Next Article in Journal
IL-7 Deficiency Exacerbates Atopic Dermatitis in NC/Nga Mice
Next Article in Special Issue
Effects of Gender and Vitamin D on Vascular Reactivity of the Carotid Artery on a Testosterone-Induced PCOS Model
Previous Article in Journal
PTEN Inhibitor Treatment Lowers Muscle Plasma Membrane Damage and Enhances Muscle ECM Homeostasis after High-Intensity Eccentric Exercise in Mice
Previous Article in Special Issue
Phosphorylcholine Monoclonal Antibody Therapy Decreases Intraplaque Angiogenesis and Intraplaque Hemorrhage in Murine Vein Grafts
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Clinical Potential of Hydrogen Sulfide in Peripheral Arterial Disease

by
Clémence Bechelli
,
Diane Macabrey
,
Sebastien Deglise
and
Florent Allagnat
*,†,‡
Department of Vascular Surgery, Lausanne University Hospital, 1005 Lausanne, Switzerland
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Current address: CHUV-Service de Chirurgie Vasculaire, Département des Sciences Biomédicales, Bugnon 7A, 1005 Lausanne, Switzerland.
Int. J. Mol. Sci. 2023, 24(12), 9955; https://doi.org/10.3390/ijms24129955
Submission received: 4 May 2023 / Revised: 1 June 2023 / Accepted: 5 June 2023 / Published: 9 June 2023

Abstract

:
Peripheral artery disease (PAD) affects more than 230 million people worldwide. PAD patients suffer from reduced quality of life and are at increased risk of vascular complications and all-cause mortality. Despite its prevalence, impact on quality of life and poor long-term clinical outcomes, PAD remains underdiagnosed and undertreated compared to myocardial infarction and stroke. PAD is due to a combination of macrovascular atherosclerosis and calcification, combined with microvascular rarefaction, leading to chronic peripheral ischemia. Novel therapies are needed to address the increasing incidence of PAD and its difficult long-term pharmacological and surgical management. The cysteine-derived gasotransmitter hydrogen sulfide (H2S) has interesting vasorelaxant, cytoprotective, antioxidant and anti-inflammatory properties. In this review, we describe the current understanding of PAD pathophysiology and the remarkable benefits of H2S against atherosclerosis, inflammation, vascular calcification, and other vasculo-protective effects.

1. Introduction

Peripheral artery disease (PAD), defined as “all arterial diseases other than coronary arteries and aorta”, affects more than 230 million people worldwide [1,2].
PAD is primarily due to the development of atherosclerotic plaques, leading to progressive narrowing of the vessel lumen. Limb symptoms include leg pain, cramps, fatigue, and muscle weakness during physical activity. At rest, blood flow remains sufficient to meet basal oxygen requirements and patients are free of symptoms. However, during exercise, the increased oxygen supply to the lower limb is impaired, leading to moderate ischemia, which the patient experiences as cramping pain. The patient usually stops walking until the pain subsides. Alternating cycles of walking and resting, known as intermittent claudication (IC), is the cardinal clinical manifestation of PAD [3]. Patients with IC have a reduced walking distance, leading to an inability to perform daily activities and a reduced quality of life [1,4,5]. However, IC may be present in only 10–35% of patients, whereas 40–50% of PAD patients have a wide range of atypical leg symptoms, and 20–50% of patients are asymptomatic [4,5,6]. The femoral and popliteal arteries are the most common sites of atherosclerotic disease in patients with PAD. Approximately 80–90% of patients with symptomatic PAD have some combination of femoropopliteal occlusive disease [4,5,7].
In late-stage PAD, ischemia worsens as the arteries become completely occluded, leading to chronic limb-threatening ischemia (CLTI). CLTI is characterized by resting muscle pain, ulceration, and gangrene, and a significant reduction in quality of life. In addition, PAD and CLTI patients are at increased risk of developing vascular occlusive disease and all-cause mortality, as atherosclerosis usually develops throughout the vasculature. Notably, the 1-year incidence of all major cardiovascular events is 30% higher in patients with PAD than in those with coronary or cerebral artery disease [8]. Without surgical revascularization, 25% of CLTI patients die within one year of initial diagnosis and 40% of CLTI patients undergo limb amputation within three years [9,10]. Venous bypass surgery and endovascular approaches such as angioplasty with or without stenting and endarterectomy are the main treatment for CLTI. The disease presentation and the patient’s general health and comorbidities determine the choice between open surgery and endovascular approaches.
Acute limb ischemia (ALI) is another severe manifestation of PAD, defined by sudden, severe hypoperfusion of the limb, usually due to thromboembolism. Symptoms may include pain, pallor, pulselessness, poikilothermia, paresthesias and paralysis, with loss of sensation and motor function in severe cases. Although ALI can occur in the absence of significant peripheral atherosclerosis due to distant plaque rupture, it is common in the setting of PAD.
Despite its prevalence, impact on quality of life, and devastating long-term clinical outcomes, PAD remains underdiagnosed and undertreated compared with other atherosclerotic diseases such as myocardial infarction and stroke [2,11].

2. Current Management of PAD and CLTI

The main risk factors for the development of PAD are age, smoking, and diabetes. Hyperlipidemia and hypertension are also risk factors for PAD, although the predictive value of these parameters does not appear to be as strong as for the primary risk factors. The presentation of PAD varies considerably and includes four categories: asymptomatic, claudication, critical limb ischemia, and ALI. PAD patients are classified according to the Fontaine or Rutherford classification systems.
  • Fontaine
  • Stage I—No symptoms
  • Stage II—Intermittent claudication subdivided into:
  • Stage IIa—Without pain on resting, but with claudication at a distance of greater than 650 feet (200 m)
  • Stage IIb—Without pain on resting, but with a claudication distance of less than 650 feet (200 m)
  • Stage III—Nocturnal and/or resting pain
  • Stage IV—Necrosis (death of tissue) and/or gangrene in the limb
  • Rutherford
  • Stage 0—Asymptomatic
  • Stage 1—Mild claudication
  • Stage 2—Moderate claudication
  • Stage 3—Severe claudication
  • Stage 4—Rest pain
  • Stage 5—Minor tissue loss with ischemic nonhealing ulcer or focal gangrene with diffuse pedal ischemia
  • Stage 6—Major tissue loss—Extending above transmetatarsal level, functional foot no longer salvageable
Asymptomatic PAD patients with evidence of atherosclerosis who do not have typical claudication symptoms (Fontaine I or Rutherford 0) are offered risk reduction strategies to decrease cardiovascular risk factors depending on symptom severity, lipid levels, and the presence of comorbidities such as diabetes, smoking and hypertension. Thus, current guidelines for the management of PAD are preventive strategies such as diet and lifestyle modification, including supervised exercise, smoking cessation and pharmacotherapy tailored to individual risk factors [1,8,12,13,14,15]. All patients with PAD should receive statin medication. Antihypertensive therapy should be administered to hypertensive patients to reduce the risk of myocardial infarction (MI), stroke, heart failure, and cardiovascular death. Antiplatelet therapy with aspirin or clopidogrel alone may be considered in asymptomatic patients, and should always be administered to symptomatic PAD patients. After assessment of bleeding risk, further anti-coagulant therapies (Rivaroxaban) may be considered for symptomatic PAD patients as they significantly reduce the risk of stroke, myocardial infarction, and ALI [1,12,13,16,17].
For patients with lifestyle-limiting claudication or CLTI (Fontaine IIb—IV; Rutherford 4–6), who are poor responders to medical and/or exercise therapy, surgical revascularization remains the only option when possible. Venous bypass surgery and endovascular approaches such as angioplasty, stenting and atherectomy are the main methods. The choice between open surgery and endovascular approaches depends on the presentation of the disease and the patient’s general health and comorbidities. Whenever possible, autogenous vein is the conduit of choice for open revascularization so that bypass surgery is limited to patients with “good” veins [7,18]. All patients with CLTI should be given antithrombotic and lipid-lowering therapies, as well as counseling on smoking cessation, diet, exercise, and preventive foot care. Additional antihypertensive, and glycemic control therapies should be given appropriately [1,12,13].
Without surgical revascularization, 25% of CLTI patients die within one year of initial diagnosis and 40% of CLTI patients undergo limb amputation within three years [9,10]. Up to 25% of CLTI patients are ineligible for revascularization and amputation is often the only option [19]. When possible, surgery may be suboptimal for symptom relief, and 20% of PAD patients have “failed revascularization”. Furthermore, PAD patients, especially those with CLTI, carry a high risk of post-op complications, including ALI, often leading to limb loss, disability, and death [13,20]. Even if the procedure is technically successful, residual microvascular disease remains and the outcomes after amputation stay poor [13,21].

3. Etiology of PAD

Atherosclerosis in lower limb arteries is the main cause of PAD [22], but emerging evidence suggests that medial calcification also contributes to the disease, especially in lower limb PAD. Microvascular disease is also emerging as a potential contributor to the progression of PAD and a clinically relevant sign of PAD severity.

3.1. Atherosclerosis

Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of fatty cholesterol streaks in arterial trees. Several pathophysiological processes are involved in this disease, including endothelial cell (EC) dysfunction, inflammation, lipid accumulation, and vascular smooth muscle cell (VSMC) proliferation and migration (reviewed in detail in [23]).
The disease is initiated by EC dysfunction. Located at the interface between the blood and the vessel wall, EC maintain a non-thrombogenic surface. In arteries, high shear stress and laminar blood flow maintain EC function and secretion of anti-thrombotic and vasodilator agents, mainly nitric oxide (NO) and prostacyclins [24]. Disturbed arterial flow patterns observed at bifurcations and curved sections of arteries create regions of low shear stress that induce EC dysfunction or “endothelial activation”. These weak points in the vasculature are the sites of primary occlusion by atherosclerotic plaques. Endothelial dysfunction or injury results in reduced production of NO and hydrogen sulfide (H2S), two gasotransmitters that maintain healthy vascular function. Impaired EC function promotes vasoconstriction, platelet aggregation and the accumulation of oxidized low-density lipoproteins (LDL) in the vessel wall. Monocytes attracted to the inflamed vessel wall differentiate into macrophages, which engulf large amounts of LDL particles and become foam cells to form the fatty streaks typical of early atherosclerotic lesions. Foam cells undergo apoptosis and form a lipid core within the vessel wall, exacerbating inflammation. The VSMC composing the media layer of vessels are highly plastic. Upon chronic inflammation, VSMC switch to a “synthetic” phenotype, characterized by a loss of contractile markers. Recent lineage-tracing studies revealed that VSMC dedifferentiate into intermediate multipotent cell type, often referred to as mesenchymal stem cells (MSC). These cells may give rise to adipocytes, myofibroblasts, macrophage-like cells and fibro/osteochondrogenic cells [25,26,27,28]. Of note, VSMC-derived macrophages perform nonprofessional phagocytosis and contribute to the population of foam cells in atherosclerotic plaques [29,30]. Altogether, proliferating immune cells and reprogrammed VSMC promote matrix remodeling and the development of a fibrous cap overlying the lipid core.
Overall, atherosclerosis is driven by dyslipidemia and vascular chronic inflammation [28,31]. Macrophages are the primary immune cells involved in atherosclerosis, but over the years evidence has accumulated of a coordinated inflammatory immune response involving T- and B-lymphocytes in the progression of atherosclerotic plaques [28]. It should also be noted that all the cell types found in atheromatous plaques can secrete pro-inflammatory cytokines, such as interleukin-1 (IL-1) and tumor necrosis factors alpha (TNFα) and chemokine monocyte chemoattractant protein-1 (MCP-1/CCL2). Activated T-helper 1 (Th1) lymphocytes produce interferon gamma (IFNγ), which promotes phagocytosis and formation of foam cells. B2 lymphocytes also secrete mediators that can aggravate atherogenesis. In contrast, other immune cells including M2 macrophages, B1 lymphocytes and Th2 lymphocytes can produce anti-inflammatory mediators to alleviate inflammation [28,31]. In addition, activated EC secrete lipid-derived pro-inflammatory molecules called eicosanoids, including prostaglandins, leukotrienes, and thromboxanes, which also play a major role in the pathophysiology of atherosclerosis [32,33].
Despite decades of research and although dyslipidemia and inflammation are known to be the major pathophysiological features leading to atherosclerosis, the exact pathways and mechanisms remain to be elucidated.

3.2. Vascular Medial Calcification

PAD is commonly described as an atherosclerotic disease. However, for lower limb artery disease, recent clinical data suggest that we underestimated the role of medial arterial calcification in PAD (recently reviewed in detail in [34,35]). Thus, the etiology of PAD, particularly in the arteries below the knee, may differ from that of the coronary and femoral arteries.
Two types of vascular calcification exist, intimal calcification (VIC) and medial calcification (VMC), also referred to as medial arterial calcification (MAC) [34,35]. VIC is a common feature of advanced atherosclerotic lesions and a risk factor for rupture. In contrast, VMC/MAC develops independently of atherosclerosis, but is a common feature of arterial disease associated with aging [36]. It is found in up to 40% of patients with advanced chronic kidney disease [37,38,39,40], and histological studies show that up to 70% of occluded arteries below the knee feature VMC and intimal thickening, but no atherosclerotis [41]. In their recent study, Jadidi et al. used machine learning to identify age, creatinine, body mass index, coronary artery disease and hypertension as the strongest predictors of calcification. They further confirmed that distal vessel segments (iliofemoral vs. aortic) calcify first. In this study of an American cohort, they estimated that up to 80% of people had VMC by the age of 40 [36].
VMC is characterized by the accumulation of calcium (Ca2+) phosphate and the formation of hydroxyapatite crystals, leading to hardening of the medial layer [38]. It is particularly prevalent in patients with chronic kidney disease, especially diabetic patients, due to impaired phosphate homeostasis [35,39,40]. Different stages/severities of arterial calcification have been described by histopathologists, ranging from punctate to nodular calcification, and finally bone formation [34].
VIC in atherosclerosis lesion is well characterized. It is due to ectopic vascular osteogenesis via phenotypic reprogramming of contractile medial VSMC into synthetic mesenchymal VSMC, which then differentiate into osteochondrogenic VSMC, leading to bone formation [35]. VMC in lower limb arteries has not been so well studied. The presence of osteogenesis vs. hydroxyapatite deposition and their respective contribution to VMC in PAD and CLTI patients remain unknown, and may differ depending on the vascular bed [38,39,40]. VMC increases the risk of complications during vascular interventions and worsens their outcomes [34,35,42]. Further work is required to define the process underlying medial calcification in the absence of atherosclerosis, evaluate its impact on PAD and CLTI, and eventually target it for treatment.

3.3. Microvascular Dysfunction

PAD is usually recognized as a macrovascular disease. However, several recent studies indicate that artery occlusion in PAD is often accompanied by microvascular disease. Microvascular dysfunction (MVD) refers to the impairment of capillary function and number. Usually, peripheral microvascular endothelial function is evaluated using laser speckle contrast imaging, which allows assessment of cutaneous microcirculation. The incidence of MVD is particularly high in diabetic patients. Thus, 20 to 30% of PAD patients, and up to 70% of CLTI patients have diabetes [10]. Of note, diabetic patients have a five-fold increased risk of developing CLTI, and diabetic CLTI patients have up to five-fold more incidence of adverse outcomes and amputations [9,10,43]. Given the strong association between diabetes complications and MVD, clinical studies also tend to define MVD as the presence of nephropathy, retinopathy, or neuropathy. Clinical studies revealed a strong association between MVD and risk of heart failure in diabetic patients, independent of traditional heart failure risk factors including coronary artery disease [44,45,46]. MVD is also a common phenomenon in PAD patients, which feature impaired cutaneous microcirculation throughout the progression of PAD, often leading to reduced capillary density in CLTI patients. In PAD patients, MVD can contribute to the progression of the disease and the development of complications such as ischemic pain, tissue hypoxia, and impaired wound healing [10]. A recent study also found a positive correlation between microvascular endothelial function and impaired cognitive performance in PAD patients [47]. MVD can also worsen the outcome of surgical procedures as it reduces the ability of the blood vessels to respond to the increased blood flow after revascularization, which impairs healing, leading to a higher risk of complications.
Additionally, recent studies suggest that MVD may be used to assess PAD severity. In a recent meta-analysis, the Chronic Kidney Disease Prognosis discovered that albuminuria, a marker of nephropathy, strongly correlates with the incidence of amputation [48]. This study advocates that even at mild-to-moderate stages, chronic kidney disease and MVD may be a major risk factor for PAD. In a similar study, a stronger association was found between retinopathy and the incidence of PAD/CLTI, than between coronary heart disease or stroke and PAD/CLTI [49].
Mechanistically, MVD is not due to the formation of atherosclerosis plaque and/or occlusion of vessels. MVD is due to EC apoptosis and progressive loss of capillaries, which plays a major role in the development and progression of diabetic complications (diabetic retinopathy, nephropathy, and neuropathy). Patients with familial hypercholesterolemia also feature impaired endothelial-dependent vasodilatation [50].
Overall, MVD contributes to PAD, but is seldom considered in diagnostic and therapeutic approaches. There is currently no specific therapy for MVD. However, the good news is that current PAD therapeutic strategies focused on optimizing risk factors (management of diabetes, and hypercholesterolemia), and lifestyle modifications (physical exercise, smoking cessation, and weight loss), improve vascular fitness, including microvascular function. For instance, several clinical studies demonstrated that exercise promotes microvascular function in disease states [51,52,53,54,55]. Although solid evidence is still lacking, statins may also provide benefits to endothelial function and against MVD [56,57]. Pre-clinical studies also showed that anti-diabetic therapies, metformin especially, may preserve/restore endothelium function [58,59,60,61]. Understanding the mechanisms underlying MVD in PAD patients and finding new treatments and therapeutics targeting MVD specifically may help reduce symptoms and improve quality of life.
Overall, PAD is due to a combination of macrovascular atherosclerosis and calcification, associated with a rarefying microvasculature, leading to impaired vascular function and a complex inter-individual response to treatment and revascularization interventions.

