Next Article in Journal
Demyelination in Multiple Sclerosis: Reprogramming Energy Metabolism and Potential PPARγ Agonist Treatment Approaches
Next Article in Special Issue
Annexins in Glaucoma
Previous Article in Journal
Antibacterial Efficacy of Silver Nanoparticles on Endometritis Caused by Prevotella melaninogenica and Arcanobacterum pyogenes in Dairy Cattle
Previous Article in Special Issue
Annexin A1: Uncovering the Many Talents of an Old Protein
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Potential of Annexin A1 in Ischemia Reperfusion Injury

1
Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71130, USA
2
Department of Neurology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71130, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2018, 19(4), 1211; https://doi.org/10.3390/ijms19041211
Submission received: 15 March 2018 / Revised: 3 April 2018 / Accepted: 4 April 2018 / Published: 16 April 2018

Abstract

:
Cardiovascular disease (CVD) continues to be the leading cause of death in the world. Increased inflammation and an enhanced thrombotic milieu represent two major complications of CVD, which can culminate into an ischemic event. Treatment for these life-threatening complications remains reperfusion and restoration of blood flow. However, reperfusion strategies may result in ischemia–reperfusion injury (I/RI) secondary to various cardiovascular pathologies, including myocardial infarction and stroke, by furthering the inflammatory and thrombotic responses and delivering inflammatory mediators to the affected tissue. Annexin A1 (AnxA1) and its mimetic peptides are endogenous anti-inflammatory and pro-resolving mediators, known to have significant effects in resolving inflammation in a variety of disease models. Mounting evidence suggests that AnxA1, which interacts with the formyl peptide receptor (FPR) family, may have a significant role in mitigating I/RI associated complications. In this review article, we focus on how AnxA1 plays a protective role in the I/R based vascular pathologies.

1. Introduction

Cardiovascular diseases (CVD) are the main cause of morbidity and mortality worldwide and were solely responsible for the death of about 17.7 million people in 2015, with the vast majority being due to either ischemic heart disease (IHD) or cerebrovascular disease [1]. This has led to the exponential increase in healthcare costs with estimated increase in total medical care costs in United States (US) alone rising from $396 billion to $918 billion between the years 2012 and 2030. Additionally, the burden of CVDs has increased tremendously in developing countries and are predicted to overtake infectious disease as the leading cause of mortality by the year 2020 [2].
Many acute cardiovascular events, such as acute ischemic stroke (AIS) and myocardial infarction (MI), are due to, e.g., unhealthy vascular states, resulting from prolonged atherosclerosis, coronary artery disease, diabetes, uncontrolled hypertension, and aging [1]. Additional studies have suggested behavioral, psychosocial including bereavement [3] and environmental factors can also trigger and contribute to acute cardiovascular events [4].
Both AIS and MI are life-threatening conditions which require prompt treatment consisting of rapid restoration of blood flow followed by subsequent management consisting of anti-inflammatory strategies and neuro- and cardio-protection for secondary prevention [5,6]. However, restoration of blood supply and re-oxygenation paradoxically lead to further exaggeration of tissue injuries and tissue destruction (termed ischemia–reperfusion injury (I/RI)). To restore homeostasis, it is essential that the ensuing inflammatory and thrombotic environment is resolved. Resolution involves a tightly orchestrated series of active events and is now accepted as one of the four major outcomes for acute inflammation, the others being progression to chronic inflammation, scarring, and fibrosis [7,8,9]. Several anti-inflammatory mediators which have pro-resolving properties have been proposed over the years, such as resolvin D1 (RvD1), RvD2, RvE1, maresins, protectin D1 and lipoxin A4 (LXA4) [10,11]. Another known anti-inflammatory and pro-resolving mediator, which is the focus of this review, is the glucocorticoid-regulated protein Annexin A1 (AnxA1). AnxA1 has a particular appeal for drug discovery programs as it is founded on endogenous anti-inflammatory molecular pathways and as such is attractive because it can mimic the body’s own pro-resolution effects while potentially having fewer side effects than existing therapeutic agents [7]. This review aims to highlight the issues associated with I/RI. We also focus on AnxA1, its history, mechanism of action and its role in I/RI in different organs including brain, heart, kidney and gastrointestinal tract. Finally, we discuss the potential of AnxA1 in drug discovery programs for the treatment of I/RI.

2. Ischemia Reperfusion Injury (I/RI)

I/RI is a fundamental vascular pathobiological paradigm that contributes to the pathophysiology of various acute CVDs such as IHD and AIS, along with other pathophysiological responses such as transplantation and acute kidney injury (AKI). I/RI occurs in two distinct phases: the initial ischemic insult caused by vascular occlusion, and the subsequent reperfusion phase manifested by a profound local and systemic inflammatory response (called “reperfusion injury”) [12,13]. The term “ischemia” refers to deficient blood supply to tissues due to obstruction of the arterial inflow. The severity of cell dysfunction is influenced by the extent and duration of obstruction. Treatment for ischemic events is always restoration of blood flow to the affected organ at the earliest time possible (with some organs such as the brain being far more susceptible to damage: “time is brain”). However, reperfusion itself can induce and exaggerate tissue injury because it results in a gamut of inflammatory mediators including reactive oxygen species (ROS), increased cell adhesion molecules (e.g., intracellular adhesion molecule-1 (ICAM-1)), heightened production of lipid mediators including leukotriene B4 (LTB4) and platelet activating factor (PAF) [14].
A wide range of pathological processes contribute to I/R-associated tissue damage. Prolonged occlusion triggers depletion of adenosine triphosphate (ATP) and decreases the intracellular pH as a result of anaerobic metabolism and lactate accumulation [13]. ATP depletion inactivates ATPases (e.g., Na+/K+ ATPase), reduces active calcium ion (Ca2+) efflux and limits the reuptake of Ca2+ by the endoplasmic reticulum (ER), thereby producing calcium overload in the cell. These changes are accompanied by opening of the mitochondrial permeability transition (MPT) pore, which dissipates the mitochondrial membrane potential and further impedes ATP production. Depletion of ATP also leads to accumulation of xanthine oxidase (XO) which is formed from xanthine dehydrogenase (XD) under hypoxic conditions, allowing for a burst of superoxide and hydrogen peroxide production and initiating the overall I/RI [15]. The re-oxygenation in reperfusion phase enhance the production ROS. During ischemic phase, electron-transport enzymes, including mitochondrial respiratory chain, are damaged. Therefore, incomplete oxidation of molecular oxygen occurs leading to generation of more ROS and exaggerating tissue injury [16].
An immune response elicits upon reperfusion and consists of both innate and adaptive immune systems. The innate immune response involves signaling events through pattern recognition molecules (especially toll like receptors (TLRs)). Ligands such as components of damaged tissue binding to TLRs lead to the activation of downstream signaling pathways, including NF-kappa-B (NF-κB), mitogen activated protein kinase (MAPK) and type I interferon pathways, resulting in the induction of pro-inflammatory cytokines and chemokines [17]. Upon reperfusion, the ischemic area acquires multiple activated leukocyte types (e.g., neutrophils, monocytes, and lymphocytes), dominated initially by neutrophil infiltration. I/RI also activates antigen-specific T cells by mechanisms that are not yet well characterized. Recent evidence suggests both antigen-specific and independent mechanisms of activation where T cells accumulate at sites of ischemia and reperfusion [18,19]. Multiple studies have shown that enhanced generation of oxidants results in the activation and deposition of complement and phospholipase A2 (PLA2)-mediated production of LTB4 and platelet activating factor (PAF). Furthermore, there is abundant evidence suggesting the role of platelets in I/RI [20] by mediating microvascular thrombosis as well as ensuing inflammation through platelet–platelet as well as platelet–leukocyte interactions. These communications lead to the release of a broad range of pro-inflammatory molecules including high-mobility group box 1 protein (HMGB1), cell differentiation 40 ligand (CD40L), PolyP, and interleukin-1 alpha/beta (IL-1α/β) furthering the thrombo-inflammatory environment and contributing in both innate and adaptive immune responses [21].

3. Annexin A1

Annexin A1 (AnxA1) is a 37-kDa member of superfamily of thirteen annexin proteins, comprising 346 amino acids with at least twelve distinct Ca2+ and phospholipid binding proteins [7]. This member of the superfamily was aptly named AnxA1 due to its capability to “annex” phospholipid membranes [7]. The protein itself was discovered in the 1970s and 1980s by its ability to inhibit PLA2 activity and eicosanoid synthesis [7]. Further anti-inflammatory actions were seen in guinea pig lung models, glucocorticoid (GC)-treated rat models and rat medulla cell experiments [7]. AnxA1 has a unique N-terminal region (active biological portion) embedded within its core, which is only exposed under elevated Ca2+ levels (≥1 mM). The N-terminal region of each member of the annexin superfamily is believed to be the “fingerprint”, endowing its biological activity, however there are other regions of interest in the core protein which are known to also possess anti-inflammatory properties, for instance, Antifammin-2 [7,22].
AnxA1 is abundantly present in cells of myeloid origin such as neutrophils, monocytes and macrophages. Under resting conditions, AnxA1 is present within the cytoplasm and is only extruded upon cell activation [7]. The molecular mechanisms responsible for secretion vary from cell to cell, e.g., in neutrophils, AnxA1 resides in the gelatinase granules and is released upon neutrophil activation (e.g., neutrophil–endothelial interaction), attaching itself to the outer leaflet of the plasma membrane and undergoing further conformational change to expose the N-terminal region [7]. In the case of macrophages, AnxA1 is released via the ATP-binding cassette (ABC) transporter system [7]. AnxA1 is regulated by GCs, with GCs increasing both the AnxA1 gene and also its secretion from existing intracellular pools by stimulating protein kinase C (PKC) activity [7].
The anti-inflammatory and pro-resolving effects of this 37 kDa protein are mediated by the shedding of l-selectin resulting in decreased neutrophil adhesion to the endothelium and restricting transmigration [7]. Pharmacological intervention of AnxA1 has been shown to decrease neutrophil rolling and adhesion to endothelium while increasing detachment of adherent and inducing neutrophil apoptosis [23]. Perretti and Flower (1993) demonstrated that AnxA1 attenuated IL-1 and IL-8 induced neutrophil migration into the murine air pouch [24]. Additionally, Getting et al. showed that both endogenous and exogenous AnxA1 were able to inhibit the neutrophil and monocyte recruitment in murine peritoneal cavity [25]. These findings suggest that AnxA1 retains its anti-inflammatory action irrespective of stimuli. Furthermore, AnxA1 may further undergo changes such as cleavage, induced by neutrophil specific proteases (e.g., Cathepsin G (CG) [26], Neutrophil elastase (NE) and Proteinase-3 [27]), although it is unknown as to whether the cleavage process occurs to inactivate AnxA1 and produce homeostasis, or to produce bioactive fragments which can act as a pro-drug [7].

