothelial growth factor and vascular homeostasis. Proc Am Thorac Soc. 2011;8:508–11.

    Article  CAS  PubMed  Google Scholar 

  55. Colotti G, Failla CM, Lacal PM, Ungarelli M, Ruffini F, Di Micco P, et al. Neuropilin-1 is required for endothelial cell adhesion to soluble vascular endothelial growth factor receptor 1. FEBS J. 2022;289:183–98.

    Article  CAS  PubMed  Google Scholar 

  56. Wilhelm K, Happel K, Eelen G, Schoors S, Oellerich MF, Lim R, et al. FOXO1 couples metabolic activity and growth state in the vascular endothelium. Nature. 2016;529:216–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Chistiakov DA, Orekhov AN, Bobryshev YV. The role of miR-126 in embryonic angiogenesis, adult vascular homeostasis, and vascular repair and its alterations in atherosclerotic disease. J Mol Cell Cardiol. 2016;97:47–55.

    Article  CAS  PubMed  Google Scholar 

  58. Lüneburg N, Siques P, Brito J, Arriaza K, Pena E, Klose H, et al. Long-term chronic intermittent hypobaric hypoxia in rats causes an imbalance in the asymmetric dimethylarginine/nitric oxide pathway and ROS activity: a possible synergistic mechanism for altitude pulmonary hypertension? Pulm Med. 2016;2016:6578578.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Papachristoforou E, Lambadiari V, Maratou E, Makrilakis K. Association of glycemic indices (hyperglycemia, glucose variability, and hypoglycemia) with oxidative stress and diabetic complications. J Diabetes Res. 2020;2020:7489795.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Romzova M, Hohenadel D, Kolostova K, Pinterova D, Fojtikova M, Ruzickova S, et al. NFkappaB and its inhibitor IkappaB in relation to type 2 diabetes and its microvascular and atherosclerotic complications. Hum Immunol. 2006;67:706–13.

    Article  CAS  PubMed  Google Scholar 

  61. Rendra E, Riabov V, Mossel DM, Sevastyanova T, Harmsen MC, Kzhyshkowska J. Reactive oxygen species (ROS) in macrophage activation and function in diabetes. Immunobiology. 2019;224:242–53.

    Article  CAS  PubMed  Google Scholar 

  62. Moldogazieva NT, Mokhosoev IM, Mel’nikova TI, Zavadskiy SP, Kuz’menko AN, Terentiev AA. Dual character of reactive oxygen, nitrogen, and halogen species: endogenous sources, interconversions and neutralization. Biochemistry (Mosc). 2020;85:S56–78.

    Article  CAS  PubMed  Google Scholar 

  63. Takahashi M, Ishida T, Traub O, Corson MA, Berk BC. Mechanotransduction in endothelial cells: temporal signaling events in response to shear stress. J Vasc Res. 1997;34:212–9.

    Article  CAS  PubMed  Google Scholar 

  64. Förstermann U, Xia N, Li H. Roles of vascular oxidative stress and nitric oxide in the pathogenesis of atherosclerosis. Circ Res. 2017;120:713–35.

    Article  PubMed  Google Scholar 

  65. Loscalzo J. The identification of nitric oxide as endothelium-derived relaxing factor. Circ Res. 2013;113:100–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Xu T, Lv Z, Chen Q, Guo M, Wang X, Huang F. Vascular endothelial growth factor over-expressed mesenchymal stem cells-conditioned media ameliorate palmitate-induced diabetic endothelial dysfunction through PI-3K/AKT/m-TOR/eNOS and p38/MAPK signaling pathway. Biomed Pharmacother. 2018;106:491–8.

    Article  CAS  PubMed  Google Scholar 

  67. Wang W, Wang Y, Long J, Wang J, Haudek SB, Overbeek P, et al. Mitochondrial fission triggered by hyperglycemia is mediated by ROCK1 activation in podocytes and endothelial cells. Cell Metab. 2012;15:186–200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Kim D, Roy S. Effects of diabetes on mitochondrial morphology and its implications in diabetic retinopathy. Invest Ophthalmol Vis Sci. 2020;61:10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Mameli E, Martello A, Caporali A. Autophagy at the interface of endothelial cell homeostasis and vascular disease. FEBS J. 2022;289:2976–91.

    Article  CAS  PubMed  Google Scholar 

  70. Spengler K, Kryeziu N, Große S, Mosig AS, Heller R. VEGF triggers transient induction of autophagy in endothelial cells via AMPKα1. Cells. 2020;9:687.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Luo E-F, Li H-X, Qin Y-H, Qiao Y, Yan G-L, Yao Y-Y, et al. Role of ferroptosis in the process of diabetes-induced endothelial dysfunction. World J Diabetes. 2021;12:124–37.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Meng Z, Liang H, Zhao J, Gao J, Liu C, Ma X, et al. HMOX1 upregulation promotes ferroptosis in diabetic atherosclerosis. Life Sci. 2021;284:119935.

    Article  CAS  PubMed  Google Scholar 

  73. Zalewski PD, Beltrame JF, Wawer AA, Abdo AI, Murgia C. Roles for endothelial zinc homeostasis in vascular physiology and coronary artery disease. Crit Rev Food Sci Nutr. 2019;59:3511–25.

    Article  CAS  PubMed  Google Scholar 

  74. Stafford N, Wilson C, Oceandy D, Neyses L, Cartwright EJ. The plasma membrane calcium ATPases and their role as major new players in human disease. Physiol Rev. 2017;97:1089–125.

    Article  CAS  PubMed  Google Scholar 

  75. Bhattacharya R, Wang E, Dutta SK, Vohra PK, Guangqi E, Prakash YS, et al. NHERF-2 maintains endothelial homeostasis. Blood. 2012;119:4798–806.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Bakker W, Eringa EC, Sipkema P, van Hinsbergh VWM. Endothelial dysfunction and diabetes: roles of hyperglycemia, impaired insulin signaling and obesity. Cell Tissue Res. 2009;335:165–89.

    Article  CAS  PubMed  Google Scholar 

  77. Ighodaro OM. Molecular pathways associated with oxidative stress in diabetes mellitus. Biomed Pharmacother. 2018;108:656–62.