3.4. Intimal Hyperplasia: The Unmet Challenge of Post-Operative PAD Management

Bypass surgery and endovascular revascularization, which includes angioplasty, stenting and atherectomy, are recommended for patients with lifestyle-limiting claudication who do not respond to medical and/or exercise therapy. Unfortunately, the vascular trauma associated with surgical revascularization eventually leads to secondary occlusion of the injured vessel, a process called restenosis. For open surgical procedures such as bypass and endarterectomy, the rate of restenosis at 1-year ranges from 20 to 30% [62]. For endovascular approaches, the rate of re-occlusion after balloon angioplasty and stenting ranges from 30 to 60% depending on the location [63]. Restenosis has various causes, such as secondary growth of atherosclerotic lesions or inward remodeling. However, the most common cause is intimal hyperplasia (IH). IH is a well-known complication of all types of vascular surgery. The progressive growth of a neointimal layer causes both an outward and inward remodeling of the vessel wall, resulting in luminal narrowing and ultimately impaired perfusion of downstream organs.
IH begins as a physiological healing response to injury to the blood vessel wall [64,65]. Like atherosclerosis, IH is initiated by EC injury, which promotes vasoconstriction, platelet aggregation and recruitment/activation of resident and circulating inflammatory cells. Inflammation leads to the reprogramming of VSMC and fibroblasts into proliferating and migrating cells that form a neointimal layer between the intima and the internal elastic lamina. This new layer is mainly composed of VSMC-derived cells expressing various markers of mesenchymal (stemness) or osteochondrogenic phenotype and secreting abundant ECM [65,66,67].
All current strategies to limit IH, such as paclitaxel and sirolimus, target cell proliferation. Paclitaxel is a chemotherapeutic agent that stabilizes microtubules, thereby preventing mitosis [68]. Sirolimus inhibits the mammalian target of rapamycin (mTOR), a master regulator of cell growth and metabolism [66]. However, targeting cell proliferation to reduce IH also impairs re-endothelialization. Endothelial repair is critical to limit inflammation, remodeling and IH. Poor endothelial repair also prolongs the need for antithrombotic therapy.
The increasing number of PAD and CLTI patients in need of surgical vascular repair, combined with difficult long-term pharmacological and surgical management, calls for novel therapies to promote endothelial repair while inhibiting VSMC phenotypic switch, fibrosis, and VMC. The gaseous vasodilator molecule H2S has interesting properties in this respect.

4. Hydrogen Sulfide

H2S is a colorless, water-soluble, flammable, and highly toxic gas with a distinctive rotten-egg odor. In the last few years, H2S has been recognized as a novel gasotransmitter, not unlike NO and carbon monoxide [69].
Under physiological conditions (pH 7.4), H2S is mostly present as HS. It acts as a reductant and undergoes a complex oxidation reaction to thiosulfate, sulfenic acids, per-sulfides, polysulfides and sulfate [70]. These oxidative products trigger post-translational modification of proteins by S-sulfhydration, also known as persulfidation, a chemical reaction that forms a persulfide group (R-SSH) on reactive cysteine residues [71]. For persulfidation to occur, cysteine residues or H2S must first be oxidized, for example in the form of polysulfides H2Sn. H2S and other forms of sulfide contribute to the homeostasis of numerous systems, including the cardiovascular, neuronal, gastrointestinal, respiratory, renal, hepatic, and reproductive systems [69]. A few high-throughput studies on the conversion of protein cysteinyl thiols (-SH) to persulfides (-SSH) showed extensive persulfidation of cysteine residues in response to H2S in different experimental designs [70,72,73,74,75,76].

4.1. Endogenous H2S Production

H2S is involved in many physiological and pathological processes [69]. In this section, we will introduce the biosynthesis of endogenous H2S and the regulation of H2S in mammalian tissues.
Endogenous H2S production in mammals results from the oxidation of the sulfur-containing amino acids cysteine and homocysteine via the reverse ‘‘transsulfuration’’ pathway. H2S is produced by two pyridoxal 5′-phosphate (PLP)-dependent enzymes: cystathionine γ-lyase (CSE) and cystathionine β-synthase (CBS). CBS catalyzes the formation of cystathionine from homocysteine, which is subsequently converted to cysteine by CSE. Two other PLP-independent enzymes, 3-mercaptopyruvate sulphurtransferase (3-MST) and cysteine aminotransferase (CAT), generate sulfur, which is further processed to H2S. CAT converts L-cysteine to 3-mercaptopyruvate (3MP), which is converted to pyruvate and H2S by 3-MST in the presence of thioredoxin [77]. It should be noted that 3-MST mainly synthesizes H2S in the mitochondria (Scheme 1).
Other mitochondrial enzymes such as persulfide dioxygenase (ETHE1), sulfide-quinone oxidoreductase (SQR), rhodanese (TST) and sulfite oxidase (SUOX) catalyze H2S oxidation to the metabolic end products sulfate and thiosulfate [78]. Moreover, cysteinyl-tRNA synthetase (CARS and CARS2) can synthesize CysSSH and cyshydropolysulfides (CysSnH), which can be further reduced to H2S [79] (Scheme 1).
Although the enzymes and pathways responsible for endogenous H2S production are well understood, little is known about their relative contributions to circulating and tissular H2S and sulfane sulfur levels (e.g., polysulfides, persulfides, and thiosulfate). Accumulating evidence indicates that the enzymes involved in H2S production are often dysregulated in pathophysiologic conditions, leading to altered endogenous H2S production. All the evidence will not be listed here but we refer the reader to the extensive review by G. Cirino, C. Szabo and A. Papapetropoulos for a detailed account of the role, cellular distribution, and regulation of CSE, CBS, and 3-MST in mammalian tissues [69].
Briefly, the basal expression of CBS had been reported to be controlled by several transcription factors, including specificity protein (SP) 1 and 3, nuclear transcription factor-Y, and upstream transcription factor-1 (USF-1) [69]. CBS is mainly expressed in the central nervous system, the liver, and the pancreas, but is also found in most other systems, including the cardiovascular system. It has been mostly reported as a cytosolic enzyme, although CBS is also found in the mitochondria. CSE is a cytosolic and mitochondrial enzyme highly expressed in the liver and kidney. In the cardiovascular system, it is mainly expressed in EC [69]. In EC, CSE expression has been shown to be under the control of the activating transcription factor 4 (ATF4), which is selectively induced via the eukaryotic initiation factor 2 alpha (eIF2α) in response to various stresses such as ER-stress or amino acid restriction [80]. S. Bibli et al. recently demonstrated that CSE expression in EC is negatively regulated by shear stress, as opposed to eNOS in the mouse aorta [81]. This is in line with a previous study showing that only disturbed flow regions show discernable CSE protein expression after carotid artery ligation in the mouse [82]. Oxidative stress (H2O2) enhances cellular H2S production through the promotion of CSE activity [83]. 3-MST is expressed both in the mitochondria and cytosol, although most studies focus on the mitochondrial role of 3-MST [69]. 3-MST is found in most mammalian cells and tissues but varies between organs. 3-MST is most abundantly expressed in the liver, kidney, testes, and brain, and 3-MST expression is lowest in the spleen, thymus, lungs, and gut. Smoking, endurance exercise training, genetic defects and down syndrome have been reported to induce 3-MST expression in various models [69].
Additional sources of H2S and related sulfur species also contribute to sulfur biology. In the gastrointestinal tract, anaerobic bacterial strains such as E. coli, S. enterica, Clostridia and E. aerogenes all convert cysteine to H2S, pyruvate and ammonia by means of cysteine desulfurases. These cysteine desulfurases are also involved in the formation of a protein-bound cysteine persulfide intermediate, which leads to the conversion of L-cysteine to L-alanine and sulfane sulfur [84].
In addition to this enzymatic production, there are several non-enzymatic pathways. Commensal bacteria use sulfite reductases to reduce sulfate or other organic oxidized sulfur compounds, resulting in the formation of H2S [85,86]. Several studies have associated these sulfate-reducing bacteria (SRB) with inflammation, inflammatory bowel syndrome and colorectal disease [87]. SRB colonize the intestines of ~50% of humans [86,87,88].

4.2. Vascular Properties of H2S and Benefits in the Context of Peripheral Arterial Disease (PAD and CLTI)

H2S participates in the homeostasis of many organs and systems. In the cardiovascular system, H2S mostly has beneficial effects, and protects against vascular diseases through several processes, including the attenuation of oxidative stress and inflammation, improving EC function and NO production and vasodilation, as well as the preservation of mitochondrial function [69]. CSE gene expression and CSE protein activity, as well as free circulating H2S, are reduced in human suffering from vascular occlusive diseases [89,90]. It was also recently demonstrated that, in patients undergoing vascular surgery, higher circulating H2S levels were associated with long-term survival [91], suggesting low H2S production as a risk factor for cardiovascular diseases. In the following sections, we will focus on the role of H2S in the vascular system and H2S properties relevant to vascular conditions.

4.2.1. H2S Is a Potent Vasodilator

H2S is commonly known as a vasodilator [92]. One of the first reports came from Hosoki et al. in 1997, showing that H2S promoted NO-induced VSMC relaxation in rat thoracic aorta [93]. Then, numerous studies showed that H2S decreases blood pressure in spontaneously hypertensive rats (SHRs) [94,95,96] and salt-sensitive hypertension in Dahl rats [97].
Mechanistically, H2S triggers endothelium-independent vasorelaxation by persulfidation/activation the ATP-dependent potassium channel (KATP) complex, specifically the regulatory sulfonylurea receptor subunit 1 and the pore-forming subunit Kir6.1 in VSMC (reviewed in [92,98]). Activation of the KATP channel and K+ export results in VSMC hyperpolarization and inhibition of voltage-dependent Ca2+ channels (VDCC), reduced [Ca2+]i and relaxation [98]. H2S may also directly inhibit VDCC in VSMC [99,100]. In EC, H2S activates Ca2+ influx through TRPV4 [101], which in turn (i) increases eNOS expression/activity and NO production and VSMC vasodilation [102,103]; (ii) increases PLA2-mediated formation of arachidonic acid metabolites and VSMC relaxation; (iii) stimulates the large-conductance Ca2+-activated potassium channels (BKCa), leading to EC hyperpolarization and subsequent hyperpolarization of adjacent VSMC, closure of VDCC and relaxation [98]. In VSMC, H2S may also enhance Ca2+ spark-induced large conductance potassium channel activation, facilitating VSMC relaxation [98]. Elevation in intracellular Ca2+ level in EC also leads to the activation of calmodulin, which in turn stimulates CSE expression to produce more H2S [99]. In addition, H2S promotes NO-dependent relaxation via enhanced eNOS activity due to persulfidation of Cys443 [70] (Scheme 2).
Although H2S is usually described as a vasodilator gasotransmitter, recent studies demonstrated that H2S can also promote vasoconstriction. Thus, while concentrations of NaHS in the µM range induced vessels vasodilation [104], NaHS concentrations in the pico-nanoM range may stimulate contraction of VSMC [105] and rat coronary artery [106]. However, it should be noted that H2S alone does not trigger vasoconstriction, but only promotes constriction of precontracted vessels, enhancing the already existing tone. Enhanced vasoconstriction seems mediated by activation of Na+, K+, 2Cl cotransport and Ca2+ influx via VDCC [105]. H2S may also act via scavenging of NO [107]. This highlights the complexity of H2S contribution to the regulation of arterial blood pressure. Additionally, H2S may differentially act on the vascular tone depending on the arterial bed (carotid vs. mesenteric artery), the vessel type and size (conduit vs. resistant; capillary vs. larger vessels) (for full review, see [69,92,108]).
Overall, and although H2S is a potent vasodilator, very little is known about the role of CSE, CBS and 3MST-mediated H2S production in the regulation of blood pressure in physiologic and pathophysiologic conditions. Of note, the expression of H2S producing enzymes and substrate-dependent H2S production are decreased in humans with hypertension [109,110]. In addition, hypertensive patients with decreased endogenous H2S level have been shown to display microvascular endothelial dysfunction and impaired endothelium-dependent vasorelaxation [109]. Furthermore, the H2S precursor N-acetylcysteine decreased systolic and diastolic blood pressures in a clinical trial with 126 hypertensive patients [111]. It was also recently shown than a 6-week antihypertensive treatment with the sulfhydryl-donating angiotensin converting enzyme (ACE) inhibitor Captopril improved cutaneous microvascular endothelium-dependent vasodilation in middle-aged adults with hypertension [112]. This evidence indicates that H2S deficiency probably contributes to the development of hypertension and that H2S-based therapies may be of use for treatment of hypertension.

4.2.2. H2S Protects against Atherosclerosis

Atherosclerosis is a chronic progressive inflammatory disease. It is characterized by the accumulation of cholesterol-rich fatty deposits in the arterial tree. This disease involves numerous pathophysiological processes. These include EC dysfunction, vascular inflammation and lipoprotein accumulation, and VMSC proliferation and migration (see Section 3.1).
Impaired H2S production in Cse−/− mice promotes atherosclerosis [113,114]. In contrast, the H2S donors NaHS [114,115,116] and GYY4137 [117] reduce the extent of vascular lesions in ApoE−/− mice under high fat diet. S-aspirin (ACS14), a H2S-releasing form of aspirin, also protects ApoE−/− mice against atherosclerosis [118].
H2S has been shown to protect against atherosclerosis mostly via anti-inflammatory (for full review, see [119]) and antioxidant effects (Scheme 3). H2S possibly reduces inflammation mainly via inhibition of nuclear factor kappa B (NF-κB) [113,117,120,121]. NF-κB is a master regulator of pro-inflammatory genes, including cytokines and cell adhesion molecules. NaHS inhibits NF-κB activity via persulfidation/stabilization of Inhibitory kinase of NFκB (IκB) [122], which prevents NF-κB translocation to the nucleus [123]. In EC, inhibition of NF-κB leads to decreased expression of adhesion molecules vascular cell adhesion molecule 1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1), thereby limiting recruitment of leukocyte to the aortic wall [113,117,121,124]. NF-κB inhibition also decreases production of pro-inflammatory cytokines and chemokines, including TNF-α, IL-1β, IL-6, and CCL2 [121,125,126]. In macrophages, H2S-mediated peroxisome proliferator activated receptor gamma (PPARγ) inhibition also inhibits C-X3-C chemokine fractalkine (CX3CL1) signaling in the context of atherosclerosis in ApoE−/− mice [118]. H2S also inhibits TNF-α expression in EC in a model high glucose-induced vascular inflammation [127].
In addition, H2S was reported to inhibit leukocyte adherence to the endothelium via activation of ATP-sensitive K+ channels between EC and monocytes [128]. Moreover, S-sulfhydration of human antigen R (on Cys13) by CSE-derived H2S prevents its homodimerization and activity, which attenuates the expression of target proteins such as E-selectin and cathepsin S, which are linked to EC activation and atherosclerosis [74]. Exogenous H2S also promotes macrophage migration and shift toward the M2, pro-resolution phenotype [129,130,131]. However, further studies are required to identify whether H2S has a direct effect on macrophage state. Moreover, the fact that H2S stimulates eNOS activity and NO production in EC has been shown to contribute to its anti-inflammatory effect in the context of atherosclerosis [113,132]. The anti-inflammatory property of H2S may also involve inhibition of cyclooxygenase (COX2) expression and secretion of prostaglandin PGE2, which stimulates the secretion of pro-inflammatory cytokines and monocyte adhesion to EC [133]. H2S has also been proposed to protect EC from inflammation by inhibiting the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome in atherosclerotic conditions [134].
H2S also protects against atherosclerosis via antioxidant effects (Scheme 3). Excessive production of reactive oxygen species (ROS), such as superoxide anions O2, H2O2, and NO, leads to cellular and molecular damages. Oxidative stress is linked to the inflammatory process and contributes to the progression of PAD [135]. H2S is an antioxidant that can directly reduce ROS. Thus, NaHS protects myocytes and contractile activity by scavenging oxygen-free radical (O2, H2O2), thereby decreasing lipid peroxidation [136]. In the context of atherosclerosis, NaHS was shown to reduce O2 formation [115]. H2S also prevents LDL oxidation and formation of oxidized LDL particles (ox-LDL), resulting in reduced foam cell formation [137,138]. Interestingly, ox-LDL triggers the hypermethylation of the CSE promoter, thus decreasing CSE expression and H2S production in murine macrophages [121,139]. Mitochondrial respiration is a major source of ROS [140,141] and H2S binds the copper center of cytochrome c oxidase (complex IV), thereby inhibiting respiration and limiting ROS production [142].
H2S also upregulates antioxidant defenses, in particular the nuclear factor erythroid 2-related factor 2 (NRF2) pathway (reviewed in [143]) (Scheme 3). NRF2 is a major transcription factor that regulates antioxidant genes including heme oxygenase 1 (HO-1), thioredoxin-1 (TRX-1) and glutathione peroxidase (GPx). H2S promotes NRF2 activity via persulfidation of Keap-1 on Cys131, leading to dissociation of the cytosolic KEAP1-NRF2 complex, and nuclear translocation of NRF2 to induce the expression of its target genes. Thus, the H2S donor GYY4137 mitigates diabetes-accelerated atherosclerosis via improved Nrf2 activation in Ldlr−/− mice, which induces Ho-1 expression and reduces superoxide formation [144]. Exogenous H2S might protect arterial EC through antioxidant proprieties by activating the NRF2 pathway [145]. H2S also increases glutathione (GSH) production via modulation of the transulfuration pathway. GSH is an antioxidant that protects cells by reducing ROS. H2S interaction with GSH has been studied in detail in the central nervous system, where GSH plays a major role in maintaining the homeostasis between antioxidant and ROS production (reviewed in detail in [146]). In the vascular system, H2S persulfidates the GPx1, which promotes GSH synthesis and results in decreased lipid peroxidation in the aortic wall in the context of atherosclerosis [147]. H2S also stimulates TRX-1 expression, via silencing the expression of inhibitory protein Trx-interacting protein (TXNIP) [148,149,150,151]. Trx-1 is instrumental in the cardioprotective effects of H2S against ischemia-induced heart failure [150]. Trx-1 has atheroprotective effects via suppression of NLRP3 expression in macrophages after ox-LDL stimulation [152]. Trx-1 also promotes the M2 pro-resolutive macrophages state in ApoE−/− mice [153]. TRX-1 also suppresses Nox4 activity and ROS production in HUVEC exposed to ox-LDL [154].
H2S biosynthesis also occurs in adipocytes. Increased adiposity-enhanced oxidative stress and obesity-related low grade adipose tissue inflammation play a crucial role in the development of atherosclerosis [155]. The perivascular adipose tissue (PVAT), in particular, has been proposed to contribute to cardiovascular pathogenesis by promoting ROS generation and inflammation. The PVAT is the fourth outer layer of vessels surrounding the vasculature, which has emerged as an active modulator of vascular homeostasis and pathogenesis of cardiovascular diseases [156,157]. The adipose tissue is a very active endocrine tissue, secreting a variety of adipokines, including leptin and adiponectin, and pro- inflammatory cytokines such as TNFα IL-1β and IL-6. Leptin has been found to promote atherosclerosis, whereas adiponectin has been shown to have anti-inflammatory and anti-atherogenic effects [158,159,160]. H2S could reduce atherosclerosis by the inhibition of adipogenesis [161]. H2S deficiency may affect the process of adipocyte maturation and lipid accumulation. 3-MST knockdown also facilitated adipocytic differentiation and lipid uptake. The 3-MST/H2S system plays a tonic role in suppressing lipid accumulation and limiting the differentiation of adipocytes [162].
Overall, H2S has been found to be cytoprotective in oxidative stress in a wide range of physiologic and pathologic conditions.

