Next Article in Journal
The Fitter I Am, the Larger I Feel—The Vicious Circle of Physical Exercise in Anorexia Nervosa
Next Article in Special Issue
The Ferroptosis Inhibitor Liproxstatin-1 Ameliorates LPS-Induced Cognitive Impairment in Mice
Previous Article in Journal
Dietary Quality and Relationships with Metabolic Dysfunction-Associated Fatty Liver Disease (MAFLD) among United States Adults, Results from NHANES 2017–2018
Previous Article in Special Issue
Appraising the Causal Association between Systemic Iron Status and Heart Failure Risk: A Mendelian Randomisation Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Ferroptosis of Endothelial Cells in Vascular Diseases

1
Department of Pharmacy, Peking University First Hospital, Beijing 100034, China
2
School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China
3
School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
4
Institute of Clinical Pharmacology, Peking University, Beijing 100191, China
*
Authors to whom correspondence should be addressed.
Nutrients 2022, 14(21), 4506; https://doi.org/10.3390/nu14214506
Submission received: 26 August 2022 / Revised: 21 October 2022 / Accepted: 23 October 2022 / Published: 26 October 2022

Abstract

:
Endothelial cells (ECs) line the inner surface of blood vessels and play a substantial role in vascular biology. Endothelial dysfunction (ED) is strongly correlated with the initiation and progression of many vascular diseases. Regulated cell death, such as ferroptosis, is one of the multiple mechanisms that lead to ED. Ferroptosis is an iron-dependent programmed cell death associated with various vascular diseases, such as cardiovascular, cerebrovascular, and pulmonary vascular diseases. This review summarized ferroptosis of ECs in vascular diseases and discussed potential therapeutic strategies for treating ferroptosis of ECs. In addition to lipid peroxidation inhibitors and iron chelators, a growing body of evidence showed that clinical drugs, natural products, and intervention of noncoding RNAs may also inhibit ferroptosis of ECs.

1. Introduction

Ferroptosis is an iron-dependent programmed cell death primarily characterized by reduced cystine uptake into cells, followed by glutathione (GSH) depletion, iron-dependent lipid peroxidation, and release of damage-associated molecular patterns [1,2]. Cell volume shrinkage and mitochondrial membrane thickening are the predominant morphologic hallmarks of ferroptosis [3]. Dixon et al. defined ferroptosis for the first time in 2012 [1]. It has since become a hot area of research. Accumulating evidence indicates that ferroptosis is linked to various pathological states, such as tumors, cardiovascular diseases, cerebrovascular disease, acute lung injury (ALI), and neurodegenerative disorders.
Endothelial cells (ECs) have direct contact with chemicals or particles in the circulatory system. The vascular endothelium is essential for maintaining multiorgan health and homeostasis, including the dynamic maintenance of vasodilation and vasoconstriction, angiogenesis and anti-angiogenesis, antithrombosis and pro-thrombosis, antiinflammation and proinflammation, and antioxidation and pro-oxidation [4,5,6,7,8]. Endothelial dysfunction (ED) is highly correlated with diverse human pan-vascular diseases, including atherosclerosis (AS), hypertension, diabetes, and ALI [9,10], and contributes to the development of neurodegenerative diseases such as Alzheimer’s disease [11]. Due to its essential role in many diseases, the endothelium is a therapeutic target for the prevention and treatment of the above pan-vascular diseases.
Regulated cell death (RCD), such as ferroptosis, occurring in ECs is one of the multiple mechanisms that lead to ED. Evidence suggests that inhibiting ferroptosis can attenuate ED [12]. Therefore, ferroptosis may be a promising target to protect the endothelium. This review aimed to highlight the effects of ferroptosis in ECs, summarize new treatments based on ferroptosis of ECs, and identify research trends and hotspots in this field.

2. Mechanism of Ferroptosis

RCD is essential in all aspects of life and contributes to various human diseases under different stresses [13,14]. A genetically encoded apparatus initiates RCD, which can be altered by pharmacologic or genetic intervention. Thus, there is a growing interest in modulating RCD to treat diseases. Due to differences in morphology, biochemistry, and genetic levels, RCD can be classified into various subtypes (necroptosis, apoptosis, autophagy, pyroptosis, ferroptosis, etc.) [14,15,16,17,18,19], as shown in Table 1.
Ferroptosis is primarily mediated by iron metabolism and lipid peroxidation [20]. It occurs as a result of iron-catalyzed lipid peroxidation, which can be initiated by both enzymatic and nonenzymatic mechanisms, such as the Fenton reaction. Two enzymes, lysophosphatidylcholine acyltransferase 3 (LPCAT3) and acyl-CoA synthetase long-chain family member 4 (ACSL4), contribute to the biosynthesis and remodeling of polyunsaturated fatty acids (PUFAs)-phosphatidylethanolamines in cellular membranes [2]. PUFAs produce lipid hydroperoxides, as well as toxic lipid free radicals, which transfer protons near PUFAs and trigger a new round of lipid oxidation reactions [21].
Ferroptosis can be caused by iron overload triggered by an increase in iron uptake via transferrin (TF) receptor 1 (TFR1) and a reduction in iron export via ferroportin 1. Ferrous ions (Fe2+) mediate the Fenton reaction, which can increase reactive oxygen species (ROS) and result in ferroptosis [21,22,23]. In addition, some Fe2+ are stored in ferritin. Ferritinophagy, which converts ferritin into intracellular iron through nuclear receptor coactivator 4 (NCOA4), also results in ferroptosis [24,25,26,27].
System Xc mediates the exchange of cystine and glutamate in a 1:1 ratio. Erastin induces ferroptosis by inhibiting system Xc. Glutathione peroxidase 4 (GPX4) is a selenoenzyme that lowers ROS via GSH and protects lipid peroxidation [28]. It is crucial in the regulation of ferroptosis and atherosclerosis [29]. Ras-selective lethal small molecule 3 (RSL3) impairs antioxidant capacity by inhibiting GPX4, ultimately causing ferroptosis. Several studies have reported that p53 can decrease solute carrier family 7 member 11 (SLC7A11) expression, inhibit GPX4 activity, and lead to ferroptosis [30]. SLC7A11 and GPX4 levels are increased by nuclear factor erythroid 2-related factor 2 (NRF2), and therefore reduces ROS. NRF2 overexpression inhibits ferroptosis, whereas Kelch-like ECH-associated protein 1 (Keap 1), which binds to NRF2 and negatively regulates it, can reverse this process [31,32]. Activating the KEAP1/NRF2 pathway can cause ferroptosis by upregulating heme oxygenase 1 (HMOX1) [33]. Moreover, the ferroptosis suppressor protein 1 (FSP1)-coenzyme Q10 (CoQ10)-nicotinamide adenine dinucleotide (phosphate) (NAD(P)H) pathway [34,35], guanosine 5′-triphosphate cyclohydrolase 1 (GCH1)-tetrahydrobiopterin (BH4)-dihydrofolate reductase (DHFR) pathway [36,37], adenosine monophosphate-activated protein kinase (AMPK) [38,39], and mitochondria [40] also play vital roles in eliminating excess lipid peroxides and regulating ferroptosis. Key molecular mechanisms of ferroptosis are summarized in Figure 1.

3. Endothelial Dysfunction

ECs perform a variety of key homeostatic functions [6]. However, ED is characterized by a change from executing physiological functions of the vascular endothelium to inflammation, hyperpermeability, leukocyte adhesion, endothelial nitric oxide synthase uncoupling, altered EC metabolism, oxidative stress, vasoconstriction, injury, cell death, senescence, and endothelial-to-mesenchymal transition [9]. In a wide range of circumstances, ED, which is usually caused by EC injury or death, is clearly associated with poor prognosis. Oxidative stress is one of the most important underlying mechanisms for ED development [41]. In cardiovascular diseases, multiple risk factors that upregulate intracellular oxidative stress and ROS initiate and accelerate the progression of atherosclerotic diseases through ED [42]. In cerebrovascular diseases, as intact EC is an important part of normal neurovascular units, the death of ECs will lead to serious damage to the blood-brain barrier and cause various pathophysiological processes, which eventually mediate more extensive programmed cell deaths [43].
EC death such as ferroptosis and pyroptosis [44] have recently been implicated in ED and vascular diseases, although the exact mechanisms remain unclear. The precise role of each mode of cell death needs further elucidation to yield new therapeutic targets that could facilitate EC survival under disease states and explore treatments of various ED-related diseases.

4. Ferroptosis of ECs

Ferroptosis occurs in ECs after fine particulate matter (PM2.5) exposure, characterized by intracellular iron overload, lipid peroxidation, GSH depletion, and redox imbalance [45]. Furthermore, iron-bearing nanoparticles, which are abundant in PM, trigger ferroptotic responses by increasing intracellular iron levels [46]. Activation of the p53-xCT (the substrate-specific subunit of system Xc)-GSH axis enhances ferroptosis in human umbilical vein endothelial cells (HUVECs). High glucose (HG) and interleukin-1β (IL-1β) induce ferroptosis with an increase in p53 and a reduction in xCT (SLC7A11) and GSH [47]. GPX4 knockdown in HUVECs increases lipid oxidation and lactate dehydrogenase (LDH) levels while reducing cellular proliferation [48]. Oxidized low-density lipoprotein (Ox-LDL) promotes ED by inducing ferroptosis with decreasing GPX4 and xCT expression [12,49]. Ox-LDL exposure could also induce mitochondrial damage [12,50]. The iron and total ROS content significantly increase, whereas the GSH level decreases [50]. Zinc oxide nanoparticles (ZnONPs) have shown promise in the field of biomedicine [51]. However, ZnONP exposure can cause ferroptosis in vascular ECs. NCOA4-mediated ferritin degradation is required for the ferroptosis of ECs induced by ZnONPs. ROS originating from mitochondria may trigger AMPK-Unc-51-like autophagy activating kinase 1 axis to cause ferritinophagy [52].
The ferroptosis of vascular ECs occurs in multiple vascular diseases (Figure 2). The molecular mechanisms of several treatments for inducing or inhibiting ferroptosis of ECs are summarized in Table 2.