AnxA1 and the Formyl Peptide Receptors (FPRs)

AnxA1 and its mimetic peptides, such as the N-terminal derived Ac2-26, bind to the formyl peptide receptor (FPR) family of seven transmembrane G-protein-coupled receptors (GPCRs) [28]. Various cell types express FPRs, especially myeloid cells, e.g., neutrophils and monocytes. Three FPR members exist in humans and they are termed: FPR1, FPR2/ALXR (also known as the LXA4 receptor), and FPR3. FPR2/ALXR shares 69% amino acid sequence homology with FPR1, and FPR3 shares 56% amino acid homology to FPR1 and 72% to FPR2/ALX [7,29]. In mice, the FPR family is more complex, consisting of at least eight members. Mouse Fpr1 shares 77% sequence homology with human FPR1, and Fpr2 has 76% homology to FPR2/ALXR [29].
The FPRs are primarily coupled to G proteins (GIα2, GIα3). Upon the binding of a ligand, such as formyl-Met-Leu-Phe (fMLP), G proteins are activated and trigger the release of several second messengers such as Ca2+ from intracellular stores, through activation of phospholipase C (PLC), PLD and PLA2 [30]. Neutrophils predominantly sense inflammatory stimuli via FPRs to perform both pro- as well as anti-inflammatory functions, depending upon the pathophysiological status and ligand binding. In inflammatory states, neutrophil FPRs participate in various biological functions including chemotaxis, degranulation, ROS production, promoting neutrophil–platelet interactions [31], and enabling apoptosis and phagocytosis [32]. The ability of FPRs to perform such wide range of biological functions is due to their ability to interact with multitude of agonists and antagonists, ranging from formylated and non-formylated proteins/peptides to small molecular weight compounds, e.g., fMLP, His-Phe-Tyr-Leu-Pro-Met (HFYLPM) (chemoattractants for monocytes and neutrophils), AnxA1, and HIV envelope proteins gp41 and gp120 [30]. A detailed description of the FPRs, their ligands and biological functions is given in Table 1 and Table 2.
Among the FPR members, FPR2/ALX is the most versatile and can interact with a multitude of ligands resulting in both anti-inflammatory and pro-resolving (AnxA1 and LXA4) as well as pro-inflammatory (serum amyloid A (SAA) and cathelicidin) functions (Table 2). These diverse effects also seem to be partly due to the ability of FPR2/ALX to facilitate biased agonism and enable different dimerization states after ligand binding [32,33]. Binding of SAA and/or the cathelicidin-associated antimicrobial peptide leucine-37 (LL37) to FPR2/ALX results in neutrophil NF-κB activation, cytokine release, increased neutrophil infiltration and lifespan [32]. However, binding of anti-neuronal nuclear antibody (AnnA1) inhibits neutrophil infiltration, promotes neutrophil apoptosis, and pushes macrophages towards a less pro-inflammatory phenotype, increasing the rate of phagocytosis by macrophages [32]. Ongoing studies are showing that FPR1 signaling is associated with neutrophil oxidative burst by inducing a rapid and reversible increase in the mitochondrial membrane potential within neutrophils [32]. In addition, mitochondrial-derived FPR1 ligands also function as chemotactic damage-associated molecular pattern molecules (DAMPs also known as alarmins or danger signals), activating the innate immune system [34].
The function of FPR3 is less well characterized, although it is expressed on eosinophils, monocytes, macrophages, and dendritic cells. Evidence suggests that FPR3 plays role in allergic reaction and dendritic cell maturation [35].

4. Acute Ischemic Stroke (AIS)

Cerebrovascular disease is the second most common cause of death after IHD, the third leading cause of disability-adjusted life-years (DALYs) and accounted for eight million deaths worldwide in 2013 [1,5]. AIS is the most common manifestation of cerebrovascular disease with limited therapeutic options to date [5], mainly due to complex pathophysiology. AIS evolves locally due to underlying atherogenesis and vasculopathy (thrombotic occlusion), or from a distant embolus arising from the heart (embolic occlusion) [36]. These events result in activation of an ischemic cascade, culminating in sudden tissue hypoxia, cellular dysfunction, cell death and subsequent disruption of blood–brain barrier [37]. The main goal in AIS management is rapid recanalization of the occluded vessel to restore blood supply and limit focal ischemic insult and global hypoxic injury. However, various animal models have demonstrated that reperfusion can actually result in tissue injury at both molecular and cellular levels [37] as translated into recurrent strokes seen in many patients [38]. As such, there are several strategies currently under development to generate neuroprotective agents to prevent I/RI and irreversible brain injury in AIS [38].
I/RI plays a significant detrimental role in AIS resulting in widespread inflammation due to immune responses. Ischemia results in the complex accumulation of various cellular mediators including activated leukocytes (neutrophils, monocytes and lymphocytes), activated platelets, alongside activation of resident cells (e.g., microglia and astrocytes) superoxide generation and cytokines (tumor necrosis factor-α (TNF-α) and interleukin-1 beta (IL-1β)) production [39]. Under hypoxia, the deficiency of vasodilator in particular endothelial nitric oxide (NO) has been shown to enlarge stroke size [40]. TLRs have also been shown to play a significant role in cerebral I/RI especially mediating responses through microglial cells, and TLR4 has been demonstrated to upregulate the expression of cellular adhesion molecules (CAMs and selectins), facilitating neuronal injury [41]. The activation of microglia and macrophages increases the expression of, e.g., ICAM-1, P and E-selectin, which enhance leukocyte rolling and sticking to the vessel surfaces [40]. Additionally, there is a crosstalk between thrombosis and inflammation in cerebral ischemia resulting in a thrombo-inflammatory state [42]. Subsequently, restoration of blood flow results in cellular and molecular responses involving innate as well as adaptive arms of immunity, activation of the complement system, and the coagulation system. These responses lead to a further damage of the affected tissue. Additional injury is caused by the production of ROS, and the involvement of other inflammatory cells such as dendritic cells [37,43].
Neutrophils play a huge role as these are first immune cells to infiltrate the ischemic region of brain [44] and accumulate within six hours of reperfusion and increase the tissue infarct size between six and twenty-four hours (the time of maximal infarct related closely with the time course of neutrophil infiltration) [44]. During reperfusion, in addition to interacting with other cells, e.g., endothelial cells and platelets, neutrophils also produce many pro-thrombotic mediators such as neutrophils extracellular traps (NETs) and serine proteases, e.g., CG and NE, which promote thrombus formation. Furthermore, matrix metalloproteinase-9 (MMP-9) and the surface integrin receptor Mac-1 (CD11b/CD18 or macrophage-1 Ag), expressed by neutrophils play significant roles in compromising blood–brain barrier (BBB) and accelerating thrombus formation by binding to fibrinogen, von Willebrand factor (vWF), and glycoprotein Ib (GPIb), respectively [42,45].

The Role of Annexin A1 in AIS

AnxA1 has been shown to act in various ways to mitigate cerebral I/RI. Our work has shown a significant effect of AnxA1 (and its peptide mimetics) on diminishing leukocyte adhesion and trafficking (mainly by acting in an in an autocrine/paracrine fashion to decrease leukocyte–endothelial interactions) [46], inhibiting the release of pro-inflammatory and pro-thrombotic cytokines, and regulating neutrophil–platelet interactions [7,31,47]. Human recombinant AnxA1 and its peptide mimetics have shown to markedly reduce the lesion size, clinical score and markers of leukocyte infiltration in murine middle cerebral artery occlusion (MCAo) model [31,46,47]. Recently, we showed that exogenous administration of AnxA1 N-terminal peptide Ac2-26 attenuated neutrophil and platelet activation and neutrophil–platelet aggregation in the murine cerebral microvasculature after induction of cerebral I/RI in mice [31]. Ac2-26 has also been shown to be protective in rats, as demonstrated by intracerebroventricular administration of the peptide reducing infarct size and cerebral edema two hours post cerebral ischemia [48]. McArthur et al. demonstrated that animals lacking the AnxA1 receptor Fpr2/ALX (i.e., Fpr2/ALX knock-out or Fpr2/ALX−/− mice) showed markedly greater BBB leakage post-ischemia than their wild-type (WT) counterparts [49]. Additionally, AnxA1 plays a role in mediating the permeability of the BBB. Cristante et al. showed that AnxA1 knock-out (AnxA1−/−) mice possess a disorganized actin cytoskeleton due to increase in activity of RhoA small GTPase and a disruption of occludin and VE-Cadherin [50]. Furthermore, it is not just blood cells that express Fpr2/ALX, but also brain resident cells such as Microglia [51] and as such are potential targets for the anti-inflammatory and pro-resolving effects of Ac2-26 and its parent compound [30].

5. Ischemic Heart Disease (IHD)

IHD is the leading cause of mortality with an estimated 8.2 million deaths worldwide in 2013 [1]. Despite significant improvement over the years in survival rates (due to advances in reperfusion therapy), secondary heart failure, due to ensuing inflammation, is a major cause of disease burden after myocardial infarction. Overproduction of ROS [52], cardiac tissue remodeling and ensuing pro-inflammatory immune response are the main causes of MI/RI [33]. The pharmacological administration of anti-oxidants and calcium channel blockers has shown the promising results under experimental settings, however both interventions failed to achieve success in clinical studies [53].
If MI lasts more than twenty minutes, cardiomyocyte death is initiated in the subendocardium, expanding towards epicardium [53]. The depletion of ATP under ischemia due to halt in oxidative phosphorylation can inhibit myocardial contractile function [53]. Ischemia leads to Ca2+ overload, which can cause damage to ultrastructure of myocardial [54]. Initially, it was thought that reperfusion can limit infarct size and preserve left ventricular (LV) systolic function, however, with growing scientific evidence, it was proven that reperfusion can itself induce cardiomyocyte death [53]. Reperfusion in IHD results in a massive outpouring into the ischemic tissue of activated leukocytes (especially neutrophils) and other mediators e.g., complement fragments, ROS and pro-inflammatory cytokines, all of which promote further injury and cardiomyocyte death [33]. Several interventions have been shown to reduce infarct size up to 50% under experimental MI/R conditions. These interventions include administration of biologics to inhibit P-selectin, CD11, CD18 and ICAM-1 and pharmacologic inhibition of complement activation and leukocyte depletion [55].
Neutrophils play a central role in the MI/RI. They are the first cells to be summoned, resulting in accelerated pro-inflammatory responses including production of inflammatory mediators (e.g., CG, NE and MMP-9 [56]) and ROS, leading to microvascular injury, cardiomyocyte death and extracellular matrix (ECM) degradation and remodeling [57]. Neutrophil activation in I/RI promotes the expression of adhesion molecules, leading to adherence of neutrophils to the endothelium followed by transmigration, and direct interaction with the cardiac myocytes [57].