4.2.3. H2S Protects against Vascular Medial Calcification

First and foremost, H2S can protect from arterial calcification indirectly. As stated in Section 3.2, chronic kidney disease and diabetes mellitus are the leading causes of VMC. H2S has been shown to provide benefits against both pathologies. These will be not discussed in this review due to space constraints. Readers interested in a more in-depth analysis of the benefits of H2S against diabetes are referred to other reviews [163,164]. H2S has been shown to decrease blood glucose, atherosclerosis, and diabetic cardiomyopathy in the context of diabetes in pre-clinical models [165,166]. H2S also provides renal protection against various injury, including models of diabetic nephropathy [167]. Below we detail the studies directly measuring the impact of H2S on the process of VMC in various experimental in vitro and in vivo models (Scheme 4).
VMC is an accumulation of Ca2+ and inorganic phosphate (Pi) in arteries with mineral deposits in the intimal or medial layer of the vessel wall [168,169]. VMC formation is a complex, controlled molecular process involving the differentiation of macrophages and VSMC into osteoclast-like cells, like that which occurs in bone formation [170,171] (see Section 3.2). In recent years, H2S supplementation has been shown to lessen VMC. In this section, we discussed these studies and their molecular insight into the potential mechanisms underlying the benefits of H2S on VMC (Scheme 4).
Using a model of VMC by administration of vitamin D3 plus nicotine (VDN), it was shown in rats that Cse expression is downregulated in the context of VMC, and that treatment with H2S donors NaHS [172] or AP39 [173] lessens VMC in that model. Similarly, exogenous NaHS treatment also restored Cse activity and expression, and inhibited aortic osteogenic transformation in a rat model of diabetic nephropathy [163]. NaHS also limits Ca2+ deposition in VSMC in in vitro models of calcification in cell culture [138,174,175].
Mechanistically, H2S has been proposed to limit VMC via reduced ER stress-induced VSMC phenotypic reprogramming [173]. H2S attenuates VSMC calcification induced by high levels of glucose and phosphate through upregulating elastin level via the inhibition of the signal transducer and activator of transcription 3 (Stat3), leading to reduced Cathepsin S expression [175]. NaHS also significantly reduced Stat3 activation, cathepsin S activity in a rat model of diabetic nephropathy [163]. In another model of VSMC calcification induced by circulating calciprotein particles, H2S was shown to mitigate VMC via activation of the antioxidant factor NRF2 [174]. Overall, H2S likely acts on several pathways improving VSMC identity to avert osteogenic transformation (Scheme 4). Of note, low plasma levels of H2S and decreased CSE enzyme activity were found in patients with chronic kidney disease receiving hemodialysis [138,176], suggesting that low H2S may contribute to VMC in patients.
From a translational point of view, it should be mentioned that the FDA-approved H2S donor Sodium thiosulfate (STS) reduces periarticular calcification in a mouse model of osteoarthritis via its effects on chondrocyte mineralization [177]. STS is already used in the clinic to treat cyanide poisoning and to increase the solubility of Ca2+ for the treatment of acute calciphylaxis, a rare vascular complication of patients with end-stage renal disease [178]. The phase III CALISTA trial of STS for acute calciphylaxis is ongoing (NCT03150420) and STS is also tested in a few clinical trials for the treatment of ectopic calcification (NCT03639779; NCT04251832; NCT02538939). Although STS has not been shown to reduce VC, it stands to reason that STS should be explored for the treatment of VMC in the context of PAD.

4.2.4. H2S Supports Endothelial Cell Function

With one simple monolayer, the endothelium regulates vascular tone, cell adhesion and vessel wall inflammation, and VSMC phenotype. Atherosclerosis and PAD preferentially develop at site of disturbed arterial flow leading to “endothelium activation”. As described in the previous sections, impaired EC-derived H2S contributes to inflammation and oxidative stress, leading to atherosclerosis. The ability of EC to proliferate and migrate to restore the endothelial barrier of the vessel is a key feature in wound healing, vascular repair, and the resolution of inflammation. In this section, we describe the effects of H2S in EC proliferation and migration, which constitute an interesting avenue of research to promote therapeutical angiogenesis for PAD patients (Scheme 5). The benefits of H2S on EC may also limit MVD, which contributes to the severity of PAD (see Section 3.3).
Preclinical studies have shown that H2S and polysulfites stimulate EC angiogenesis and arteriogenesis. Thus, H2S donors stimulate the growth, motility, and organization of EC into a vascular structure in vitro [179]. Conversely, inhibition of H2S biosynthesis, either by pharmacological inhibitors or by silencing CSE, CBS or 3MST, reduces EC growth and migration in vitro [180,181]. Cse−/− mice also show reduced vascular endothelial growth factor (VEGF)-induced sprouting angiogenesis in the mouse aortic ring assay ex vivo [182]. In vivo studies on chicken chorioallantoic membranes treated with the CSE inhibitor propargylglycine (PAG) also indicate that CSE is important for vascular branching [182]. In vivo, there is no adequate PAD model. Most studies are conducted using the hindlimb ischemia (HLI) model, which can be applied to rodent and pigs alike. In this model, transection or occlusion of the femoral or iliac artery leads to ALI. Recovery from ALI is then followed for 2 to 4 weeks via angiographic scores, return of hind limb blood flow, and capillary density in the gastrocnemius muscle. As such, the model allows for assessment of arteriogenesis and angiogenesis-mediated neovascularization. Using this model, it was shown that whole-body Cse−/− mice with impaired H2S production displayed impaired neovascularization [114,183]. Conversely, we recently showed that Cse overexpression in transgenic mice is sufficient to promote neovascularization following HLI [184]. Various H2S donors such as NaHS, GYY4137, ZYZ-803, a hybrid NO and H2S donor were also shown to improve capillary density, angiographic scores, and hind limb blood flow in rodent models [179,185,186]. Fu et al. also reported that H2S-saturated water accelerates perfusion recovery through improved arteriogenesis in the abductor muscle and increased capillary density in the gastrocnemius muscle in the mouse [187]. Diallyl trisulfide, S-allylcysteine and S-propyl-L-cysteine, organosulfur compounds found in garlic, were also shown to improve blood flow recovery after HLI in mice in various context [188,189,190,191,192,193]. Rushing et al. also showed that SG1002, a H2S-releasing pro-drug, increases leg revascularization and collateral vessel number after occlusion of the external iliac artery in the minipig [194]. We also recently showed that the H2S donor STS promotes EC proliferation and migration in vitro, and VEGF-induced angiogenesis in vivo. STS also accelerates neovascularization in the HLI model in WT and Ldlr−/− male mice [195].
Several mechanisms have been proposed to explain H2S-induced angiogenesis (Scheme 5). Most studies report that H2S promotes VEGF-driven sprouting angiogenesis. Thus, overexpression of CSE, CBS and 3-MST leads to an increase in VEGF expression and decrease in anti-angiogenic factor endostatin [196]. Similarly, NaHS increases VEGF expression while reducing the levels of anti-angiogenic factors [197]. In EC, H2S induces the VEGF receptor VEGFR2 persulfidation, which facilitates dimerization, autophosphorylation and activation [198]. Interestingly, short-term exposure of human EC to VEGF increases H2S production [182], suggesting a positive feedback loop of VEGF signaling through H2S. Matrigel plug angiogenesis assay also confirmed the importance of CSE and H2S in VEGF-induced angiogenesis [195,199,200]. CSE overexpression is also sufficient to stimulate VEGF-dependent EC migration in vitro, and capillary formation using an aortic ring assay ex vivo [184]. CSE and H2S are also required for VEGF-dependent EC migration and angiogenesis in response to amino acid restriction [80]. Exogenous H2S donors have also been shown to stimulate the growth pathways Akt, p38 and ERK1/2, which all promote EC proliferation and migration [182,200,201]. EC migration is also activated by exogenous H2S through KATP channels/MAPK pathways in vitro [182]. CSE overexpression has also been reported to increase cGMP level [199], which fuels capillary tube formation [187]. In addition, H2S promotes angiogenesis via interactions with NO, which is essential for EC survival and growth during VEGF- or bFGF-induced angiogenesis [202]). Finally, H2S is proposed to promote angiogenesis by inhibiting mitochondrial electron transport and oxidative phosphorylation, increasing glucose uptake and glycolytic ATP production required to rapidly power EC migration [80]. Indeed, under hypoxia when mitochondrial respiration is not possible, glycolysis fuels EC migration and proliferation during angiogenesis [203,204]. H2S promotes the metabolic switch in EC to favor glycolysis, which drives VEGF-induced EC migration [80,205] (Scheme 5).

4.2.5. H2S Inhibits Intimal Hyperplasia: Post-Operative Management of CLTI Patients

The revascularization procedure in CLTI patients is plagued by restenosis of the operated area, a progressive reduction of the vessel lumen at the site of angioplasty, or at the anastomosis of a bypass graft. Restenosis is mainly related to a complex phenomenon called IH (see Section 3.4). IH is characterized by a thickened wall due to VSMC proliferation and deposition of a proteoglycan-rich ECM between the endothelium and the internal elastic lamina.
Mice lacking Cse show a significant increase in IH formation as compared to WT mice in a model of carotid artery ligation [205,206]. On the contrary, Cse overexpression decreases IH formation in a murine model of vein graft by carotid-interposition cuff technique [207]. We and others demonstrated that systemic treatment using diverse H2S donors inhibit IH in vivo in various models in rats [208], rabbits [209] and mice [205,206,210]. We also showed that various H2S donors inhibit IH ex vivo in a model of vein graft IH [205,210,211]. Recently, it was shown that a locally applicable gel containing the hydrogen sulfide releasing prodrug (GYY4137) mitigates graft failure and improves arterial remodeling in a model of vein graft surgery in the mouse [212]. We also recently showed that a H2S-releasing biodegradable hydrogel inhibited VSMC proliferation while facilitating EC proliferation and migration, which inhibited IH in an ex vivo model of human vein graft disease [211].
H2S probably reduces IH mainly via inhibition of VSMC proliferation (Scheme 6). Indeed, several studies demonstrated that H2S supplementation using various donors, or CSE overexpression, decreases VSMC proliferation [205,209,210,211,213]. H2S also specifically inhibits VSMC migration. Thus, Several H2S donors have also been shown to reduce VSMC migration in vitro [205,210,211]. VSMC isolated from Cse−/− mice also migrate faster than wild type VSMC, and blocking CSE activity using PAG increases VSMC migration [206,214].
The mechanisms whereby H2S affects VSMC proliferation and migration are not fully understood (Scheme 6). In mouse VSMC, H2S has been shown to modulate the MAPK pathway, especially ERK1,2 [208], and Ca2+-sensing receptors [215,216]. H2S may also limit MMP2 expression and ECM degradation, preventing VSMC migration from the media to the intima [206,214]. In human VSMC, we reported that the H2S donor Zofenopril decreases the activity of the MAPK and mTOR pathways, which correlates with reduced VSMC proliferation and migration [210]. We also showed that the H2S donors NaHS and Sodium thiosulfate (STS; Na2S2O3) inhibit microtubule polymerization, which results in cell cycle arrest and inhibition of proliferation and migration in primary human VSMC [205]. Interestingly, an ongoing clinical study aims to evaluate the efficacy and safety of STS compared to placebo on myocardial infarct size in ST-segment elevation myocardial infarction (STEMI) patients treated with percutaneous coronary intervention (NCT02899364). The anti-inflammatory properties of H2S may also contribute to reduced IH [217,218] and it was recently shown that NaHS prevents IH through activation of the Nrf2/HIF1α pathway [219].

4.3. Further Directions and Limitations

Although H2S research is still in its early stages, there is considerable evidence to suggest that this gas plays a protective role in the development of cardiovascular disease. As mentioned throughout the review, H2S acts in concert with NO, and the vascular effects of NO and H2S are mutually supportive and intertwined (for a complete review, see [69]). Due to poor tolerability and uncontrolled hypotensive effects, all therapeutic strategies based on NO have failed. Whether H2S-based solutions can succeed where NO has failed remains to be seen. There is currently no clinically approved molecule that exploits the therapeutic potential of H2S. Most compounds available for research have poor translational potential due to their pharmacokinetic properties. Developing stable H2S donors that allow slow and sustained H2S release over months/years will be the first challenge. Given the instability and short half-life of H2S, such molecules are difficult to design. Another challenge for systemic or local H2S release is the delivery system, as H2S donors may require a carrier system. Gels, nanoparticles, multilayer coatings, and biodegradable scaffolds were invented for sustained release. Applying this knowledge to H2S donors will be interesting. Another strategy to harness the benefits of H2S is to conjugate the H2S-releasing moiety with well-established parent compounds. For example, the sulphhydrylated ACEi zofenopril has been shown to improve clinical outcomes in patients with various cardiovascular diseases such as acute myocardial infarction and congestive heart failure [220,221,222]. S-aspirin (ACS14), an H2S-releasing form of aspirin, and otenaproxesul, an H2S-releasing non-steroidal anti-inflammatory drug developed by Antibe Therapeutics Inc, may also prove beneficial for vascular patients. Further work is needed to evaluate the therapeutic potential of these molecules against atherosclerosis, but also against VMC and MVD in PAD. H2S-eluting balloons and stents would be interesting tools to limit VSMC proliferation while promoting EC recovery to limit IH in PAD/CLTI patients requiring surgery.
Strategies to increase endogenous H2S production using small molecules or diet are also explored. However, further animal studies are needed to understand and leverage endogenous H2S production and to test the potential and safety of new H2S-based therapies.

5. Conclusions

PAD is a chronic, recurrent disease with a major impact on quality of life and devastating long-term clinical outcomes. PAD remains underdiagnosed and undertreated compared to other atherosclerotic diseases such as myocardial infarction and stroke. Emerging evidence suggests that PAD has different pathological features in peripheral vessels compared to the well-characterized coronary arteries, in particular media calcification and microvascular dysfunction. In addition, the incidence of restenosis following surgical revascularization remains high. The increasing number of PAD and CLTI patients, combined with difficult long-term pharmacological and surgical management, warrants further research to better understand the molecular mechanisms of PAD.
Although still in its early stages, research into H2S suggests its potential to protect against cardiovascular disease. The success of H2S-based solutions remains uncertain and there are currently no clinically approved molecules exploiting its therapeutic potential. The development of stable H2S donor molecules for sustained release is challenging due to the instability of the gas. Delivery systems such as gels, nanoparticles and biodegradable scaffolds designed for sustained release could be applied to H2S donors. H2S-eluting balloons and stents may be useful in limiting VSMC proliferation and promoting EC recovery in patients with PAD. Another strategy is to combine H2S donors with established drugs. Strategies to increase endogenous H2S production using small molecules or diet are also investigated. Further studies are needed to explore the therapeutic potential and safety of these molecules against atherosclerosis, vascular calcification, and microvascular dysfunction. The advancement of this knowledge will contribute to the development of successful H2S-based therapies in the future.