4.1. Ferroptosis of ECs in Cardiovascular Disease

Many serious vascular diseases have AS as their basic pathological state [69]. AS is mainly caused by lipid deposition, EC damage, vascular smooth muscle cell proliferation, macrophage transformation, and so on [70]. During the onset and progression of AS, ferroptosis may occur. In high-fat diet-induced apolipoprotein E knockout (ApoE−/−) mice, ferroptosis inhibitor ferrostatin-1 (Fer-1) alleviated AS lesions, ameliorated lipid peroxidation, reduced iron accumulation, and suppressed cell death and angiogenesis in the thoracic aorta. The effects of inhibition of ferroptosis were also evaluated in ox-LDL-induced mouse aortic EC (MAECs) in vitro. EC injury, lipid peroxidation, cell death, angiogenesis, and ED induced by ox-LDL can all be reduced by Fer-1 [12]. Ox-LDL exposure caused the mitochondrial damage of human coronary artery ECs (HCAECs) and increased iron and total ROS levels, which promoted ferroptosis. Decaprenyl diphosphate synthase subunit 2 (PDSS2) overexpression can suppress the ferroptosis of HCAECs by activating NRF2 pathways [50].
As ED in diabetes leads to the development of cardiovascular complications [71], it is vital to investigate the function and regulatory mechanisms of ferroptosis in diabetes-induced ED. The aorta of db/db mice exhibited increased lipid peroxidation production and iron accumulation. Due to the impairment of xCT expression by p53 activation, there is a reduction in intracellular cystine and GSH production, leading to ferroptosis and ED [47]. A bioinformatic assay identified that ferroptosis and HMOX1 were key factors in atherosclerotic vascular diseases. Additionally, an in vivo study revealed that diabetes greatly contributed to ferroptosis and increased HMOX1 levels. Fer-1 could effectively rescue them. In MAECs and HUVECs, Fer-1 ameliorated HG/high lipid-induced redox imbalance, GSH depletion, and lipid peroxidation. HMOX1 knockdown reduced Fe2+ overload, iron content, and ROS and alleviated lipid peroxidation, which attenuated ferroptosis in diabetic human ECs. Genetic/pharmacologic HMOX1 inhibition and other interventions that block ferroptosis, such as Fer-1 administration, may represent potential approaches to treat diabetic AS [55]. Moreover, iron overload may contribute to EC calcification and be related to cell ferroptosis. Cellular calcification can be alleviated with ferroptosis inhibitors [59].
Ferroptosis of ECs also contributes to other cardiovascular diseases, such as cardiac hypertrophy [56,57] and hypertension [58]. The underlying mechanisms of treatments associated with ferroptosis of ECs are shown in Table 2.

4.2. Ferroptosis of ECs in Cerebrovascular Disease

The cerebrovascular diseases, intracerebral hemorrhage (ICH) and subarachnoid hemorrhage (SAH), have high morbidity and mortality rates [72,73,74]. The pathological process of hemorrhagic stroke has lately been linked to ferroptosis [75,76,77,78,79]. Additionally, this association was demonstrated in several studies on ECs (Table 2) [61,62,63]. Mouse brain microvascular ECs (BMVECs) bEnd.3 and rat BMVECs are commonly used to study chronic cerebrovascular disease. Gao et al. observed that ferroptosis occurred in the microglia and endothelium after SAH, and this process was facilitated by increasing arachidonate 15-lipoxygenase (ALOX15) levels [63]. Long noncoding RNA (lncRNA) H19 was overexpressed in ICH model cells. H19 knockdown promoted cell proliferation and suppressed ferroptosis of BMVECs by regulating the miR-106b-5p/ACSL4 axis [61]. Individuals with diabetes were not only at higher risk of stroke, but also experienced functional impairment and poor recovery after ischemic brain injury [80,81]. Therefore, several studies focused on ferroptosis of ECs in diabetic cerebrovascular diseases [64,65]. The functional and regulatory roles of maternally expressed gene 3 (Meg3) in diabetic brain ischemic injuries were studied in vitro. By inhibiting GPX4 activity, the Meg3-p53 signaling pathway mediated the ferroptosis of rat BMVECs, induced by oxygen and glucose deprivation (OGD), combined with hyperglycemic reperfusion [64]. In BMVECs derived from male type 2 diabetic rats, deferoxamine (DFO) significantly reduced ferroptosis markers and prevented iron-mediated decline in cell viability [65].

4.3. Ferroptosis of ECs in Pulmonary Vascular Disease

ALI is a life-threatening inflammatory disease linked to the dysfunction of the alveolar-capillary barrier caused by various factors [82,83]. Ferroptosis was reported to be crucial in radiation-induced lung injury by mediating the release of inflammatory cytokines [84]. Ferroptosis of lung ECs modulated by piezo-type mechanosensitive ion channel component 1 (PIEZO1)/calcium (Ca2+)/calpain signaling was a potential therapeutic mechanism. Vascular endothelial-cadherin (VE-cadherin) knockdown caused ferroptosis-like phenomena in human pulmonary microvascular endothelial cells (HULEC-5a), whereas VE-cadherin overexpression partly decreased ferroptosis [66]. In addition to ALI, ferroptosis of pulmonary artery ECs (PAECs) is also involved in pulmonary hypertension (PH). Pulmonary artery endothelial ferroptosis triggers inflammatory responses through the high-mobility group box 1/Toll-like receptor 4/NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome signaling pathway. A ferroptosis inhibitor (such as Fer-1) may attenuate PH progression [67].

5. Potential Ferroptosis Inhibitors

As ferroptosis of ECs is associated with multiple vascular diseases, ferroptosis inhibition may provide a novel treatment strategy. In addition to lipid peroxidation inhibitors and iron chelators, growing evidence shows that clinical drugs, natural products, and ncRNA intervention may also inhibit ferroptosis of ECs, as summarized in Table 3.

5.1. Lipid Peroxidation Inhibitors and Iron Chelators

Lipid peroxidation inhibitors (e.g., Fer-1 and liproxstatin-1) and iron chelators (e.g., DFO and deferiprone (DFP)) can pharmacologically inhibit ferroptosis. Growing evidence shows that Fer-1 rescues lipid peroxidation and ED in several ECs, such as hemangioma ECs, corneal ECs, MAECs, PAECs, and HUVECs [12,45,47,52,67,92,93]. Liproxstatin-1 reverses ferroptosis by scavenging free radicals to prevent PUFA oxidation, whose therapeutic effects were similar to Fer-1 [94,95].
Several studies showed that DFO and DFP could reduce intracellular iron levels and cell ferroptosis [12,45,47,49,52,58,62,65]. DFOs are clinically approved iron chelators that can remove iron by binding with free or protein-bound Fe3+ [95] and prevent the onset of lipid peroxidation by inhibiting lipoxygenases (LOXs) [96]. Moreover, they reduced inflammation and the development of atherosclerotic lesions in ApoE-deficient mice, showing cardiovascular protective effects [97]. Iron chelation and possibly ferroptosis inhibition may provide a novel disease-modifying treatment approach in preventing post-stroke cognitive impairment in diabetes and alleviating AS [58,65]. However, DFO is ototoxic and neurotoxic [98]. Therefore, more attention should be paid to compounds with fewer side effects.

5.2. Clinical Drugs

In addition to the compounds mentioned above, which are closely related to the crucial mechanism of ferroptosis, some clinical drugs have also shown the potential to inhibit ferroptosis of ECs (Table 3). Fluvastatin may protect ECs from ferroptosis by counteracting the ox-LDL-induced inhibition of GPX4 and xCT expression [49]. Dexmedetomidine and tongxinluo can also protect ECs from ferroptosis [68,85].

5.3. Natural Products

Natural products are common resources for drug discovery [99]. Some have been used to manage human diseases [100,101]. Moreover, medical nutritional therapy with healthy foods and their bioactive ingredients has been considered an alternative strategy for the treatment of certain chronic diseases [102]. With the rapid development of studies on ferroptosis, increasing evidence has revealed that natural products can regulate ferroptosis, and are related to cancers, Parkinson’s disease, intracerebral hemorrhage, and other diseases [103,104,105,106]. In terms of ferroptosis in ECs, the potential role of natural products in regulating ferroptosis-mediated diseases requires further study and may be a hot area for future research.
The ethyl acetate-extracted fraction of the total Ginkgo biloba flower extract showed an anti-ferroptosis effect in vascular ECs, and luteolin was the active compound in the extract. Luteolin downregulated ACSL4 and upregulated GPX4 in erastin-induced ferroptosis of HUVECs [86]. Astragaloside IV significantly reversed decreased cell activity, elevated iron ion and lipid ROS levels, enhanced cell senescence, and reversed the change in mitochondrial morphology caused by lysophosphatidylcholine treatment. The mechanism may be due to the partial upregulation of SLC7A11 and GPX4 expression levels [87]. A new flavonoid glycoside isolated from Clematis tangutica, Apigenin-7-O-β-D-(-6″-p-coumaroyl)-glucopyranoside, attenuated intestinal ischemia-reperfusion injury (IIRI)-induced ROS generation, Fe2+ accumulation, and mitochondrial damage, and could inhibit ferroptosis through HMOX1 and monoamine oxidase B (MAO-B) [88]. The active lipophilic ingredient, tanshinone IIA (TSA), which was isolated from the root of Salvia miltorrhiza [107], protected cells by modulating intracellular redox status and has been extensively used to treat cardiovascular diseases [108]. He et al. found that TSA protected HCAECs from ferroptosis by activating the NRF2 pathway [60]. After SAH, ferroptosis in the microglia and endothelium was facilitated by treatment with cepharanthine, a type of bisbenzylisoquinoline alkaloid, by downregulating ALOX15 levels [63]. In addition, vitamin E-rich food, such as brown rice, can compensate for the loss of GPX4 by protecting cells from lipid peroxidation [48]. Docosahexaenoic acid (DHA), the primary ingredient of Omega (ω)-3 polyunsaturated fatty acids, can provide numerous benefits for human health [109]. DHA treatment has been identified to upregulate the expression of interferon regulating factor 3 and reduce cellular ferroptosis [57].
Importantly, natural products have the advantages of stable structure, higher safety, low cost, and easy availability, compared with classical ferroptosis inhibitors. Therefore, it is a worthwhile and promising endeavor to explore natural products as ferroptosis inhibitors. Further clinical investigations are urgently required to assess the efficacy and viability of natural products as dietary supplements or prospective medications in vivo because the exactly regulatory targets and molecular mechanisms regulating ferroptosis by natural products remain unclear.

5.4. Novel Regulators Associated with Molecular Biology

5.4.1. ncRNAs

lncRNAs are RNAs >200 nt in length, which act as regulators of almost every cellular process, physiological condition, and various human diseases [110]. Some lncRNAs act as miRNA sponges and negatively regulate miRNAs. miRNAs with a size of ~20 nt can negatively regulate gene expression by binding a target mRNA and inhibiting its translation or promoting its degradation [111,112].
Links between ncRNAs and endothelial ferroptosis have opened up a new realm of therapeutic opportunities. lncRNA H19 knockdown promoted cell viability and inhibited ferroptosis of BMVECs by regulating the miR-106b-5p/ACSL4 axis, which may be a potential therapeutic target for ICH [61]. By modulating GPX4 transcription and expression, lncRNA Meg3 mediated ferroptosis induced by OGD combined with hyperglycemia in rat BMVECs via the p53/GPX4 axis. Therefore, Meg3 may be an attractive target for treating diabetic brain ischemia [64]. Targeting the lncRNA Meg8/miR-497-5p/Notch receptor 2 signaling pathway may inhibit the ferroptosis of hemangioma ECs [93].
For miRNAs, miR-132 promotes AS by inducing mitochondrial oxidative stress-mediated ferroptosis and downregulating GPX4 [53]. miR-17-92 targeting the zinc lipoprotein A20-ACSL4 axis protects HUVEC from erastin-induced ferroptosis [89].