The Involvement of AnxA1 in IHD

AnxA1 and its N-terminal peptides have both been shown to be cardioprotective in various I/RI models [58,59]. D’Amico et al. demonstrated a decrease in infarct size (50%) upon infusion of recombinant AnxA1 and the protective effects of AnxA1 were further confirmed by La et al. as evidenced by the administration of the AnxA1 N-terminal peptide, Ac2-26, to decrease infarct size and reduce MPO and IL-1β content in infarcted hearts [58]. Treatment with AnxA1 also attenuated loss of fiber organization, decreased MPO activity, reduced TNF-α and macrophage inflammatory protein (MIP-1α) levels and leukocyte extravagation in cardiac tissues [60]. Peptide Ac2-26 has been shown to preserve cardiomyocyte contractile function and reduce cardiac myocyte injury, with protection, at least in part, being associated with activation of PKC, p38-MAPK and KATP channels [61]. Additionally, Qin et al. demonstrated that exogenous administration of Ac2-26 at the onset of reperfusion increased cardiomyocyte viability and recovery of LV function, which was associated with FPR1/Akt signaling [62].
Hematopoietic progenitor stem cell (HPSC) mobilization and differentiation has been shown to be regulated by AnxA1 [63], e.g., AnxA1 deficiency results in increased cardiac necrosis, inflammation, hypertrophy and fibrosis resulting in exaggerated cardiac infarct size after eight days of myocardial infarction [63]. These effects were linked to a greater expansion and altered mobilization of HPSC, with increased circulating neutrophils and platelets and altered macrophage inflammatory phenotype and function [63].
More recently, there has been a large focus on the pharmacological concept of biased-agonism (multiple active conformations of a receptor exist and elicit distinct signals yielding to multiple functional outcomes). The small-molecule FPR1/FPR2/ALX agonist, Compound 17b (Cmpd17b), has been shown to have biased agonism, by enabling FPR based pro-resolving biology without changing FPR1/2-mediated calcium mobilization, hence providing superior cardio-protection. Cmpd17b, similar to AnxA1, attenuates both early and late inflammatory responses after reperfusion in MI [64], with ERK1/2–Akt kinases seeming to play a significant role [33,64].

6. I/RI in the Kidney

Kidney transplantation is the leading cause of kidney I/RI. At the end of 2016, there were approximately 100,000 people waiting for a kidney transplant. Due to greater availability, the vast majority of kidney transplants which take place use organ(s) from a cadaveric donor. For instance, out of 17,107 kidney transplants that took place in 2014, 11,570 organs came from cadaveric donors and only 5537 came from living donors [65]. However, cadaveric donor kidneys are more susceptible to I/RI due to prolonged ischemia under cold conditions [66]. In the case of living donors, although ischemic period is shorter, the organ is still subjected to warm ischemia during harvesting which may also result in I/RI [66]. Finally, a more severe injury occurs during the reperfusion phase in the recipient due to the activation of innate and adaptive immune responses. Indeed, many renal transplant recipients are under immunosuppressant therapy to prevent graft dysfunction. Figures show that 3% of kidney transplants will fail within a month of transplantation, 7% within a year and 17% within three years. Furthermore, each year, more than 20% of total kidney transplants are re-transplanted due to failure of the transplanted kidney [67]. Therefore, there is an urgent unmet clinical need to find better therapies to prevent graft rejection.
Renal I/RI is characterized by a decrease in glomerular filtration rate (GFR, which is the volume of the fluid filtered from the kidney each minute). Ischemia leads to the depletion of ATP which in turn inhibits the activity of tubular Na +K+ ATPase and the redistribution of the Na+/H+ exchanger. Both conditions cause a decrease in the reabsorption of Na+ by tubular cells. Due to increased Na+ filtration, GFR is limited via tubuloglomerular feedback mechanism to avoid Na+ and water loss [68]. Additionally, ROS production is increased during I/RI causing renal tubule injury via oxidation of proteins and lipids on membrane, DNA damage and thereby triggering apoptosis of tubular epithelial cells which can shed into tubular lumen causing tubular obstruction [69,70]. The paracellular diffusion of water, ions and macromolecules becomes unregulated upon the loss of tubular cells and this increases the back-leak into the interstitium and blood vessels, which also decreases GFR. Thus, it can be said that GFR can also be decreased by the synergetic effect of cell surface area and tubular obstruction. Additionally, apoptosis leads to the decrease in cell–cell interactions increasing the vascular permeability [69,71]. The depletion of ATP also inhibits the activity of Ca2+ ATPase causing an increase in intracellular calcium levels which can induce cytoskeletal damage and increase in vascular permeability [69].
Inflammation plays a major role in renal I/RI due to enhanced production of pro-inflammatory cytokines, e.g., IL-6 and TNF-α, which have been shown to work mainly through JAK/STAT signaling pathways such that inhibition of the JAK/STAT signaling pathway has been shown to improve kidney I/RI outcomes. Other cytokines, e.g., MCP-1, IL-1β, IL-6, and IL-18, are released upon renal tubular cell activation and further the damage due to I/RI [72]. As with other I/RIs, in renal I/RI, neutrophils are the first cells to respond to the injury site, which results in the production of multiple mediators including NE, tissue type plasminogen activator, hepatocyte growth factor and CD44 expression. Neutrophils also cause renal microvasculature obstruction as they bind to endothelial cells and platelets. Additionally, adhesion molecules, e.g., ICAM-1 and P-selectin, which are major regulators of neutrophil recruitment have been shown to be involved in kidney I/RI. More recently, Kelly et al. showed that kidneys from ICAM-1−/− mice displayed protection against I/RI, which was further confirmed by blocking ICAM-1 with an ICAM-1 specific antibody [73,74]. Takada et al. showed that soluble P-selectin ligand decreased the neutrophil transmigration in rat ischemic kidneys [75].

The Association of AnxA1 in Renal I/RI

The role of AnxA1 in kidney I/RI is in its nascent stage. However, some studies have shown that AnxA1 may exert a protective role, e.g., in a rat model of renal I/RI, exogenous administration of peptide Ac2-26 reduced: (a) tubular necrosis; (b) the influx of phagocytic cells (neutrophil and monocytes); (c) sodium and potassium excretion; and (d) GFR, thereby improving functional recovery and outcome [76].
Cyclosporine is an immunosuppressant and is used to avoid organ transplant rejection. The major side effect of Cyclosporine treatment is nephrotoxicity, as the compound decreases renal blood flow and increases renal vascular resistance (both common conditions in renal I/RI). However, exogenous administration of Ac2-26 has been shown to reverse these side effects [77].
Diabetes is metabolic syndrome which affects many organs including the kidneys, with 30–40% of diabetic patients developing nephropathy [78]. AnxA1 has been shown to afford protection in murine diabetic nephropathy by decreasing p38, ERK and JNK, and activating Akt signaling, suggesting that AnxA1 could be a potential therapeutic strategy for treating renal dysfunction caused by diseases such as diabetes [79].

7. I/RI in Gastrointestinal (GI) System

I/RI in the GI system is mainly seen with abdominal and thoracic vascular surgery, but it can also be associated with small bowel transplantation and hemorrhagic shock [80]. Most of these GI complications lead to mesenteric ischemia and I/RI. The mortality rate associated with mesenteric I/RI is in the range of 40–60% [81]. Enterocytes are the main intestinal cells and are resistant to short term hypoxia, however prolonged hypoxia can cause irreversible cell death resulting in the production of various DAMPs, e.g., nucleic acids and heat shock proteins. Ischemia causes inactivation of enzymes such as cytochrome oxidase and superoxide dismutase, so upon re-oxygenation, oxidative phosphorylation does not occur, resulting in ROS production which can further trigger apoptosis of enterocytes further causing intestinal barrier disruption. There is also increase in intracellular calcium concentration leading to changes in cytoskeleton, which further damages cell–cell junctions, thereby increasing vascular permeability [82].
Systemic inflammation is a significant component of intestinal I/RI. The loss of barrier function facilitates movement of microbiota and their products from the gut to the blood stream. The molecular patterns associated with these microbes and their products, known as pathogen-associated molecules pattern (PAMPs). PAMPS and DAMPs can bind TLRs on immune cells, activating local inflammatory response such as production of cytokines, cell adhesion molecules, and chemokine receptors via induction of NF-κB. These steps lead to recruitment and infiltration of leukocytes causing tissue damage [82]. The inflamed intestinal mucosa also produces as whole plethora of different inflammatory cytokines and chemokines, e.g., TNF-α, IL-12, IL-15 and IL-23, which can significantly increase leukocyte infiltration and exacerbate inflammation and disease [83].

The Protective Role of AnxA1 in GI-Associated I/RI

AnxA1 has been shown to play a protective role in gastrointestinal I/RI. We have shown that intravenous administration of Ac2-26 inhibits leukocyte adhesion and emigration in a mouse mesenteric I/RI model, as induced by clamping of the superior mesenteric artery, followed by reperfusion [84]. The loss of epithelial cell barrier is major characteristic of intestinal I/RI. The effect of AnxA1 or its peptides on epithelial cell barrier has not been studied in intestinal I/RI models but they have afforded protection in other gastrointestinal inflammatory murine models including colitis and gastric ulcers. Leoni et al. demonstrated that Ac2-26, when delivered locally in the intestine using polymeric nanoparticles, enhanced the epithelial cell restoration in colitis [85]. Martin et al. showed the positive effect of peptide Ac2-26 in enhancing the healing process of gastric mucosal damage and reducing the ulcer areas. Furthermore, this same group also demonstrated that hemorrhagic lesions were greater in AnxA1−/− mice upon induction of gastric ulcers [86]. Leoni et al. suggested that AnxA1 exhibits an FPR1/NOX1-dependent positive role in intestinal wound healing: upon binding to FPR1, AnxA1 activates Src and downstream NOX-1 [87]. ROS produced by NOX-1 causes rapid and reversible oxidation and subsequent inactivation of phosphatase PTEN and PTP-PEST. This inactivation of phosphatases leads to activation of focal adhesion protein involved in cell movement, FAX and Paxillin, thereby increasing the intestinal epithelial cell movement and wound repair [87].
Intestinal I/RI is also a major cause of acute lung injury (ALI). Treatment with exogenous Ac2-26 has been shown to reduce pulmonary vascular leakage, decrease neutrophil infiltration and MPO content in lung tissues following intestinal I/RI, possibly by inducing release of anti-inflammatory cytokine IL-10 and decreasing the release of pro-inflammatory cytokine TNF-α [88]. Babbin et al. also showed in a dextran-sulfate sodium (DSS)-induced colitis model that AnxA1 was able to regulate intestinal mucosal injury and inflammation via engagement with FPR2/ALX. In addition, AnxA1 deficient mice failed to regain weight and showed no improvement in disease activity index and mucosal injury upon withdrawal of disease in the DSS-induced colitis model [89], advocating the crucial and beneficial role of AnxA1.