Author Contributions

Conceptualization, F.A. and S.D.; writing—original draft preparation, F.A., D.M. and C.B.; writing—review and editing, F.A., C.B. and S.D.; visualization, F.A. and C.B.; supervision, F.A. and S.D.; funding acquisition, F.A. and S.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Aboyans, V.; Ricco, J.B.; Bartelink, M.E.L.; Bjorck, M.; Brodmann, M.; Cohnert, T.; Collet, J.P.; Czerny, M.; De Carlo, M.; Debus, S.; et al. 2017 ESC Guidelines on the Diagnosis and Treatment of Peripheral Arterial Diseases, in collaboration with the European Society for Vascular Surgery (ESVS): Document covering atherosclerotic disease of extracranial carotid and vertebral, mesenteric, renal, upper and lower extremity arteriesEndorsed by: The European Stroke Organization (ESO)The Task Force for the Diagnosis and Treatment of Peripheral Arterial Diseases of the European Society of Cardiology (ESC) and of the European Society for Vascular Surgery (ESVS). Eur. Heart J. 2018, 39, 763–816. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Aday, A.W.; Matsushita, K. Epidemiology of Peripheral Artery Disease and Polyvascular Disease. Circ. Res. 2021, 128, 1818–1832. [Google Scholar] [CrossRef] [PubMed]
  3. Hamburg, N.M.; Creager, M.A. Pathophysiology of Intermittent Claudication in Peripheral Artery Disease. Circ. J. 2017, 81, 281–289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Shan, L.L.; Yang, L.S.; Tew, M.; Westcott, M.J.; Spelman, T.D.; Choong, P.F.; Davies, A.H. Quality of Life in Chronic Limb Threatening Ischaemia: Systematic Review and Meta-Analysis. Eur. J. Vasc. Endovasc. Surg. 2022, 64, 666–683. [Google Scholar] [CrossRef]
  5. Porras, C.P.; Bots, M.L.; Teraa, M.; van Doorn, S.; Vernooij, R.W.M. Differences in Symptom Presentation in Women and Men with Confirmed Lower Limb Peripheral Artery Disease: A Systematic Review and Meta-Analysis. Eur. J. Vasc. Endovasc. Surg. 2022, 63, 602–612. [Google Scholar] [CrossRef]
  6. Hiatt, W.R.; Armstrong, E.J.; Larson, C.J.; Brass, E.P. Pathogenesis of the limb manifestations and exercise limitations in peripheral artery disease. Circ. Res. 2015, 116, 1527–1539. [Google Scholar] [CrossRef] [Green Version]
  7. Thukkani, A.K.; Kinlay, S. Endovascular intervention for peripheral artery disease. Circ. Res. 2015, 116, 1599–1613. [Google Scholar] [CrossRef] [Green Version]
  8. Fowkes, F.G.; Aboyans, V.; Fowkes, F.J.; McDermott, M.M.; Sampson, U.K.; Criqui, M.H. Peripheral artery disease: Epidemiology and global perspectives. Nat. Rev. Cardiol. 2017, 14, 156–170. [Google Scholar] [CrossRef]
  9. Ying, A.F.; Tang, T.Y.; Jin, A.; Chong, T.T.; Hausenloy, D.J.; Koh, W.P. Diabetes and other vascular risk factors in association with the risk of lower extremity amputation in chronic limb-threatening ischemia: A prospective cohort study. Cardiovasc. Diabetol. 2022, 21, 7. [Google Scholar] [CrossRef]
  10. Barnes, J.A.; Eid, M.A.; Creager, M.A.; Goodney, P.P. Epidemiology and Risk of Amputation in Patients With Diabetes Mellitus and Peripheral Artery Disease. Atheroscler. Thromb. Vasc. Biol. 2020, 40, 1808–1817. [Google Scholar] [CrossRef]
  11. Criqui, M.H.; Matsushita, K.; Aboyans, V.; Hess, C.N.; Hicks, C.W.; Kwan, T.W.; McDermott, M.M.; Misra, S.; Ujueta, F.; on behalf of the American Heart Association Council on Epidemiology and Prevention; et al. Lower Extremity Peripheral Artery Disease: Contemporary Epidemiology, Management Gaps, and Future Directions: A Scientific Statement From the American Heart Association. Circulation 2021, 144, e171–e191. [Google Scholar] [CrossRef] [PubMed]
  12. Abola, M.T.B.; Golledge, J.; Miyata, T.; Rha, S.W.; Yan, B.P.; Dy, T.C.; Ganzon, M.S.V.; Handa, P.K.; Harris, S.; Zhisheng, J.; et al. Asia-Pacific Consensus Statement on the Management of Peripheral Artery Disease: A Report from the Asian Pacific Society of Atherosclerosis and Vascular Disease Asia-Pacific Peripheral Artery Disease Consensus Statement Project Committee. J. Atheroscler. Thromb. 2020, 27, 809–907. [Google Scholar] [CrossRef] [PubMed]
  13. Conte, M.S.; Bradbury, A.W.; Kolh, P.; White, J.V.; Dick, F.; Fitridge, R.; Mills, J.L.; Ricco, J.B.; Suresh, K.R.; Murad, M.H.; et al. Global vascular guidelines on the management of chronic limb-threatening ischemia. Eur. J. Vasc. Endovasc. Surg. 2019, 58, S1–S109.e33. [Google Scholar] [CrossRef] [Green Version]
  14. Gerhard-Herman, M.D.; Gornik, H.L.; Barrett, C.; Barshes, N.R.; Corriere, M.A.; Drachman, D.E.; Fleisher, L.A.; Fowkes, F.G.R.; Hamburg, N.M.; Kinlay, S.; et al. 2016 AHA/ACC Guideline on the Management of Patients With Lower Extremity Peripheral Artery Disease: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J. Am. Coll. Cardiol. 2017, 69, e71–e126. [Google Scholar] [CrossRef] [PubMed]
  15. Fowkes, F.G.; Rudan, D.; Rudan, I.; Aboyans, V.; Denenberg, J.O.; McDermott, M.M.; Norman, P.E.; Sampson, U.K.; Williams, L.J.; Mensah, G.A.; et al. Comparison of global estimates of prevalence and risk factors for peripheral artery disease in 2000 and 2010: A systematic review and analysis. Lancet 2013, 382, 1329–1340. [Google Scholar] [CrossRef]
  16. Morrison, J.T.; Govsyeyev, N.; Hess, C.N.; Bonaca, M.P. Vorapaxar for Prevention of Major Adverse Cardiovascular and Limb Events in Peripheral Artery Disease. J. Cardiovasc. Pharmacol. Ther. 2022, 27, 10742484211056115. [Google Scholar] [CrossRef]
  17. Golledge, J. Update on the pathophysiology and medical treatment of peripheral artery disease. Nat. Rev. Cardiol. 2022, 19, 456–474. [Google Scholar] [CrossRef]
  18. Almasri, J.; Adusumalli, J.; Asi, N.; Lakis, S.; Alsawas, M.; Prokop, L.J.; Bradbury, A.; Kolh, P.; Conte, M.S.; Murad, M.H. A systematic review and meta-analysis of revascularization outcomes of infrainguinal chronic limb-threatening ischemia. J. Vasc. Surg. 2019, 69, 126S–136S. [Google Scholar] [CrossRef] [Green Version]
  19. Iyer, S.R.; Annex, B.H. Therapeutic Angiogenesis for Peripheral Artery Disease: Lessons Learned in Translational Science. JACC Basic Transl. Sci. 2017, 2, 503–512. [Google Scholar] [CrossRef]
  20. Bager, L.G.V.; Petersen, J.K.; Havers-Borgersen, E.; Resch, T.; Smolderen, K.G.; Mena-Hurtado, C.; Eiberg, J.; Kober, L.; Fosbol, E.L. The Use of Evidence-Based Medical Therapy in Patients with Critical Limb-Threatening Ischemia. Eur. J. Prev. Cardiol. 2023, zwad022. [Google Scholar] [CrossRef]
  21. Govsyeyev, N.; Nehler, M.R.; Low Wang, C.C.; Kavanagh, S.; Hiatt, W.R.; Long, C.; Jones, W.S.; Fowkes, F.G.R.; Berger, J.S.; Baumgartner, I.; et al. Etiology and outcomes of amputation in patients with peripheral artery disease in the EUCLID trial. J. Vasc. Surg. 2022, 75, 660–670.e3. [Google Scholar] [CrossRef] [PubMed]
  22. Shu, J.; Santulli, G. Update on peripheral artery disease: Epidemiology and evidence-based facts. Atherosclerosis 2018, 275, 379–381. [Google Scholar] [CrossRef] [PubMed]
  23. Libby, P.; Buring, J.E.; Badimon, L.; Hansson, G.K.; Deanfield, J.; Bittencourt, M.S.; Tokgozoglu, L.; Lewis, E.F. Atherosclerosis. Nat. Rev. Dis. Primers 2019, 5, 56. [Google Scholar] [CrossRef] [PubMed]
  24. Stone, J.R. Chapter 4—Diseases of Small and Medium-sized Blood Vessels. In Cardiovascular Pathology, 4th ed.; Academic Press: Cambridge, MA, USA, 2016; pp. 125–168. [Google Scholar] [CrossRef]
  25. Hartmann, F.; Gorski, D.J.; Newman, A.A.C.; Homann, S.; Petz, A.; Owsiany, K.M.; Serbulea, V.; Zhou, Y.Q.; Deaton, R.A.; Bendeck, M.; et al. SMC-Derived Hyaluronan Modulates Vascular SMC Phenotype in Murine Atherosclerosis. Circ. Res. 2021, 129, 992–1005. [Google Scholar] [CrossRef] [PubMed]
  26. Pan, H.; Xue, C.; Auerbach, B.J.; Fan, J.; Bashore, A.C.; Cui, J.; Yang, D.Y.; Trignano, S.B.; Liu, W.; Shi, J.; et al. Single-Cell Genomics Reveals a Novel Cell State During Smooth Muscle Cell Phenotypic Switching and Potential Therapeutic Targets for Atherosclerosis in Mouse and Human. Circulation 2020, 142, 2060–2075. [Google Scholar] [CrossRef]
  27. Brandt, K.J.; Burger, F.; Baptista, D.; Roth, A.; Fernandes da Silva, R.; Montecucco, F.; Mach, F.; Miteva, K. Single-Cell Analysis Uncovers Osteoblast Factor Growth Differentiation Factor 10 as Mediator of Vascular Smooth Muscle Cell Phenotypic Modulation Associated with Plaque Rupture in Human Carotid Artery Disease. Int. J. Mol. Sci. 2022, 23, 1796. [Google Scholar] [CrossRef] [PubMed]
  28. Kong, P.; Cui, Z.Y.; Huang, X.F.; Zhang, D.D.; Guo, R.J.; Han, M. Inflammation and atherosclerosis: Signaling pathways and therapeutic intervention. Signal. Transduct. Target Ther. 2022, 7, 131. [Google Scholar] [CrossRef]
  29. Vengrenyuk, Y.; Nishi, H.; Long, X.; Ouimet, M.; Savji, N.; Martinez, F.O.; Cassella, C.P.; Moore, K.J.; Ramsey, S.A.; Miano, J.M.; et al. Cholesterol loading reprograms the microRNA-143/145-myocardin axis to convert aortic smooth muscle cells to a dysfunctional macrophage-like phenotype. Arterioscler. Thromb. Vasc. Biol. 2015, 35, 535–546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Wang, Y.; Dubland, J.A.; Allahverdian, S.; Asonye, E.; Sahin, B.; Jaw, J.E.; Sin, D.D.; Seidman, M.A.; Leeper, N.J.; Francis, G.A. Smooth Muscle Cells Contribute the Majority of Foam Cells in ApoE (Apolipoprotein E)-Deficient Mouse Atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2019, 39, 876–887. [Google Scholar] [CrossRef]
  31. Libby, P. The changing landscape of atherosclerosis. Nature 2021, 592, 524–533. [Google Scholar] [CrossRef]
  32. Gleim, S.; Stitham, J.; Tang, W.H.; Martin, K.A.; Hwa, J. An eicosanoid-centric view of atherothrombotic risk factors. Cell. Mol. Life Sci. 2012, 69, 3361–3380. [Google Scholar] [CrossRef] [Green Version]
  33. Yamaguchi, A.; Botta, E.; Holinstat, M. Eicosanoids in inflammation in the blood and the vessel. Front. Pharmacol. 2022, 13, 997403. [Google Scholar] [CrossRef] [PubMed]
  34. Lanzer, P.; Hannan, F.M.; Lanzer, J.D.; Janzen, J.; Raggi, P.; Furniss, D.; Schuchardt, M.; Thakker, R.; Fok, P.W.; Saez-Rodriguez, J.; et al. Medial Arterial Calcification: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2021, 78, 1145–1165. [Google Scholar] [CrossRef]
  35. Kim, T.I.; Guzman, R.J. Medial artery calcification in peripheral artery disease. Front. Cardiovasc. Med. 2023, 10, 1093355. [Google Scholar] [CrossRef] [PubMed]
  36. Jadidi, M.; Poulson, W.; Aylward, P.; MacTaggart, J.; Sanderfer, C.; Marmie, B.; Pipinos, M.; Kamenskiy, A. Calcification prevalence in different vascular zones and its association with demographics, risk factors, and morphometry. Am. J. Physiol. Heart Circ. Physiol. 2021, 320, H2313–H2323. [Google Scholar] [CrossRef]
  37. Sorensen, I.M.H.; Saurbrey, S.A.K.; Hjortkjaer, H.O.; Brainin, P.; Carlson, N.; Ballegaard, E.L.F.; Kamper, A.L.; Christoffersen, C.; Feldt-Rasmussen, B.; Kofoed, K.F.; et al. Regional distribution and severity of arterial calcification in patients with chronic kidney disease stages 1-5: A cross-sectional study of the Copenhagen chronic kidney disease cohort. BMC Nephrol. 2020, 21, 534. [Google Scholar] [CrossRef] [PubMed]
  38. Chen, Y.; Zhao, X.; Wu, H. Arterial Stiffness: A Focus on Vascular Calcification and Its Link to Bone Mineralization. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 1078–1093. [Google Scholar] [CrossRef]
  39. Zununi Vahed, S.; Mostafavi, S.; Hosseiniyan Khatibi, S.M.; Shoja, M.M.; Ardalan, M. Vascular Calcification: An Important Understanding in Nephrology. Vasc. Health Risk Manag. 2020, 16, 167–180. [Google Scholar] [CrossRef]
  40. Singh, A.; Tandon, S.; Tandon, C. An update on vascular calcification and potential therapeutics. Mol. Biol. Rep. 2021, 48, 887–896. [Google Scholar] [CrossRef]
  41. Narula, N.; Dannenberg, A.J.; Olin, J.W.; Bhatt, D.L.; Johnson, K.W.; Nadkarni, G.; Min, J.; Torii, S.; Poojary, P.; Anand, S.S.; et al. Pathology of Peripheral Artery Disease in Patients With Critical Limb Ischemia. J. Am. Coll. Cardiol. 2018, 72, 2152–2163. [Google Scholar] [CrossRef]
  42. Skolnik, J.; Weiss, R.; Meyr, A.J.; Dhanisetty, R.; Choi, E.T.; Cunningham-Hill, M.; Rubin, D.; Oresanya, L. Evaluating the Impact of Medial Arterial Calcification on Outcomes of Infrageniculate Endovascular Interventions for Treatment of Diabetic Foot Ulcers. Vasc. Endovasc. Surg. 2021, 55, 382–388. [Google Scholar] [CrossRef] [PubMed]
  43. Belur, A.D.; Shah, A.J.; Virani, S.S.; Vorla, M.; Kalra, D.K. Role of Lipid-Lowering Therapy in Peripheral Artery Disease. J. Clin. Med. 2022, 11, 4872. [Google Scholar] [CrossRef] [PubMed]
  44. Crea, F.; Camici, P.G.; Bairey Merz, C.N. Coronary microvascular dysfunction: An update. Eur. Heart J. 2014, 35, 1101–1111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Gallinoro, E.; Paolisso, P.; Candreva, A.; Bermpeis, K.; Fabbricatore, D.; Esposito, G.; Bertolone, D.; Fernandez Peregrina, E.; Munhoz, D.; Mileva, N.; et al. Microvascular Dysfunction in Patients With Type II Diabetes Mellitus: Invasive Assessment of Absolute Coronary Blood Flow and Microvascular Resistance Reserve. Front. Cardiovasc. Med. 2021, 8, 765071. [Google Scholar] [CrossRef] [PubMed]
  46. Kaze, A.D.; Santhanam, P.; Erqou, S.; Ahima, R.S.; Bertoni, A.; Echouffo-Tcheugui, J.B. Microvascular Disease and Incident Heart Failure among Individuals with Type 2 Diabetes Mellitus. J. Am. Heart Assoc. 2021, 10, e018998. [Google Scholar] [CrossRef]
  47. Owens, C.D.; Mukli, P.; Csipo, T.; Lipecz, A.; Silva-Palacios, F.; Dasari, T.W.; Tarantini, S.; Gardner, A.W.; Montgomery, P.S.; Waldstein, S.R.; et al. Microvascular dysfunction and neurovascular uncoupling are exacerbated in peripheral artery disease, increasing the risk of cognitive decline in older adults. Am. J. Physiol. Heart Circ. Physiol. 2022, 322, H924–H935. [Google Scholar] [CrossRef]
  48. Matsushita, K.; Ballew, S.H.; Coresh, J.; Arima, H.; Arnlov, J.; Cirillo, M.; Ebert, N.; Hiramoto, J.S.; Kimm, H.; Shlipak, M.G.; et al. Measures of chronic kidney disease and risk of incident peripheral artery disease: A collaborative meta-analysis of individual participant data. Lancet Diabetes Endocrinol. 2017, 5, 718–728. [Google Scholar] [CrossRef] [Green Version]
  49. Yang, C.; Kwak, L.; Ballew, S.H.; Jaar, B.G.; Deal, J.A.; Folsom, A.R.; Heiss, G.; Sharrett, A.R.; Selvin, E.; Sabanayagam, C.; et al. Retinal microvascular findings and risk of incident peripheral artery disease: An analysis from the Atherosclerosis Risk in Communities (ARIC) Study. Atherosclerosis 2020, 294, 62–71. [Google Scholar] [CrossRef] [Green Version]
  50. De Lorenzo, A.; Moreira, A.S.B.; Muccillo, F.B.; Assad, M.; Tibirica, E.V. Microvascular Function and Endothelial Progenitor Cells in Patients with Severe Hypercholesterolemia and the Familial Hypercholesterolemia Phenotype. Cardiology 2017, 137, 231–236. [Google Scholar] [CrossRef]