5.4.2. Exosomes (Exos) or Extracellular Vesicles (EVs)

Endothelial progenitor cells (EPCs) are involved in maintaining endothelial homeostasis and forming new vessels [113]. EVs are small membrane-bound vesicles secreted by almost all cell types and mediate intercellular communication by transferring bioactive molecules, such as miRNAs [114]. Exos are EVs that deliver miRNAs, mRNAs, and proteins, which are important for communication from organ to organ and cell to cell [115]. EPC-derived Exos (EPC-Exos) had a protective effect on ECs in a mouse model of diabetes [116]. In irradiated fibroblasts, one study identified the regulation of Exos in cell ferroptosis [117]. Moreover, miRNA itself can also relieve ferroptosis [118]. Therefore, the role of EPC-EVs or EPC-Exos carrying a miRNA in ferroptosis of ECs deserves further investigation and may confer novel insights for clinical management.
EPC-EVs transferred miR-199a-3p to inhibit specificity protein 1 (SP1), further repress ferroptosis of ECs, and alleviate AS occurrence [54]. EPC-derived Exos transferred miR-30e-5p, which inhibited SP1 and activated the AMPK pathway to regulate erastin-induced ferroptosis in HUVECs [90]. As a widely expressed transcription factor that regulates the inflammatory repair process, SP1 is a key target for treating atherosclerotic vascular diseases [119,120].

5.4.3. Other Targets

NRF2, a transcription factor, regulates several metabolic mechanisms, including proteostasis, xenobiotic/drug metabolism, iron/heme metabolism, carbohydrate and lipid metabolism, apoptosis, and ferroptosis, as well as the cellular antioxidant response [121]. Two of the most important targets inhibiting ferroptosis, xCT, and GPX4, are established downstream transcriptional targets of NRF2. Therefore, NRF2 is essential for reducing lipid peroxidation and ferroptosis [122]. By promoting the activation of NRF2 pathways, PDSS2 overexpression prevents HCAECs from ferroptosis. Thus, PDSS2 may be protective in AS [50].
In addition, HMOX1 upregulation promotes ferroptosis in diabetic AS. HMOX1 inhibition via genetic or pharmacological means may have the potential to attenuate ferroptosis [55]. PIEZO1/Ca2+/calpain signaling modulates ferroptosis of lung ECs, which is a potential therapeutic mechanism for treating radiation-induced lung injury [66]. Tripartite motif-containing 46 and GPX4 form a regulatory pathway that controls HG-induced ferroptosis of human retinal capillary ECs. Thus, inhibiting this pathway or maintaining GPX4 expression enables cells to resist damage caused by HG [91]. Moreover, elabela, a second endogenous ligand for the apelin receptor, can antagonize angiotensin II-mediated cardiac microvascular ECs (CMVECs) ferroptosis by modulating the IL-6/signal transducer and activator of transcription 3/GPX4 pathway [58].

5.5. Nanomaterials

Nanoparticles have already been used as drug delivery systems and have greatly extended the range of drug carriers. For enhancing in vivo siRNA delivery, nanotechnology has shown great potential [98].
lncRNA AAB sponged and sequestered miR-30b-5p to induce the imbalance of matrix metalloproteinase 9/TIMP metallopeptidase inhibitor 1, which enhanced TFR1 activation and eventually caused the ferroptosis of CMVECs. Shi et al. developed a delivery system that effectively transported silenced lncRNA AAB (si-AAB) using the neutrophil membrane (NM)-camouflaged mesoporous silica nanocomplex (MSN). NM+si-AAB+MSN inhibited ferroptosis of CMVECs by miR-30b-5p upregulation. An in vivo study showed that it could repair the cardiac microvascular endothelial injury in cardiac hypertrophy. The MSN core has a high siRNA loading capacity and effectively protects the nanocomplex from degradation in body fluids. At the endothelial injury site, the si-AAB is released long-term to prevent ferroptosis of CMVECs, which improves cardiac microvascular function [56].

6. Conclusions

Ferroptosis occurs in ECs, according to a growing body of studies. In this review, ferroptosis of ECs, their role in vascular diseases, and potential regulators were summarized. Ferroptosis of ECs has an intimate connection with cardiovascular, cerebrovascular, and pulmonary vascular diseases. Lipid peroxidation inhibitors, iron chelators, and some clinical drugs have shown their potential as inhibitors of ferroptosis in ECs. Natural products, ncRNAs, and other novel targets involved in the ferroptosis pathway also conferred new insights for clinical management. Exos or EVs and nanomaterials that deliver key regulators require further investigation. More studies on regulatory ferroptosis of ECs will broaden the pathogenesis of vascular diseases and provide novel treatment strategies for multiple diseases.

Author Contributions

Investigation, H.Z., S.Z., and M.S.; visualization, H.Z., M.S., and M.H.; writing—original draft, H.Z. and M.S.; writing—review and editing, S.Z., Z.L., G.M., Z.W., and Q.X.; conceptualization, supervision, and funding acquisition, Q.X, and Y.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by National High Level Hospital Clinical Research Funding (Scientific and Technological Achievements Transformation Incubation Guidance Fund Project of Peking University First Hospital, no. 2022CX14) and National Natural Science Foundation of China (no. 81872940, 81973395, and 82073935).

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ΔΨm: mitochondrial membrane potential; 4-HNE: 4-hydroxy-2-nonenal; A20: zinc lipoprotein A20; AAB: long non-coding RNA AABR07017145.1; ACC: acetyl coenzyme A carboxylase; ACSL4: acyl-CoA synthetase long-chain family member 4; ALI: acute lung injury; ALOX12: arachidonate 12-lipoxygenase, 12S Type; ALOX15: arachidonate 15-lipoxygenase; AMPK: adenosine monophosphate-activated protein kinase; Ang II: angiotensin II; ApoE: apolipoprotein E; AS: atherosclerosis; bEnd.3: mouse brain microvascular endothelial cell; BH2: dihydrobiopterin; BH4: tetrahydrobiopterin; BMVEC: brain microvascular EC; Ca2+: calcium; CMVEC: cardiac microvascular endothelial cell; CoA: coenzyme A; COPD: chronic obstructive pulmonary disease; CoQ10: coenzyme Q10; CoQ10H2: the reduced form of coenzyme Q10, ubiquinol; DFO: deferoxamine; DFP: deferiprone; DHA: docosahexaenoic acid; DHFR: dihydrofolate reductase; EC: Endothelial cell; EPC: endothelial progenitor cell; EPC-Exo: EPC-derived exosome; EPC-EV: EPC-secreted extracellular vesicle; EV: extracellular vesicle; Exo: exosome; Fe2+: ferrous ion; Fer-1: ferrostatin-1; FSP1: ferroptosis suppressor protein 1; GCH1: guanosine 5’-triphosphate cyclohydrolase 1; GPX4: glutathione peroxidase 4; GSH: glutathione; GSSG: glutathione disulfide; HCAEC: human coronary artery endothelial cell; HMGB1: high-mobility group box 1; HMOX1: heme oxygenase 1; HPMEC: human pulmonary microvascular endothelial cell; HRCEC: human retinal capillary endothelial cell; HULEC-5a: human pulmonary microvascular endothelial cell; HUVEC: human umbilical vein endothelial cell; ICH: intracerebral hemorrhage; IIRI: intestinal ischemia-reperfusion injury; IL-6: interleukin-6; IRF3: interferon regulating factor 3; Keap 1: Kelch-like ECH-associated protein 1; L: lipid; LDH: lactate dehydrogenase; lncRNA: long noncoding RNA; LOX: lipoxygenase; LPCAT3: lysophosphatidylcholine acyltransferase 3; LPS: lipopolysaccharide; MAEC: mouse aortic endothelial cell; MAO-B: monoamine oxidase B; MDAs: malondialdehydes; Meg3: maternally expressed gene 3; MLKL: mixed lineage kinase domain-like pseudokinase; MSN: mesoporous silica nanocomplex; NADH: nicotinamide adenine dinucleotide; NADPH: nicotinamide adenine dinucleotide phosphate; NCOA4: nuclear receptor coactivator 4; NLRP3: NACHT, LRR and PYD domains-containing protein 3; NM: neutrophil membrane; NRF2: nuclear factor erythroid 2-related factor 2; -OHs: alcohols; -OOHs: hydroperoxides; Ox-LDL: oxidized low-density lipoprotein; OXPHOX: oxidative phosphorylation; PAEC: pulmonary artery endothelial cell; PDSS2: prenyldiphosphate synthase subunit 2; PEG-HCC: poly(ethylene glycol)-conjugated hydrophilic carbon clusters; PH: pulmonary hypertension; PIEZO1: piezo type mechanosensitive ion channel component 1; PL: phospholipid; PM2.5: particulate matter <2.5 mum in diameter; POR: cytochrome P450 reductase; PUFA: polyunsaturated fatty acid; RBMVEC: rat brain microvascular endothelial cell; RCD: regulated cell death; RIPK: receptor-interacting serine/threonine kinase; ROS: reactive oxygen species; RPVEC: rat pulmonary vein endothelial cell; RSL3: Ras-selective lethal small molecule 3; SAH: subarachnoid hemorrhage; si-AAB: silenced lncRNA AAB; siRNA: small interfering RNA; SLC3A2: solute carrier family 3 member 2; SLC7A11: solute carrier family 7 member 11; SP1: specificity protein 1; STAT3: signal transducer and activator of transcription 3; STEAP3: six-transmembrane epithelial antigen of prostate 3; TAC: tricarboxylic acid; TF: transferrin; TFR1: transferrin receptor 1; TIMP1: TIMP metallopeptidase inhibitor 1; TLR4: toll-like receptor 4; TRIM46: tripartite motif containing 46; TSA: tanshinone IIA; VE-cadherin: vascular endothelial-cadherin; ZnONPs: zinc oxide nanoparticles.