8. Concluding Remarks

In summary, CVDs are the leading cause of morbidity and mortality worldwide, with MI and IS being the primary causes of death. The majority of CVD related complications are due to I/RI. Hence, it has become imperative to develop preventative as well as therapeutic strategies to mitigate these life-threatening complications. In this review, we focus on AnxA1 and its peptide mimetics as possible therapeutic strategies for the treatment of I/RI, based on their ability to alleviate thrombosis and inflammation and promote resolution (Figure 1). Indeed, drug development programs focused on endogenous anti-inflammatory and pro-resolving agents, such as AnxA1-based pharmacologic strategies, offer great therapeutic potential as they are devoid of metabolic side effects because they mimic the way inflammation naturally subsides in the body. However, the development of novel therapeutics should also be mindful of ascertaining not only if the drug is anti-inflammatory, but also if it is resolution-toxic (i.e., deranges or impairs timely and/or complete resolution), which could ultimately outweigh the overall therapeutic efficacy in treating I/RI and other inflammatory disorders.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Benjamin, E.J.; Blaha, M.J.; Chiuve, S.E.; Cushman, M.; Das, S.R.; Deo, R.; de Ferranti, S.D.; Floyd, J.; Fornage, M.; Gillespie, C.; et al. Heart Disease and Stroke Statistics-2017 Update: A Report From the American Heart Association. Circulation 2017, 135, e146–e603. [Google Scholar] [CrossRef] [PubMed]
  2. Stewart, J.; Manmathan, G.; Wilkinson, P. Primary prevention of cardiovascular disease: A review of contemporary guidance and literature. JRSM Cardiovasc. Dis. 2017, 6, 2048004016687211. [Google Scholar] [CrossRef] [PubMed]
  3. Carey, I.M.; Shah, S.M.; DeWilde, S.; Harris, T.; Victor, C.R.; Cook, D.G. Increased risk of acute cardiovascular events after partner bereavement: A matched cohort study. JAMA Intern. Med. 2014, 174, 598–605. [Google Scholar] [CrossRef] [PubMed]
  4. Mittleman, M.A.; Mostofsky, E. Physical, psychological and chemical triggers of acute cardiovascular events: Preventive strategies. Circulation 2011, 124, 346–354. [Google Scholar] [CrossRef] [PubMed]
  5. Hankey, G.J. Stroke. Lancet 2017, 389, 641–654. [Google Scholar] [CrossRef]
  6. O’Gara, P.T.; Kushner, F.G.; Ascheim, D.D.; Casey, D.E., Jr.; Chung, M.K.; de Lemos, J.A.; Ettinger, S.M.; Fang, J.C.; Fesmire, F.M.; Franklin, B.A.; et al. 2013 ACCF/AHA guideline for the management of ST-elevation myocardial infarction: A report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. Circulation 2013, 127, e362–e425. [Google Scholar] [CrossRef] [PubMed]
  7. Gavins, F.N.; Hickey, M.J. Annexin A1 and the regulation of innate and adaptive immunity. Front. Immunol. 2012, 3, 354. [Google Scholar] [CrossRef] [PubMed]
  8. Kumar, V.A.; Fausto, N. Robbins Pathologic Basis of Disease; Elsevier Saunders: Philadelphia, PA, USA, 1999. [Google Scholar]
  9. Kohli, P.; Levy, B.D. Resolvins and protectins: Mediating solutions to inflammation. Br. J. Pharmacol. 2009, 158, 960–971. [Google Scholar] [CrossRef] [PubMed]
  10. Serhan, C.N.; Chiang, N.; Van Dyke, T.E. Resolving inflammation: Dual anti-inflammatory and pro-resolution lipid mediators. Nat. Rev. Immunol. 2008, 8, 349–361. [Google Scholar] [CrossRef] [PubMed]
  11. Cash, J.L.; Norling, L.V.; Perretti, M. Resolution of inflammation: Targeting GPCRs that interact with lipids and peptides. Drug Discov. Today 2014, 19, 1186–1192. [Google Scholar] [CrossRef] [PubMed]
  12. Carden, D.L.; Granger, D.N. Pathophysiology of ischaemia-reperfusion injury. J. Pathol. 2000, 190, 255–266. [Google Scholar] [CrossRef]
  13. Kalogeris, T.; Baines, C.P.; Krenz, M.; Korthuis, R.J. Cell biology of ischemia/reperfusion injury. Int. Rev. Cell. Mol. Biol. 2012, 298, 229–317. [Google Scholar] [CrossRef] [PubMed]
  14. Kerrigan, C.L.; Stotland, M.A. Ischemia reperfusion injury: A review. Microsurgery 1993, 14, 165–175. [Google Scholar] [CrossRef] [PubMed]
  15. Sanada, S.; Komuro, I.; Kitakaze, M. Pathophysiology of myocardial reperfusion injury: Preconditioning, postconditioning, and translational aspects of protective measures. Am. J. Physiol. Heart Circ. Physiol. 2011, 301, H1723–H1741. [Google Scholar] [CrossRef] [PubMed]
  16. De Groot, H.; Rauen, U. Ischemia-reperfusion injury: Processes in pathogenetic networks: A review. Transplant. Proc. 2007, 39, 481–484. [Google Scholar] [CrossRef] [PubMed]
  17. Chen, G.Y.; Nuñez, G. Sterile inflammation: Sensing and reacting to damage. Nat. Rev. Immunol. 2010, 10, 826–837. [Google Scholar] [CrossRef] [PubMed]
  18. Satpute, S.R.; Park, J.M.; Jang, H.R.; Agreda, P.; Liu, M.; Gandolfo, M.T.; Racusen, L.; Rabb, H. The role for T cell repertoire/antigen-specific interactions in experimental kidney ischemia reperfusion injury. J. Immunol. 2009, 183, 984–992. [Google Scholar] [CrossRef] [PubMed]
  19. Shen, X.; Wang, Y.; Gao, F.; Ren, F.; Busuttil, R.W.; Kupiec-Weglinski, J.W.; Zhai, Y. CD4 T cells promote tissue inflammation via CD40 signaling without de novo activation in a murine model of liver ischemia/reperfusion injury. Hepatology 2009, 50, 1537–1546. [Google Scholar] [CrossRef] [PubMed]
  20. Xu, Y.; Huo, Y.; Toufektsian, M.C.; Ramos, S.I.; Ma, Y.; Tejani, A.D.; French, B.A.; Yang, Z. Activated platelets contribute importantly to myocardial reperfusion injury. Am. J. Physiol. Heart Circ. Physiol. 2006, 290, H692–H699. [Google Scholar] [CrossRef] [PubMed]
  21. Maiocchi, S.; Alwis, I.; Wu, M.C.L.; Yuan, Y.; Jackson, S.P. Thromboinflammatory Functions of Platelets in Ischemia-Reperfusion Injury and Its Dysregulation in Diabetes. Semin. Thromb. Hemost. 2018. [Google Scholar] [CrossRef] [PubMed]
  22. Kamal, A.M.; Hayhoe, R.P.; Paramasivam, A.; Cooper, D.; Flower, R.J.; Solito, E.; Perretti, M. Antiflammin-2 activates the human formyl-peptide receptor like 1. Sci. World J. 2006, 6, 1375–1384. [Google Scholar] [CrossRef] [PubMed]
  23. Sugimoto, M.A.; Vago, J.P.; Teixeira, M.M.; Sousa, L.P. Annexin A1 and the Resolution of Inflammation: Modulation of Neutrophil Recruitment, Apoptosis, and Clearance. J. Immunol. Res. 2016, 2016, 8239258. [Google Scholar] [CrossRef] [PubMed]
  24. Perretti, M.; Flower, R.J. Modulation of IL-1-induced neutrophil migration by dexamethasone and lipocortin 1. J. Immunol. 1993, 150, 992–999. [Google Scholar] [PubMed]
  25. Getting, S.J.; Flower, R.J.; Perretti, M. Inhibition of neutrophil and monocyte recruitment by endogenous and exogenous lipocortin 1. Br. J. Pharmacol. 1997, 120, 1075–1082. [Google Scholar] [CrossRef] [PubMed]
  26. Vago, J.P.; Tavares, L.P.; Sugimoto, M.A.; Lima, G.L.; Galvao, I.; de Caux, T.R.; Lima, K.M.; Ribeiro, A.L.; Carneiro, F.S.; Nunes, F.F.; et al. Proresolving Actions of Synthetic and Natural Protease Inhibitors Are Mediated by Annexin A1. J. Immunol. 2016, 196, 1922–1932. [Google Scholar] [CrossRef] [PubMed]
  27. Vong, L.; D’Acquisto, F.; Pederzoli-Ribeil, M.; Lavagno, L.; Flower, R.J.; Witko-Sarsat, V.; Perretti, M. Annexin 1 cleavage in activated neutrophils: A pivotal role for proteinase 3. J. Biol. Chem. 2007, 282, 29998–30004. [Google Scholar] [CrossRef] [PubMed]
  28. Walther, A.; Riehemann, K.; Gerke, V. A novel ligand of the formyl peptide receptor: Annexin I regulates neutrophil extravasation by interacting with the FPR. Mol. Cell 2000, 5, 831–840. [Google Scholar] [CrossRef]
  29. Dufton, N.; Perretti, M. Therapeutic anti-inflammatory potential of formyl-peptide receptor agonists. Pharmacol. Ther. 2010, 127, 175–188. [Google Scholar] [CrossRef] [PubMed]
  30. Gavins, F.N. Are formyl peptide receptors novel targets for therapeutic intervention in ischaemia-reperfusion injury? Trends Pharmacol. Sci. 2010, 31, 266–276. [Google Scholar] [CrossRef] [PubMed]
  31. Vital, S.A.; Becker, F.; Holloway, P.M.; Russell, J.; Perretti, M.; Granger, D.N.; Gavins, F.N. Formyl-Peptide Receptor 2/3/Lipoxin A4 Receptor Regulates Neutrophil-Platelet Aggregation and Attenuates Cerebral Inflammation: Impact for Therapy in Cardiovascular Disease. Circulation 2016, 133, 2169–2179. [Google Scholar] [CrossRef] [PubMed]
  32. Dorward, D.A.; Lucas, C.D.; Chapman, G.B.; Haslett, C.; Dhaliwal, K.; Rossi, A.G. The role of formylated peptides and formyl peptide receptor 1 in governing neutrophil function during acute inflammation. Am. J. Pathol. 2015, 185, 1172–1184. [Google Scholar] [CrossRef] [PubMed]
  33. Qin, C.; Yang, Y.H.; May, L.; Gao, X.; Stewart, A.G.; Tu, Y.; Woodman, O.L.; Ritchie, R.H. Cardioprotective potential of annexin-A1 mimetics in myocardial infarction. Pharmacol. Ther. 2015, 148, 47–65. [Google Scholar] [CrossRef] [PubMed]
  34. Wenceslau, C.F.; McCarthy, C.G.; Szasz, T.; Spitler, K.; Goulopoulou, S.; Webb, R.C.; Working Group on DAMPs in Cardiovascular Disease. Mitochondrial damage-associated molecular patterns and vascular function. Eur. Heart J. 2014, 35, 1172–1177. [Google Scholar] [CrossRef] [PubMed]