    Article  CAS  PubMed  Google Scholar 

  78. Clyne AM. Endothelial response to glucose: dysfunction, metabolism, and transport. Biochem Soc T. 2021;49:313–25.

    Article  CAS  Google Scholar 

  79. Shen C-Y, Lu C-H, Wu C-H, Li K-J, Kuo Y-M, Hsieh S-C, et al. The development of maillard reaction, and advanced glycation end product (AGE)-receptor for AGE (RAGE) signaling inhibitors as novel therapeutic strategies for patients with AGE-related diseases. Molecules. 2020;25:E5591.

    Article  Google Scholar 

  80. Tang G, Li S, Zhang C, Chen H, Wang N, Feng Y. Clinical efficacies, underlying mechanisms and molecular targets of Chinese medicines for diabetic nephropathy treatment and management. Acta Pharm Sin B. 2021;11:2749–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Fujii EY, Nakayama M. The measurements of RAGE, VEGF, and AGEs in the plasma and follicular fluid of reproductive women: the influence of aging. Fertil Steril. 2010;94:694–700.

    Article  CAS  PubMed  Google Scholar 

  82. Gong DJ, Wang L, Yang YY, Zhang JJ, Liu XH. Diabetes aggravates renal ischemia and reperfusion injury in rats by exacerbating oxidative stress, inflammation, and apoptosis. Ren Fail. 2019;41:750–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Kaur R, Kaur M, Singh J. Endothelial dysfunction and platelet hyperactivity in type 2 diabetes mellitus: molecular insights and therapeutic strategies. Cardiovasc Diabetol. 2018;17:121.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Wu F, Wang H, Li J, Liang J, Ma S. Homoplantaginin modulates insulin sensitivity in endothelial cells by inhibiting inflammation. Biol Pharm Bull. 2012;35:1171–7.

    Article  CAS  PubMed  Google Scholar 

  85. Cheng C-I, Lee Y-H, Chen P-H, Lin Y-C, Chou M-H, Kao Y-H. Free fatty acids induce autophagy and LOX-1 upregulation in cultured aortic vascular smooth muscle cells. J Cell Biochem. 2017;118:1249–61.

    Article  CAS  PubMed  Google Scholar 

  86. Patella F, Schug ZT, Persi E, Neilson LJ, Erami Z, Avanzato D, et al. Proteomics-based metabolic modeling reveals that fatty acid oxidation (FAO) controls endothelial cell (EC) permeability. Mol Cell Proteomics. 2015;14:621–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Kalucka J, Bierhansl L, Conchinha NV, Missiaen R, Elia I, Brüning U, et al. Quiescent endothelial cells upregulate fatty acid β-oxidation for vasculoprotection via redox homeostasis. Cell Metab. 2018;28:881–894.e13.

    Article  CAS  PubMed  Google Scholar 

  88. Akhter F, Khan MS, Alatar AA, Faisal M, Ahmad S. Antigenic role of the adaptive immune response to d-ribose glycated LDL in diabetes, atherosclerosis and diabetes atherosclerotic patients. Life Sci. 2016;151:139–46.

    Article  CAS  PubMed  Google Scholar 

  89. Jin J-L, Zhang H-W, Cao Y-X, Liu H-H, Hua Q, Li Y-F, et al. Association of small dense low-density lipoprotein with cardiovascular outcome in patients with coronary artery disease and diabetes: a prospective, observational cohort study. Cardiovasc Diabetol. 2020;19:45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Zhang Q, Liu J, Duan H, Li R, Peng W, Wu C. Activation of Nrf2/HO-1 signaling: an important molecular mechanism of herbal medicine in the treatment of atherosclerosis via the protection of vascular endothelial cells from oxidative stress. J Adv Res. 2021;34:43–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Agus A, Clément K, Sokol H. Gut microbiota-derived metabolites as central regulators in metabolic disorders. Gut. 2021;70:1174–82.

    Article  CAS  PubMed  Google Scholar 

  92. Canyelles M, Tondo M, Cedó L, Farràs M, Escolà-Gil JC, Blanco-Vaca F. Trimethylamine N-oxide: a link among diet, gut microbiota, gene regulation of liver and intestine Cholesterol homeostasis and HDL function. Int J Mol Sci. 2018;19:E3228.

    Article  Google Scholar 

  93. Matsumoto T, Kojima M, Takayanagi K, Taguchi K, Kobayashi T. Role of S-equol, indoxyl sulfate, and trimethylamine N-oxide on vascular function. Am J Hypertens. 2020;33:793–803.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Wang B, Wu L, Chen J, Dong L, Chen C, Wen Z, et al. Metabolism pathways of arachidonic acids: mechanisms and potential therapeutic targets. Signal Transduct Tar. 2021;6:94.

    Article  CAS  Google Scholar 

  95. Félétou M, Huang Y, Vanhoutte PM. Endothelium-mediated control of vascular tone: COX-1 and COX-2 products. Brit J Pharmacol. 2011;164:894–912.

    Article  Google Scholar 

  96. Csanyi G, Lepran I, Flesch T, Telegdy G, Szabo G, Mezei Z. Lack of endothelium-derived hyperpolarizing factor (EDHF) up-regulation in endothelial dysfunction in aorta in diabetic rats. Pharmacol Rep. 2007;59:447–55.

    CAS  PubMed  Google Scholar 

  97. Imig JD. Eicosanoid blood vessel regulation in physiological and pathological states. Clin Sci (Lond). 2020;134:2707–27.

    Article  CAS  PubMed  Google Scholar 

  98. Pinheiro Júnior JEG, Moraes PZ, Rodriguez MD, Simões MR, Cibin F, Pinton S, et al. Cadmium exposure activates NADPH oxidase, renin-angiotensin system and cyclooxygenase 2 pathways in arteries, inducing hypertension and vascular damage. Toxicol Lett. 2020;333:80–9.

    Article  PubMed  Google Scholar 

  99. Ma T, Li X, Zhu Y, Yu S, Liu T, Zhang X, et al. Excessive activation of notch signaling in macrophages promote kidney inflammation, fibrosis, and necroptosis. Front Immunol. 2022;13:835879.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Xue J, Schmidt SV, Sander J, Draffehn A, Krebs W, Quester I, et al. Transcriptome-based network analysis reveals a spectrum model of human macrophage activation. Immunity. 2014;40:274–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Torres-Castro I, Arroyo-Camarena ÚD, Martínez-Reyes CP, Gómez-Arauz AY, Dueñas-Andrade Y, Hernández-Ruiz J, et al. Human monocytes and macrophages undergo M1-type inflammatory polarization in response to high levels of glucose. Immunol Lett. 2016;176:81–9.