References

  1. Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef] [Green Version]
  2. Stockwell, B.R.; Friedmann Angeli, J.P.; Bayir, H.; Bush, A.I.; Conrad, M.; Dixon, S.J.; Fulda, S.; Gascón, S.; Hatzios, S.K.; Kagan, V.E.; et al. Ferroptosis: A Regulated Cell Death Nexus Linking Metabolism, Redox Biology, and Disease. Cell 2017, 171, 273–285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Yu, H.; Guo, P.; Xie, X.; Wang, Y.; Chen, G. Ferroptosis, a new form of cell death, and its relationships with tumourous diseases. J. Cell. Mol. Med. 2017, 21, 648–657. [Google Scholar] [CrossRef] [Green Version]
  4. Vallance, P.; Collier, J.; Moncada, S. Effects of endothelium-derived nitric oxide on peripheral arteriolar tone in man. Lancet 1989, 2, 997–1000. [Google Scholar] [CrossRef]
  5. Vanhoutte, P.M. Endothelium and control of vascular function. State of the Art lecture. Hypertension 1989, 13, 658–667. [Google Scholar] [CrossRef] [Green Version]
  6. Pober, J.S.; Min, W.; Bradley, J.R. Mechanisms of endothelial dysfunction, injury, and death. Annu. Rev. Pathol. 2009, 4, 71–95. [Google Scholar] [CrossRef] [PubMed]
  7. Vita, J.A. Endothelial function. Circulation 2011, 124, e906–e912. [Google Scholar] [CrossRef] [Green Version]
  8. Aird, W.C. Phenotypic heterogeneity of the endothelium: II. Representative vascular beds. Circ. Res. 2007, 100, 174–190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Xu, S.; Ilyas, I.; Little, P.J.; Li, H.; Kamato, D.; Zheng, X.; Luo, S.; Li, Z.; Liu, P.; Han, J.; et al. Endothelial Dysfunction in Atherosclerotic Cardiovascular Diseases and Beyond: From Mechanism to Pharmacotherapies. Pharmacol. Rev. 2021, 73, 924–967. [Google Scholar] [CrossRef] [PubMed]
  10. Matthay, M.A.; Ware, L.B.; Zimmerman, G.A. The acute respiratory distress syndrome. J. Clin. Investig. 2012, 122, 2731–2740. [Google Scholar] [CrossRef]
  11. Grammas, P. Neurovascular dysfunction, inflammation and endothelial activation: Implications for the pathogenesis of Alzheimer’s disease. J. Neuroinflamm. 2011, 8, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Bai, T.; Li, M.; Liu, Y.; Qiao, Z.; Wang, Z. Inhibition of ferroptosis alleviates atherosclerosis through attenuating lipid peroxidation and endothelial dysfunction in mouse aortic endothelial cell. Free Radic. Biol. Med. 2020, 160, 92–102. [Google Scholar] [CrossRef] [PubMed]
  13. Galluzzi, L.; Vitale, I.; Aaronson, S.A.; Abrams, J.M.; Adam, D.; Agostinis, P.; Alnemri, E.S.; Altucci, L.; Amelio, I.; Andrews, D.W.; et al. Molecular mechanisms of cell death: Recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018, 25, 486–541. [Google Scholar] [CrossRef] [PubMed]
  14. Tang, D.; Kang, R.; Berghe, T.V.; Vandenabeele, P.; Kroemer, G. The molecular machinery of regulated cell death. Cell Res. 2019, 29, 347–364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Lin, L.; Zhang, M.X.; Zhang, L.; Zhang, D.; Li, C.; Li, Y.L. Autophagy, Pyroptosis, and Ferroptosis: New Regulatory Mechanisms for Atherosclerosis. Front. Cell Dev. Biol. 2021, 9, 809955. [Google Scholar] [CrossRef]
  16. Xie, Y.; Hou, W.; Song, X.; Yu, Y.; Huang, J.; Sun, X.; Kang, R.; Tang, D. Ferroptosis: Process and function. Cell Death Differ. 2016, 23, 369–379. [Google Scholar] [CrossRef] [Green Version]
  17. Kroemer, G.; Galluzzi, L.; Vandenabeele, P.; Abrams, J.; Alnemri, E.S.; Baehrecke, E.H.; Blagosklonny, M.V.; El-Deiry, W.S.; Golstein, P.; Green, D.R.; et al. Classification of cell death: Recommendations of the Nomenclature Committee on Cell Death 2009. Cell Death Differ. 2009, 16, 3–11. [Google Scholar] [CrossRef]
  18. Galluzzi, L.; Vitale, I.; Abrams, J.M.; Alnemri, E.S.; Baehrecke, E.H.; Blagosklonny, M.V.; Dawson, T.M.; Dawson, V.L.; El-Deiry, W.S.; Fulda, S.; et al. Molecular definitions of cell death subroutines: Recommendations of the Nomenclature Committee on Cell Death 2012. Cell Death Differ. 2012, 19, 107–120. [Google Scholar] [CrossRef] [Green Version]
  19. Kroemer, G.; El-Deiry, W.S.; Golstein, P.; Peter, M.E.; Vaux, D.; Vandenabeele, P.; Zhivotovsky, B.; Blagosklonny, M.V.; Malorni, W.; Knight, R.A.; et al. Classification of cell death: Recommendations of the Nomenclature Committee on Cell Death. Cell Death Differ. 2005, 12 (Suppl. S2), 1463–1467. [Google Scholar] [CrossRef] [Green Version]
  20. Dixon, S.J.; Stockwell, B.R. The role of iron and reactive oxygen species in cell death. Nat. Chem. Biol. 2014, 10, 9–17. [Google Scholar] [CrossRef]
  21. Yang, W.S.; Kim, K.J.; Gaschler, M.M.; Patel, M.; Shchepinov, M.S.; Stockwell, B.R. Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc. Natl. Acad. Sci. USA 2016, 113, E4966–E4975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Yang, W.S.; Stockwell, B.R. Synthetic lethal screening identifies compounds activating iron-dependent, nonapoptotic cell death in oncogenic-RAS-harboring cancer cells. Chem. Biol. 2008, 15, 234–245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Gao, M.; Monian, P.; Quadri, N.; Ramasamy, R.; Jiang, X. Glutaminolysis and Transferrin Regulate Ferroptosis. Mol. Cell 2015, 59, 298–308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Buccarelli, M.; Marconi, M.; Pacioni, S.; De Pascalis, I.; D’Alessandris, Q.G.; Martini, M.; Ascione, B.; Malorni, W.; Larocca, L.M.; Pallini, R.; et al. Inhibition of autophagy increases susceptibility of glioblastoma stem cells to temozolomide by igniting ferroptosis. Cell Death Dis. 2018, 9, 841. [Google Scholar] [CrossRef] [Green Version]
  25. Torii, S.; Shintoku, R.; Kubota, C.; Yaegashi, M.; Torii, R.; Sasaki, M.; Suzuki, T.; Mori, M.; Yoshimoto, Y.; Takeuchi, T.; et al. An essential role for functional lysosomes in ferroptosis of cancer cells. Biochem. J. 2016, 473, 769–777. [Google Scholar] [CrossRef] [Green Version]
  26. Hou, W.; Xie, Y.; Song, X.; Sun, X.; Lotze, M.T.; Zeh, H.J., 3rd; Kang, R.; Tang, D. Autophagy promotes ferroptosis by degradation of ferritin. Autophagy 2016, 12, 1425–1428. [Google Scholar] [CrossRef]
  27. Gao, M.; Monian, P.; Pan, Q.; Zhang, W.; Xiang, J.; Jiang, X. Ferroptosis is an autophagic cell death process. Cell Res. 2016, 26, 1021–1032. [Google Scholar] [CrossRef] [Green Version]
  28. Ingold, I.; Berndt, C.; Schmitt, S.; Doll, S.; Poschmann, G.; Buday, K.; Roveri, A.; Peng, X.; Porto Freitas, F.; Seibt, T.; et al. Selenium Utilization by GPX4 Is Required to Prevent Hydroperoxide-Induced Ferroptosis. Cell 2018, 172, 409–422.e421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Ouyang, S.; You, J.; Zhi, C.; Li, P.; Lin, X.; Tan, X.; Ma, W.; Li, L.; Xie, W. Ferroptosis: The potential value target in atherosclerosis. Cell Death Dis. 2021, 12, 782. [Google Scholar] [CrossRef]
  30. Jiang, L.; Kon, N.; Li, T.; Wang, S.J.; Su, T.; Hibshoosh, H.; Baer, R.; Gu, W. Ferroptosis as a p53-mediated activity during tumour suppression. Nature 2015, 520, 57–62. [Google Scholar] [CrossRef]
  31. Song, X.; Long, D. Nrf2 and Ferroptosis: A New Research Direction for Neurodegenerative Diseases. Front. Neurosci. 2020, 14, 267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Fan, Z.; Wirth, A.K.; Chen, D.; Wruck, C.J.; Rauh, M.; Buchfelder, M.; Savaskan, N. Nrf2-Keap1 pathway promotes cell proliferation and diminishes ferroptosis. Oncogenesis 2017, 6, e371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Conrad, M.; Proneth, B. Broken hearts: Iron overload, ferroptosis and cardiomyopathy. Cell Res. 2019, 29, 263–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Doll, S.; Freitas, F.P.; Shah, R.; Aldrovandi, M.; da Silva, M.C.; Ingold, I.; Goya Grocin, A.; Xavier da Silva, T.N.; Panzilius, E.; Scheel, C.H.; et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature 2019, 575, 693–698. [Google Scholar] [CrossRef]
  35. Bersuker, K.; Hendricks, J.M.; Li, Z.; Magtanong, L.; Ford, B.; Tang, P.H.; Roberts, M.A.; Tong, B.; Maimone, T.J.; Zoncu, R.; et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature 2019, 575, 688–692. [Google Scholar] [CrossRef]
  36. Kraft, V.A.N.; Bezjian, C.T.; Pfeiffer, S.; Ringelstetter, L.; Müller, C.; Zandkarimi, F.; Merl-Pham, J.; Bao, X.; Anastasov, N.; Kössl, J.; et al. GTP Cyclohydrolase 1/Tetrahydrobiopterin Counteract Ferroptosis through Lipid Remodeling. ACS Cent. Sci. 2020, 6, 41–53. [Google Scholar] [CrossRef] [Green Version]