  51. Romero, S.A.; Moralez, G.; Jaffery, M.F.; Huang, M.U.; Engelland, R.E.; Cramer, M.N.; Crandall, C.G. Exercise Training Improves Microvascular Function in Burn Injury Survivors. Med. Sci. Sports Exerc. 2020, 52, 2430–2436. [Google Scholar] [CrossRef]
  52. Bauer, C.J.; Findlay, M.; Koliamitra, C.; Zimmer, P.; Schick, V.; Ludwig, S.; Gurtner, G.C.; Riedel, B.; Schier, R. Preoperative exercise induces endothelial progenitor cell mobilisation in patients undergoing major surgery—A prospective randomised controlled clinical proof-of-concept trial. Heliyon 2022, 8, e10705. [Google Scholar] [CrossRef]
  53. Schier, R.; El-Zein, R.; Cortes, A.; Liu, M.; Collins, M.; Rafat, N.; Teschendorf, P.; Wu, H.K.; Heymach, J.; Mehran, R.; et al. Endothelial progenitor cell mobilization by preoperative exercise: A bone marrow response associated with postoperative outcome. Br. J. Anaesth. 2014, 113, 652–660. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Hill, J.M.; Zalos, G.; Halcox, J.P.; Schenke, W.H.; Waclawiw, M.A.; Quyyumi, A.A.; Finkel, T. Circulating endothelial progenitor cells, vascular function, and cardiovascular risk. N. Engl. J. Med. 2003, 348, 593–600. [Google Scholar] [CrossRef] [PubMed]
  55. Hurley, D.M.; Williams, E.R.; Cross, J.M.; Riedinger, B.R.; Meyer, R.A.; Abela, G.S.; Slade, J.M. Aerobic Exercise Improves Microvascular Function in Older Adults. Med. Sci. Sports Exerc. 2019, 51, 773–781. [Google Scholar] [CrossRef]
  56. Dillon, G.A.; Stanhewicz, A.E.; Serviente, C.; Flores, V.A.; Stachenfeld, N.; Alexander, L.M. Seven days of statin treatment improves nitric-oxide mediated endothelial-dependent cutaneous microvascular function in women with endometriosis. Microvasc. Res. 2022, 144, 104421. [Google Scholar] [CrossRef] [PubMed]
  57. Pajkowski, M.; Dudziak, M.; Chlebus, K.; Hellmann, M. Assessment of microvascular function and pharmacological regulation in genetically confirmed familial hypercholesterolemia. Microvasc. Res. 2021, 138, 104216. [Google Scholar] [CrossRef]
  58. Tentolouris, A.; Eleftheriadou, I.; Tzeravini, E.; Tsilingiris, D.; Paschou, S.A.; Siasos, G.; Tentolouris, N. Endothelium as a Therapeutic Target in Diabetes Mellitus: From Basic Mechanisms to Clinical Practice. Curr. Med. Chem. 2020, 27, 1089–1131. [Google Scholar] [CrossRef]
  59. Love, K.M.; Barrett, E.J.; Horton, W.B. Metformin’s Impact on the Microvascular Response to Insulin. Endocrinology 2022, 163, bqac162. [Google Scholar] [CrossRef]
  60. Liu, J.; Aylor, K.W.; Chai, W.; Barrett, E.J.; Liu, Z. Metformin prevents endothelial oxidative stress and microvascular insulin resistance during obesity development in male rats. Am. J. Physiol. Endocrinol. Metab. 2022, 322, E293–E306. [Google Scholar] [CrossRef]
  61. Silva, C.; Rodrigues, I.; Andrade, S.; Costa, R.; Soares, R. Metformin Reduces Vascular Assembly in High Glucose-Treated Human Microvascular Endothelial Cells in An AMPK-Independent Manner. Cell J. 2021, 23, 174–183. [Google Scholar] [CrossRef]
  62. Simpson, E.L.; Kearns, B.; Stevenson, M.D.; Cantrell, A.J.; Littlewood, C.; Michaels, J.A. Enhancements to angioplasty for peripheral arterial occlusive disease: Systematic review, cost-effectiveness assessment and expected value of information analysis. Health Technol. Assess. 2014, 18, 1–252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Buccheri, D.; Piraino, D.; Andolina, G.; Cortese, B. Understanding and managing in-stent restenosis: A review of clinical data, from pathogenesis to treatment. J. Thorac. Dis. 2016, 8, E1150–E1162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Nakano, M.; Otsuka, F.; Yahagi, K.; Sakakura, K.; Kutys, R.; Ladich, E.R.; Finn, A.V.; Kolodgie, F.D.; Virmani, R. Human autopsy study of drug-eluting stents restenosis: Histomorphological predictors and neointimal characteristics. Eur. Heart J. 2013, 34, 3304–3313. [Google Scholar] [CrossRef] [Green Version]
  65. Deglise, S.; Bechelli, C.; Allagnat, F. Vascular smooth muscle cells in intimal hyperplasia, an update. Front. Physiol. 2022, 13, 1081881. [Google Scholar] [CrossRef]
  66. Chakraborty, R.; Chatterjee, P.; Dave, J.M.; Ostriker, A.C.; Greif, D.M.; Rzucidlo, E.M.; Martin, K.A. Targeting smooth muscle cell phenotypic switching in vascular disease. JVS Vasc. Sci. 2021, 2, 79–94. [Google Scholar] [CrossRef] [PubMed]
  67. Li, F.; Yan, K.; Wu, L.; Zheng, Z.; Du, Y.; Liu, Z.; Zhao, L.; Li, W.; Sheng, Y.; Ren, L.; et al. Single-cell RNA-seq reveals cellular heterogeneity of mouse carotid artery under disturbed flow. Cell Death Discov. 2021, 7, 180. [Google Scholar] [CrossRef]
  68. Yu-Wei, D.; Li, Z.S.; Xiong, S.M.; Huang, G.; Luo, Y.F.; Huo, T.Y.; Zhou, M.H.; Zheng, Y.W. Paclitaxel induces apoptosis through the TAK1-JNK activation pathway. FEBS Open Bio 2020, 10, 1655–1667. [Google Scholar] [CrossRef]
  69. Cirino, G.; Szabo, C.; Papapetropoulos, A. Physiological roles of hydrogen sulfide in mammalian cells, tissues and organs. Physiol. Rev. 2022, 103, 31–276. [Google Scholar] [CrossRef]
  70. Filipovic, M.R.; Zivanovic, J.; Alvarez, B.; Banerjee, R. Chemical Biology of H2S Signaling through Persulfidation. Chem. Rev. 2018, 118, 1253–1337. [Google Scholar] [CrossRef]
  71. Sen, N. Functional and Molecular Insights of Hydrogen Sulfide Signaling and Protein Sulfhydration. J. Mol. Biol. 2017, 429, 543–561. [Google Scholar] [CrossRef] [Green Version]
  72. Bibli, S.I.; Hu, J.; Looso, M.; Weigert, A.; Ratiu, C.; Wittig, J.; Drekolia, M.K.; Tombor, L.; Randriamboavonjy, V.; Leisegang, M.S.; et al. Mapping the Endothelial Cell S-Sulfhydrome Highlights the Crucial Role of Integrin Sulfhydration in Vascular Function. Circulation 2021, 143, 935–948. [Google Scholar] [CrossRef] [PubMed]
  73. Fu, L.; Liu, K.; He, J.; Tian, C.; Yu, X.; Yang, J. Direct Proteomic Mapping of Cysteine Persulfidation. Antioxid. Redox Signal. 2020, 33, 1061–1076. [Google Scholar] [CrossRef] [PubMed]
  74. Bibli, S.I.; Hu, J.; Sigala, F.; Wittig, I.; Heidler, J.; Zukunft, S.; Tsilimigras, D.I.; Randriamboavonjy, V.; Wittig, J.; Kojonazarov, B.; et al. Cystathionine gamma Lyase Sulfhydrates the RNA Binding Protein Human Antigen R to Preserve Endothelial Cell Function and Delay Atherogenesis. Circulation 2019, 139, 101–114. [Google Scholar] [CrossRef]
  75. Zivanovic, J.; Kouroussis, E.; Kohl, J.B.; Adhikari, B.; Bursac, B.; Schott-Roux, S.; Petrovic, D.; Miljkovic, J.L.; Thomas-Lopez, D.; Jung, Y.; et al. Selective Persulfide Detection Reveals Evolutionarily Conserved Antiaging Effects of S-Sulfhydration. Cell Metab. 2019, 30, 1152–1170.e13. [Google Scholar] [CrossRef] [PubMed]
  76. Mustafa, A.K.; Gadalla, M.M.; Sen, N.; Kim, S.; Mu, W.; Gazi, S.K.; Barrow, R.K.; Yang, G.; Wang, R.; Snyder, S.H. H2S signals through protein S-sulfhydration. Sci. Signal. 2009, 2, ra72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Tanito, M.; Agbaga, M.P.; Anderson, R.E. Upregulation of thioredoxin system via Nrf2-antioxidant responsive element pathway in adaptive-retinal neuroprotection in vivo and in vitro. Free Radic. Biol. Med. 2007, 42, 1838–1850. [Google Scholar] [CrossRef] [PubMed]
  78. Kolluru, G.K.; Shackelford, R.E.; Shen, X.; Dominic, P.; Kevil, C.G. Sulfide regulation of cardiovascular function in health and disease. Nat. Rev. Cardiol. 2022, 20, 109–125. [Google Scholar] [CrossRef]
  79. Akaike, T.; Ida, T.; Wei, F.Y.; Nishida, M.; Kumagai, Y.; Alam, M.M.; Ihara, H.; Sawa, T.; Matsunaga, T.; Kasamatsu, S.; et al. Cysteinyl-tRNA synthetase governs cysteine polysulfidation and mitochondrial bioenergetics. Nat. Commun. 2017, 8, 1177. [Google Scholar] [CrossRef] [Green Version]
  80. Longchamp, A.; Mirabella, T.; Arduini, A.; MacArthur, M.R.; Das, A.; Trevino-Villarreal, J.H.; Hine, C.; Ben-Sahra, I.; Knudsen, N.H.; Brace, L.E.; et al. Amino Acid Restriction Triggers Angiogenesis via GCN2/ATF4 Regulation of VEGF and H2S Production. Cell 2018, 173, 117–129.e4. [Google Scholar] [CrossRef] [Green Version]
  81. Bibli, S.I.; Hu, J.; Leisegang, M.S.; Wittig, J.; Zukunft, S.; Kapasakalidi, A.; Fisslthaler, B.; Tsilimigras, D.; Zografos, G.; Filis, K.; et al. Shear stress regulates cystathionine gamma lyase expression to preserve endothelial redox balance and reduce membrane lipid peroxidation. Redox Biol. 2020, 28, 101379. [Google Scholar] [CrossRef]
  82. Yuan, S.; Yurdagul, A., Jr.; Peretik, J.M.; Alfaidi, M.; Al Yafeai, Z.; Pardue, S.; Kevil, C.G.; Orr, A.W. Cystathionine gamma-Lyase Modulates Flow-Dependent Vascular Remodeling. Arterioscler. Thromb. Vasc. Biol. 2018, 38, 2126–2136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Wang, J.; Jia, G.; Li, H.; Yan, S.; Qian, J.; Guo, X.; Li, G.; Qin, H.; Zhu, Z.; Wu, Y.; et al. H2O2-Mediated Oxidative Stress Enhances Cystathionine γ-Lyase-Derived H2S Synthesis Via a Sulfenic Acid Intermediate. Antioxidants 2021, 10, 1488. [Google Scholar] [CrossRef]
  84. Das, M.; Dewan, A.; Shee, S.; Singh, A. The Multifaceted Bacterial Cysteine Desulfurases: From Metabolism to Pathogenesis. Antioxidants 2021, 10, 997. [Google Scholar] [CrossRef] [PubMed]
  85. Kushkevych, I.; Cejnar, J.; Treml, J.; Dordevic, D.; Kollar, P.; Vitezova, M. Recent Advances in Metabolic Pathways of Sulfate Reduction in Intestinal Bacteria. Cells 2020, 9, 698. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Figliuolo, V.R.; Coutinho-Silva, R.; Coutinho, C. Contribution of sulfate-reducing bacteria to homeostasis disruption during intestinal inflammation. Life Sci. 2018, 215, 145–151. [Google Scholar] [CrossRef] [PubMed]
  87. Singh, S.B.; Lin, H.C. Hydrogen Sulfide in Physiology and Diseases of the Digestive Tract. Microorganisms 2015, 3, 866–889. [Google Scholar] [CrossRef] [Green Version]
  88. Rey, F.E.; Gonzalez, M.D.; Cheng, J.; Wu, M.; Ahern, P.P.; Gordon, J.I. Metabolic niche of a prominent sulfate-reducing human gut bacterium. Proc. Natl. Acad. Sci. USA 2013, 110, 13582–13587. [Google Scholar] [CrossRef] [Green Version]
  89. Islam, K.N.; Polhemus, D.J.; Donnarumma, E.; Brewster, L.P.; Lefer, D.J. Hydrogen Sulfide Levels and Nuclear Factor-Erythroid 2-Related Factor 2 (NRF2) Activity Are Attenuated in the Setting of Critical Limb Ischemia (CLI). J. Am. Heart Assoc. 2015, 4, e001986. [Google Scholar] [CrossRef] [Green Version]
  90. Beard, R.S., Jr.; Bearden, S.E. Vascular complications of cystathionine beta-synthase deficiency: Future directions for homocysteine-to-hydrogen sulfide research. Am. J. Physiol. Heart Circ. Physiol. 2011, 300, H13–H26. [Google Scholar] [CrossRef]
  91. Longchamp, A.; MacArthur, M.R.; Trocha, K.; Ganahl, J.; Mann, C.G.; Kip, P.; King, W.W.; Sharma, G.; Tao, M.; Mitchell, S.J.; et al. Plasma Hydrogen Sulfide Is Positively Associated With Post-operative Survival in Patients Undergoing Surgical Revascularization. Front. Cardiovasc. Med. 2021, 8, 750926. [Google Scholar] [CrossRef]
  92. Wang, R. Roles of Hydrogen Sulfide in Hypertension Development and Its Complications: What, So What, Now What. Hypertension 2023, 80, 936–944. [Google Scholar] [CrossRef] [PubMed]
  93. Hosoki, R.; Matsuki, N.; Kimura, H. The possible role of hydrogen sulfide as an endogenous smooth muscle relaxant in synergy with nitric oxide. Biochem. Biophys. Res. Commun. 1997, 237, 527–531. [Google Scholar] [CrossRef] [PubMed]
  94. Tain, Y.L.; Hsu, C.N.; Lu, P.C. Early short-term treatment with exogenous hydrogen sulfide postpones the transition from prehypertension to hypertension in spontaneously hypertensive rat. Clin. Exp. Hypertens. 2018, 40, 58–64. [Google Scholar] [CrossRef] [PubMed]
  95. Zhao, X.; Zhang, L.K.; Zhang, C.Y.; Zeng, X.J.; Yan, H.; Jin, H.F.; Tang, C.S.; Du, J.B. Regulatory effect of hydrogen sulfide on vascular collagen content in spontaneously hypertensive rats. Hypertens. Res. 2008, 31, 1619–1630. [Google Scholar] [CrossRef] [Green Version]
  96. Sun, Y.; Huang, Y.; Zhang, R.; Chen, Q.; Chen, J.; Zong, Y.; Liu, J.; Feng, S.; Liu, A.D.; Holmberg, L.; et al. Hydrogen sulfide upregulates KATP channel expression in vascular smooth muscle cells of spontaneously hypertensive rats. J. Mol. Med. 2015, 93, 439–455. [Google Scholar] [CrossRef]
  97. Huang, P.; Chen, S.; Wang, Y.; Liu, J.; Yao, Q.; Huang, Y.; Li, H.; Zhu, M.; Wang, S.; Li, L.; et al. Down-regulated CBS/H2S pathway is involved in high-salt-induced hypertension in Dahl rats. Nitric Oxide 2015, 46, 192–203. [Google Scholar] [CrossRef]
  98. Liu, X.Y.; Qian, L.L.; Wang, R.X. Hydrogen Sulfide-Induced Vasodilation: The Involvement of Vascular Potassium Channels. Front. Pharmacol. 2022, 13, 911704. [Google Scholar] [CrossRef] [PubMed]
  99. Yang, G.; Wu, L.; Jiang, B.; Yang, W.; Qi, J.; Cao, K.; Meng, Q.; Mustafa, A.K.; Mu, W.; Zhang, S.; et al. H2S as a physiologic vasorelaxant: Hypertension in mice with deletion of cystathionine gamma-lyase. Science 2008, 322, 587–590. [Google Scholar] [CrossRef] [Green Version]
  100. Mustafa, A.K.; Sikka, G.; Gazi, S.K.; Steppan, J.; Jung, S.M.; Bhunia, A.K.; Barodka, V.M.; Gazi, F.K.; Barrow, R.K.; Wang, R.; et al. Hydrogen sulfide as endothelium-derived hyperpolarizing factor sulfhydrates potassium channels. Circ. Res. 2011, 109, 1259–1268. [Google Scholar] [CrossRef]
  101. Naik, J.S.; Osmond, J.M.; Walker, B.R.; Kanagy, N.L. Hydrogen sulfide-induced vasodilation mediated by endothelial TRPV4 channels. Am. J. Physiol. Heart Circ. Physiol. 2016, 311, H1437–H1444. [Google Scholar] [CrossRef] [Green Version]
  102. Szabo, C.; Papapetropoulos, A. Hydrogen sulphide and angiogenesis: Mechanisms and applications. Br. J. Pharmacol. 2011, 164, 853–865. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Moccia, F.; Bertoni, G.; Pla, A.F.; Dragoni, S.; Pupo, E.; Merlino, A.; Mancardi, D.; Munaron, L.; Tanzi, F. Hydrogen sulfide regulates intracellular Ca2+ concentration in endothelial cells from excised rat aorta. Curr. Pharm. Biotechnol. 2011, 12, 1416–1426. [Google Scholar] [CrossRef] [PubMed]
  104. d’Emmanuele di Villa Bianca, R.; Sorrentino, R.; Coletta, C.; Mitidieri, E.; Rossi, A.; Vellecco, V.; Pinto, A.; Cirino, G.; Sorrentino, R. Hydrogen sulfide-induced dual vascular effect involves arachidonic acid cascade in rat mesenteric arterial bed. J. Pharmacol. Exp. Ther. 2011, 337, 59–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Orlov, S.N.; Gusakova, S.V.; Smaglii, L.V.; Koltsova, S.V.; Sidorenko, S.V. Vasoconstriction triggered by hydrogen sulfide: Evidence for Na+,K+,2Cl cotransport and L-type Ca2+ channel-mediated pathway. Biochem. Biophys. Rep. 2017, 12, 220–227. [Google Scholar] [CrossRef] [PubMed]
  106. Ping, N.N.; Li, S.; Mi, Y.N.; Cao, L.; Cao, Y.X. Hydrogen sulphide induces vasoconstriction of rat coronary artery via activation of Ca2+ influx. Acta Physiol. 2015, 214, 88–96. [Google Scholar] [CrossRef]
  107. Szijarto, I.A.; Marko, L.; Filipovic, M.R.; Miljkovic, J.L.; Tabeling, C.; Tsvetkov, D.; Wang, N.; Rabelo, L.A.; Witzenrath, M.; Diedrich, A.; et al. Cystathionine gamma-Lyase-Produced Hydrogen Sulfide Controls Endothelial NO Bioavailability and Blood Pressure. Hypertension 2018, 71, 1210–1217. [Google Scholar] [CrossRef]