    Article  CAS  PubMed  Google Scholar 

  102. Bajpai A, Tilley DG. The role of leukocytes in diabetic cardiomyopathy. Front Physiol. 2018;9:1547.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Vasamsetti SB, Karnewar S, Kanugula AK, Thatipalli AR, Kumar JM, Kotamraju S. Metformin inhibits monocyte-to-macrophage differentiation via AMPK-mediated inhibition of STAT3 activation: potential role in atherosclerosis. Diabetes. 2015;64:2028–41.

    Article  CAS  PubMed  Google Scholar 

  104. Marfella R, Esposito K, Giunta R, Coppola G, De Angelis L, Farzati B, et al. Circulating adhesion molecules in humans: role of hyperglycemia and hyperinsulinemia. Circulation. 2000;101:2247–51.

    Article  CAS  PubMed  Google Scholar 

  105. Lv Y, Kim K, Sheng Y, Cho J, Qian Z, Zhao Y-Y, et al. YAP controls endothelial activation and vascular inflammation through TRAF6. Circ Res. 2018;123:43–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Yang S, Yuan H-Q, Hao Y-M, Ren Z, Qu S-L, Liu L-S, et al. Macrophage polarization in atherosclerosis. Clin Chim Acta. 2020;501:142–6.

    Article  CAS  PubMed  Google Scholar 

  107. Linton MF, Moslehi JJ, Babaev VR. Akt signaling in macrophage polarization, survival, and atherosclerosis. Int J Mol Sci. 2019;20:E2703.

    Article  Google Scholar 

  108. Wågsäter D, Zhu C, Björkegren J, Skogsberg J, Eriksson P. MMP-2 and MMP-9 are prominent matrix metalloproteinases during atherosclerosis development in the Ldlr(-/-)Apob(100/100) mouse. Int J Mol Med. 2011;28:247–53.

    PubMed  Google Scholar 

  109. Kolaczkowska E, Kubes P. Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol. 2013;13:159–75.

    Article  CAS  PubMed  Google Scholar 

  110. Bajpai A, Nadkarni S, Neidrauer M, Weingarten MS, Lewin PA, Spiller KL. Effects of non-thermal, non-cavitational ultrasound exposure on human diabetic ulcer healing and inflammatory gene expression in a pilot study. Ultrasound Med Biol. 2018;44:2043–9.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Yu Y, Lin Q, Ye D, Wang Y, He B, Li Y, et al. Neutrophil count as a reliable marker for diabetic kidney disease in autoimmune diabetes. BMC Endocr Disord. 2020;20:158.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Veglia F, Sanseviero E, Gabrilovich DI. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat Rev Immunol. 2021;21:485–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Wierusz-Wysocka B, Wykretowicz A, Byks H, Sadurska K, Wysocki H. Polymorphonuclear neutrophils adherence, superoxide anion (O2) production and HBA1 level in diabetic patients. Diabetes Res Clin Pract. 1993;21:109–14.

    Article  CAS  PubMed  Google Scholar 

  114. Fardon NJM, Wilkinson R, Thomas TH. Abnormalities in primary granule exocytosis in neutrophils from type I diabetic patients with nephropathy. Clin Sci (Lond). 2002;102:69–75.

    Article  CAS  PubMed  Google Scholar 

  115. Cappellari R, D’Anna M, Menegazzo L, Bonora BM, Albiero M, Avogaro A, et al. Diabetes mellitus impairs circulating proangiogenic granulocytes. Diabetologia. 2020;63:1872–84.

    Article  CAS  PubMed  Google Scholar 

  116. Massena S, Christoffersson G, Vågesjö E, Seignez C, Gustafsson K, Binet F, et al. Identification and characterization of VEGF-A—responsive neutrophils expressing CD49d, VEGFR1, and CXCR4 in mice and humans. Blood. 2015;126:2016–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Fukui M, Tanaka M, Hamaguchi M, Senmaru T, Sakabe K, Shiraishi E, et al. Eosinophil count is positively correlated with albumin excretion rate in men with type 2 diabetes. Clin J Am Soc Nephrol. 2009;4:1761–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Wheelock KM, Saulnier P-J, Tanamas SK, Vijayakumar P, Weil EJ, Looker HC, et al. White blood cell fractions correlate with lesions of diabetic kidney disease and predict loss of kidney function in type 2 diabetes. Nephrol Dial Transplant. 2018;33:1001–9.

    Article  CAS  PubMed  Google Scholar 

  119. Castanheira FVS, Kubes P. Neutrophils and NETs in modulating acute and chronic inflammation. Blood. 2019;133:2178–85.

    Article  CAS  PubMed  Google Scholar 

  120. Rada B, Jendrysik MA, Pang L, Hayes CP, Yoo D-G, Park JJ, et al. Pyocyanin-enhanced neutrophil extracellular trap formation requires the NADPH oxidase. PLoS ONE. 2013;8:e54205.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Westerterp M, Fotakis P, Ouimet M, Bochem AE, Zhang H, Molusky MM, et al. Cholesterol efflux pathways suppress inflammasome activation, NETosis, and atherogenesis. Circulation. 2018;138:898–912.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Pleskovič A, Vraspir-Porenta O, Zorc-Pleskovič R, Petrovič D, Zorc M, Milutinović A. Deficiency of mast cells in coronary artery endarterectomy of male patients with type 2 diabetes. Cardiovasc Diabetol. 2011;10:40.

    Article  PubMed  PubMed Central  Google Scholar 

  123. McNeil HP, Adachi R, Stevens RL. Mast cell-restricted tryptases: structure and function in inflammation and pathogen defense. J Biol Chem. 2007;282:20785–9.

    Article  CAS  PubMed  Google Scholar 

  124. Abraham SN, St John AL. Mast cell-orchestrated immunity to pathogens. Nat Rev Immunol. 2010;10:440–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Théry C, Amigorena S. The cell biology of antigen presentation in dendritic cells. Curr Opin Immunol. 2001;13:45–51.

    Article  PubMed  Google Scholar 

  126. Gardner A, de Mingo PÁ, Ruffell B. Dendritic cells and their role in immunotherapy. Front Immunol. 2020;11:924.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Kim H, Kim M, Lee HY, Park HY, Jhun H, Kim S. Role of dendritic cell in diabetic nephropathy. Int J Mol Sci. 2021;22(14):7554. 