  37. Soula, M.; Weber, R.A.; Zilka, O.; Alwaseem, H.; La, K.; Yen, F.; Molina, H.; Garcia-Bermudez, J.; Pratt, D.A.; Birsoy, K. Metabolic determinants of cancer cell sensitivity to canonical ferroptosis inducers. Nat. Chem. Biol. 2020, 16, 1351–1360. [Google Scholar] [CrossRef]
  38. Lee, H.; Zandkarimi, F.; Zhang, Y.; Meena, J.K.; Kim, J.; Zhuang, L.; Tyagi, S.; Ma, L.; Westbrook, T.F.; Steinberg, G.R.; et al. Energy-stress-mediated AMPK activation inhibits ferroptosis. Nat. Cell Biol. 2020, 22, 225–234. [Google Scholar] [CrossRef]
  39. Song, X.; Zhu, S.; Chen, P.; Hou, W.; Wen, Q.; Liu, J.; Xie, Y.; Liu, J.; Klionsky, D.J.; Kroemer, G.; et al. AMPK-Mediated BECN1 Phosphorylation Promotes Ferroptosis by Directly Blocking System X(c)(-) Activity. Curr. Biol. CB 2018, 28, 2388–2399.e2385. [Google Scholar] [CrossRef] [Green Version]
  40. Garcia-Bermudez, J.; Birsoy, K. A mitochondrial gatekeeper that helps cells escape death by ferroptosis. Nature 2021, 593, 514–515. [Google Scholar] [CrossRef]
  41. Higashi, Y.; Maruhashi, T.; Noma, K.; Kihara, Y. Oxidative stress and endothelial dysfunction: Clinical evidence and therapeutic implications. Trends Cardiovasc. Med. 2014, 24, 165–169. [Google Scholar] [CrossRef] [PubMed]
  42. Sitia, S.; Tomasoni, L.; Atzeni, F.; Ambrosio, G.; Cordiano, C.; Catapano, A.; Tramontana, S.; Perticone, F.; Naccarato, P.; Camici, P.; et al. From endothelial dysfunction to atherosclerosis. Autoimmun. Rev. 2010, 9, 830–834. [Google Scholar] [CrossRef] [PubMed]
  43. Fang, Y.; Gao, S.; Wang, X.; Cao, Y.; Lu, J.; Chen, S.; Lenahan, C.; Zhang, J.H.; Shao, A.; Zhang, J. Programmed Cell Deaths and Potential Crosstalk with Blood-Brain Barrier Dysfunction after Hemorrhagic Stroke. Front. Cell. Neurosci. 2020, 14, 68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Jia, C.; Zhang, J.; Chen, H.; Zhuge, Y.; Chen, H.; Qian, F.; Zhou, K.; Niu, C.; Wang, F.; Qiu, H.; et al. Endothelial cell pyroptosis plays an important role in Kawasaki disease via HMGB1/RAGE/cathespin B signaling pathway and NLRP3 inflammasome activation. Cell Death Dis. 2019, 10, 778. [Google Scholar] [CrossRef] [Green Version]
  45. Wang, Y.; Tang, M. PM2.5 induces ferroptosis in human endothelial cells through iron overload and redox imbalance. Environ. Pollut. 2019, 254, 112937. [Google Scholar] [CrossRef] [PubMed]
  46. Liu, Z.; Xia, X.; Lv, X.; Song, E.; Song, Y. Iron-bearing nanoparticles trigger human umbilical vein endothelial cells ferroptotic responses by promoting intracellular iron level. Environ. Pollut. 2021, 287, 117345. [Google Scholar] [CrossRef]
  47. Luo, E.F.; Li, H.X.; Qin, Y.H.; Qiao, Y.; Yan, G.L.; Yao, Y.Y.; Li, L.Q.; Hou, J.T.; Tang, C.C.; Wang, D. Role of ferroptosis in the process of diabetes-induced endothelial dysfunction. World J. Diabetes 2021, 12, 124–137. [Google Scholar] [CrossRef]
  48. Sakai, O.; Yasuzawa, T.; Sumikawa, Y.; Ueta, T.; Imai, H.; Sawabe, A.; Ueshima, S. Role of GPx4 in human vascular endothelial cells, and the compensatory activity of brown rice on GPx4 ablation condition. Pathophysiol. Off. J. Int. Soc. Pathophysiol. 2017, 24, 9–15. [Google Scholar] [CrossRef]
  49. Li, Q.; Liu, C.; Deng, L.; Xie, E.; Yadav, N.; Tie, Y.; Cheng, Z.; Deng, J. Novel function of fluvastatin in attenuating oxidized low-density lipoprotein-induced endothelial cell ferroptosis in a glutathione peroxidase4- and cystine-glutamate antiporter-dependent manner. Exp. Ther. Med. 2021, 22, 1275. [Google Scholar] [CrossRef]
  50. Yang, K.; Song, H.; Yin, D. PDSS2 Inhibits the Ferroptosis of Vascular Endothelial Cells in Atherosclerosis by Activating Nrf2. J. Cardiovasc. Pharmacol. 2021, 77, 767–776. [Google Scholar] [CrossRef]
  51. Xiong, H.M. ZnO nanoparticles applied to bioimaging and drug delivery. Adv. Mater. 2013, 25, 5329–5335. [Google Scholar] [CrossRef] [PubMed]
  52. Qin, X.; Zhang, J.; Wang, B.; Xu, G.; Yang, X.; Zou, Z.; Yu, C. Ferritinophagy is involved in the zinc oxide nanoparticles-induced ferroptosis of vascular endothelial cells. Autophagy 2021, 17, 4266–4285. [Google Scholar] [CrossRef] [PubMed]
  53. Liu, Z.; Cao, S.; Chen, Q.; Fu, F.; Cheng, M.; Huang, X. MicroRNA-132 promotes atherosclerosis by inducing mitochondrial oxidative stressmediated ferroptosis. Nan Fang Yi Ke Da Xue Xue Bao = J. South. Med. Univ. 2022, 42, 143–149. [Google Scholar]
  54. Li, L.; Wang, H.; Zhang, J.; Chen, X.; Zhang, Z.; Li, Q. Effect of endothelial progenitor cell-derived extracellular vesicles on endothelial cell ferroptosis and atherosclerotic vascular endothelial injury. Cell Death Discov. 2021, 7, 235. [Google Scholar] [CrossRef]
  55. Meng, Z.; Liang, H.; Zhao, J.; Gao, J.; Liu, C.; Ma, X.; Liu, J.; Liang, B.; Jiao, X.; Cao, J.; et al. HMOX1 upregulation promotes ferroptosis in diabetic atherosclerosis. Life Sci. 2021, 284, 119935. [Google Scholar] [CrossRef] [PubMed]
  56. Shi, P.; Li, M.; Song, C.; Qi, H.; Ba, L.; Cao, Y.; Zhang, M.; Xie, Y.; Ren, J.; Wu, J.; et al. Neutrophil-like cell membrane-coated siRNA of lncRNA AABR07017145.1 therapy for cardiac hypertrophy via inhibiting ferroptosis of CMECs. Mol. Ther. Nucleic Acids 2022, 27, 16–36. [Google Scholar] [CrossRef]
  57. Shi, P.; Song, C.; Qi, H.; Ren, J.; Ren, P.; Wu, J.; Xie, Y.; Zhang, M.; Sun, H.; Cao, Y. Up-regulation of IRF3 is required for docosahexaenoic acid suppressing ferroptosis of cardiac microvascular endothelial cells in cardiac hypertrophy rat. J. Nutr. Biochem. 2022, 104, 108972. [Google Scholar] [CrossRef]
  58. Zhang, Z.; Tang, J.; Song, J.; Xie, M.; Liu, Y.; Dong, Z.; Liu, X.; Li, X.; Zhang, M.; Chen, Y.; et al. Elabela alleviates ferroptosis, myocardial remodeling, fibrosis and heart dysfunction in hypertensive mice by modulating the IL-6/STAT3/GPX4 signaling. Free Radic. Biol. Med. 2022, 181, 130–142. [Google Scholar] [CrossRef]
  59. Zhao, L.; Yang, N.; Song, Y.; Si, H.; Qin, Q.; Guo, Z. Effect of iron overload on endothelial cell calcification and its mechanism. Ann. Transl. Med. 2021, 9, 1658. [Google Scholar] [CrossRef]
  60. He, L.; Liu, Y.Y.; Wang, K.; Li, C.; Zhang, W.; Li, Z.Z.; Huang, X.Z.; Xiong, Y. Tanshinone IIA protects human coronary artery endothelial cells from ferroptosis by activating the NRF2 pathway. Biochem. Biophys. Res. Commun. 2021, 575, 1–7. [Google Scholar] [CrossRef]
  61. Chen, B.; Wang, H.; Lv, C.; Mao, C.; Cui, Y. Long non-coding RNA H19 protects against intracerebral hemorrhage injuries via regulating microRNA-106b-5p/acyl-CoA synthetase long chain family member 4 axis. Bioengineered 2021, 12, 4004–4015. [Google Scholar] [CrossRef]
  62. Dharmalingam, P.; Talakatta, G.; Mitra, J.; Wang, H.; Derry, P.J.; Nilewski, L.G.; McHugh, E.A.; Fabian, R.H.; Mendoza, K.; Vasquez, V.; et al. Pervasive Genomic Damage in Experimental Intracerebral Hemorrhage: Therapeutic Potential of a Mechanistic-Based Carbon Nanoparticle. ACS Nano 2020, 14, 2827–2846. [Google Scholar] [CrossRef] [PubMed]
  63. Gao, S.; Zhou, L.; Lu, J.; Fang, Y.; Wu, H.; Xu, W.; Pan, Y.; Wang, J.; Wang, X.; Zhang, J.; et al. Cepharanthine Attenuates Early Brain Injury after Subarachnoid Hemorrhage in Mice via Inhibiting 15-Lipoxygenase-1-Mediated Microglia and Endothelial Cell Ferroptosis. Oxid. Med. Cell. Longev. 2022, 2022, 4295208. [Google Scholar] [CrossRef]
  64. Chen, C.; Huang, Y.; Xia, P.; Zhang, F.; Li, L.; Wang, E.; Guo, Q.; Ye, Z. Long noncoding RNA Meg3 mediates ferroptosis induced by oxygen and glucose deprivation combined with hyperglycemia in rat brain microvascular endothelial cells, through modulating the p53/GPX4 axis. Eur. J. Histochem. EJH 2021, 65, 3224. [Google Scholar] [CrossRef]
  65. Abdul, Y.; Li, W.; Ward, R.; Abdelsaid, M.; Hafez, S.; Dong, G.; Jamil, S.; Wolf, V.; Johnson, M.H.; Fagan, S.C.; et al. Deferoxamine Treatment Prevents Post-Stroke Vasoregression and Neurovascular Unit Remodeling Leading to Improved Functional Outcomes in Type 2 Male Diabetic Rats: Role of Endothelial Ferroptosis. Transl. Stroke Res. 2021, 12, 615–630. [Google Scholar] [CrossRef] [PubMed]
  66. Guo, X.W.; Zhang, H.; Huang, J.Q.; Wang, S.N.; Lu, Y.; Cheng, B.; Dong, S.H.; Wang, Y.Y.; Li, F.S.; Li, Y.W. PIEZO1 Ion Channel Mediates Ionizing Radiation-Induced Pulmonary Endothelial Cell Ferroptosis via Ca(2+)/Calpain/VE-Cadherin Signaling. Front. Mol. Biosci. 2021, 8, 725274. [Google Scholar] [CrossRef] [PubMed]
  67. Xie, S.S.; Deng, Y.; Guo, S.L.; Li, J.Q.; Zhou, Y.C.; Liao, J.; Wu, D.D.; Lan, W.F. Endothelial cell ferroptosis mediates monocrotaline-induced pulmonary hypertension in rats by modulating NLRP3 inflammasome activation. Sci. Rep. 2022, 12, 3056. [Google Scholar] [CrossRef]