  108. Kanagy, N.L.; Szabo, C.; Papapetropoulos, A. Vascular biology of hydrogen sulfide. Am. J. Physiol. Cell Physiol. 2017, 312, C537–C549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Greaney, J.L.; Kutz, J.L.; Shank, S.W.; Jandu, S.; Santhanam, L.; Alexander, L.M. Impaired Hydrogen Sulfide-Mediated Vasodilation Contributes to Microvascular Endothelial Dysfunction in Hypertensive Adults. Hypertension 2017, 69, 902–909. [Google Scholar] [CrossRef] [Green Version]
  110. Aminzadeh, M.A.; Vaziri, N.D. Downregulation of the renal and hepatic hydrogen sulfide (H2S)-producing enzymes and capacity in chronic kidney disease. Nephrol. Dial. Transplant. 2012, 27, 498–504. [Google Scholar] [CrossRef] [Green Version]
  111. Khaledifar, A.; Mobasheri, M.; Kheiri, S.; Zamani, Z. Comparison of N-acetylcysteine and angiotensin converting enzyme inhibitors in blood pressure regulation in hypertensive patients. ARYA Atheroscler. 2015, 11, 5–13. [Google Scholar]
  112. Dillon, G.A.; Stanhewicz, A.E.; Serviente, C.; Greaney, J.L.; Alexander, L.M. Hydrogen sulfide-dependent microvascular vasodilation is improved following chronic sulfhydryl-donating antihypertensive pharmacotherapy in adults with hypertension. Am. J. Physiol. Heart Circ. Physiol. 2021, 321, H728–H734. [Google Scholar] [CrossRef] [PubMed]
  113. Wang, Y.; Zhao, X.; Jin, H.; Wei, H.; Li, W.; Bu, D.; Tang, X.; Ren, Y.; Tang, C.; Du, J. Role of hydrogen sulfide in the development of atherosclerotic lesions in apolipoprotein E knockout mice. Arterioscler. Thromb. Vasc. Biol. 2009, 29, 173–179. [Google Scholar] [CrossRef]
  114. Mani, S.; Li, H.; Untereiner, A.; Wu, L.; Yang, G.; Austin, R.C.; Dickhout, J.G.; Lhotak, S.; Meng, Q.H.; Wang, R. Decreased endogenous production of hydrogen sulfide accelerates atherosclerosis. Circulation 2013, 127, 2523–2534. [Google Scholar] [CrossRef] [Green Version]
  115. Ford, A.; Al-Magableh, M.; Gaspari, T.A.; Hart, J.L. Chronic NaHS Treatment Is Vasoprotective in High-Fat-Fed ApoE−/− Mice. Int. J.Vasc. Med. 2013, 2013, 915983. [Google Scholar] [CrossRef] [Green Version]
  116. Zhang, H.; Guo, C.; Wu, D.; Zhang, A.; Gu, T.; Wang, L.; Wang, C. Hydrogen sulfide inhibits the development of atherosclerosis with suppressing CX3CR1 and CX3CL1 expression. PLoS ONE 2012, 7, e41147. [Google Scholar] [CrossRef]
  117. Liu, Z.; Han, Y.; Li, L.; Lu, H.; Meng, G.; Li, X.; Shirhan, M.; Peh, M.T.; Xie, L.; Zhou, S.; et al. The hydrogen sulfide donor, GYY4137, exhibits anti-atherosclerotic activity in high fat fed apolipoprotein E−/− mice. Br. J. Pharmacol. 2013, 169, 1795–1809. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Zhang, H.; Guo, C.; Zhang, A.; Fan, Y.; Gu, T.; Wu, D.; Sparatore, A.; Wang, C. Effect of S-aspirin, a novel hydrogen-sulfide-releasing aspirin (ACS14), on atherosclerosis in apoE-deficient mice. Eur. J. Pharmacol. 2012, 697, 106–116. [Google Scholar] [CrossRef]
  119. Pan, L.L.; Qin, M.; Liu, X.H.; Zhu, Y.Z. The Role of Hydrogen Sulfide on Cardiovascular Homeostasis: An Overview with Update on Immunomodulation. Front. Pharmacol. 2017, 8, 686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Du, J.; Huang, Y.; Yan, H.; Zhang, Q.; Zhao, M.; Zhu, M.; Liu, J.; Chen, S.X.; Bu, D.; Tang, C.; et al. Hydrogen sulfide suppresses oxidized low-density lipoprotein (ox-LDL)-stimulated monocyte chemoattractant protein 1 generation from macrophages via the nuclear factor kappaB (NF-kappaB) pathway. J. Biol. Chem. 2014, 289, 9741–9753. [Google Scholar] [CrossRef] [Green Version]
  121. Wang, X.H.; Wang, F.; You, S.J.; Cao, Y.J.; Cao, L.D.; Han, Q.; Liu, C.F.; Hu, L.F. Dysregulation of cystathionine gamma-lyase (CSE)/hydrogen sulfide pathway contributes to ox-LDL-induced inflammation in macrophage. Cell Signal. 2013, 25, 2255–2262. [Google Scholar] [CrossRef]
  122. Sen, N.; Paul, B.D.; Gadalla, M.M.; Mustafa, A.K.; Sen, T.; Xu, R.; Kim, S.; Snyder, S.H. Hydrogen sulfide-linked sulfhydration of NF-kappaB mediates its antiapoptotic actions. Mol. Cell 2012, 45, 13–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Jin, H.F.; Liang, C.; Liang, J.M.; Tang, C.S.; Du, J.B. Effects of hydrogen sulfide on vascular inflammation in pulmonary hypertension induced by high pulmonary blood flow: Experiment with rats. Zhonghua Yi Xue Za Zhi 2008, 88, 2235–2239. [Google Scholar] [PubMed]
  124. Brancaleone, V.; Mitidieri, E.; Flower, R.J.; Cirino, G.; Perretti, M. Annexin A1 mediates hydrogen sulfide properties in the control of inflammation. J. Pharmacol. Exp. Ther. 2014, 351, 96–104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Du, C.; Lin, X.; Xu, W.; Zheng, F.; Cai, J.; Yang, J.; Cui, Q.; Tang, C.; Cai, J.; Xu, G.; et al. Sulfhydrated Sirtuin-1 Increasing Its Deacetylation Activity Is an Essential Epigenetics Mechanism of Anti-Atherogenesis by Hydrogen Sulfide. Antioxid. Redox Signal. 2019, 30, 184–197. [Google Scholar] [CrossRef] [PubMed]
  126. Liu, Y.H.; Lu, M.; Hu, L.F.; Wong, P.T.; Webb, G.D.; Bian, J.S. Hydrogen sulfide in the mammalian cardiovascular system. Antioxid. Redox Signal. 2012, 17, 141–185. [Google Scholar] [CrossRef]
  127. Martelli, A.; Piragine, E.; Gorica, E.; Citi, V.; Testai, L.; Pagnotta, E.; Lazzeri, L.; Pecchioni, N.; Ciccone, V.; Montanaro, R.; et al. The H2S-Donor Erucin Exhibits Protective Effects against Vascular Inflammation in Human Endothelial and Smooth Muscle Cells. Antioxidants 2021, 10, 961. [Google Scholar] [CrossRef]
  128. Wang, R.; Szabo, C.; Ichinose, F.; Ahmed, A.; Whiteman, M.; Papapetropoulos, A. The role of H2S bioavailability in endothelial dysfunction. Trends Pharmacol. Sci. 2015, 36, 568–578. [Google Scholar] [CrossRef] [Green Version]
  129. Dufton, N.; Natividad, J.; Verdu, E.F.; Wallace, J.L. Hydrogen sulfide and resolution of acute inflammation: A comparative study utilizing a novel fluorescent probe. Sci. Rep. 2012, 2, 499. [Google Scholar] [CrossRef] [Green Version]
  130. Miao, L.; Xin, X.; Xin, H.; Shen, X.; Zhu, Y.Z. Hydrogen Sulfide Recruits Macrophage Migration by Integrin beta1-Src-FAK/Pyk2-Rac Pathway in Myocardial Infarction. Sci. Rep. 2016, 6, 22363. [Google Scholar] [CrossRef] [Green Version]
  131. Zhou, X.; Chu, X.; Xin, D.; Li, T.; Bai, X.; Qiu, J.; Yuan, H.; Liu, D.; Wang, D.; Wang, Z. L-Cysteine-Derived H2S Promotes Microglia M2 Polarization via Activation of the AMPK Pathway in Hypoxia-Ischemic Neonatal Mice. Front. Mol. Neurosci. 2019, 12, 58. [Google Scholar] [CrossRef]
  132. Lin, Y.; Chen, Y.; Zhu, N.; Zhao, S.; Fan, J.; Liu, E. Hydrogen sulfide inhibits development of atherosclerosis through up-regulating protein S-nitrosylation. Biomed. Pharmacother. 2016, 83, 466–476. [Google Scholar] [CrossRef] [PubMed]
  133. Ang, S.F.; Sio, S.W.; Moochhala, S.M.; MacAry, P.A.; Bhatia, M. Hydrogen sulfide upregulates cyclooxygenase-2 and prostaglandin E metabolite in sepsis-evoked acute lung injury via transient receptor potential vanilloid type 1 channel activation. J. Immunol. 2011, 187, 4778–4787. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Zheng, Q.; Pan, L.; Ji, Y. H 2S protects against diabetes-accelerated atherosclerosis by preventing the activation of NLRP3 inflammasome. J. Biomed. Res. 2019, 34, 94–102. [Google Scholar] [CrossRef] [PubMed]
  135. Steven, S.; Daiber, A.; Dopheide, J.F.; Munzel, T.; Espinola-Klein, C. Peripheral artery disease, redox signaling, oxidative stress—Basic and clinical aspects. Redox. Biol. 2017, 12, 787–797. [Google Scholar] [CrossRef]
  136. Geng, B.; Chang, L.; Pan, C.; Qi, Y.; Zhao, J.; Pang, Y.; Du, J.; Tang, C. Endogenous hydrogen sulfide regulation of myocardial injury induced by isoproterenol. Biochem. Biophys. Res. Commun. 2004, 318, 756–763. [Google Scholar] [CrossRef]
  137. Muellner, M.K.; Schreier, S.M.; Laggner, H.; Hermann, M.; Esterbauer, H.; Exner, M.; Gmeiner, B.M.; Kapiotis, S. Hydrogen sulfide destroys lipid hydroperoxides in oxidized LDL. Biochem. J. 2009, 420, 277–281. [Google Scholar] [CrossRef] [Green Version]
  138. Zavaczki, E.; Jeney, V.; Agarwal, A.; Zarjou, A.; Oros, M.; Katko, M.; Varga, Z.; Balla, G.; Balla, J. Hydrogen sulfide inhibits the calcification and osteoblastic differentiation of vascular smooth muscle cells. Kidney Int. 2011, 80, 731–739. [Google Scholar] [CrossRef] [Green Version]
  139. Du, H.P.; Li, J.; You, S.J.; Wang, Y.L.; Wang, F.; Cao, Y.J.; Hu, L.F.; Liu, C.F. DNA methylation in cystathionine-gamma-lyase (CSE) gene promoter induced by ox-LDL in macrophages and in apoE knockout mice. Biochem. Biophys. Res. Commun. 2016, 469, 776–782. [Google Scholar] [CrossRef]
  140. Balaban, R.S.; Nemoto, S.; Finkel, T. Mitochondria, oxidants, and aging. Cell 2005, 120, 483–495. [Google Scholar] [CrossRef] [Green Version]
  141. Turrens, J.F. Mitochondrial formation of reactive oxygen species. J. Physiol. 2003, 552, 335–344. [Google Scholar] [CrossRef]
  142. Paul, B.D.; Snyder, S.H.; Kashfi, K. Effects of hydrogen sulfide on mitochondrial function and cellular bioenergetics. Redox. Biol. 2021, 38, 101772. [Google Scholar] [CrossRef] [PubMed]
  143. Corsello, T.; Komaravelli, N.; Casola, A. Role of Hydrogen Sulfide in NRF2- and Sirtuin-Dependent Maintenance of Cellular Redox Balance. Antioxidants 2018, 7, 129. [Google Scholar] [CrossRef] [PubMed]
  144. Xie, L.; Gu, Y.; Wen, M.; Zhao, S.; Wang, W.; Ma, Y.; Meng, G.; Han, Y.; Wang, Y.; Liu, G.; et al. Hydrogen Sulfide Induces Keap1 S-sulfhydration and Suppresses Diabetes-Accelerated Atherosclerosis via Nrf2 Activation. Diabetes 2016, 65, 3171–3184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Liu, J.; Wu, J.; Sun, A.; Sun, Y.; Yu, X.; Liu, N.; Dong, S.; Yang, F.; Zhang, L.; Zhong, X.; et al. Hydrogen sulfide decreases high glucose/palmitate-induced autophagy in endothelial cells by the Nrf2-ROS-AMPK signaling pathway. Cell Biosci. 2016, 6, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Shefa, U.; Kim, M.S.; Jeong, N.Y.; Jung, J. Antioxidant and Cell-Signaling Functions of Hydrogen Sulfide in the Central Nervous System. Oxid. Med. Cell Longev. 2018, 2018, 1873962. [Google Scholar] [CrossRef] [Green Version]
  147. Cheung, S.H.; Lau, J.Y.W. Hydrogen sulfide mediates athero-protection against oxidative stress via S-sulfhydration. PLoS ONE 2018, 13, e0194176. [Google Scholar] [CrossRef] [Green Version]
  148. Xie, Z.Z.; Liu, Y.; Bian, J.S. Hydrogen Sulfide and Cellular Redox Homeostasis. Oxid. Med. Cell Longev. 2016, 2016, 6043038. [Google Scholar] [CrossRef] [Green Version]
  149. Mao, Z.; Huang, Y.; Zhang, Z.; Yang, X.; Zhang, X.; Huang, Y.; Sawada, N.; Mitsui, T.; Takeda, M.; Yao, J. Pharmacological levels of hydrogen sulfide inhibit oxidative cell injury through regulating the redox state of thioredoxin. Free Radic. Biol. Med. 2019, 134, 190–199. [Google Scholar] [CrossRef]
  150. Nicholson, C.K.; Lambert, J.P.; Molkentin, J.D.; Sadoshima, J.; Calvert, J.W. Thioredoxin 1 is essential for sodium sulfide-mediated cardioprotection in the setting of heart failure. Arterioscler. Thromb. Vasc. Biol. 2013, 33, 744–751. [Google Scholar] [CrossRef] [Green Version]
  151. Tian, D.; Dong, J.; Jin, S.; Teng, X.; Wu, Y. Endogenous hydrogen sulfide-mediated MAPK inhibition preserves endothelial function through TXNIP signaling. Free Radic. Biol. Med. 2017, 110, 291–299. [Google Scholar] [CrossRef]
  152. Wang, Y.; Ji, N.; Gong, X.; Ni, S.; Xu, L.; Zhang, H. Thioredoxin-1 attenuates atherosclerosis development through inhibiting NLRP3 inflammasome. Endocrine 2020, 70, 65–70. [Google Scholar] [CrossRef] [PubMed]
  153. El Hadri, K.; Mahmood, D.F.; Couchie, D.; Jguirim-Souissi, I.; Genze, F.; Diderot, V.; Syrovets, T.; Lunov, O.; Simmet, T.; Rouis, M. Thioredoxin-1 promotes anti-inflammatory macrophages of the M2 phenotype and antagonizes atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2012, 32, 1445–1452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Feng, L.; Zhao, G.; Chen, B. Protective Mechanism of Thioredoxin-1 against Atherosclerotic Endothelial Injury Induced by Ox-LDL. J. Cardiol. Cardiovasc. Sci. 2018, 2, 13–16. [Google Scholar] [CrossRef] [Green Version]
  155. Swiatkiewicz, I.; Wroblewski, M.; Nuszkiewicz, J.; Sutkowy, P.; Wroblewska, J.; Wozniak, A. The Role of Oxidative Stress Enhanced by Adiposity in Cardiometabolic Diseases. Int. J. Mol. Sci. 2023, 24, 6382. [Google Scholar] [CrossRef]
  156. Cheng, C.K.; Ding, H.; Jiang, M.; Yin, H.; Gollasch, M.; Huang, Y. Perivascular adipose tissue: Fine-tuner of vascular redox status and inflammation. Redox. Biol. 2023, 62, 102683. [Google Scholar] [CrossRef]
  157. Kim, H.W.; Shi, H.; Winkler, M.A.; Lee, R.; Weintraub, N.L. Perivascular Adipose Tissue and Vascular Perturbation/Atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 2569–2576. [Google Scholar] [CrossRef]
  158. Liu, L.; Shi, Z.; Ji, X.; Zhang, W.; Luan, J.; Zahr, T.; Qiang, L. Adipokines, adiposity, and atherosclerosis. Cell. Mol. Life Sci. 2022, 79, 272. [Google Scholar] [CrossRef]
  159. Raman, P.; Khanal, S. Leptin in Atherosclerosis: Focus on Macrophages, Endothelial and Smooth Muscle Cells. Int. J. Mol. Sci. 2021, 22, 5446. [Google Scholar] [CrossRef]
  160. Maeda, N.; Funahashi, T.; Matsuzawa, Y.; Shimomura, I. Adiponectin, a unique adipocyte-derived factor beyond hormones. Atherosclerosis 2020, 292, 1–9. [Google Scholar] [CrossRef]
  161. Zhu, L.; Yang, B.; Ma, D.; Wang, L.; Duan, W. Hydrogen Sulfide, Adipose Tissue and Diabetes Mellitus. Diabetes Metab. Syndr. Obes. 2020, 13, 1873–1886. [Google Scholar] [CrossRef]
  162. Casili, G.; Randi, E.; Panagaki, T.; Zuhra, K.; Petrosino, M.; Szabo, C. Inhibition of the 3-mercaptopyruvate sulfurtransferase-hydrogen sulfide system promotes cellular lipid accumulation. Geroscience 2022, 44, 2271–2289. [Google Scholar] [CrossRef] [PubMed]
  163. Wang, F.Z.; Zhou, H.; Wang, H.Y.; Dai, H.B.; Gao, Q.; Qian, P.; Zhou, Y.B. Hydrogen sulfide prevents arterial medial calcification in rats with diabetic nephropathy. BMC Cardiovasc. Disord. 2021, 21, 495. [Google Scholar] [CrossRef] [PubMed]
  164. Szabo, C. Roles of hydrogen sulfide in the pathogenesis of diabetes mellitus and its complications. Antioxid. Redox Signal. 2012, 17, 68–80. [Google Scholar] [CrossRef] [PubMed]
  165. Xue, R.; Hao, D.D.; Sun, J.P.; Li, W.W.; Zhao, M.M.; Li, X.H.; Chen, Y.; Zhu, J.H.; Ding, Y.J.; Liu, J.; et al. Hydrogen sulfide treatment promotes glucose uptake by increasing insulin receptor sensitivity and ameliorates kidney lesions in type 2 diabetes. Antioxid. Redox Signal. 2013, 19, 5–23. [Google Scholar] [CrossRef] [Green Version]
  166. Zhao, S.; Li, X.; Li, X.; Wei, X.; Wang, H. Hydrogen Sulfide Plays an Important Role in Diabetic Cardiomyopathy. Front. Cell Dev. Biol. 2021, 9, 627336. [Google Scholar] [CrossRef]