  128. Kaunisto MA, Sjölind L, Sallinen R, Pettersson-Fernholm K, Saraheimo M, Fröjdö S, et al. Elevated MBL concentrations are not an indication of association between the MBL2 gene and type 1 diabetes or diabetic nephropathy. Diabetes. 2009;58:1710–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Tang SCW, Yiu WH. Innate immunity in diabetic kidney disease. Nat Rev Nephrol. 2020;16:206–22.

    Article  CAS  PubMed  Google Scholar 

  130. Li L, Chen L, Zang J, Tang X, Liu Y, Zhang J, et al. C3a and C5a receptor antagonists ameliorate endothelial-myofibroblast transition via the Wnt/β-catenin signaling pathway in diabetic kidney disease. Metabolism. 2015;64:597–610.

    Article  CAS  PubMed  Google Scholar 

  131. Portilla D, Xavier S. Role of intracellular complement activation in kidney fibrosis. Brit J Pharmacol. 2021;178:2880–91.

    Article  CAS  Google Scholar 

  132. Golec E, Ekström A, Noga M, Omar-Hmeadi M, Lund P-E, Villoutreix BO, et al. Alternative splicing encodes functional intracellular CD59 isoforms that mediate insulin secretion and are down-regulated in diabetic islets. Proc Natl Acad Sci U S A. 2022;119:e2120083119.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Clark M, Kroger CJ, Tisch RM. Type 1 diabetes: a chronic anti-self-inflammatory response. Front Immunol. 2017;8:1898.

    Article  PubMed  PubMed Central  Google Scholar 

  134. Burhans MS, Hagman DK, Kuzma JN, Schmidt KA, Kratz M. Contribution of adipose tissue inflammation to the development of type 2 diabetes mellitus. Compr Physiol. 2018;9:1–58.

    PubMed  PubMed Central  Google Scholar 

  135. Alrabiah M, Al-Aali KA, Al-Sowygh ZH, Binmahfooz AM, Mokeem SA, Abduljabbar T. Association of advanced glycation end products with peri-implant inflammation in prediabetes and type 2 diabetes mellitus patients. Clin Implant Dent Relat Res. 2018;20:535–40.

    Article  PubMed  Google Scholar 

  136. Miya A, Nakamura A, Miyoshi H, Takano Y, Sunagoya K, Hayasaka K, et al. Impact of glucose loading on variations in CD4+ and CD8+ T cells in Japanese participants with or without type 2 diabetes. Front Endocrinol (Lausanne). 2018;9:81.

    Article  PubMed  PubMed Central  Google Scholar 

  137. Liang C, Yang KY, Chan VW, Li X, Fung THW, Wu Y, et al. CD8+ T-cell plasticity regulates vascular regeneration in type-2 diabetes. Theranostics. 2020;10:4217–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Martin S, Tesse A, Hugel B, Martínez MC, Morel O, Freyssinet J-M, et al. Shed membrane particles from T lymphocytes impair endothelial function and regulate endothelial protein expression. Circulation. 2004;109:1653–9.

    Article  PubMed  Google Scholar 

  139. Wu C-C, Sytwu H-K, Lu K-C, Lin Y-F. Role of T cells in type 2 diabetic nephropathy. Exp Diabetes Res. 2011;2011:514738.

    Article  PubMed  PubMed Central  Google Scholar 

  140. Smith MJ, Simmons KM, Cambier JC. B cells in type 1 diabetes mellitus and diabetic kidney disease. Nat Rev Nephrol. 2017. p. 712–20.

  141. Jetten N, Verbruggen S, Gijbels MJ, Post MJ, De Winther MPJ, Donners MMPC. Anti-inflammatory M2, but not pro-inflammatory M1 macrophages promote angiogenesis in vivo. Angiogenesis. 2014;17:109–18.

    Article  CAS  PubMed  Google Scholar 

  142. Naylor D, Sharma A, Li Z, Monteith G, Mallard BA, Bergeron R, et al. Endotoxin-induced cytokine, chemokine and white blood cell profiles of variable stress-responding sheep. Stress. 2021;24:888–97.

    Article  CAS  PubMed  Google Scholar 

  143. Salvador B, Arranz A, Francisco S, Córdoba L, Punzón C, Llamas MÁ, et al. Modulation of endothelial function by Toll like receptors. Pharmacol Res. 2016;108:46–56.

    Article  CAS  PubMed  Google Scholar 

  144. Fuentes-Antrás J, Ioan AM, Tuñón J, Egido J, Lorenzo O. Activation of Toll-like receptors and inflammasome complexes in the diabetic cardiomyopathy-associated inflammation. Int J Endocrinol. 2014;2014:847827.

    Article  PubMed  PubMed Central  Google Scholar 

  145. Baroja-Mazo A, Martín-Sánchez F, Gomez AI, Martínez CM, Amores-Iniesta J, Compan V, et al. The NLRP3 inflammasome is released as a particulate danger signal that amplifies the inflammatory response. Nat Immunol. 2014;15:738–48.

    Article  CAS  PubMed  Google Scholar 

  146. Dasu MR, Devaraj S, Park S, Jialal I. Increased toll-like receptor (TLR) activation and TLR ligands in recently diagnosed type 2 diabetic subjects. Diabetes Care. 2010;33:861–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Hu R, Wang M-Q, Ni S-H, Wang M, Liu L-Y, You H-Y, et al. Salidroside ameliorates endothelial inflammation and oxidative stress by regulating the AMPK/NF-κB/NLRP3 signaling pathway in AGEs-induced HUVECs. Eur J Pharmacol. 2020;867:172797.

    Article  CAS  PubMed  Google Scholar 

  148. Gupta A, Singh K, Fatima S, Ambreen S, Zimmermann S, Younis R, et al. Neutrophil extracellular traps promote NLRP3 inflammasome activation and glomerular endothelial dysfunction in diabetic kidney disease. Nutrients. 2022;14:2965.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Lee J, Lee S, Zhang H, Hill MA, Zhang C, Park Y. Interaction of IL-6 and TNF-α contributes to endothelial dysfunction in type 2 diabetic mouse hearts. PLoS ONE. 2017;12:e0187189.

    Article  PubMed  PubMed Central  Google Scholar 

  150. Garbers C, Aparicio-Siegmund S, Rose-John S. The IL-6/gp130/STAT3 signaling axis: recent advances towards specific inhibition. Curr Opin Immunol. 2015;34:75–82.