  35. Devosse, T.; Guillabert, A.; D’Haene, N.; Berton, A.; De Nadai, P.; Noel, S.; Brait, M.; Franssen, J.D.; Sozzani, S.; Salmon, I.; et al. Formyl peptide receptor-like 2 is expressed and functional in plasmacytoid dendritic cells, tissue-specific macrophage subpopulations, and eosinophils. J. Immunol. 2009, 182, 4974–4984. [Google Scholar] [CrossRef] [PubMed]
  36. Deb, P.; Sharma, S.; Hassan, K.M. PathoPhysiol.ogic mechanisms of acute ischemic stroke: An overview with emphasis on therapeutic significance beyond thrombolysis. Pathophysiology 2010, 17, 197–218. [Google Scholar] [CrossRef] [PubMed]
  37. Nour, M.; Scalzo, F.; Liebeskind, D.S. Ischemia-reperfusion injury in stroke. Interv. Neurol. 2013, 1, 185–199. [Google Scholar] [CrossRef] [PubMed]
  38. Onwuekwe, I.; Ezeala-Adikaibe, B. Ischemic stroke and neuroprotection. Ann. Med. Health Sci. Res. 2012, 2, 186–190. [Google Scholar] [CrossRef] [PubMed]
  39. Jin, R.; Yang, G.; Li, G. Inflammatory mechanisms in ischemic stroke: Role of inflammatory cells. J. Leukoc. Biol. 2010, 87, 779–789. [Google Scholar] [CrossRef] [PubMed]
  40. Moskowitz, M.A.; Lo, E.H.; Iadecola, C. The science of stroke: Mechanisms in search of treatments. Neuron 2010, 67, 181–198. [Google Scholar] [CrossRef] [PubMed]
  41. Arumugam, T.V.; Okun, E.; Tang, S.C.; Thundyil, J.; Taylor, S.M.; Woodruff, T.M. Toll-like receptors in ischemia-reperfusion injury. Shock 2009, 32, 4–16. [Google Scholar] [CrossRef] [PubMed]
  42. De Meyer, S.F.; Denorme, F.; Langhauser, F.; Geuss, E.; Fluri, F.; Kleinschnitz, C. Thromboinflammation in Stroke Brain Damage. Stroke 2016, 47, 1165–1172. [Google Scholar] [CrossRef] [PubMed]
  43. Felger, J.C.; Abe, T.; Kaunzner, U.W.; Gottfried-Blackmore, A.; Gal-Toth, J.; McEwen, B.S.; Iadecola, C.; Bulloch, K. Brain dendritic cells in ischemic stroke: Time course, activation state, and origin. Brain Behav. Immun. 2010, 24, 724–737. [Google Scholar] [CrossRef] [PubMed]
  44. Zhang, R.L.; Chopp, M.; Chen, H.; Garcia, J.H. Temporal profile of ischemic tissue damage, neutrophil response, and vascular plugging following permanent and transient (2H) middle cerebral artery occlusion in the rat. J. Neurol. Sci. 1994, 125, 3–10. [Google Scholar] [CrossRef]
  45. Rosell, A.; Cuadrado, E.; Ortega-Aznar, A.; Hernandez-Guillamon, M.; Lo, E.H.; Montaner, J. MMP-9-positive neutrophil infiltration is associated to blood-brain barrier breakdown and basal lamina type IV collagen degradation during hemorrhagic transformation after human ischemic stroke. Stroke 2008, 39, 1121–1126. [Google Scholar] [CrossRef] [PubMed]
  46. Smith, H.K.; Gil, C.D.; Oliani, S.M.; Gavins, F.N. Targeting formyl peptide receptor 2 reduces leukocyte-endothelial interactions in a murine model of stroke. FASEB J. 2015, 29, 2161–2171. [Google Scholar] [CrossRef] [PubMed]
  47. Gavins, F.N.; Dalli, J.; Flower, R.J.; Granger, D.N.; Perretti, M. Activation of the annexin 1 counter-regulatory circuit affords protection in the mouse brain microcirculation. FASEB J. 2007, 21, 1751–1758. [Google Scholar] [CrossRef] [PubMed]
  48. Relton, J.K.; Strijbos, P.J.; O’Shaughnessy, C.T.; Carey, F.; Forder, R.A.; Tilders, F.J.; Rothwell, N.J. Lipocortin-1 is an endogenous inhibitor of ischemic damage in the rat brain. J. Exp. Med. 1991, 174, 305–310. [Google Scholar] [CrossRef] [PubMed]
  49. McArthur, S.; Loiola, R.A.; Maggioli, E.; Errede, M.; Virgintino, D.; Solito, E. The restorative role of annexin A1 at the blood-brain barrier. Fluids Barriers CNS 2016, 13, 17. [Google Scholar] [CrossRef] [PubMed]
  50. Cristante, E.; McArthur, S.; Mauro, C.; Maggioli, E.; Romero, I.A.; Wylezinska-Arridge, M.; Couraud, P.O.; Lopez-Tremoleda, J.; Christian, H.C.; Weksler, B.B.; et al. Identification of an essential endogenous regulator of blood-brain barrier integrity, and its pathological and therapeutic implications. Proc. Natl. Acad. Sci. USA 2013, 110, 832–841. [Google Scholar] [CrossRef] [PubMed]
  51. Cui, Y.H.; Le, Y.; Gong, W.; Proost, P.; Van Damme, J.; Murphy, W.J.; Wang, J.M. Bacterial lipopolysaccharide selectively up-regulates the function of the chemotactic peptide receptor formyl peptide receptor 2 in murine microglial cells. J. Immunol. 2002, 168, 434–442. [Google Scholar] [CrossRef] [PubMed]
  52. Qin, C.; Yap, S.; Woodman, O.L. Antioxidants in the prevention of myocardial ischemia/reperfusion injury. Expert Rev. Clin. Pharmacol. 2009, 2, 673–695. [Google Scholar] [CrossRef] [PubMed]
  53. Hausenloy, D.J.; Yellon, D.M. Myocardial ischemia-reperfusion injury: A neglected therapeutic target. J. Clin. Investig. 2013, 123, 92–100. [Google Scholar] [CrossRef] [PubMed]
  54. Turer, A.T.; Hill, J.A. Pathogenesis of myocardial ischemia-reperfusion injury and rationale for therapy. Am. J. Cardiol. 2010, 106, 360–368. [Google Scholar] [CrossRef] [PubMed]
  55. Yellon, D.M.; Hausenloy, D.J. Myocardial reperfusion injury. N. Engl. J. Med. 2007, 357, 1121–1135. [Google Scholar] [CrossRef] [PubMed]
  56. Lindsey, M.; Wedin, K.; Brown, M.D.; Keller, C.; Evans, A.J.; Smolen, J.; Burns, A.R.; Rossen, R.D.; Michael, L.; Entman, M. Matrix-dependent mechanism of neutrophil-mediated release and activation of matrix metalloproteinase 9 in myocardial ischemia/reperfusion. Circulation 2001, 103, 2181–2187. [Google Scholar] [CrossRef] [PubMed]
  57. Jordan, J.E.; Zhao, Z.Q.; Vinten-Johansen, J. The role of neutrophils in myocardial ischemia-reperfusion injury. Cardiovasc. Res. 1999, 43, 860–878. [Google Scholar] [CrossRef]
  58. La, M.; D’Amico, M.; Bandiera, S.; Di Filippo, C.; Oliani, S.M.; Gavins, F.N.; Flower, R.J.; Perretti, M. Annexin 1 peptides protect against experimental myocardial ischemia-reperfusion: Analysis of their mechanism of action. FASEB J. 2001, 15, 2247–2256. [Google Scholar] [CrossRef] [PubMed]
  59. Gavins, F.N.; Kamal, A.M.; D’Amico, M.; Oliani, S.M.; Perretti, M. Formyl-peptide receptor is not involved in the protection afforded by annexin 1 in murine acute myocardial infarct. FASEB J. 2005, 19, 100–102. [Google Scholar] [CrossRef] [PubMed]
  60. D’Amico, M.; Di Filippo, C.; La, M.; Solito, E.; McLean, P.G.; Flower, R.J.; Oliani, S.M.; Perretti, M. Lipocortin 1 reduces myocardial ischemia-reperfusion injury by affecting local leukocyte recruitment. FASEB J. 2000, 14, 1867–1869. [Google Scholar] [CrossRef] [PubMed]
  61. Ritchie, R.H.; Gordon, J.M.; Woodman, O.L.; Cao, A.H.; Dusting, G.J. Annexin-1 peptide Anx-1(2-26) protects adult rat cardiac myocytes from cellular injury induced by simulated ischaemia. Br. J. Pharmacol. 2005, 145, 495–502. [Google Scholar] [CrossRef] [PubMed]
  62. Qin, C.; Buxton, K.D.; Pepe, S.; Cao, A.H.; Venardos, K.; Love, J.E.; Kaye, D.M.; Yang, Y.H.; Morand, E.F.; Ritchie, R.H. Reperfusion-induced myocardial dysfunction is prevented by endogenous annexin-A1 and its N-terminal-derived peptide Ac-ANX-A1(2–26). Br. J. Pharmacol. 2013, 168, 238–252. [Google Scholar] [CrossRef] [PubMed]
  63. Qin, C.X.; Finlayson, S.B.; Ai-Sharea, A.; Tate, M.; De Blasio, M.J.; Deo, M.; Rosli, S.; Prakoso, D.; Thomas, C.J.; Kiriazis, H.; et al. Endogenous Annexin-A1 Regulates Haematopoietic Stem Cell Mobilisation and Inflammatory Response Post Myocardial Infarction in Mice In Vivo. Sci. Rep. 2017, 7, 16615. [Google Scholar] [CrossRef] [PubMed]
  64. Qin, C.X.; May, L.T.; Li, R.; Cao, N.; Rosli, S.; Deo, M.; Alexander, A.E.; Horlock, D.; Bourke, J.E.; Yang, Y.H.; et al. Small-molecule-biased formyl peptide receptor agonist compound 17b protects against myocardial ischaemia-reperfusion injury in mice. Nat. Commun. 2017, 8, 14232. [Google Scholar] [CrossRef] [PubMed]
  65. Organ Donation and Transplantation Statistics. Available online: https://www.kidney.org/news/newsroom/factsheets/Organ-Donation-and-Transplantation-Stats (accessed on 12 March 2018).
  66. Salvadori, M.; Rosso, G.; Bertoni, E. Update on ischemia-reperfusion injury in kidney transplantation: Pathogenesis and treatment. World J. Transplant. 2015, 5, 52–67. [Google Scholar] [CrossRef] [PubMed]
  67. When a Transplant Fails. Available online: https://www.kidney.org/transplantation/transaction/TC/summer09/TCsm09_TransplantFails (accessed on 12 March 2018).
  68. Barin-Le Guellec, C.; Largeau, B.; Bon, D.; Marquet, P.; Hauet, T. Ischemia/reperfusion-associated tubular cells injury in renal transplantation: Can metabolomics inform about mechanisms and help identify new therapeutic targets? Pharmacol. Res. 2018, 129, 34–43. [Google Scholar] [CrossRef] [PubMed]
  69. Devarajan, P. Update on mechanisms of ischemic acute kidney injury. J. Am. Soc. Nephrol. 2006, 17, 1503–1520. [Google Scholar] [CrossRef] [PubMed]
  70. Havasi, A.; Borkan, S.C. Apoptosis and acute kidney injury. Kidney Int. 2011, 80, 29–40. [Google Scholar] [CrossRef] [PubMed]
  71. Versteilen, A.M.; Di Maggio, F.; Leemreis, J.R.; Groeneveld, A.B.; Musters, R.J.; Sipkema, P. Molecular mechanisms of acute renal failure following ischemia/reperfusion. Int. J. Artif. Organs 2004, 27, 1019–1029. [Google Scholar] [CrossRef] [PubMed]