  167. Zhang, H.; Zhao, H.; Guo, N. Protective effect of hydrogen sulfide on the kidney (Review). Mol. Med. Rep. 2021, 24, 696. [Google Scholar] [CrossRef]
  168. Abedin, M.; Tintut, Y.; Demer, L.L. Vascular calcification: Mechanisms and clinical ramifications. Arterioscler. Thromb. Vasc. Biol. 2004, 24, 1161–1170. [Google Scholar] [CrossRef] [Green Version]
  169. Karwowski, W.; Naumnik, B.; Szczepanski, M.; Mysliwiec, M. The mechanism of vascular calcification—A systematic review. Med. Sci. Monit. 2012, 18, RA1–RA11. [Google Scholar] [CrossRef] [Green Version]
  170. Sage, A.P.; Tintut, Y.; Demer, L.L. Regulatory mechanisms in vascular calcification. Nat. Rev. Cardiol. 2010, 7, 528–536. [Google Scholar] [CrossRef]
  171. Shao, J.S.; Cheng, S.L.; Sadhu, J.; Towler, D.A. Inflammation and the osteogenic regulation of vascular calcification: A review and perspective. Hypertension 2010, 55, 579–592. [Google Scholar] [CrossRef] [Green Version]
  172. Wu, S.Y.; Pan, C.S.; Geng, B.; Zhao, J.; Yu, F.; Pang, Y.Z.; Tang, C.S.; Qi, Y.F. Hydrogen sulfide ameliorates vascular calcification induced by vitamin D3 plus nicotine in rats. Acta Pharmacol. Sin. 2006, 27, 299–306. [Google Scholar] [CrossRef]
  173. Yang, R.; Teng, X.; Li, H.; Xue, H.M.; Guo, Q.; Xiao, L.; Wu, Y.M. Hydrogen Sulfide Improves Vascular Calcification in Rats by Inhibiting Endoplasmic Reticulum Stress. Oxidative Med. Cell. Longev. 2016, 2016, 9095242. [Google Scholar] [CrossRef] [Green Version]
  174. Aghagolzadeh, P.; Radpour, R.; Bachtler, M.; van Goor, H.; Smith, E.R.; Lister, A.; Odermatt, A.; Feelisch, M.; Pasch, A. Hydrogen sulfide attenuates calcification of vascular smooth muscle cells via KEAP1/NRF2/NQO1 activation. Atherosclerosis 2017, 265, 78–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Zhou, Y.B.; Zhou, H.; Li, L.; Kang, Y.; Cao, X.; Wu, Z.Y.; Ding, L.; Sethi, G.; Bian, J.S. Hydrogen Sulfide Prevents Elastin Loss and Attenuates Calcification Induced by High Glucose in Smooth Muscle Cells through Suppression of Stat3/Cathepsin S Signaling Pathway. Int. J. Mol. Sci. 2019, 20, 4202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Perna, A.F.; Luciano, M.G.; Ingrosso, D.; Pulzella, P.; Sepe, I.; Lanza, D.; Violetti, E.; Capasso, R.; Lombardi, C.; De Santo, N.G. Hydrogen sulphide-generating pathways in haemodialysis patients: A study on relevant metabolites and transcriptional regulation of genes encoding for key enzymes. Nephrol. Dial. Transplant. 2009, 24, 3756–3763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Nasi, S.; Ea, H.K.; Liote, F.; So, A.; Busso, N. Sodium Thiosulfate Prevents Chondrocyte Mineralization and Reduces the Severity of Murine Osteoarthritis. PLoS ONE 2016, 11, e0158196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Peng, T.; Zhuo, L.; Wang, Y.; Jun, M.; Li, G.; Wang, L.; Hong, D. Systematic review of sodium thiosulfate in treating calciphylaxis in chronic kidney disease patients. Nephrology 2018, 23, 669–675. [Google Scholar] [CrossRef]
  179. Wang, M.J.; Cai, W.J.; Li, N.; Ding, Y.J.; Chen, Y.; Zhu, Y.C. The hydrogen sulfide donor NaHS promotes angiogenesis in a rat model of hind limb ischemia. Antioxid. Redox Signal. 2010, 12, 1065–1077. [Google Scholar] [CrossRef]
  180. Altaany, Z.; Yang, G.; Wang, R. Crosstalk between hydrogen sulfide and nitric oxide in endothelial cells. J. Cell Mol. Med. 2013, 17, 879–888. [Google Scholar] [CrossRef]
  181. Coletta, C.; Papapetropoulos, A.; Erdelyi, K.; Olah, G.; Modis, K.; Panopoulos, P.; Asimakopoulou, A.; Gero, D.; Sharina, I.; Martin, E.; et al. Hydrogen sulfide and nitric oxide are mutually dependent in the regulation of angiogenesis and endothelium-dependent vasorelaxation. Proc. Natl. Acad. Sci. USA 2012, 109, 9161–9166. [Google Scholar] [CrossRef] [Green Version]
  182. Papapetropoulos, A.; Pyriochou, A.; Altaany, Z.; Yang, G.; Marazioti, A.; Zhou, Z.; Jeschke, M.G.; Branski, L.K.; Herndon, D.N.; Wang, R.; et al. Hydrogen sulfide is an endogenous stimulator of angiogenesis. Proc. Natl. Acad. Sci. USA 2009, 106, 21972–21977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Kolluru, G.K.; Bir, S.C.; Yuan, S.; Shen, X.; Pardue, S.; Wang, R.; Kevil, C.G. Cystathionine gamma-lyase regulates arteriogenesis through NO-dependent monocyte recruitment. Cardiovasc. Res. 2015, 107, 590–600. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Kiesworo, K.; MacArthur, M.R.; Kip, P.; Agius, T.; Macabrey, D.; Lambelet, M.; Hamard, L.; Ozaki, C.K.; Mitchell, J.R.; Deglise, S.; et al. Cystathionine-gamma-lyase overexpression modulates oxidized nicotinamide adenine dinucleotide biosynthesis and enhances neovascularization. JVS Vasc. Sci. 2023, 4, 100095. [Google Scholar] [CrossRef] [PubMed]
  185. Majumder, A.; Singh, M.; George, A.K.; Behera, J.; Tyagi, N.; Tyagi, S.C. Hydrogen sulfide improves postischemic neoangiogenesis in the hind limb of cystathionine-beta-synthase mutant mice via PPAR-gamma/VEGF axis. Physiol. Rep. 2018, 6, e13858. [Google Scholar] [CrossRef]
  186. Xiong, Y.; Chang, L.L.; Tran, B.; Dai, T.; Zhong, R.; Mao, Y.C.; Zhu, Y.Z. ZYZ-803, a novel hydrogen sulfide-nitric oxide conjugated donor, promotes angiogenesis via cross-talk between STAT3 and CaMKII. Acta Pharmacol. Sin. 2020, 41, 218–228. [Google Scholar] [CrossRef]
  187. Fu, J.; Zou, J.; Chen, C.; Li, H.; Wang, L.; Zhou, Y. Hydrogen molecules (H2) improve perfusion recovery via antioxidant effects in experimental peripheral arterial disease. Mol. Med. Rep. 2018, 18, 5009–5015. [Google Scholar] [CrossRef] [Green Version]
  188. Cheng, Z.; Garikipati, V.N.; Nickoloff, E.; Wang, C.; Polhemus, D.J.; Zhou, J.; Benedict, C.; Khan, M.; Verma, S.K.; Rabinowitz, J.E.; et al. Restoration of Hydrogen Sulfide Production in Diabetic Mice Improves Reparative Function of Bone Marrow Cells. Circulation 2016, 134, 1467–1483. [Google Scholar] [CrossRef]
  189. Hayashida, R.; Kondo, K.; Morita, S.; Unno, K.; Shintani, S.; Shimizu, Y.; Calvert, J.W.; Shibata, R.; Murohara, T. Diallyl Trisulfide Augments Ischemia-Induced Angiogenesis via an Endothelial Nitric Oxide Synthase-Dependent Mechanism. Circ. J. 2017, 81, 870–878. [Google Scholar] [CrossRef] [Green Version]
  190. Pardue, S.; Kolluru, G.K.; Shen, X.; Lewis, S.E.; Saffle, C.B.; Kelley, E.E.; Kevil, C.G. Hydrogen sulfide stimulates xanthine oxidoreductase conversion to nitrite reductase and formation of NO. Redox Biol. 2020, 34, 101447. [Google Scholar] [CrossRef]
  191. Yang, H.B.; Liu, H.M.; Yan, J.C.; Lu, Z.Y. Effect of Diallyl Trisulfide on Ischemic Tissue Injury and Revascularization in a Diabetic Mouse Model. J. Cardiovasc. Pharmacol. 2018, 71, 367–374. [Google Scholar] [CrossRef]
  192. Syu, J.N.; Yang, M.D.; Tsai, S.Y.; Chiang, E.I.; Chiu, S.C.; Chao, C.Y.; Rodriguez, R.L.; Tang, F.Y. S-allylcysteine Improves Blood Flow Recovery and Prevents Ischemic Injury by Augmenting Neovasculogenesis. Cell Transplant. 2017, 26, 1636–1647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Kan, J.; Guo, W.; Huang, C.; Bao, G.; Zhu, Y.; Zhu, Y.Z. S-propargyl-cysteine, a novel water-soluble modulator of endogenous hydrogen sulfide, promotes angiogenesis through activation of signal transducer and activator of transcription 3. Antioxid. Redox Signal. 2014, 20, 2303–2316. [Google Scholar] [CrossRef] [Green Version]
  194. Rushing, A.M.; Donnarumma, E.; Polhemus, D.J.; Au, K.R.; Victoria, S.E.; Schumacher, J.D.; Li, Z.; Jenkins, J.S.; Lefer, D.J.; Goodchild, T.T. Effects of a novel hydrogen sulfide prodrug in a porcine model of acute limb ischemia. J. Vasc. Surg. 2019, 69, 1924–1935. [Google Scholar] [CrossRef]
  195. Macabrey, D.; Joniova, J.; Gasser, Q.; Bechelli, C.; Longchamp, A.; Urfer, S.; Lambelet, M.; Fu, C.Y.; Schwarz, G.; Wagnieres, G.; et al. Sodium thiosulfate, a source of hydrogen sulfide, stimulates endothelial cell proliferation and neovascularization. Front. Cardiovasc. Med. 2022, 9, 965965. [Google Scholar] [CrossRef]
  196. Sen, U.; Sathnur, P.B.; Kundu, S.; Givvimani, S.; Coley, D.M.; Mishra, P.K.; Qipshidze, N.; Tyagi, N.; Metreveli, N.; Tyagi, S.C. Increased endogenous H2S generation by CBS, CSE, and 3MST gene therapy improves ex vivo renovascular relaxation in hyperhomocysteinemia. Am. J. Physiol. Cell Physiol. 2012, 303, C41–C51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Qipshidze, N.; Metreveli, N.; Mishra, P.K.; Lominadze, D.; Tyagi, S.C. Hydrogen sulfide mitigates cardiac remodeling during myocardial infarction via improvement of angiogenesis. Int. J. Biol. Sci. 2012, 8, 430–441. [Google Scholar] [CrossRef] [Green Version]
  198. Tao, B.B.; Liu, S.Y.; Zhang, C.C.; Fu, W.; Cai, W.J.; Wang, Y.; Shen, Q.; Wang, M.J.; Chen, Y.; Zhang, L.J.; et al. VEGFR2 functions as an H2S-targeting receptor protein kinase with its novel Cys1045-Cys1024 disulfide bond serving as a specific molecular switch for hydrogen sulfide actions in vascular endothelial cells. Antioxid. Redox Signal. 2013, 19, 448–464. [Google Scholar] [CrossRef] [Green Version]
  199. Katsouda, A.; Bibli, S.I.; Pyriochou, A.; Szabo, C.; Papapetropoulos, A. Regulation and role of endogenously produced hydrogen sulfide in angiogenesis. Pharmacol. Res. 2016, 113, 175–185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  200. Cai, W.J.; Wang, M.J.; Moore, P.K.; Jin, H.M.; Yao, T.; Zhu, Y.C. The novel proangiogenic effect of hydrogen sulfide is dependent on Akt phosphorylation. Cardiovasc. Res. 2007, 76, 29–40. [Google Scholar] [CrossRef]
  201. Jang, H.; Oh, M.Y.; Kim, Y.J.; Choi, I.Y.; Yang, H.S.; Ryu, W.S.; Lee, S.H.; Yoon, B.W. Hydrogen sulfide treatment induces angiogenesis after cerebral ischemia. J. Neurosci. Res. 2014, 92, 1520–1528. [Google Scholar] [CrossRef] [PubMed]
  202. Szabo, C. Hydrogen sulfide, an enhancer of vascular nitric oxide signaling: Mechanisms and implications. Am. J. Physiol. Cell Physiol. 2017, 312, C3–C15. [Google Scholar] [CrossRef] [PubMed]
  203. Zecchin, A.; Kalucka, J.; Dubois, C.; Carmeliet, P. How Endothelial Cells Adapt Their Metabolism to Form Vessels in Tumors. Front. Immunol. 2017, 8, 1750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Eelen, G.; de Zeeuw, P.; Treps, L.; Harjes, U.; Wong, B.W.; Carmeliet, P. Endothelial Cell Metabolism. Physiol. Rev. 2018, 98, 3–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Macabrey, D.; Longchamp, A.; MacArthur, M.R.; Lambelet, M.; Urfer, S.; Deglise, S.; Allagnat, F. Sodium thiosulfate acts as a hydrogen sulfide mimetic to prevent intimal hyperplasia via inhibition of tubulin polymerisation. EBioMedicine 2022, 78, 103954. [Google Scholar] [CrossRef]
  206. Yang, G.; Li, H.; Tang, G.; Wu, L.; Zhao, K.; Cao, Q.; Xu, C.; Wang, R. Increased neointimal formation in cystathionine gamma-lyase deficient mice: Role of hydrogen sulfide in alpha5beta1-integrin and matrix metalloproteinase-2 expression in smooth muscle cells. J. Mol. Cell Cardiol. 2012, 52, 677–688. [Google Scholar] [CrossRef]
  207. Trocha, K.M.; Kip, P.; Tao, M.; MacArthur, M.R.; Trevino-Villarreal, J.H.; Longchamp, A.; Toussaint, W.; Lambrecht, B.N.; de Vries, M.R.; Quax, P.H.A.; et al. Short-term preoperative protein restriction attenuates vein graft disease via induction of cystathionine gamma-lyase. Cardiovasc. Res. 2020, 116, 416–428. [Google Scholar] [CrossRef]
  208. Meng, Q.H.; Yang, G.; Yang, W.; Jiang, B.; Wu, L.; Wang, R. Protective effect of hydrogen sulfide on balloon injury-induced neointima hyperplasia in rat carotid arteries. Am. J. Pathol. 2007, 170, 1406–1414. [Google Scholar] [CrossRef] [Green Version]
  209. Ma, B.; Liang, G.; Zhang, F.; Chen, Y.; Zhang, H. Effect of hydrogen sulfide on restenosis of peripheral arteries after angioplasty. Mol. Med. Rep. 2012, 5, 1497–1502. [Google Scholar] [CrossRef] [Green Version]
  210. Macabrey, D.; Deslarzes-Dubuis, C.; Longchamp, A.; Lambelet, M.; Ozaki, C.K.; Corpataux, J.M.; Allagnat, F.; Deglise, S. Hydrogen Sulphide Release via the Angiotensin Converting Enzyme Inhibitor Zofenopril Prevents Intimal Hyperplasia in Human Vein Segments and in a Mouse Model of Carotid Artery Stenosis. Eur. J. Vasc. Endovasc. Surg. 2022, 63, 336–346. [Google Scholar] [CrossRef]
  211. Longchamp, A.; Kaur, K.; Macabrey, D.; Dubuis, C.; Corpataux, J.M.; Deglise, S.; Matson, J.B.; Allagnat, F. Hydrogen sulfide-releasing peptide hydrogel limits the development of intimal hyperplasia in human vein segments. Acta Biomater. 2019, 97, 374–384. [Google Scholar] [CrossRef]
  212. Kip, P.; Tao, M.; Trocha, K.M.; MacArthur, M.R.; Peters, H.A.B.; Mitchell, S.J.; Mann, C.G.; Sluiter, T.J.; Jung, J.; Patterson, S.; et al. Periprocedural Hydrogen Sulfide Therapy Improves Vascular Remodeling and Attenuates Vein Graft Disease. J. Am. Heart Assoc. 2020, 9, e016391. [Google Scholar] [CrossRef] [PubMed]
  213. Yang, G.; Wu, L.; Wang, R. Pro-apoptotic effect of endogenous H2S on human aorta smooth muscle cells. FASEB J. 2006, 20, 553–555. [Google Scholar] [CrossRef] [PubMed]
  214. Yang, G.; Wu, L.; Bryan, S.; Khaper, N.; Mani, S.; Wang, R. Cystathionine gamma-lyase deficiency and overproliferation of smooth muscle cells. Cardiovasc. Res. 2010, 86, 487–495. [Google Scholar] [CrossRef] [Green Version]
  215. Wang, Y.; Wang, X.; Liang, X.; Wu, J.; Dong, S.; Li, H.; Jin, M.; Sun, D.; Zhang, W.; Zhong, X. Inhibition of hydrogen sulfide on the proliferation of vascular smooth muscle cells involved in the modulation of calcium sensing receptor in high homocysteine. Exp. Cell Res. 2016, 347, 184–191. [Google Scholar] [CrossRef]
  216. Zhong, X.; Wang, Y.; Wu, J.; Sun, A.; Yang, F.; Zheng, D.; Li, T.; Dong, S.; Zhao, Y.; Yang, G.; et al. Calcium sensing receptor regulating smooth muscle cells proliferation through initiating cystathionine-gamma-lyase/hydrogen sulfide pathway in diabetic rat. Cell Physiol. Biochem. 2015, 35, 1582–1598. [Google Scholar] [CrossRef] [PubMed]
  217. Wu, B.; Werlin, E.C.; Chen, M.; Mottola, G.; Chatterjee, A.; Lance, K.D.; Bernards, D.A.; Sansbury, B.E.; Spite, M.; Desai, T.A.; et al. Perivascular delivery of resolvin D1 inhibits neointimal hyperplasia in a rabbit vein graft model. J. Vasc. Surg. 2018, 68, 188S–200S.e4. [Google Scholar] [CrossRef] [Green Version]
  218. Razavi, M.K.; Donohoe, D.; D’Agostino, R.B., Jr.; Jaff, M.R.; Adams, G.; Investigators, D. Adventitial Drug Delivery of Dexamethasone to Improve Primary Patency in the Treatment of Superficial Femoral and Popliteal Artery Disease: 12-Month Results From the DANCE Clinical Trial. JACC Cardiovasc. Interv. 2018, 11, 921–931. [Google Scholar] [CrossRef]
  219. Ling, K.; Xu, A.; Chen, Y.; Chen, X.; Li, Y.; Wang, W. Protective effect of a hydrogen sulfide donor on balloon injury-induced restenosis via the Nrf2/HIF-1alpha signaling pathway. Int. J. Mol. Med. 2019, 43, 1299–1310. [Google Scholar] [CrossRef] [Green Version]