    Article  CAS  PubMed  Google Scholar 

  151. Rose-John S. Interleukin-6 family cytokines. Cold Spring Harb Perspect Biol. 2018;10:a028415.

    Article  PubMed  PubMed Central  Google Scholar 

  152. Singh S, Anshita D, Ravichandiran V. MCP-1: function, regulation, and involvement in disease. Int Immunopharmacol. 2021;101:107598.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Conti P, DiGioacchino M. MCP-1 and RANTES are mediators of acute and chronic inflammation. Allergy Asthma Proc. 2001;22:133–7.

    Article  CAS  PubMed  Google Scholar 

  154. Viedt C, Orth SR. Monocyte chemoattractant protein-1 (MCP-1) in the kidney: does it more than simply attract monocytes? Nephrol Dial Transplant. 2002;17:2043–7.

    Article  CAS  PubMed  Google Scholar 

  155. Ma P, Zhang P, Chen S, Shi W, Ye J, Chen S, et al. Immune cell landscape of patients with diabetic macular edema by single-cell RNA analysis. Front Pharmacol. 2021;12:754933.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Li Y, Baccouche B, Olayinka O, Serikbaeva A, Kazlauskas A. The role of the Wnt pathway in VEGF/anti-VEGF-dependent control of the endothelial cell barrier. Invest Ophthalmol Vis Sci. 2021;62:17.

    PubMed  PubMed Central  Google Scholar 

  157. Kida T, Oku H, Osuka S, Horie T, Ikeda T. Hyperglycemia-induced VEGF and ROS production in retinal cells is inhibited by the mTOR inhibitor, rapamycin. Sci Rep. 2021;11:1885.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Shi S, Song L, Yu H, Feng S, He J, Liu Y, et al. Knockdown of LncRNA-H19 ameliorates kidney fibrosis in diabetic mice by suppressing miR-29a-mediated EndMT. Front Pharmacol. 2020;11:586895.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Son M, Oh S, Jang JT, Son KH, Byun K. Pyrogallol-phloroglucinol-6 6-bieckol on attenuates high-fat diet-induced hypertension by modulating endothelial-to-mesenchymal transition in the aorta of mice. Oxid Med Cell Longev. 2021;2021:8869085.

    Article  PubMed  PubMed Central  Google Scholar 

  160. Tian J, Zhang M, Suo M, Liu D, Wang X, Liu M, et al. Dapagliflozin alleviates cardiac fibrosis through suppressing EndMT and fibroblast activation via AMPKα/TGF-β/Smad signalling in type 2 diabetic rats. J Cell Mol Med. 2021;25:7642–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Thomas AA, Biswas S, Feng B, Chen S, Gonder J, Chakrabarti S. lncRNA H19 prevents endothelial-mesenchymal transition in diabetic retinopathy. Diabetologia. 2019;62:517–30.

    Article  CAS  PubMed  Google Scholar 

  162. Tuleta I, Frangogiannis NG. Diabetic fibrosis. Biochim Biophys Acta Mol Basis Dis. 2021;1867:166044.

    Article  CAS  PubMed  Google Scholar 

  163. Wu M-Y, Yiang G-T, Lai T-T, Li C-J. The oxidative stress and mitochondrial dysfunction during the pathogenesis of diabetic retinopathy. Oxid Med Cell Longev. 2018;2018:3420187.

    Article  PubMed  PubMed Central  Google Scholar 

  164. Odegaard AO, Jacobs DR, Sanchez OA, Goff DC, Reiner AP, Gross MD. Oxidative stress, inflammation, endothelial dysfunction and incidence of type 2 diabetes. Cardiovasc Diabetol. 2016;15:51.

    Article  PubMed  PubMed Central  Google Scholar 

  165. El Assar M, Angulo J, Rodríguez-Mañas L. Oxidative stress and vascular inflammation in aging. Free Radical Bio Med. 2013;65:380–401.

    Article  Google Scholar 

  166. Grassi D, Desideri G, Ferri L, Aggio A, Tiberti S, Ferri C. Oxidative stress and endothelial dysfunction: say NO to cigarette smoking! Curr Pharm Design. 2010;16:2539–50.

    Article  CAS  Google Scholar 

  167. Meng T, Qin W, Liu B. SIRT1 antagonizes oxidative stress in diabetic vascular complication. Front Endocrinol. 2020;11:568861.

    Article  Google Scholar 

  168. McMahon M, Lamont DJ, Beattie KA, Hayes JD. Keap1 perceives stress via three sensors for the endogenous signaling molecules nitric oxide, zinc, and alkenals. Proc Natl Acad Sci U S A. 2010;107:18838–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Andrade J, Shi C, Costa ASH, Choi J, Kim J, Doddaballapur A, et al. Control of endothelial quiescence by FOXO-regulated metabolites. Nat Cell Biol. 2021;23:413–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Ku S-K, Bae J-S. Vicenin-2 and scolymoside inhibit high-glucose-induced vascular inflammation in vitro and in vivo. Can J Physiol Pharmacol. 2016;94:287–95.

    Article  CAS  PubMed  Google Scholar 

  171. Gnudi L, Benedetti S, Woolf AS, Long DA. Vascular growth factors play critical roles in kidney glomeruli. Clin Sci (Lond). 2015;129:1225–36.

    Article  CAS  PubMed  Google Scholar 

  172. Liu J, Zhao Z, Willcox MDP, Xu B, Shi B. Multiplex bead analysis of urinary cytokines of type 2 diabetic patients with normo- and microalbuminuria. J Immunoassay Immunochem. 2010;31:279–89.

    Article  PubMed  Google Scholar 

  173. Zhu JJ, Wang BF, Hong YZ, Yang XC. Effect of triptolide on the expression of RANTES in the renal tissue of diabetic nephropathy rats. Zhongguo Zhong Xi Yi Jie He Za Zhi. 2014;34:1231–7.