  72. Yang, N.; Luo, M.; Li, R.; Huang, Y.; Zhang, R.; Wu, Q.; Wang, F.; Li, Y.; Yu, X. Blockage of JAK/STAT signalling attenuates renal ischaemia-reperfusion injury in rat. Nephrol. Dial. Transplant. 2008, 23, 91–100. [Google Scholar] [CrossRef] [PubMed]
  73. Kelly, K.J.; Williams, W.W., Jr.; Colvin, R.B.; Meehan, S.M.; Springer, T.A.; Gutierrez-Ramos, J.C.; Bonventre, J.V. Intercellular adhesion molecule-1-deficient mice are protected against ischemic renal injury. J. Clin. Investig. 1996, 97, 1056–1063. [Google Scholar] [CrossRef] [PubMed]
  74. Kelly, K.J.; Williams, W.W., Jr.; Colvin, R.B.; Bonventre, J.V. Antibody to intercellular adhesion molecule 1 protects the kidney against ischemic injury. Proc. Natl. Acad. Sci. USA 1994, 91, 812–816. [Google Scholar] [CrossRef] [PubMed]
  75. Takada, M.; Nadeau, K.C.; Shaw, G.D.; Marquette, K.A.; Tilney, N.L. The cytokine-adhesion molecule cascade in ischemia/reperfusion injury of the rat kidney. Inhibition by a soluble P-selectin ligand. J. Clin. Investig. 1997, 99, 2682–2690. [Google Scholar] [CrossRef] [PubMed]
  76. Facio, F.N., Jr.; Sena, A.A.; Araujo, L.P.; Mendes, G.E.; Castro, I.; Luz, M.A.; Yu, L.; Oliani, S.M.; Burdmann, E.A. Annexin 1 mimetic peptide protects against renal ischemia/reperfusion injury in rats. J. Mol. Med. 2011, 89, 51–63. [Google Scholar] [CrossRef] [PubMed]
  77. Araujo, L.P.; Truzzi, R.R.; Mendes, G.E.; Luz, M.A.; Burdmann, E.A.; Oliani, S.M. Annexin A1 protein attenuates cyclosporine-induced renal hemodynamics changes and macrophage infiltration in rats. Inflamm. Res. 2012, 61, 189–196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Umanath, K.; Lewis, J.B. Update on Diabetic Nephropathy: Core Curriculum 2018. Am. J. Kidney Dis. 2018. [Google Scholar] [CrossRef] [PubMed]
  79. Purvis, G.S.D.; Chiazza, F.; Chen, J.; Azevedo-Loiola, R.; Martin, L.; Kusters, D.H.M.; Reutelingsperger, C.; Fountoulakis, N.; Gnudi, L.; Yaqoob, M.M.; et al. Annexin A1 attenuates microvascular complications through restoration of Akt signalling in a murine model of type 1 diabetes. Diabetologia 2018, 61, 482–495. [Google Scholar] [CrossRef] [PubMed]
  80. Grootjans, J.; Lenaerts, K.; Derikx, J.P.; Matthijsen, R.A.; de Bruine, A.P.; van Bijnen, A.A.; van Dam, R.M.; Dejong, C.H.; Buurman, W.A. Human intestinal ischemia-reperfusion-induced inflammation characterized: Experiences from a new translational model. Am. J. Pathol. 2010, 176, 2283–2291. [Google Scholar] [CrossRef] [PubMed]
  81. Pope, M.R.; Fleming, S.D. TLR2 modulates antibodies required for intestinal ischemia/reperfusion-induced damage and inflammation. J. Immunol. 2015, 194, 1190–1198. [Google Scholar] [CrossRef] [PubMed]
  82. Doster, D.L.; Jensen, A.R.; Khaneki, S.; Markel, T.A. Mesenchymal stromal cell therapy for the treatment of intestinal ischemia: Defining the optimal cell isolate for maximum therapeutic benefit. Cytotherapy 2016, 18, 1457–1470. [Google Scholar] [CrossRef] [PubMed]
  83. Zhou, G.X.; Liu, Z.J. Potential roles of neutrophils in regulating intestinal mucosal inflammation of inflammatory bowel disease. J. Dig. Dis. 2017, 18, 495–503. [Google Scholar] [CrossRef] [PubMed]
  84. Gavins, F.N.; Yona, S.; Kamal, A.M.; Flower, R.J.; Perretti, M. Leukocyte antiadhesive actions of annexin 1: ALXR- and FPR-related anti-inflammatory mechanisms. Blood 2003, 101, 4140–4147. [Google Scholar] [CrossRef] [PubMed]
  85. Leoni, G.; Neumann, P.A.; Kamaly, N.; Quiros, M.; Nishio, H.; Jones, H.R.; Sumagin, R.; Hilgarth, R.S.; Alam, A.; Fredman, G.; et al. Annexin A1-containing extracellular vesicles and polymeric nanoparticles promote epithelial wound repair. J. Clin. Investig. 2015, 125, 1215–1227. [Google Scholar] [CrossRef] [PubMed]
  86. Martin, G.R.; Perretti, M.; Flower, R.J.; Wallace, J.L. Annexin-1 modulates repair of gastric mucosal injury. Am. J. Physiol. Gastrointest. Liver Physiol. 2008, 294, G764–G769. [Google Scholar] [CrossRef] [PubMed]
  87. Leoni, G.; Alam, A.; Neumann, P.A.; Lambeth, J.D.; Cheng, G.; McCoy, J.; Hilgarth, R.S.; Kundu, K.; Murthy, N.; Kusters, D.; et al. Annexin A1, formyl peptide receptor, and NOX1 orchestrate epithelial repair. J. Clin. Investig. 2013, 123, 443–454. [Google Scholar] [CrossRef] [PubMed]
  88. Guido, B.C.; Zanatelli, M.; Tavares-de-Lima, W.; Oliani, S.M.; Damazo, A.S. Annexin-A1 peptide down-regulates the leukocyte recruitment and up-regulates interleukin-10 release into lung after intestinal ischemia-reperfusion in mice. J. Inflamm. 2013, 10, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Babbin, B.A.; Laukoetter, M.G.; Nava, P.; Koch, S.; Lee, W.Y.; Capaldo, C.T.; Peatman, E.; Severson, E.A.; Flower, R.J.; Perretti, M.; et al. Annexin A1 regulates intestinal mucosal injury, inflammation, and repair. J. Immunol. 2008, 181, 5035–5044. [Google Scholar] [CrossRef] [PubMed]
  90. Le, Y.; Oppenheim, J.J.; Wang, J.M. Pleiotropic roles of formyl peptide receptors. Cytokine Growth Factor Rev. 2001, 12, 91–105. [Google Scholar] [CrossRef]
  91. Liao, W.I.; Wu, S.Y.; Wu, G.C.; Pao, H.P.; Tang, S.E.; Huang, K.L.; Chu, S.J. Ac2-26, an Annexin A1 Peptide, Attenuates Ischemia-Reperfusion-Induced Acute Lung Injury. Int. J. Mol. Sci. 2017, 18, 1771. [Google Scholar] [CrossRef] [PubMed]
  92. Karlsson, J.; Fu, H.; Boulay, F.; Dahlgren, C.; Hellstrand, K.; Movitz, C. Neutrophil NADPH-oxidase activation by an annexin AI peptide is transduced by the formyl peptide receptor (FPR), whereas an inhibitory signal is generated independently of the FPR family receptors. J. Leukoc. Biol. 2005, 78, 762–771. [Google Scholar] [CrossRef] [PubMed]
  93. Luo, Z.Z.; Gao, Y.; Sun, N.; Zhao, Y.; Wang, J.; Tian, B.; Shi, J. Enhancing the interaction between annexin-1 and formyl peptide receptors regulates microglial activation to protect neurons from ischemia-like injury. J. NeuroImmunol. 2014, 276, 24–36. [Google Scholar] [CrossRef] [PubMed]
  94. Faraday, N.; Schunke, K.; Saleem, S.; Fu, J.; Wang, B.; Zhang, J.; Morrell, C.; Dore, S. Cathepsin G-dependent modulation of platelet thrombus formation in vivo by blood neutrophils. PLoS ONE 2013, 8, e71447. [Google Scholar] [CrossRef] [PubMed]
  95. Sun, R.; Iribarren, P.; Zhang, N.; Zhou, Y.; Gong, W.; Cho, E.H.; Lockett, S.; Chertov, O.; Bednar, F.; Rogers, T.J.; et al. Identification of neutrophil granule protein cathepsin G as a novel chemotactic agonist for the G protein-coupled formyl peptide receptor. J. Immunol. 2004, 173, 428–436. [Google Scholar] [CrossRef] [PubMed]
  96. Kurosaka, K.; Chen, Q.; Yarovinsky, F.; Oppenheim, J.J.; Yang, D. Mouse cathelin-related antimicrobial peptide chemoattracts leukocytes using formyl peptide receptor-like 1/mouse formyl peptide receptor-like 2 as the receptor and acts as an immune adjuvant. J. Immunol. 2005, 174, 6257–6265. [Google Scholar] [CrossRef] [PubMed]
  97. Furlan, F.; Orlando, S.; Laudanna, C.; Resnati, M.; Basso, V.; Blasi, F.; Mondino, A. The soluble D2D3(88-274) fragment of the urokinase receptor inhibits monocyte chemotaxis and integrin-dependent cell adhesion. J. Cell Sci. 2004, 117, 2909–2916. [Google Scholar] [CrossRef] [PubMed]
  98. Perez, H.D.; Kelly, E.; Elfman, F.; Armitage, G.; Winkler, J. Defective polymorphonuclear leukocyte formyl peptide receptor(s) in juvenile periodontitis. J. Clin. Investig. 1991, 87, 971–976. [Google Scholar] [CrossRef] [PubMed]
  99. Shin, M.K.; Jang, Y.H.; Yoo, H.J.; Kang, D.W.; Park, M.H.; Kim, M.K.; Song, J.H.; Kim, S.D.; Min, G.; You, H.K.; et al. N-formyl-methionyl-leucyl-phenylalanine (fMLP) promotes osteoblast differentiation via the N-formyl peptide receptor 1-mediated signaling pathway in human mesenchymal stem cells from bone marrow. J. Biol. Chem. 2011, 286, 17133–17143. [Google Scholar] [CrossRef] [PubMed]
  100. Harada, M.; Habata, Y.; Hosoya, M.; Nishi, K.; Fujii, R.; Kobayashi, M.; Hinuma, S. N-Formylated humanin activates both formyl peptide receptor-like 1 and 2. Biochem. Biophys. Res. Commun. 2004, 324, 255–261. [Google Scholar] [CrossRef] [PubMed]
  101. Su, S.B.; Gong, W.H.; Gao, J.L.; Shen, W.P.; Grimm, M.C.; Deng, X.; Murphy, P.M.; Oppenheim, J.J.; Wang, J.M. T20/DP178, an ectodomain peptide of human immunodeficiency virus type 1 gp41, is an activator of human phagocyte N-formyl peptide receptor. Blood 1999, 93, 3885–3892. [Google Scholar] [PubMed]
  102. Hartt, J.K.; Liang, T.; Sahagun-Ruiz, A.; Wang, J.M.; Gao, J.L.; Murphy, P.M. The HIV-1 cell entry inhibitor T-20 potently chemoattracts neutrophils by specifically activating the N-formylpeptide receptor. Biochem. Biophys. Res. Commun. 2000, 272, 699–704. [Google Scholar] [CrossRef] [PubMed]
  103. Su, S.B.; Gao, J.; Gong, W.; Dunlop, N.M.; Murphy, P.M.; Oppenheim, J.J.; Wang, J.M. T21/DP107, A synthetic leucine zipper-like domain of the HIV-1 envelope gp41, attracts and activates human phagocytes by using G-protein-coupled formyl peptide receptors. J. Immunol. 1999, 162, 5924–5930. [Google Scholar] [PubMed]
  104. Le, Y.; Jiang, S.; Hu, J.; Gong, W.; Su, S.; Dunlop, N.M.; Shen, W.; Li, B.; Ming Wang, J. N36, a synthetic N-terminal heptad repeat domain of the HIV-1 envelope protein gp41, is an activator of human phagocytes. Clin. Immunol. 2000, 96, 236–242. [Google Scholar] [CrossRef] [PubMed]
  105. Bannenberg, G.; Moussignac, R.L.; Gronert, K.; Devchand, P.R.; Schmidt, B.A.; Guilford, W.J.; Bauman, J.G.; Subramanyam, B.; Perez, H.D.; Parkinson, J.F.; et al. Lipoxins and novel 15-epi-lipoxin analogs display potent anti-inflammatory actions after oral administration. Br. J. Pharmacol. 2004, 143, 43–52. [Google Scholar] [CrossRef] [PubMed]
  106. De, Y.; Chen, Q.; Schmidt, A.P.; Anderson, G.M.; Wang, J.M.; Wooters, J.; Oppenheim, J.J.; Chertov, O. LL-37, the neutrophil granule- and epithelial cell-derived cathelicidin, utilizes formyl peptide receptor-like 1 (FPRL1) as a receptor to chemoattract human peripheral blood neutrophils, monocytes, and T cells. J. Exp. Med. 2000, 192, 1069–1074. [Google Scholar]
  107. Hu, J.Y.; Le, Y.; Gong, W.; Dunlop, N.M.; Gao, J.L.; Murphy, P.M.; Wang, J.M. Synthetic peptide MMK-1 is a highly specific chemotactic agonist for leukocyte FPRL1. J. Leukoc. Biol. 2001, 70, 155–161. [Google Scholar] [PubMed]
  108. Rabiet, M.J.; Huet, E.; Boulay, F. Human mitochondria-derived N-formylated peptides are novel agonists equally active on FPR and FPRL1, while Listeria monocytogenes-derived peptides preferentially activate FPR. Eur. J. Immunol. 2005, 35, 2486–2495. [Google Scholar] [CrossRef] [PubMed]
  109. Kim, Y.; Lee, B.D.; Kim, O.; Bae, Y.S.; Lee, T.; Suh, P.G.; Ryu, S.H. Pituitary adenylate cyclase-activating polypeptide 27 is a functional ligand for formyl peptide receptor-like 1. J. Immunol. 2006, 176, 2969–2975. [Google Scholar] [CrossRef] [PubMed]
  110. Brandenburg, L.O.; Koch, T.; Sievers, J.; Lucius, R. Internalization of PrP106-126 by the formyl-peptide-receptor-like-1 in glial cells. J. Neurochem. 2007, 101, 718–728. [Google Scholar] [CrossRef] [PubMed]
  111. Nanamori, M.; Cheng, X.; Mei, J.; Sang, H.; Xuan, Y.; Zhou, C.; Wang, M.W.; Ye, R.D. A novel nonpeptide ligand for formyl peptide receptor-like 1. Mol. Pharmacol. 2004, 66, 1213–1222. [Google Scholar] [CrossRef] [PubMed]
  112. Chen, X.; Mellon, R.D.; Yang, L.; Dong, H.; Oppenheim, J.J.; Howard, O.M. Regulatory effects of deoxycholic acid, a component of the anti-inflammatory traditional Chinese medicine Niuhuang, on human leukocyte response to chemoattractants. Biochem. Pharmacol. 2002, 63, 533–541. [Google Scholar] [CrossRef]
  113. Gargiulo, L.; Longanesi-Cattani, I.; Bifulco, K.; Franco, P.; Raiola, R.; Campiglia, P.; Grieco, P.; Peluso, G.; Stoppelli, M.P.; Carriero, M.V. Cross-talk between fMLP and vitronectin receptors triggered by urokinase receptor-derived SRSRY peptide. J. Biol. Chem. 2005, 280, 25225–25232. [Google Scholar] [CrossRef] [PubMed]
  114. Su, S.B.; Gong, W.; Gao, J.L.; Shen, W.; Murphy, P.M.; Oppenheim, J.J.; Wang, J.M. A seven-transmembrane, G protein-coupled receptor, FPRL1, mediates the chemotactic activity of serum amyloid A for human phagocytic cells. J. Exp. Med. 1999, 189, 395–402. [Google Scholar] [CrossRef] [PubMed]
  115. Shen, W.; Proost, P.; Li, B.; Gong, W.; Le, Y.; Sargeant, R.; Murphy, P.M.; Van Damme, J.; Wang, J.M. Activation of the chemotactic peptide receptor FPRL1 in monocytes phosphorylates the chemokine receptor CCR5 and attenuates cell responses to selected chemokines. Biochem. Biophys. Res. Commun. 2000, 272, 276–283. [Google Scholar] [CrossRef] [PubMed]
  116. Le, Y.; Gong, W.; Li, B.; Dunlop, N.M.; Shen, W.; Su, S.B.; Ye, R.D.; Wang, J.M. Utilization of two seven-transmembrane, G protein-coupled receptors, formyl peptide receptor-like 1 and formyl peptide receptor, by the synthetic hexapeptide WKYMVm for human phagocyte activation. J. Immunol. 1999, 163, 6777–6784. [Google Scholar] [PubMed]
  117. Stenfeldt, A.L.; Karlsson, J.; Wenneras, C.; Bylund, J.; Fu, H.; Dahlgren, C.; Cyclosporin, H. Boc-MLF and Boc-FLFLF are antagonists that preferentially inhibit activity triggered through the formyl peptide receptor. Inflammation 2007, 30, 224–229. [Google Scholar] [CrossRef] [PubMed]
  118. Chen, X.; Yang, D.; Shen, W.; Dong, H.F.; Wang, J.M.; Oppenheim, J.J.; Howard, M.Z. Characterization of chenodeoxycholic acid as an endogenous antagonist of the G-coupled formyl peptide receptors. Inflamm. Res. 2000, 49, 744–755. [Google Scholar] [CrossRef] [PubMed]
  119. Haas, P.J.; de Haas, C.J.; Kleibeuker, W.; Poppelier, M.J.; van Kessel, K.P.; Kruijtzer, J.A.; Liskamp, R.M.; van Strijp, J.A. N-terminal residues of the chemotaxis inhibitory protein of Staphylococcus aureus are essential for blocking formylated peptide receptor but not C5a receptor. J. Immunol. 2004, 173, 5704–5711. [Google Scholar] [CrossRef] [PubMed]
  120. Mills, J.S. Peptides derived from HIV-1, HIV-2, Ebola virus, SARS coronavirus and coronavirus 229E exhibit high affinity binding to the formyl peptide receptor. Biochim. Biophys. Acta 2006, 1762, 693–703. [Google Scholar] [CrossRef] [PubMed]
  121. Yan, P.; Nanamori, M.; Sun, M.; Zhou, C.; Cheng, N.; Li, N.; Zheng, W.; Xiao, L.; Xie, X.; Ye, R.D.; et al. The immunosuppressant cyclosporin A antagonizes human formyl peptide receptor through inhibition of cognate ligand binding. J. Immunol. 2006, 177, 7050–7058. [Google Scholar] [CrossRef] [PubMed]
  122. Prat, C.; Bestebroer, J.; de Haas, C.J.; van Strijp, J.A.; van Kessel, K.P. A new staphylococcal anti-inflammatory protein that antagonizes the formyl peptide receptor-like 1. J. Immunol. 2006, 177, 8017–8026. [Google Scholar] [CrossRef] [PubMed]
  123. Higgins, J.D., 3rd; Bridger, G.J.; Derian, C.K.; Beblavy, M.J.; Hernandez, P.E.; Gaul, F.E.; Abrams, M.J.; Pike, M.C.; Solomon, H.F. N-terminus urea-substituted chemotactic peptides: New potent agonists and antagonists toward the neutrophil fMLF receptor. J. Med. Chem. 1996, 39, 1013–1015. [Google Scholar] [CrossRef] [PubMed]
  124. Liang, T.S.; Gao, J.L.; Fatemi, O.; Lavigne, M.; Leto, T.L.; Murphy, P.M. The endogenous opioid spinorphin blocks fMet-Leu-Phe-induced neutrophil chemotaxis by acting as a specific antagonist at the N-formylpeptide receptor subtype FPR. J. Immunol. 2001, 167, 6609–6614. [Google Scholar] [CrossRef] [PubMed]
  125. Bae, Y.S.; Lee, H.Y.; Jo, E.J.; Kim, J.I.; Kang, H.K.; Ye, R.D.; Kwak, J.Y.; Ryu, S.H. Identification of peptides that antagonize formyl peptide receptor-like 1-mediated signaling. J. Immunol. 2004, 173, 607–614. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of the protective effects of AnxA1 in I/RI. By binding to members of the FPR family, AnxA1 and its peptide mimetics can (A) decrease rolling, adhesion and emigration of leukocytes (monocytes and neutrophils) [31,46,47,53,76,84,88]. In addition, platelet aggregates are diminished [7,31,47] and pro-inflammatory mediators (e.g., myeloperoxidase (MPO), reactive oxygen species (ROS) and cytokines) are moderated [88,90]. (B) AnxA1 promotes the balance between pro-inflammatory [7,31,47,58,60,88,90] and anti-inflammatory cytokines, thereby fostering homeostasis in the host. (C) The disruption of barrier function, due to the loss of junctional molecules and apoptosis/necrosis of endothelial/epithelial cells, is one of the main characteristics of I/RI. AnxA1 has a positive effect on maintaining barrier function [49] and reducing edema [48] by inhibiting the loss of junction molecules, in particular occludin, VE-cadherin and actin cytoskeleton [50,59]. Thus, AnxA1 regulates the inflammatory responses elicited during I/RI leading to the resolution of inflammation and decreased tissue injury in (D) different vascular beds.
Figure 1. Schematic representation of the protective effects of AnxA1 in I/RI. By binding to members of the FPR family, AnxA1 and its peptide mimetics can (A) decrease rolling, adhesion and emigration of leukocytes (monocytes and neutrophils) [31,46,47,53,76,84,88]. In addition, platelet aggregates are diminished [7,31,47] and pro-inflammatory mediators (e.g., myeloperoxidase (MPO), reactive oxygen species (ROS) and cytokines) are moderated [88,90]. (B) AnxA1 promotes the balance between pro-inflammatory [7,31,47,58,60,88,90] and anti-inflammatory cytokines, thereby fostering homeostasis in the host. (C) The disruption of barrier function, due to the loss of junctional molecules and apoptosis/necrosis of endothelial/epithelial cells, is one of the main characteristics of I/RI. AnxA1 has a positive effect on maintaining barrier function [49] and reducing edema [48] by inhibiting the loss of junction molecules, in particular occludin, VE-cadherin and actin cytoskeleton [50,59]. Thus, AnxA1 regulates the inflammatory responses elicited during I/RI leading to the resolution of inflammation and decreased tissue injury in (D) different vascular beds.
Ijms 19 01211 g001
Table 1. FPR nomenclature and cellular and tissue distribution in human and mouse.
Table 1. FPR nomenclature and cellular and tissue distribution in human and mouse.
SpeciesIUPHAR NomenclatureCellular DistributionTissue Distribution
HUMANFPR1Adrenal cortical cells, astrocytes, carcinoma cells, endothelial cell, epithelial cells, fibroblasts, kupffer cells, macrophages, microglial cells, monocytes, neuroblastoma cells, neutrophil, platelets, immature dendritic cells, hepatocytesAdrenal glands, bone marrow, central nervous system, colon, eye, heart, kidney, liver, lung, ovary, placenta, spleen
FPR2/ALXAstrocytes, endothelial cells, epithelial cells, fibroblasts, hepatocytes, immature dendritic cells, microglial cells, macrophages, monocytes, neuroblastoma cells, neutrophils, T and B lymphocytes,Bone marrow, brain, lung, placenta, spleen, testis
FPR3Dendritic cells, HL-60 cells, macrophages, monocytes, eosinophilsAdrenal gland, liver, lung, lymph nodes, placenta, small intestine, spleen, trachea
MURINEFpr1Dendritic cells, microglia, mononuclear cells, neutrophilsAdrenal gland, anterior pituitary, hippocampus, hypothalamus, liver, lung, spleen
Fpr2Dendritic cells, microglia, neutrophilsAnterior pituitary, adrenal gland, hippocampus, hypothalamus, lungs, spleen
Fpr3Microglia, neutrophilsAdrenal gland, anterior pituitary, heart, hippocampus, hypothalamus, liver, lung, spleen
Fpr-rs3 Skeletal muscle
Fpr-rs4 unknown
Fpr-rs5 unknown
Fpr-rs6 Brain, skeletal muscle, spleen, testis
Fpr-rs7Smooth muscleHeart, liver, lung, pancreas, spleen, smooth muscle
Fpr-rs8 unknown
Table 2. Non-exhaustive list of FPR ligands and their biological actions.
Table 2. Non-exhaustive list of FPR ligands and their biological actions.
LigandBiological ActionDisease StateRefs
FPR agonists
42 (FPR2/ALX)Chemotaxis of mononuclear cellsBacterial pathogenesis[90]
Ac2-12 (FPR1)Cardioprotection in experimental MI/RMI [58][58]
Ac2-26
(FPR1, FPR2/ALX)
Decreases neutrophil–endothelium interactions in flow chamber
Regulates leukocyte–platelet response in the cerebral microvasculature
Decreases pulmonary arterial pressure
Decreases inflammatory cytokine production
Decreases lung tissue damage following I/R
Decreases infarct size post myocardial I/R
Reduces myeloperoxidase activity
Prevents IFN-γ and endotoxin induced inotropic and cyclooxygenase 2 gene expression
Decreases adhesion and transmigration in inflammatory mesentery in vivo
AIS [31]
I/R induced lung injury [91]
MI [33,58]
Mesenteric ischemia [84]
[31,33,58,84,91]
Ac9–25 (FPR1)Activates neutrophil NADPH oxidaseInflammation[92]
Annexin A1 (FPR2/ALX)Regulates microglial efferocytosis and phagocytosis
Decreases neutrophil–endothelium interactions in flow chamber
Cardioprotection in experimental MI/R
Prevents leukocyte migration to the inflamed tissue
AIS [93]
MI [58]
[58,93]
Antiflammin 2 (FPR2/ALX)Decreases neutrophil–endothelium interactionsInflammation[22]
Cathepsin G (FPR1)Chemoattractant for phagocytic leukocytes
Promotes platelet aggregation and hemostasis
Promotes thrombus formation
Promotes middle cerebral artery occlusion and brain injury in ischemic stroke model
AIS [94][94,95]
CRAMP (FPR2/ALX)Chemotactic activator of mouse and human leukocytes
Calcium flux, MAPK activation
Bacterial pathogenesis[96]
Compound 17b (FPR1, FPR2/ALX)Attenuates early as well as late inflammatory responses after via ERK1/2–Akt kinase systemMI [64][64]
D2D388–274 (FPR2/ALX)Inhibits monocyte chemotaxis and integrin-dependent cell adhesionInflammation[97]
fMLP and analogues (FPR2/ALX)Defective PMN chemotaxis in juvenile peridontitis in vivo
Lineage specific differentiation of mesenchymal stem cells especially osteoblasts
Juvenile periodontitis [98]
Osteoporosis [99]
[98,99]
Formylated humanin (FPR2/ALX)Chemotaxis of human FPR2/ALX-transfected CHO cellsInflammation[100]
Humanin (FPR2/ALX)Chemotaxis of human FPR2/ALX-transfected CHO cellsInflammation[100]
HIV-1 T20 (DP178) (FPR1, FPR2/ALX)Chemoattractant and activator of peripheral phagocytes, hence promoting host immune responses against HIV-1 replicationHIV/AIDS [101,102][101,102]
HIV-1 T21 (DP107) (FPR1, FPR2/ALX)Chemoattractant and activator of peripheral phagocytes (high affinity towards FPR2/ALX)HIV/AIDS [103][103]
HIV gp41 (N36) (FPR2/ALX)Induces directional migration and calcium mobilization in human monocytes and neutrophilsHIV/AIDS [104][104]
Lipoxin A4 and ATL (aspirin triggered lipoxin)Inhibition of lung inflammation after hind-limb IR
Downregulation of neutrophil accumulation Regulates neutrophil–platelet aggregates
ATL is required for ASA protection in AIS
Hind limb ischemia [105]
AIS [31]
[31,105]
LL-37 (cathelicidin peptide) (FPR2/ALX)Enhances phagocytosis of IgG-opsonized Gram negative and Gram-positive bacteria Chemoattractant for human peripheral blood neutrophils, monocytes and T cells Calcium mobilizationBacterial pathogenesis[106]
MMK-1 (FPR2/ALX)Potent chemoattractant and calcium mobilizing agent agonist for human monocytes, neutrophils and FPR2/ALX transfected human embryonic kidney (HEK) 293 cells.Bacterial pathogenesis[107]
N36 peptide (FPR2/ALX)Chemotaxis and calcium mobilization in monocytes and neutrophilsViral pathogenesis[104]
NADH dehydrogenase (FPR2/ALX)Chemotaxis and calcium mobilization in human FPR2/ALX-expressing HL-60 cellsInflammation[108]
PACAP27 (FPR2/ALX)Neutrophil chemotaxis and upregulation of CD11b
Intracellular calcium mobilization
ERK phosphorylation
Inflammation[109]
PRP106-126 (FPR2/ALX)Endocytosis in glial cellsNeurodegenerative diseases[110]
Quin-C1 (FPR2/ALX)Neutrophil chemotaxis, stimulates calcium mobilization, and MAP kinase phosphorylation Inflammation[111]
Rana-6 (FPR2/ALX)Chemoattractant of phagocytesInflammation[112]
SRSRY (FPR1)Directional cell migration on vitronectin-coated filtersInflammation[113]
Serum amyloid A (SAA) (FPR2/ALX)Potent leukocyte chemoattractantInflammation[114]
Temporin A (FPR2/ALX)Chemoattractant and activator of peripheral phagocytesBacterial pathogenesis[96]
uPAR84–95 (FPR2/ALX)Chemoattractant and activator of peripheral phagocytes (high affinity)Inflammmation[115]
V3 peptideChemoattractant of phagocytes
Inhibits monocytic response to chemokines
Inflammation[115]
W peptide (FPR2/ALX)Activates phagocyte chemotaxis and calcium fluxInflammation[116]
FPR antagonists
BOC2 (FPR1, FPR2/ALX)Decreased neutrophil activationInflammation[117]
CDCA (FPR1)Inhibits neutrophil chemoattraction and migration
Inhibits calcium flux
Leukocyte migration/Inflammation[118]
CHIPS (FPR1)Inhibits chemotaxis in S. aureus infectionBacterial pathogenesis[119]
Coronavirus 229E peptides (FPR2/ALX)Ligand binding studies using transfected CHO cells demonstrated antagonism of FPR2/ALXViral/bacterial pathogenesis[120]
Coronavirus peptides (FPR2/ALX)Inhibits fMLP interaction in CHO cellsViral/bacterial pathogenesis[120]
Cyclosporine A (FPR1)Inhibits fMLF-stimulated degranulation, chemotaxis, calcium mobilization of neutrophilsInflammation[121]
Cyclosporine H (FPR1)Decreased neutrophil activationInflammation[117]
DCA (FPR1)Inhibits fMLP-induced monocyte and neutrophil chemotaxis and calcium mobilizationInflammation[112]
Ebola peptides (FPR1)Inhibits fMLP interaction in CHO cellsViral pathogenesis[120]
FLIPr (FPR2/ALX)FPR2/ALX inhibitory protein (FLIPr) exerts anti-inflammatory activity by inhibiting calcium mobilization and cell migration toward chemoattractants.Inflammation[122]
HIV-2 peptides (FPR1)Inhibits fMLP interaction in CHO cellsViral pathogenesis[120]
Isopropylureido-FLFLF (FPR1)Inhibits chemotaxisInflammation[123]
Spinorphin (FPR1)Inhibits calcium mobilization and fMLP induced neutrophil chemotaxisInflammation[124]
WRW4 (FPR2/ALX)Inhibits chemotaxis, calcium flux, superoxide generation and ERK phosphorylationNeurodegenerative diseases, AIS[125]
MI, myocardial infarction; MI/R, myocardial ischemia reperfusion; AIS, acute ischemic stroke; IFN-γ, interferon gamma; MAPK, Mitogen-activated protein kinase; ERK, extracellular signal–regulated kinase; AKT, serine/threonine-protein kinase; PMN, polymorphonuclear leukocytes; CHO cells, Chinese hamster ovary; ATL, aspirin triggered lipoxin; ASA, aspirin ; IgG, Immunoglobulin G; fMLP, formyl-Met-Leu-Phe (fMLP), G proteins; FLIPr, FPR2/ALX inhibitory protein

Share and Cite

MDPI and ACS Style

Ansari, J.; Kaur, G.; Gavins, F.N.E. Therapeutic Potential of Annexin A1 in Ischemia Reperfusion Injury. Int. J. Mol. Sci. 2018, 19, 1211. https://doi.org/10.3390/ijms19041211

AMA Style

Ansari J, Kaur G, Gavins FNE. Therapeutic Potential of Annexin A1 in Ischemia Reperfusion Injury. International Journal of Molecular Sciences. 2018; 19(4):1211. https://doi.org/10.3390/ijms19041211

Chicago/Turabian Style

Ansari, Junaid, Gaganpreet Kaur, and Felicity N. E. Gavins. 2018. "Therapeutic Potential of Annexin A1 in Ischemia Reperfusion Injury" International Journal of Molecular Sciences 19, no. 4: 1211. https://doi.org/10.3390/ijms19041211

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