  220. Borghi, C.; Bacchelli, S.; Esposti, D.D.; Bignamini, A.; Magnani, B.; Ambrosioni, E. Effects of the administration of an angiotensin-converting enzyme inhibitor during the acute phase of myocardial infarction in patients with arterial hypertension. SMILE Study Investigators. Survival of Myocardial Infarction Long-term Evaluation. Am. J. Hypertens. 1999, 12, 665–672. [Google Scholar] [CrossRef] [Green Version]
  221. Borghi, C.; Omboni, S.; Novo, S.; Vinereanu, D.; Ambrosio, G.; Ambrosioni, E. Efficacy and Safety of Zofenopril Versus Ramipril in the Treatment of Myocardial Infarction and Heart Failure: A Review of the Published and Unpublished Data of the Randomized Double-Blind SMILE-4 Study. Adv. Ther. 2018, 35, 604–618. [Google Scholar] [CrossRef] [Green Version]
  222. Ambrosioni, E.; Borghi, C.; Magnani, B. The effect of the angiotensin-converting-enzyme inhibitor zofenopril on mortality and morbidity after anterior myocardial infarction. The Survival of Myocardial Infarction Long-Term Evaluation (SMILE) Study Investigators. N. Engl. J. Med. 1995, 332, 80–85. [Google Scholar] [CrossRef] [PubMed]
Scheme 1. Endogenous H2S production. L-cysteine and homocysteine are the essential substrates for H2S generation by CBS and CSE in the cytosol. In the mitochondria, CAT metabolizes L-cysteine to 3-MP, which is used by 3MST to release H2S and pyruvate. SQR oxidizes H2S to hydropersulfides (R-SSH), which are then oxidized by ETHE1 in thiosulfate (SO32−). SO32− can be oxidized to sulfate (SO42−) by SUOX or S2O32− by TST. SQR enhances the activity of the complex 2 of the electron transport chain. Moreover, CARS1 and 2 enzymes reconstitute Cys-SSH, which can be reduced to release H2S. CBS: cystathionine β-synthase; CSE: cystathionine γ-lyase; CAT: cysteine aminotransferase; 3-MST: 3-mercaptopyruvate sulphurtransferase; SQR: Sulfide-quinone oxidoreductase; ETHE1: ethylmalonic encephalopathy 1 protein; SUOX: sulfite oxidase; TST: rhodanese; CARS1 and 2: cyteinyl-tRNA 1 and 2; GSH: Gluthathione; CysSSH: Cysteine hydropersulfide; CysSH: Cysteine; Cyt c: cytochrome c; CoQ: coenzyme Q.
Scheme 1. Endogenous H2S production. L-cysteine and homocysteine are the essential substrates for H2S generation by CBS and CSE in the cytosol. In the mitochondria, CAT metabolizes L-cysteine to 3-MP, which is used by 3MST to release H2S and pyruvate. SQR oxidizes H2S to hydropersulfides (R-SSH), which are then oxidized by ETHE1 in thiosulfate (SO32−). SO32− can be oxidized to sulfate (SO42−) by SUOX or S2O32− by TST. SQR enhances the activity of the complex 2 of the electron transport chain. Moreover, CARS1 and 2 enzymes reconstitute Cys-SSH, which can be reduced to release H2S. CBS: cystathionine β-synthase; CSE: cystathionine γ-lyase; CAT: cysteine aminotransferase; 3-MST: 3-mercaptopyruvate sulphurtransferase; SQR: Sulfide-quinone oxidoreductase; ETHE1: ethylmalonic encephalopathy 1 protein; SUOX: sulfite oxidase; TST: rhodanese; CARS1 and 2: cyteinyl-tRNA 1 and 2; GSH: Gluthathione; CysSSH: Cysteine hydropersulfide; CysSH: Cysteine; Cyt c: cytochrome c; CoQ: coenzyme Q.
Ijms 24 09955 sch001
Scheme 2. H2S promotes vasorelaxation. In VSMC, H2S induces vasodilation mainly via persulfidation/opening of the KATP channels, leading to hyperpolarization, closing of VDCC and VSMC relaxation. Moreover, persulfidation of eNOS in EC will allow the production of NO, which will cause sGC/GMP-dependent vasodilation. H2S also promotes TRPV4-mediated Ca2+-influx in EC, which enhances eNOS/NO expression/production, PLA-2-dependent production of arachidonic-acid metabolites, and CSE/H2S expression/production. Ca2+ entry also activates Ca2+-sensitive K+ channels in EC, leading to EC hyperpolarization, which travels through myoendothelial gap junctions to hyperpolarize nearby VSMC. AA: arachidonic-acid; NO: nitric oxide; VSMC: vascular smooth muscle cells; eNOS: endothelial nitric oxide synthase; VDCC: voltage-dependent Ca2+ channels; PLA-2: phospholipase A-2; sGC: soluble guanylate cyclase; GMP: cyclic guanosine-3′,5′-monophosphate; PKG: protein kinase G; PDE5: Phosphodiesterase 5; EC: endothelial cell; CSE: cystathionine γ-lyase; KATP: ATP-sensitive potassium channel. TRPV4: transient receptor potential vanilloid 4.
Scheme 2. H2S promotes vasorelaxation. In VSMC, H2S induces vasodilation mainly via persulfidation/opening of the KATP channels, leading to hyperpolarization, closing of VDCC and VSMC relaxation. Moreover, persulfidation of eNOS in EC will allow the production of NO, which will cause sGC/GMP-dependent vasodilation. H2S also promotes TRPV4-mediated Ca2+-influx in EC, which enhances eNOS/NO expression/production, PLA-2-dependent production of arachidonic-acid metabolites, and CSE/H2S expression/production. Ca2+ entry also activates Ca2+-sensitive K+ channels in EC, leading to EC hyperpolarization, which travels through myoendothelial gap junctions to hyperpolarize nearby VSMC. AA: arachidonic-acid; NO: nitric oxide; VSMC: vascular smooth muscle cells; eNOS: endothelial nitric oxide synthase; VDCC: voltage-dependent Ca2+ channels; PLA-2: phospholipase A-2; sGC: soluble guanylate cyclase; GMP: cyclic guanosine-3′,5′-monophosphate; PKG: protein kinase G; PDE5: Phosphodiesterase 5; EC: endothelial cell; CSE: cystathionine γ-lyase; KATP: ATP-sensitive potassium channel. TRPV4: transient receptor potential vanilloid 4.
Ijms 24 09955 sch002
Scheme 3. Anti-atherosclerotic effects of H2S. H2S persulfides/stabilize IκB, which prevent nucleus translocation of NFκB, leading to decreased production of pro-inflammatory genes. H2S also persulfides Keap1, leading to the translocation of Nrf2 in the nucleus and overexpression of antioxidant factors. This leads to reduced expression of adhesion molecules (ICAM and VCAM) in EC, thereby reducing monocyte adhesion and infiltration. In EC, H2S also promotes eNOS/NO, which inhibits pro-inflammatory signals. In macrophages, NFκB inhibition and Nrf2 activation favor the M2 phenotype. Overall, NRF2 activation and NF-κB inhibition in EC and macrophages leads to reduced secretion of pro-inflammatory factors promoting VSMC dedifferentiation and pathological phenotypic change. Moreover, H2S prevents lipid peroxidation, leading to decreased LDL oxidation and formation of foam cells. H2S also reduces cell apoptosis, limits myofibroblast proliferation and ECM remodeling, and may reduce vascular intimal calcification ICAM: Intercellular Adhesion Molecule 1; VCAM: vascular cell adhesion molecule1; IκB: Inhibitory kinase of NFκB; NFκB: Nuclear factor kappa B; COX2: cyclooxygenase-2; PGE2: Prostaglandin E2; NRF2: nuclear factor erythroid 2–related factor 2; Keap1: Kelch-like ECH-associated protein 1; LDL: low-density lipoprotein; M1: Type 1 macrophages; M2: Type 2 macrophages; ROS: reactive oxygen species; ECM: extracellular matrix; EC: endothelial cells; VSMC: vascular smooth muscle cells.
Scheme 3. Anti-atherosclerotic effects of H2S. H2S persulfides/stabilize IκB, which prevent nucleus translocation of NFκB, leading to decreased production of pro-inflammatory genes. H2S also persulfides Keap1, leading to the translocation of Nrf2 in the nucleus and overexpression of antioxidant factors. This leads to reduced expression of adhesion molecules (ICAM and VCAM) in EC, thereby reducing monocyte adhesion and infiltration. In EC, H2S also promotes eNOS/NO, which inhibits pro-inflammatory signals. In macrophages, NFκB inhibition and Nrf2 activation favor the M2 phenotype. Overall, NRF2 activation and NF-κB inhibition in EC and macrophages leads to reduced secretion of pro-inflammatory factors promoting VSMC dedifferentiation and pathological phenotypic change. Moreover, H2S prevents lipid peroxidation, leading to decreased LDL oxidation and formation of foam cells. H2S also reduces cell apoptosis, limits myofibroblast proliferation and ECM remodeling, and may reduce vascular intimal calcification ICAM: Intercellular Adhesion Molecule 1; VCAM: vascular cell adhesion molecule1; IκB: Inhibitory kinase of NFκB; NFκB: Nuclear factor kappa B; COX2: cyclooxygenase-2; PGE2: Prostaglandin E2; NRF2: nuclear factor erythroid 2–related factor 2; Keap1: Kelch-like ECH-associated protein 1; LDL: low-density lipoprotein; M1: Type 1 macrophages; M2: Type 2 macrophages; ROS: reactive oxygen species; ECM: extracellular matrix; EC: endothelial cells; VSMC: vascular smooth muscle cells.
Ijms 24 09955 sch003
Scheme 4. H2S and vascular medial calcification. In VSMC, H2S inhibits STAT3 and cathepsin S, which will stop the elastin degradation and improves the resolution of vascular calcification. Moreover, H2S increases the production of antioxidant genes Nrf2, which inhibits vascular calcification. Vascular calcification is also reduced by the inhibition of ER-stress by H2S. STAT3: Signal transducer and activator of transcription 3; ER-stress: Endoplasmic reticulum-stress; IκB: Inhibitory kinase of NFκB; NFκB: Nuclear factor kappa B; NRF2: nuclear factor erythroid 2–related factor 2; KEAP1: Kelch-like ECH-associated protein 1;CBFα1: core-binding factor alpha1; ECM: extracellular matrix; EC: endothelial cells; VSMC: vascular smooth muscle cells.
Scheme 4. H2S and vascular medial calcification. In VSMC, H2S inhibits STAT3 and cathepsin S, which will stop the elastin degradation and improves the resolution of vascular calcification. Moreover, H2S increases the production of antioxidant genes Nrf2, which inhibits vascular calcification. Vascular calcification is also reduced by the inhibition of ER-stress by H2S. STAT3: Signal transducer and activator of transcription 3; ER-stress: Endoplasmic reticulum-stress; IκB: Inhibitory kinase of NFκB; NFκB: Nuclear factor kappa B; NRF2: nuclear factor erythroid 2–related factor 2; KEAP1: Kelch-like ECH-associated protein 1;CBFα1: core-binding factor alpha1; ECM: extracellular matrix; EC: endothelial cells; VSMC: vascular smooth muscle cells.
Ijms 24 09955 sch004
Scheme 5. H2S promotes angiogenesis. H2S promotes VEGF signaling via persulfidation/activation of the VEGFR2, leading to (i) increased eNOS/NO expression/production; (ii) increased MAPK signaling; (iii) increased CSE/H2S expression/production in a positive feedback loop. H2S further enhances NO production via eNOS persulfidation. Altogether, these effects facilitate VEGF-induced sprouting angiogenesis. In addition, H2S inhibits mitochondrial respiration, which promotes glycolysis and ATP production, proliferation, and migration in hypoxic condition. Akt: Protein kinase B, EC: endothelial cells; ERK: Extracellular signal-regulated kinase; ETC: electron transport chain; GLUT: glucose transporter; IP3: inositol triphosphate; MKK: mitogen-activated kinase kinase; PI3K: phosphor-inositol 3 kinase; PKC: protein kinase C; VEGFR2: Vascular endothelial growth factor receptor 2; NO: nitric oxide; eNOS: endothelial NO synthase; persulf: perfulfidation; EC: endothelial cells; CSE: cystathionine γ-lyase.
Scheme 5. H2S promotes angiogenesis. H2S promotes VEGF signaling via persulfidation/activation of the VEGFR2, leading to (i) increased eNOS/NO expression/production; (ii) increased MAPK signaling; (iii) increased CSE/H2S expression/production in a positive feedback loop. H2S further enhances NO production via eNOS persulfidation. Altogether, these effects facilitate VEGF-induced sprouting angiogenesis. In addition, H2S inhibits mitochondrial respiration, which promotes glycolysis and ATP production, proliferation, and migration in hypoxic condition. Akt: Protein kinase B, EC: endothelial cells; ERK: Extracellular signal-regulated kinase; ETC: electron transport chain; GLUT: glucose transporter; IP3: inositol triphosphate; MKK: mitogen-activated kinase kinase; PI3K: phosphor-inositol 3 kinase; PKC: protein kinase C; VEGFR2: Vascular endothelial growth factor receptor 2; NO: nitric oxide; eNOS: endothelial NO synthase; persulf: perfulfidation; EC: endothelial cells; CSE: cystathionine γ-lyase.
Ijms 24 09955 sch005
Scheme 6. H2S decreases intimal hyperplasia. H2S decreases the activity of the MAPK and mTOR pathways, which correlates with reduced VSMC proliferation and migration. H2S inhibits the microtubule polymerization leading to an arrest of cell-cycle inhibition of proliferation and migration of VSMC. H2S also reduces MMP2 expression and ECM degradation, inhibiting VSMC migration from the media to the intima. MEK1/2: mitogen-activated protein kinases; ERK: Extracellular signal-regulated kinase; NFκB: Nuclear factor kappa B; mTOR: mammalian target of rapamycin; MMP2: matrix metalloproteinase-2; TLR4: toll-like receptor 4; GPCR: G protein-coupled receptors; IGF-1: insulin-like Growth Factor 1; IGF-1R: IGF-1 receptor; IRS-1: insulin receptor substrate 1; PDGF-BB: Platelet-derived growth factor BB; FGF: fibroblast growth factor; EGF: epidermal growth factor; TNFα: tumour necrosis factor alpha; IL-1β: interleukin-1 beta; MCP-1: monocyte chemoattractant protein-1; ECM: extracellular matrix; EC: endothelial cells; VSMC: vascular smooth muscle cell; CSE: cystathionine γ-lyase.
Scheme 6. H2S decreases intimal hyperplasia. H2S decreases the activity of the MAPK and mTOR pathways, which correlates with reduced VSMC proliferation and migration. H2S inhibits the microtubule polymerization leading to an arrest of cell-cycle inhibition of proliferation and migration of VSMC. H2S also reduces MMP2 expression and ECM degradation, inhibiting VSMC migration from the media to the intima. MEK1/2: mitogen-activated protein kinases; ERK: Extracellular signal-regulated kinase; NFκB: Nuclear factor kappa B; mTOR: mammalian target of rapamycin; MMP2: matrix metalloproteinase-2; TLR4: toll-like receptor 4; GPCR: G protein-coupled receptors; IGF-1: insulin-like Growth Factor 1; IGF-1R: IGF-1 receptor; IRS-1: insulin receptor substrate 1; PDGF-BB: Platelet-derived growth factor BB; FGF: fibroblast growth factor; EGF: epidermal growth factor; TNFα: tumour necrosis factor alpha; IL-1β: interleukin-1 beta; MCP-1: monocyte chemoattractant protein-1; ECM: extracellular matrix; EC: endothelial cells; VSMC: vascular smooth muscle cell; CSE: cystathionine γ-lyase.
Ijms 24 09955 sch006
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bechelli, C.; Macabrey, D.; Deglise, S.; Allagnat, F. Clinical Potential of Hydrogen Sulfide in Peripheral Arterial Disease. Int. J. Mol. Sci. 2023, 24, 9955. https://doi.org/10.3390/ijms24129955

AMA Style

Bechelli C, Macabrey D, Deglise S, Allagnat F. Clinical Potential of Hydrogen Sulfide in Peripheral Arterial Disease. International Journal of Molecular Sciences. 2023; 24(12):9955. https://doi.org/10.3390/ijms24129955

Chicago/Turabian Style

Bechelli, Clémence, Diane Macabrey, Sebastien Deglise, and Florent Allagnat. 2023. "Clinical Potential of Hydrogen Sulfide in Peripheral Arterial Disease" International Journal of Molecular Sciences 24, no. 12: 9955. https://doi.org/10.3390/ijms24129955

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