  68. Wang, Y.; Kuang, X.; Yin, Y.; Han, N.; Chang, L.; Wang, H.; Hou, Y.; Li, H.; Li, Z.; Liu, Y.; et al. Tongxinluo prevents chronic obstructive pulmonary disease complicated with atherosclerosis by inhibiting ferroptosis and protecting against pulmonary microvascular barrier dysfunction. Biomed. Pharmacother. 2022, 145, 112367. [Google Scholar] [CrossRef] [PubMed]
  69. Weber, C.; Noels, H. Atherosclerosis: Current pathogenesis and therapeutic options. Nat. Med. 2011, 17, 1410–1422. [Google Scholar] [CrossRef]
  70. Vinchi, F.; Porto, G.; Simmelbauer, A.; Altamura, S.; Passos, S.T.; Garbowski, M.; Silva, A.M.N.; Spaich, S.; Seide, S.E.; Sparla, R.; et al. Atherosclerosis is aggravated by iron overload and ameliorated by dietary and pharmacological iron restriction. Eur. Heart J. 2020, 41, 2681–2695. [Google Scholar] [CrossRef]
  71. Widlansky, M.E.; Gokce, N.; Keaney, J.F., Jr.; Vita, J.A. The clinical implications of endothelial dysfunction. J. Am. Coll. Cardiol. 2003, 42, 1149–1160. [Google Scholar] [CrossRef]
  72. Etminan, N.; Chang, H.S.; Hackenberg, K.; de Rooij, N.K.; Vergouwen, M.D.I.; Rinkel, G.J.E.; Algra, A. Worldwide Incidence of Aneurysmal Subarachnoid Hemorrhage According to Region, Time Period, Blood Pressure, and Smoking Prevalence in the Population: A Systematic Review and Meta-analysis. JAMA Neurol. 2019, 76, 588–597. [Google Scholar] [CrossRef] [PubMed]
  73. Campbell, B.C.V.; Khatri, P. Stroke. Lancet 2020, 396, 129–142. [Google Scholar] [CrossRef]
  74. Hostettler, I.C.; Seiffge, D.J.; Werring, D.J. Intracerebral hemorrhage: An update on diagnosis and treatment. Expert Rev. Neurother. 2019, 19, 679–694. [Google Scholar] [CrossRef]
  75. Karuppagounder, S.S.; Alin, L.; Chen, Y.; Brand, D.; Bourassa, M.W.; Dietrich, K.; Wilkinson, C.M.; Nadeau, C.A.; Kumar, A.; Perry, S.; et al. N-acetylcysteine targets 5 lipoxygenase-derived, toxic lipids and can synergize with prostaglandin E(2) to inhibit ferroptosis and improve outcomes following hemorrhagic stroke in mice. Ann. Neurol. 2018, 84, 854–872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Chen, B.; Chen, Z.; Liu, M.; Gao, X.; Cheng, Y.; Wei, Y.; Wu, Z.; Cui, D.; Shang, H. Inhibition of neuronal ferroptosis in the acute phase of intracerebral hemorrhage shows long-term cerebroprotective effects. Brain Res. Bull. 2019, 153, 122–132. [Google Scholar] [CrossRef]
  77. Cao, Y.; Li, Y.; He, C.; Yan, F.; Li, J.R.; Xu, H.Z.; Zhuang, J.F.; Zhou, H.; Peng, Y.C.; Fu, X.J.; et al. Selective Ferroptosis Inhibitor Liproxstatin-1 Attenuates Neurological Deficits and Neuroinflammation after Subarachnoid Hemorrhage. Neurosci. Bull. 2021, 37, 535–549. [Google Scholar] [CrossRef]
  78. Li, Y.; Liu, Y.; Wu, P.; Tian, Y.; Liu, B.; Wang, J.; Bihl, J.; Shi, H. Inhibition of Ferroptosis Alleviates Early Brain Injury after Subarachnoid Hemorrhage In Vitro and In Vivo via Reduction of Lipid Peroxidation. Cell. Mol. Neurobiol. 2021, 41, 263–278. [Google Scholar] [CrossRef] [PubMed]
  79. Qu, X.F.; Liang, T.Y.; Wu, D.G.; Lai, N.S.; Deng, R.M.; Ma, C.; Li, X.; Li, H.Y.; Liu, Y.Z.; Shen, H.T.; et al. Acyl-CoA synthetase long chain family member 4 plays detrimental role in early brain injury after subarachnoid hemorrhage in rats by inducing ferroptosis. CNS Neurosci. Ther. 2021, 27, 449–463. [Google Scholar] [CrossRef]
  80. Virani, S.S.; Alonso, A.; Benjamin, E.J.; Bittencourt, M.S.; Callaway, C.W.; Carson, A.P.; Chamberlain, A.M.; Chang, A.R.; Cheng, S.; Delling, F.N.; et al. Heart Disease and Stroke Statistics-2020 Update: A Report from the American Heart Association. Circulation 2020, 141, e139–e596. [Google Scholar] [CrossRef]
  81. Li, W.; Ward, R.; Valenzuela, J.P.; Dong, G.; Fagan, S.C.; Ergul, A. Diabetes Worsens Functional Outcomes in Young Female Rats: Comparison of Stroke Models, Tissue Plasminogen Activator Effects, and Sexes. Transl. Stroke Res. 2017, 8, 429–439. [Google Scholar] [CrossRef] [PubMed]
  82. Butt, Y.; Kurdowska, A.; Allen, T.C. Acute Lung Injury: A Clinical and Molecular Review. Arch. Pathol. Lab. Med. 2016, 140, 345–350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Hughes, K.T.; Beasley, M.B. Pulmonary Manifestations of Acute Lung Injury: More than Just Diffuse Alveolar Damage. Arch. Pathol. Lab. Med. 2017, 141, 916–922. [Google Scholar] [CrossRef] [Green Version]
  84. Li, X.; Zhuang, X.; Qiao, T. Role of ferroptosis in the process of acute radiation-induced lung injury in mice. Biochem. Biophys. Res. Commun. 2019, 519, 240–245. [Google Scholar] [CrossRef]
  85. She, H.; Hu, Y.; Zhou, Y.; Tan, L.; Zhu, Y.; Ma, C.; Wu, Y.; Chen, W.; Wang, L.; Zhang, Z.; et al. Protective Effects of Dexmedetomidine on Sepsis-Induced Vascular Leakage by Alleviating Ferroptosis via Regulating Metabolic Reprogramming. J. Inflamm. Res. 2021, 14, 6765–6782. [Google Scholar] [CrossRef]
  86. Li, M.; Zhang, D.; Zhang, L.; Wang, L.; Tian, Y.; Li, B.; Dong, J.; Xiao, F. The inhibitory effect of chemical components from Ginkgo biloba flower on ferroptosis in vascular endothelial cells. J. Int. Pharm. Res. 2020, 47, 857–862. [Google Scholar]
  87. Sheng, S.; Xu, J.; Liang, Q.; Hong, L.; Zhang, L. Astragaloside IV Inhibits Bleomycin-Induced Ferroptosis in Human Umbilical Vein Endothelial Cells by Mediating LPC. Oxid. Med. Cell. Longev. 2021, 2021, 6241242. [Google Scholar] [CrossRef]
  88. Feng, Y.D.; Ye, W.; Tian, W.; Meng, J.R.; Zhang, M.; Sun, Y.; Zhang, H.N.; Wang, S.J.; Wu, K.H.; Liu, C.X.; et al. Old targets, new strategy: Apigenin-7-O-β-d-(-6″-p-coumaroyl)-glucopyranoside prevents endothelial ferroptosis and alleviates intestinal ischemia-reperfusion injury through HO-1 and MAO-B inhibition. Free Radic. Biol. Med. 2022, 184, 74–88. [Google Scholar] [CrossRef]
  89. Xiao, F.J.; Zhang, D.; Wu, Y.; Jia, Q.H.; Zhang, L.; Li, Y.X.; Yang, Y.F.; Wang, H.; Wu, C.T.; Wang, L.S. miRNA-17-92 protects endothelial cells from erastin-induced ferroptosis through targeting the A20-ACSL4 axis. Biochem. Biophys. Res. Commun. 2019, 515, 448–454. [Google Scholar] [CrossRef]
  90. Xia, J.; Song, X.; Meng, J.; Lou, D. Endothelial progenitor cells-derived exosomes transfer microRNA-30e-5p to regulate Erastin-induced ferroptosis in human umbilical vein endothelial cells via the specificity protein 1/adenosine monophosphate-activated protein kinase axis. Bioengineered 2022, 13, 3566–3580. [Google Scholar] [CrossRef]
  91. Zhang, J.; Qiu, Q.; Wang, H.; Chen, C.; Luo, D. TRIM46 contributes to high glucose-induced ferroptosis and cell growth inhibition in human retinal capillary endothelial cells by facilitating GPX4 ubiquitination. Exp. Cell Res. 2021, 407, 112800. [Google Scholar] [CrossRef]
  92. Lovatt, M.; Adnan, K.; Kocaba, V.; Dirisamer, M.; Peh, G.S.L.; Mehta, J.S. Peroxiredoxin-1 regulates lipid peroxidation in corneal endothelial cells. Redox Biol. 2020, 30, 101417. [Google Scholar] [CrossRef]
  93. Ma, Q.; Dai, X.; Lu, W.; Qu, X.; Liu, N.; Zhu, C. Silencing long non-coding RNA MEG8 inhibits the proliferation and induces the ferroptosis of hemangioma endothelial cells by regulating miR-497-5p/NOTCH2 axis. Biochem. Biophys. Res. Commun. 2021, 556, 72–78. [Google Scholar] [CrossRef]
  94. Zilka, O.; Shah, R.; Li, B.; Friedmann Angeli, J.P.; Griesser, M.; Conrad, M.; Pratt, D.A. On the Mechanism of Cytoprotection by Ferrostatin-1 and Liproxstatin-1 and the Role of Lipid Peroxidation in Ferroptotic Cell Death. ACS Cent. Sci. 2017, 3, 232–243. [Google Scholar] [CrossRef]
  95. Dayani, P.N.; Bishop, M.C.; Black, K.; Zeltzer, P.M. Desferoxamine (DFO)--mediated iron chelation: Rationale for a novel approach to therapy for brain cancer. J. Neuro-Oncol. 2004, 67, 367–377. [Google Scholar] [CrossRef]
  96. Barradas, M.A.; Jeremy, J.Y.; Kontoghiorghes, G.J.; Mikhailidis, D.P.; Hoffbrand, A.V.; Dandona, P. Iron chelators inhibit human platelet aggregation, thromboxane A2 synthesis and lipoxygenase activity. FEBS Lett. 1989, 245, 105–109. [Google Scholar] [CrossRef] [Green Version]
  97. Zhang, W.J.; Wei, H.; Frei, B. The iron chelator, desferrioxamine, reduces inflammation and atherosclerotic lesion development in experimental mice. Exp. Biol. Med. 2010, 235, 633–641. [Google Scholar] [CrossRef] [Green Version]
  98. Shi, J.; Kantoff, P.W.; Wooster, R.; Farokhzad, O.C. Cancer nanomedicine: Progress, challenges and opportunities. Nat. Rev. Cancer 2017, 17, 20–37. [Google Scholar] [CrossRef]