    CAS  PubMed  Google Scholar 

  174. Meng X-M, Nikolic-Paterson DJ, Lan HY. Inflammatory processes in renal fibrosis. Nat Rev Nephrol. 2014;10:493–503.

    Article  CAS  PubMed  Google Scholar 

  175. Mack M, Yanagita M. Origin of myofibroblasts and cellular events triggering fibrosis. Kidney Int. 2015;87:297–307.

    Article  PubMed  Google Scholar 

  176. Lechner J, O’Leary OE, Stitt AW. The pathology associated with diabetic retinopathy. Vision Res. 2017;139:7–14.

    Article  PubMed  Google Scholar 

  177. Pan WW, Lin F, Fort PE. The innate immune system in diabetic retinopathy. Prog Retin Eye Res. 2021;84:100940.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Xu H, Chen M. Diabetic retinopathy and dysregulated innate immunity. Vision Res. 2017;139:39–46.

    Article  PubMed  Google Scholar 

  179. Dick AD, Carter D, Robertson M, Broderick C, Hughes E, Forrester JV, et al. Control of myeloid activity during retinal inflammation. J Leukoc Biol. 2003;74:161–6.

    Article  CAS  PubMed  Google Scholar 

  180. Combadière C, Feumi C, Raoul W, Keller N, Rodéro M, Pézard A, et al. CX3CR1-dependent subretinal microglia cell accumulation is associated with cardinal features of age-related macular degeneration. J Clin Invest. 2007;117:2920–8.

    Article  PubMed  PubMed Central  Google Scholar 

  181. Forrester JV, Kuffova L, Delibegovic M. The role of inflammation in diabetic retinopathy. Front Immunol. 2020;11:583687.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. van de Weijer T, Schrauwen-Hinderling VB, Schrauwen P. Lipotoxicity in type 2 diabetic cardiomyopathy. Cardiovasc Res. 2011;92:10–8.

    Article  PubMed  Google Scholar 

  183. Sowton AP, Griffin JL, Murray AJ. Metabolic profiling of the diabetic heart: toward a richer picture. Front Physiol. 2019;10:639.

    Article  PubMed  PubMed Central  Google Scholar 

  184. Peng M-L, Fu Y, Wu C-W, Zhang Y, Ren H, Zhou S-S. Signaling pathways related to oxidative stress in diabetic cardiomyopathy. Front Endocrinol (Lausanne). 2022;13:907757.

    Article  PubMed  Google Scholar 

  185. Dabravolski SA, Sadykhov NK, Kartuesov AG, Borisov EE, Sukhorukov VN, Orekhov AN. The role of mitochondrial abnormalities in diabetic cardiomyopathy. Int J Mol Sci. 2022;23:7863.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Li S-Y, Yang X, Ceylan-Isik AF, Du M, Sreejayan N, Ren J. Cardiac contractile dysfunction in Lep/Lep obesity is accompanied by NADPH oxidase activation, oxidative modification of sarco(endo)plasmic reticulum Ca2+-ATPase and myosin heavy chain isozyme switch. Diabetologia. 2006;49:1434–46.

    Article  CAS  PubMed  Google Scholar 

  187. Erickson JR, Pereira L, Wang L, Han G, Ferguson A, Dao K, et al. Diabetic hyperglycaemia activates CaMKII and arrhythmias by O-linked glycosylation. Nature. 2013;502:372–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Alnaim L, Altuwaym RA, Aldehan SM, Alquraishi NM. Assessment of knowledge among caregivers of diabetic patients in insulin dosage regimen and administration. Saudi Pharm J. 2021;29:1137–42.

    Article  PubMed  PubMed Central  Google Scholar 

  189. Haller H, Ito S, Izzo JL, Januszewicz A, Katayama S, Menne J, et al. Olmesartan for the delay or prevention of microalbuminuria in type 2 diabetes. N Engl J Med. 2011;364:907–17.

    Article  CAS  PubMed  Google Scholar 

  190. Umemura S, Arima H, Arima S, Asayama K, Dohi Y, Hirooka Y, et al. The Japanese Society of Hypertension Guidelines for the management of hypertension (JSH 2019). Hypertens Res. 2019;42:1235–481.

    Article  PubMed  Google Scholar 

  191. Bhatt DL, Eikelboom JW, Connolly SJ, Steg PG, Anand SS, Verma S, et al. Role of combination antiplatelet and anticoagulation therapy in diabetes mellitus and cardiovascular disease: insights from the COMPASS trial. Circulation. 2020;141:1841–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Banerjee S, Brown A, McGahan L, Asakawa K, Hutton B, Clark M, et al. Clopidogrel versus other antiplatelet agents for secondary prevention of vascular events in adults with acute coronary syndrome or peripheral vascular disease: clinical and cost-effectiveness analyses. CADTH Technol Overv. 2012;2:e2102.

    CAS  PubMed  PubMed Central  Google Scholar 

  193. Rajbhandari J, Fernandez CJ, Agarwal M, Yeap BXY, Pappachan JM. Diabetic heart disease: a clinical update. World J Diabetes. 2021;12:383–406.

    Article  PubMed  PubMed Central  Google Scholar 

  194. Ravera M, Ratto E, Vettoretti S, Parodi D, Deferrari G. Prevention and treatment of diabetic nephropathy: the program for irbesartan mortality and morbidity evaluation. JASN. 2005;16:S48–52.

    Article  CAS  PubMed  Google Scholar 

  195. Han S-Y, Kim C-H, Kim H-S, Jee Y-H, Song H-K, Lee M-H, et al. Spironolactone prevents diabetic nephropathy through an anti-inflammatory mechanism in type 2 diabetic rats. JASN. 2006;17:1362–72.

    Article  CAS  PubMed  Google Scholar 

  196. Vessal G, Akmali M, Najafi P, Moein MR, Sagheb MM. Silymarin and milk thistle extract may prevent the progression of diabetic nephropathy in streptozotocin-induced diabetic rats. Ren Fail. 2010;32:733–9.

    Article  CAS  PubMed  Google Scholar 

  197. Rabbani N, Thornalley PJ. Emerging role of thiamine therapy for prevention and treatment of early-stage diabetic nephropathy. Diabetes Obes Metab. 2011;13:577–83.

    Article  CAS  PubMed  Google Scholar 

  198. Cai T, Wu X-Y, Zhang X-Q, Shang H-X, Zhang Z-W, Liao L, et al. Calcium dobesilate prevents diabetic kidney disease by decreasing bim and inhibiting apoptosis of renal proximal tubular epithelial cells. DNA Cell Biol. 2017;36:249–55.

    Article  CAS  PubMed  Google Scholar 

  199. Do MH, Lee JH, Cho K, Kang MC, Subedi L, Parveen A, et al. Therapeutic potential of Lespedeza bicolor to prevent methylglyoxal-induced glucotoxicity in familiar diabetic nephropathy. J Clin Med. 2019;8:E1138.

    Article  Google Scholar 

  200. Renganathan S, Pillai RG. Antioxidant activities of Dhanwantaram Kashayam—an Ayurvedic poly herbal formulation alleviates diabetic complications in rats. J Diabetes Metab Disord. 2020;19:1345–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Fioretto P, Vettor R, Pontremoli R. SGLT2 inhibitors to prevent diabetic kidney disease. Lancet Diabetes Endocrinol. 2020;8:4–5.

    Article  CAS  PubMed  Google Scholar 

  202. Sun H-J, Xiong S-P, Cao X, Cao L, Zhu M-Y, Wu Z-Y, et al. Polysulfide-mediated sulfhydration of SIRT1 prevents diabetic nephropathy by suppressing phosphorylation and acetylation of p65 NF-κB and STAT3. Redox Biol. 2021;38:101813.

    Article  CAS  PubMed  Google Scholar 

  203. Yu X, Su Q, Geng J, Liu H, Liu Y, Liu J, et al. Ginkgo biloba leaf extract prevents diabetic nephropathy through the suppression of tissue transglutaminase. Exp Ther Med. 2021;21:333.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Chang J, Zheng J, Gao X, Dong H, Yu H, Huang M, et al. TangShenWeiNing formula prevents diabetic nephropathy by protecting podocytes through the SIRT1/HIF-1α pathway. Front Endocrinol. 2022;13:888611.

    Article  Google Scholar 

  205. Cheng X, Zhou T, He Y, Xie Y, Xu Y, Huang W. The role and mechanism of butyrate in the prevention and treatment of diabetic kidney disease. Front Microbiol. 2022;13:961536.

    Article  PubMed  PubMed Central  Google Scholar 

  206. Wang Z, Chen Z, Wang X, Hu Y, Kong J, Lai J, et al. Sappanone a prevents diabetic kidney disease by inhibiting kidney inflammation and fibrosis via the NF-κB signaling pathway. Front Pharmacol. 2022;13:953004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Zhou K, Zi X, Song J, Zhao Q, Liu J, Bao H, et al. Molecular mechanistic pathways targeted by natural compounds in the prevention and treatment of diabetic kidney disease. Molecules. 2022;27:6221.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Bapputty R, Talahalli R, Zarini S, Samuels I, Murphy R, Gubitosi-Klug R. Montelukast prevents early diabetic retinopathy in mice. Diabetes. 2019;68:2004–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Millán I, Desco MDC, Torres-Cuevas I, Pérez S, Pulido I, Mena-Mollá S, et al. Pterostilbene prevents early diabetic retinopathy alterations in a rabbit experimental model. Nutrients. 2019;12:E82.

    Article  Google Scholar 

  210. Yin M-S, Zhang Y-C, Xu S-H, Liu J-J, Sun X-H, Liang C, et al. Puerarin prevents diabetic cardiomyopathy in vivo and in vitro by inhibition of inflammation. J Asian Nat Prod Res. 2019;21:476–93.

    Article  CAS  PubMed  Google Scholar 

  211. Tamoli SM, Kohli KR, Kaikini AA, Muke SA, Shaikh AA, Sathaye S. Vasant Kusmakar Ras, an ayurvedic herbo-mineral formulation prevents the development of diabetic retinopathy in rats. J Ayurveda Integr Med. 2020;11:270–6.

    Article  PubMed  PubMed Central  Google Scholar 

  212. Di S, An X, Pang B, Wang T, Wu H, Wang J, et al. Yiqi Tongluo Fang could preventive and delayed development and formation of diabetic retinopathy through antioxidant and anti-inflammatory effects. Biomed Pharmacother. 2022;148:112254.

    Article  CAS  PubMed  Google Scholar 

  213. Wang J, Song Y, Elsherif L, Song Z, Zhou G, Prabhu SD, et al. Cardiac metallothionein induction plays the major role in the prevention of diabetic cardiomyopathy by zinc supplementation. Circulation. 2006;113:544–54.

    Article  CAS  PubMed  Google Scholar 

  214. Wenmeng W, Qizhu T. Early administration of trimetazidine may prevent or ameliorate diabetic cardiomyopathy. Med Hypotheses. 2011;76:181–3.

    Article  PubMed  Google Scholar 

  215. Malek V, Gaikwad AB. Telmisartan and thiorphan combination treatment attenuates fibrosis and apoptosis in preventing diabetic cardiomyopathy. Cardiovasc Res. 2019;115:373–84.

    Article  CAS  PubMed  Google Scholar 

  216. Li N, Zhou H. SGLT2 inhibitors: a novel player in the treatment and prevention of diabetic cardiomyopathy. Drug Des Devel Ther. 2020;14:4775–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Veitch CR, Power AS, Erickson JR. CaMKII inhibition is a novel therapeutic strategy to prevent diabetic cardiomyopathy. Front Pharmacol. 2021;12:695401.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  218. Wang D, Yin Y, Wang S, Zhao T, Gong F, Zhao Y, et al. FGF1ΔHBS prevents diabetic cardiomyopathy by maintaining mitochondrial homeostasis and reducing oxidative stress via AMPK/Nur77 suppression. Signal Transduct Target Ther. 2021;6:133.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  219. Tofte N, Lindhardt M, Adamova K, Bakker SJL, Beige J, Beulens JWJ, et al. Early detection of diabetic kidney disease by urinary proteomics and subsequent intervention with spironolactone to delay progression (PRIORITY): a prospective observational study and embedded randomised placebo-controlled trial. Lancet Diabetes Endocrinol. 2020;8:301–12.

    Article  CAS  PubMed  Google Scholar 

  220. Anderson SL, Marrs JC. Diabetes: how to manage cardiovascular risk in secondary prevention patients. Drugs Context. 2022;11:1.

    Article  Google Scholar 

  221. Archundia Herrera MC, Subhan FB, Chan CB. Dietary patterns and cardiovascular disease risk in people with type 2 diabetes. Curr Obes Rep. 2017;6:405–13.

    Article  PubMed  Google Scholar 

  222. Ojo O. Dietary intake and type 2 diabetes. Nutrients. 2019;11:E2177.

    Article  Google Scholar 

  223. Cowie MR, Fisher M. SGLT2 inhibitors: mechanisms of cardiovascular benefit beyond glycaemic control. Nat Rev Cardiol. 2020;17:761–72.