  99. Kibble, M.; Saarinen, N.; Tang, J.; Wennerberg, K.; Mäkelä, S.; Aittokallio, T. Network pharmacology applications to map the unexplored target space and therapeutic potential of natural products. Nat. Prod. Rep. 2015, 32, 1249–1266. [Google Scholar] [CrossRef]
  100. Kim, C.; Kim, B. Anti-Cancer Natural Products and Their Bioactive Compounds Inducing ER Stress-Mediated Apoptosis: A Review. Nutrients 2018, 10, 1021. [Google Scholar] [CrossRef] [Green Version]
  101. Choudhury, H.; Pandey, M.; Hua, C.K.; Mun, C.S.; Jing, J.K.; Kong, L.; Ern, L.Y.; Ashraf, N.A.; Kit, S.W.; Yee, T.S.; et al. An update on natural compounds in the remedy of diabetes mellitus: A systematic review. J. Tradit. Complement. Med. 2018, 8, 361–376. [Google Scholar] [CrossRef]
  102. Michel, J.; Abd Rani, N.Z.; Husain, K. A Review on the Potential Use of Medicinal Plants from Asteraceae and Lamiaceae Plant Family in Cardiovascular Diseases. Front. Pharmacol. 2020, 11, 852. [Google Scholar] [CrossRef]
  103. Wu, L.; Liu, M.; Liang, J.; Li, N.; Yang, D.; Cai, J.; Zhang, Y.; He, Y.; Chen, Z.; Ma, T. Ferroptosis as a New Mechanism in Parkinson’s Disease Therapy Using Traditional Chinese Medicine. Front. Pharmacol. 2021, 12, 659584. [Google Scholar] [CrossRef]
  104. Duan, L.; Zhang, Y.; Yang, Y.; Su, S.; Zhou, L.; Lo, P.C.; Cai, J.; Qiao, Y.; Li, M.; Huang, S.; et al. Baicalin Inhibits Ferroptosis in Intracerebral Hemorrhage. Front. Pharmacol. 2021, 12, 629379. [Google Scholar] [CrossRef]
  105. Ge, C.; Zhang, S.; Mu, H.; Zheng, S.; Tan, Z.; Huang, X.; Xu, C.; Zou, J.; Zhu, Y.; Feng, D.; et al. Emerging Mechanisms and Disease Implications of Ferroptosis: Potential Applications of Natural Products. Front. Cell Dev. Biol. 2021, 9, 774957. [Google Scholar] [CrossRef]
  106. Zheng, K.; Dong, Y.; Yang, R.; Liang, Y.; Wu, H.; He, Z. Regulation of ferroptosis by bioactive phytochemicals: Implications for medical nutritional therapy. Pharmacol. Res. 2021, 168, 105580. [Google Scholar] [CrossRef]
  107. Ansari, M.A.; Khan, F.B.; Safdari, H.A.; Almatroudi, A.; Alzohairy, M.A.; Safdari, M.; Amirizadeh, M.; Rehman, S.; Equbal, M.J.; Hoque, M. Prospective therapeutic potential of Tanshinone IIA: An updated overview. Pharmacol. Res. 2021, 164, 105364. [Google Scholar] [CrossRef]
  108. Wen, J.; Chang, Y.; Huo, S.; Li, W.; Huang, H.; Gao, Y.; Lin, H.; Zhang, J.; Zhang, Y.; Zuo, Y.; et al. Tanshinone IIA attenuates atherosclerosis via inhibiting NLRP3 inflammasome activation. Aging 2020, 13, 910–932. [Google Scholar] [CrossRef]
  109. Li, Z.; Zhao, F.; Cao, Y.; Zhang, J.; Shi, P.; Sun, X.; Zhang, F.; Tong, L. DHA attenuates hepatic ischemia reperfusion injury by inhibiting pyroptosis and activating PI3K/Akt pathway. Eur. J. Pharm. 2018, 835, 1–10. [Google Scholar] [CrossRef]
  110. Wapinski, O.; Chang, H.Y. Long noncoding RNAs and human disease. Trends Cell Biol. 2011, 21, 354–361. [Google Scholar] [CrossRef]
  111. Beermann, J.; Piccoli, M.T.; Viereck, J.; Thum, T. Non-coding RNAs in Development and Disease: Background, Mechanisms, and Therapeutic Approaches. Physiol. Rev. 2016, 96, 1297–1325. [Google Scholar] [CrossRef] [Green Version]
  112. Huang, Y. The novel regulatory role of lncRNA-miRNA-mRNA axis in cardiovascular diseases. J. Cell. Mol. Med. 2018, 22, 5768–5775. [Google Scholar] [CrossRef]
  113. Yang, J.X.; Pan, Y.Y.; Wang, X.X.; Qiu, Y.G.; Mao, W. Endothelial progenitor cells in age-related vascular remodeling. Cell Transplant. 2018, 27, 786–795. [Google Scholar] [CrossRef]
  114. Yáñez-Mó, M.; Siljander, P.R.; Andreu, Z.; Zavec, A.B.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef] [Green Version]
  115. Wang, J.; Chen, S.; Zhang, W.; Chen, Y.; Bihl, J.C. Exosomes from miRNA-126-modified endothelial progenitor cells alleviate brain injury and promote functional recovery after stroke. CNS Neurosci. Ther. 2020, 26, 1255–1265. [Google Scholar] [CrossRef]
  116. Bai, S.; Yin, Q.; Dong, T.; Dai, F.; Qin, Y.; Ye, L.; Du, J.; Zhang, Q.; Chen, H.; Shen, B. Endothelial progenitor cell-derived exosomes ameliorate endothelial dysfunction in a mouse model of diabetes. Biomed. Pharmacother. 2020, 131, 110756. [Google Scholar] [CrossRef]
  117. Gan, F.; Wang, R.; Lyu, P.; Li, Y.; Fu, R.; Du, Y.; Gong, P.; Yao, Y. Plasma-Derived Exosomes Boost the Healing of Irradiated Wound by Regulating Cell Proliferation and Ferroptosis. J. Biomed. Nanotechnol. 2021, 17, 100–114. [Google Scholar] [CrossRef]
  118. Zhao, H.; Li, X.; Yang, L.; Zhang, L.; Jiang, X.; Gao, W.; Chen, P.; Cheng, Y.; Wang, F.; Liu, J. Isorhynchophylline Relieves Ferroptosis-Induced Nerve Damage after Intracerebral Hemorrhage via miR-122-5p/TP53/SLC7A11 Pathway. Neurochem. Res. 2021, 46, 1981–1994. [Google Scholar] [CrossRef]
  119. Yang, S.; Yin, J.; Hou, X. Inhibition of miR-135b by SP-1 promotes hypoxia-induced vascular endothelial cell injury via HIF-1α. Exp. Cell Res. 2018, 370, 31–38. [Google Scholar] [CrossRef]
  120. Lee, W.R.; Kim, K.H.; An, H.J.; Park, Y.Y.; Kim, K.S.; Lee, C.K.; Min, B.K.; Park, K.K. Effects of chimeric decoy oligodeoxynucleotide in the regulation of transcription factors NF-κB and Sp1 in an animal model of atherosclerosis. Basic Clin. Pharmacol. Toxicol. 2013, 112, 236–243. [Google Scholar] [CrossRef]
  121. Dodson, M.; de la Vega, M.R.; Cholanians, A.B.; Schmidlin, C.J.; Chapman, E.; Zhang, D.D. Modulating NRF2 in Disease: Timing Is Everything. Annu. Rev. Pharmacol. Toxicol. 2019, 59, 555–575. [Google Scholar] [CrossRef] [PubMed]
  122. Dodson, M.; Castro-Portuguez, R.; Zhang, D.D. NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biol. 2019, 23, 101107. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Key mechanisms of ferroptosis. The regulatory pathways of ferroptosis are mainly divided into three metabolisms (iron, lipid, and amino acid). By binding TFR1, cells take iron into endosomes as Fe3+. Six-transmembrane epithelial antigen of prostate 3 (STEAP3) helps reduce Fe3+ to Fe2+. Most Fe2+ can be stored in ferritin, utilized for enzyme synthesis, or exported by ferroportin (oxidized to Fe3+). The rest of Fe2+ may mediate Fenton reaction producing ROS. The accumulation of ROS will induce ferroptosis because it oxidizes PL-PUFA-OH into PL-PUFA-OOH, which can decompose to 4-hydroxy-2-nonenals (4-HNEs) or malondialdehydes (MDAs) that cause cellular damage. Another important cause of ferroptosis is reduced antioxidant capacity. The system Xc transporter, consisting of solute carrier family 7 member 11 (SLC7A11) and solute carrier family 3 member 2 (SLC3A2), releases glutamate in exchange for cystine, which can be further transformed into cysteine, a crucial component of GSH. With the help of GPX4, GSH can deoxidize PL-PUFA-OOH back into PL-PUFA-OH. GPX4 can inhibit ferroptosis by reducing ROS production and protecting cells from lipid hydroperoxides. In addition, the FSP1-CoQ10-NAD(P)H pathway, GCH1-BH4-DHFR pathway, mitochondria, and energy stress are involved in the progress of ferroptosis as well. GSSG: glutathione disulfide; L: lipid; -OHs: alcohols; -OOHs: hydroperoxides; CoA: coenzyme A; ACC: acetyl coenzyme A carboxylase; PL: phospholipid; POR: cytochrome P450 reductase; LOXs: lipoxygenases; TAC: tricarboxylic acid; OXPHOX: oxidative phosphorylation; CoQ10H2: the reduced form of coenzyme Q10, ubiquinol; NADH: nicotinamide adenine dinucleotide; NADPH: nicotinamide adenine dinucleotide phosphate; BH2: dihydrobiopterin.
Figure 1. Key mechanisms of ferroptosis. The regulatory pathways of ferroptosis are mainly divided into three metabolisms (iron, lipid, and amino acid). By binding TFR1, cells take iron into endosomes as Fe3+. Six-transmembrane epithelial antigen of prostate 3 (STEAP3) helps reduce Fe3+ to Fe2+. Most Fe2+ can be stored in ferritin, utilized for enzyme synthesis, or exported by ferroportin (oxidized to Fe3+). The rest of Fe2+ may mediate Fenton reaction producing ROS. The accumulation of ROS will induce ferroptosis because it oxidizes PL-PUFA-OH into PL-PUFA-OOH, which can decompose to 4-hydroxy-2-nonenals (4-HNEs) or malondialdehydes (MDAs) that cause cellular damage. Another important cause of ferroptosis is reduced antioxidant capacity. The system Xc transporter, consisting of solute carrier family 7 member 11 (SLC7A11) and solute carrier family 3 member 2 (SLC3A2), releases glutamate in exchange for cystine, which can be further transformed into cysteine, a crucial component of GSH. With the help of GPX4, GSH can deoxidize PL-PUFA-OOH back into PL-PUFA-OH. GPX4 can inhibit ferroptosis by reducing ROS production and protecting cells from lipid hydroperoxides. In addition, the FSP1-CoQ10-NAD(P)H pathway, GCH1-BH4-DHFR pathway, mitochondria, and energy stress are involved in the progress of ferroptosis as well. GSSG: glutathione disulfide; L: lipid; -OHs: alcohols; -OOHs: hydroperoxides; CoA: coenzyme A; ACC: acetyl coenzyme A carboxylase; PL: phospholipid; POR: cytochrome P450 reductase; LOXs: lipoxygenases; TAC: tricarboxylic acid; OXPHOX: oxidative phosphorylation; CoQ10H2: the reduced form of coenzyme Q10, ubiquinol; NADH: nicotinamide adenine dinucleotide; NADPH: nicotinamide adenine dinucleotide phosphate; BH2: dihydrobiopterin.