    Article  CAS  PubMed  Google Scholar 

  224. Xiao L, Yang Y-J, Liu Q, Peng J, Yan J-F, Peng Q-H. Visualizing the intellectual structure and recent research trends of diabetic retinopathy. Int J Ophthalmol. 2021;14:1248–59.

    Article  PubMed  PubMed Central  Google Scholar 

  225. Sun H, Luo G, Chen D, Xiang Z. A comprehensive and system review for the pharmacological mechanism of action of rhein, an active anthraquinone ingredient. Front Pharmacol. 2016;7:247.

    Article  PubMed  PubMed Central  Google Scholar 

  226. Junren C, Xiaofang X, Huiqiong Z, Gangmin L, Yanpeng Y, Xiaoyu C, et al. Pharmacological activities and mechanisms of hirudin and its derivatives—a review. Front Pharmacol. 2021;12:660757.

    Article  PubMed  PubMed Central  Google Scholar 

  227. Zheng Y, Bai L, Zhou Y, Tong R, Zeng M, Li X, et al. Polysaccharides from Chinese herbal medicine for anti-diabetes recent advances. Int J Biol Macromol. 2019;121:1240–53.

    Article  CAS  PubMed  Google Scholar 

  228. Zhang F, Liu H, Liu D, Liu Y, Li H, Tan X, et al. Effects of RAAS inhibitors in patients with kidney disease. Curr Hypertens Rep. 2017;19:72.

    Article  PubMed  Google Scholar 

  229. An J, Niu F, Sim JJ. Cardiovascular and kidney outcomes of spironolactone or eplerenone in combination with ACEI/ARBs in patients with diabetic kidney disease. Pharmacotherapy. 2021;41:998–1008.

    Article  CAS  PubMed  Google Scholar 

  230. Weil EJ, Fufaa G, Jones LI, Lovato T, Lemley KV, Hanson RL, et al. Effect of losartan on prevention and progression of early diabetic nephropathy in American Indians with type 2 diabetes. Diabetes. 2013;62:3224–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  231. Mann JFE, Green D, Jamerson K, Ruilope LM, Kuranoff SJ, Littke T, et al. Avosentan for overt diabetic nephropathy. JASN. 2010;21:527–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  232. de Zeeuw D, Coll B, Andress D, Brennan JJ, Tang H, Houser M, et al. The endothelin antagonist atrasentan lowers residual albuminuria in patients with type 2 diabetic nephropathy. J Am Soc Nephrol. 2014;25:1083–93.

    Article  PubMed  PubMed Central  Google Scholar 

  233. Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, Charytan DM, et al. Canagliflozin and renal outcomes in type 2 diabetes and nephropathy. N Engl J Med. 2019;380:2295–306.

    Article  CAS  PubMed  Google Scholar 

  234. Nakamura S, Morimoto N, Tsuruma K, Izuta H, Yasuda Y, Kato N, et al. Tissue kallikrein inhibits retinal neovascularization via the cleavage of vascular endothelial growth factor-165. Arterioscler Thromb Vasc Biol. 2011;31:1041–8.

    Article  CAS  PubMed  Google Scholar 

  235. Cheng Y, Yu X, Zhang J, Chang Y, Xue M, Li X, et al. Pancreatic kallikrein protects against diabetic retinopathy in KK Cg-Ay/J and high-fat diet/streptozotocin-induced mouse models of type 2 diabetes. Diabetologia. 2019;62:1074–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Härma M-A, Dahlström EH, Sandholm N, Forsblom C, Groop P-H, Lehto M, et al. Decreased plasma kallikrein activity is associated with reduced kidney function in individuals with type 1 diabetes. Diabetologia. 2020;63:1349–54.

    Article  PubMed  PubMed Central  Google Scholar 

  237. Du J-K, Yu Q, Liu Y-J, Du S-F, Huang L-Y, Xu D-H, et al. A novel role of kallikrein-related peptidase 8 in the pathogenesis of diabetic cardiac fibrosis. Theranostics. 2021;11:4207–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  238. Kato N, Hou Y, Lu Z, Lu C, Nagano H, Suzuma K, et al. Kallidinogenase normalizes retinal vasopermeability in streptozotocin-induced diabetic rats: potential roles of vascular endothelial growth factor and nitric oxide. Eur J Pharmacol. 2009;606:187–90.

    Article  CAS  PubMed  Google Scholar 

  239. Savvatis K, Westermann D, Schultheiss H-P, Tschöpe C. Kinins in cardiac inflammation and regeneration: insights from ischemic and diabetic cardiomyopathy. Neuropeptides. 2010;44:119–25.

    Article  CAS  PubMed  Google Scholar 

  240. Zhu D, Zhang L, Cheng L, Ren L, Tang J, Sun D. Pancreatic Kininogenase ameliorates renal fibrosis in streptozotocin induced-diabetic nephropathy rat. Kidney Blood Press Res. 2016;41:9–17.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the China Academy of Chinese Medical Sciences for supporting the study. We also appreciate the involvement of all authors in this study.

Funding

This work was supported by Innovation Team and Talents Cultivation Program of National Administration of Traditional Chinese Medicine (Nos.ZYYCXTD-D-202001), and Scientific and Technological innovation project of China Academy of Chinese Medical Sciences (Nos.CI2021A01602).

Author information

Authors and Affiliations

Authors

Contributions

CX, KC, and ZG were co-responsible for the collection, collation, and writing of the original manuscript. TB and LD read through and corrected the manuscript. LZ, XT, and XL designed and guided the writing of this article. All authors contributed to the article and approved the submitted version.

Corresponding authors

Correspondence to Linhua Zhao, Xiaolin Tong or Xiuyang Li.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

All authors have read and approved the version to be published.

Competing interests

The authors confirm that there are no conflicts of interest. All the figures were created by Figdraw (www.figdraw.com) and ScienceSlides plug-in.

Additional information

Publisher's Note

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

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xue, C., Chen, K., Gao, Z. et al. Common mechanisms underlying diabetic vascular complications: focus on the interaction of metabolic disorders, immuno-inflammation, and endothelial dysfunction. Cell Commun Signal 21, 298 (2023). https://doi.org/10.1186/s12964-022-01016-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12964-022-01016-w

Keywords