Nutrients 14 04506 g001
Figure 2. Ferroptosis of ECs is related to multiple vascular diseases, such as AS, stroke, ALI, and so on. Modified from Figdraw (www.figdraw.com, accessed on 22 May 2022).
Figure 2. Ferroptosis of ECs is related to multiple vascular diseases, such as AS, stroke, ALI, and so on. Modified from Figdraw (www.figdraw.com, accessed on 22 May 2022).
Nutrients 14 04506 g002
Table 1. The main features of various RCD subtypes.
Table 1. The main features of various RCD subtypes.
TypeMorphological FeaturesBiochemical FeaturesImmune Features
NecroptosisCell swelling; plasma membrane rupture; moderate chromatin condensationRIPK1, RIPK3, and MLKL activation; cytosolic necrosome formationMostly pro-inflammatory
ApoptosisCell rounding-up; pseudopode retraction; cellular and nuclear volume reduction; plasma membrane blebbing; nuclear fragmentation; apoptotic body formationCaspase activation; DNA fragmentation; ΔΨm dissipation; phosphatidylserine exposureOften anti-inflammatory and immune silent;
sometimes elicits an immune response
AutophagyDouble-membraned autophagic vacuole formationIncreased lysosomal activityMostly anti-inflammatory
PyroptosisCell swelling; pore formation; membrane rupture; massive leakage of cytoplasmic componentsCaspase-1/4/5/11 activation and proinflammatory cytokine releasePro-inflammatory
FerroptosisSmaller mitochondria; mitochondria crista reduction; mitochondrial membrane density elevation; increased mitochondrial membrane ruptureIron accumulation; lipid peroxidationPro-inflammatory
RIPK: receptor-interacting serine/threonine kinase; MLKL: mixed lineage kinase domain-like pseudokinase; ΔΨm: mitochondrial membrane potential.
Table 2. Treatment associated with ferroptosis of ECs in vascular diseases.
Table 2. Treatment associated with ferroptosis of ECs in vascular diseases.
CategoryVascular DiseaseCellsTreatment Leading to FerroptosisTreatment Inhibiting Ferroptosis and Its MechanismRef.
Cardiovascular diseaseAtherosclerosisHCAECOx-LDLOverexpression of PDSS2: ↑ NRF2 pathway[50]
AtherosclerosisHUVECMiR-132/[53]
AtherosclerosisMAECOx-LDLFer-1: ↑ SCL7A11 and GPX4
DFO: ↓ iron
[12]
AtherosclerosisMAECOx-LDLEPC-EVs carrying miR-199a-3p targeting SP1[54]
Diabetic atherosclerosisHUVEC; MAEC obtained from ApoE−/− miceHigh glucose/high lipidsFer-1; knockdown of HMOX1[55]
Cardiac hypertrophyCMVECAng IINeutrophil-like cell membrane-coated siRNA of lncRNA AABR07017145.1: ↑ miR-30b-5p targeting TIMP1, ↓ TFR-1[56]
Cardiac hypertrophyCMVECAng IIDHA: ↑ IRF3, ↑ SLC7A11/ALOX12[57]
HypertensionCMVECAng IIElabela: ↓ IL-6/STAT3 pathway, ↑ xCT/GPX4 pathway[58]
Endothelial cell calcificationHUVECFeSO4DFO: ↓ iron; ferrostatin[59]
/HCAEC/Tanshinone IIA: ↑ NRF2 pathway[60]
Cerebrovascular diseaseICHRBMVECLncRNA H19Knockdown of lncRNA H19: ↑ miR-106b-5p, ↓ ACSL4[61]
ICHbEnd.3IronDFO-HCC-PEG: combined iron chelator therapy[62]
Early brain injury after SAHbEnd.3RSL3Cepharanthine: ↓ ALOX15[63]
Diabetic brain strokeRBMVECOxygen and glucose deprivation + hyperglycemic reperfusionKnockdown of lncRNA Meg3: ↓ p53, ↑ GPX4[64]
Post-stroke cognitive impairment in diabetesRBMVECIron(III) sulfateDeferoxamine: iron chelator therapy[65]
Pulmonary vascular diseaseLung injury: radiation-induced lung injuryHULEC-5aIonizing radiation: ↑ PIEZO1, ↑ intracellular calcium concentration and calpain activity, ↓ VE-cadherinGsMTx4, a specific inhibitor of PIEZO1;
PD151746, a selective calpain inhibitor;
overexpression of VE-cadherin
[66]
Pulmonary hypertensionPAECPAEC from monocrotaline-induced pulmonary hypertension ratsFer-1: ↓ HMGB1 and TLR4, ↓ NLRP3 inflammasome and proinflammatory cytokines[67]
COPD complicated with atherosclerosisHPMECCigarette smoke extract; homocysteineTongxinluo: ↑ GPX4 and FSP1, ↓ ACSL4[68]
↑ indicates elevation or activation; ↓indicates reduction or suppression. ICH: intracerebral hemorrhage; SAH: subarachnoid hemorrhage; LPS: lipopolysaccharide; COPD: chronic obstructive pulmonary disease; HCAEC: human coronary artery endothelial cell; MAEC: mouse aortic endothelial cell; CMVEC: cardiac microvascular endothelial cell; RBMVEC: rat brain microvascular endothelial cell; bEnd.3: mouse brain microvascular endothelial cell; HULEC-5a: human pulmonary microvascular endothelial cell; PAEC: pulmonary artery endothelial cell; HPMEC: human pulmonary microvascular endothelial cell; Ang II: angiotensin II; LncRNA: long non-coding RNA; PIEZO1: piezo type mechanosensitive ion channel component 1; VE-cadherin: vascular endothelial-cadherin; PDSS2: prenyldiphosphate synthase subunit 2; Fer-1: ferrostatin-1; DFO: deferoxamine; EPC-EVs: endothelial progenitor cells-secreted extracellular vesicles; siRNA: small interfering RNA; SP1: specificity protein 1; TIMP1: TIMP metallopeptidase inhibitor 1; DHA: docosahexaenoic acid; IRF3: interferon regulating factor 3; ALOX12: arachidonate 12-lipoxygenase, 12S Type; IL-6: interleukin-6; STAT3: signal transducer and activator of transcription 3; PEG-HCC: poly(ethylene glycol)-conjugated hydrophilic carbon clusters; ALOX15: arachidonate 15-lipoxygenase; Meg3: maternally expressed gene 3; HMGB1: high-mobility group box 1; TLR4: toll-like receptor 4; NLRP3: NACHT, LRR and PYD domains-containing protein 3.
Table 3. Potential inhibitors of ferroptosis in ECs.
Table 3. Potential inhibitors of ferroptosis in ECs.
Potential InhibitorsCellsInhibitorsMechanismsRef.
Clinical drugsHUVECFluvastatin↑ GPX4, ↑ xCT[49]
RPVECDexmedetomidine↑ NRF2, ↓ mitochondrial fission[85]
HPMECTongxinluo↑ GPX4 and FSP1, ↓ ACSL4[68]
Natural productsHUVECα-tocopherol (vitamin E) and extract of brown rice↑ GPX4[48]
HUVECLuteolin↓ ACSL4, ↑ GPX4[86]
HUVECAstragaloside IVPartially ↑ SLC7A11 and ↑ GPX4[87]
HUVECApigenin-7-O-β-D-(-6”-p-coumaroyl)-glucopyranoside↓ HMOX1, ↓ MAO-B[88]
HCAECTanshinone IIA↑ NRF2[60]
CMVECDHA↑ IRF3[57]
bEnd.3Cepharanthine↓ ALOX15[63]
NanomaterialsCMVECNeutrophil membrane + si-AAB + mesoponanocomplex↑ miR-30b-5p, ↓ TIMP1, ↓ TFR-1[56]
ncRNAsBMVECLncRNA H19 knockdown↑ miR-106b-5p,↓ ACSL4[61]
BMVECLncRNA Meg3-siRNA↓ Meg3, ↓ p53, ↑ GPX4[64]
HUVECOverexpression of miRNA-17-92↓ A20, ↓ ACSL4[89]
EVs or ExosMAECMouse EPC-EVs↑ miR199a-3p, ↓ SP1[54]
HUVECHuman umbilical vein blood EPC-Exos↑ miR-30e-5p, ↓ SP1, ↑ AMPK[90]
Other novel targetsHCAECOverexpression of PDSS2↑ NRF2[50]
HUVECKnockdown of HMOX1↓ iron, ↓ ROS, ↓ lipid peroxidation, ↑SLC7A11, ↑GPX4[55]
HULEC-5aGsMTx4, a specific inhibitor of PIEZO1;
PD151746, a selective calpain inhibitor;
Overexpression of VE-cadherin
↓ PIEZO1/Ca2+/calpain, ↑ VE-Cadherin[66]
HRCECKnockdown of TRIM46↑ GPX4[91]
CMVECElabela↓ IL-6/STAT3 pathway, ↑ xCT/GPX4 pathway[58]
↑ indicates elevation or activation; ↓indicates reduction or suppression. RPVEC: rat pulmonary vein endothelial cell; HRCEC: human retinal capillary endothelial cell; AAB: long non-coding RNA AABR07017145.1; Exo: exosome; TRIM46: tripartite motif containing 46; MAO-B: monoamine oxidase B; A20: zinc lipoprotein A20; SP1: specificity protein 1.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zhang, H.; Zhou, S.; Sun, M.; Hua, M.; Liu, Z.; Mu, G.; Wang, Z.; Xiang, Q.; Cui, Y. Ferroptosis of Endothelial Cells in Vascular Diseases. Nutrients 2022, 14, 4506. https://doi.org/10.3390/nu14214506

AMA Style

Zhang H, Zhou S, Sun M, Hua M, Liu Z, Mu G, Wang Z, Xiang Q, Cui Y. Ferroptosis of Endothelial Cells in Vascular Diseases. Nutrients. 2022; 14(21):4506. https://doi.org/10.3390/nu14214506

Chicago/Turabian Style

Zhang, Hanxu, Shuang Zhou, Minxue Sun, Manqi Hua, Zhiyan Liu, Guangyan Mu, Zhe Wang, Qian Xiang, and Yimin Cui. 2022. "Ferroptosis of Endothelial Cells in Vascular Diseases" Nutrients 14, no. 21: 4506. https://doi.org/10.3390/nu14214506

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop