Abstract
Obesity-induced insulin resistance and metabolic syndrome continue to pose an important public health challenge worldwide as they significantly increase the risk of type 2 diabetes and atherosclerotic cardiovascular disease. Advances in the pathophysiologic understanding of this process has identified that chronic inflammation plays a pivotal role. In this regard, given that both animal models and human studies have demonstrated that the interaction of P-selectin glycoprotein ligand-1 (PSGL-1) with P-selectin are not only critical for normal immune response, but also are upregulated in the setting of metabolic syndrome, PSGL-1/P-selectin interactions provide a novel target for preventing and treating resultant disease. Current approaches of interfering with PSGL-1/P-selectin interactions include targeted antibodies, recombinant immunoglobulins that competitively bind P-selectin, and synthetic molecular therapies. Experimental models as well as clinical trials assessing the role of these modalities in a variety of diseases have continued to contribute to the understanding of PSGL-1/P-selectin interactions and have demonstrated the difficulty in creating clinically relevant therapeutics. Most recently, however, computational simulations have further enhanced our understanding of the structural features of PSGL-1 and related glycomimetics, which are responsible for high affinity selectin interactions. Leveraging these insights for the design of next generation agents has thus led to development of a promising synthetic method for generating PSGL-1 glycosulfopeptide mimetics for the treatment of metabolic syndrome.
INTRODUCTION
The metabolic syndrome, characterized as a collection of risk factors for atherosclerotic cardiovascular disease and type 2 diabetes, is driven by excess energy intake and obesity [1]. The five interrelated factors comprising the syndrome are atherogenic dyslipidemia, elevated blood pressure, glucose intolerance and insulin resistance, a pro-thrombotic state, and a pro-inflammatory state [2]. Primarily, management of metabolic syndrome focuses on lifestyle modifications, such as weight reduction and increased physical activity [3]. In patients with persistent risk factors, further treatment with lipid lowering agents, anti-hypertensives, and antiplatelet agents help reduce the risk of cardiovascular disease, whereas drugs to reduce serum glucose and improve insulin sensitivity can be used to treat resultant diabetes [2]. Currently, despite a prevalence of 20–30%, therapies to prevent the development of cardiovascular disease and diabetes due to obesity-induced metabolic syndrome are lacking [2].
Mechanistically, a state of chronic inflammation has been suggested to underlie metabolic syndrome [4]. Specifically, obesity-induced immune cell infiltration of adipose tissue has been found to be a significant factor in the development of insulin resistance, type 2 diabetes, hepatosteatosis, and atherosclerosis [5–11]. Broadly, the inflammatory response includes monocytes [8, 12–16], neutrophils [17, 18], T cells [19–22], B cells [23, 24], mast cells [25], and eosinophils [26], with the extent of metabolic dysfunction directly correlating with the activation of pro-inflammatory cytokines and chemokines [27–29], as well as the modulation of inflammatory pathways such as the c-Jun N-terminal kinases (JNK) and nuclear factor-κB (NF-κB) transcription factor [30, 31].
In view of this, attempts to develop targeted therapies that modulate the inflammatory cascade as it pertains to metabolic syndrome, are ongoing [4]. Examples of such anti-inflammatory agents include statins and angiotensin converting enzyme inhibitors (ACE-I), which suppress the production of the pro-inflammatory Th1 and Th17 cells [32, 33]; apolipoprotein C-III inhibitors that prevent toll-like receptor 2 (TLR2) activation [4]; omega-3 fatty acids that can be converted to specialized pro-resolving mediators (SPMs) [34, 35]; and peroxisome proliferator-activated receptor alpha (PPAR-α) agonists, which promote suppression of monocyte chemoattractant protein 1 (MCP-1), intracellular adhesion molecule 1 (ICAM), vascular cell adhesion protein 1 (VCAM) [36], and NF-κB [37]. Additionally, in randomized clinical trials, the anti-inflammatory drug salsalate has been found to improve insulin sensitivity and inflammatory parameters [38], as well as glucose and triglyceride levels [39]. In a subsequent multicenter trial, a reduction in blood glucose, diabetes medication, and markers of cardiovascular risk were noted over a 48-week interval in patients with type 2 diabetes [40]. A sustained improvement in insulin sensitivity, along with a reduction in markers of systemic inflammation have also been reported in response to an IL-1 receptor antagonist [41]. Although the magnitude of glucose lowering has been modest in response to both salsalate and IL-1β blockade, these studies suggest that targeting inflammation is a valid strategy for the prevention and treatment of the adverse metabolic effects of obesity. With the inflammatory pathway continuing to evolve as a focus for the prevention and treatment of obesity-induced insulin resistance, diabetes, and cardiovascular disease, new promising targets have been identified and warrant review.
In this article, targeting the interaction of P-selectin glycoprotein ligand-1 (PSGL-1) with selectin will be discussed as a novel therapeutic strategy for metabolic syndrome. Specifically, PSGL-1 and selectin interactions in inflammation will be reviewed, with a specific emphasis on their role in the pathophysiology of obesity-induced metabolic syndrome. Importantly, current strategies of blocking PSGL-1/P-selectin interactions will be discussed and next generation synthetic approaches of creating PSGL-1 glycosulfopeptide (GSP) mimetics will be considered.
PSGL-1/P-SELECTIN INTERACTIONS AS MEDIATORS OF OBESITY-INDUCED INFLAMMATORY RESPONSES
PSGL-1 is a glycoprotein that is expressed on the surface of all leukocytes and supports leukocyte recruitment as a component of a range of inflammatory responses [42–46]. Structurally, PSGL-1 is a membrane protein with a disulfide-linked homodimer that has a mucin ectodomain with serine, threonine, and proline repeats that are sites for potential O-glycan modification [46]. PSGL-1 is a ligand for endothelium-selectin (E-selectin, CD62L), platelet-selectin (P-selectin, CD62P) and leukocyte-selectin (L-selectin, CD62L), but binds with highest affinity to P-selectin [47, 48].
P-selectin, which as its name implies, was originally isolated form platelets but later noted to be expressed by endothelial cells, has a membrane bound and a soluble form [48, 49], both of which bind leukocytes [50]. P-selectin is stored in the α-granules in platelets and in the Weibel-Palade bodies of endothelial cells. As a cell adhesion molecule, in the setting of inflammation, P-selectin translocates to the plasma membrane where it can interact with ligands. P-selectin is structurally made up of three characteristic protein motifs including a lectin, an epidermal growth factor (EGF) domain, and a number of short consensus repeats (SCRs)[51]. The soluble form of P-selectin may be produced from alternatively spliced mRNA that does not code for the transmembrane domain [49], and may also arise from damaged platelet membranes expressing P-selectin [52, 53]. The interaction of PSGL-1 with P-selectin on activated platelets leads to leukocyte-platelet aggregates that promote the adhesion and infiltration of inflammatory cells [54–57]. Similarly, the ligation of P-selectin on endothelial cells by PSGL-1 comprises the initial capture and rolling step in leukocyte-endothelial cell interactions (Figure 1) [58, 59]. Given this critical role in inflammatory response, and in turn its importance in the pathophysiology of obesity-induced metabolic syndrome, animal and clinical studies have been performed to better characterize PSGL-1/P-selectin interactions in this context.
Figure 1.
Interaction of P-selectin on activated endothelial-cells and platelets with PSGL-1 promotes capture and rolling of inflammatory cells. PSGL-1: P-selectin glycoprotein ligand-1. Modified from Kolaczkowska E, Kubes P. Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol. 2013;13:159–75.
In animal models of obesity, the influx of leukocytes into visceral adipose tissue is mediated by P-selectin [60] and PSGL-1 [12]. Particularly, in high-fat diets, excess free fatty acid formation increases endothelial cell (EC) expression of P-selectin [61–63]. Myeloperoxidase (MPO), which is released upon adherence of leukocytes to P-selectin, is also increased in obese mice [17]. MPO impairs adiponectin signaling [64], which increases leukocyte trafficking to visceral fat and further increases inflammation [65–67]. It is noteworthy that genetic deletion of PSGL-1 is protective against visceral fat inflammation in both diet-induced and leptin receptor mutant obese mice [68]. Leukocyte-EC interactions and macrophage content are both reduced in these animal models, as are circulating levels of P-selectin and MCP-1 [68]. These results confirm that PSGL-1/P-selectin interactions mediate leukocyte-EC and leukocyte-platelet interactions, which contribute to the adverse metabolic consequences of obesity. Furthermore, obese mice that are deficient in PSGL-1 display enhanced insulin sensitivity, improved lipid metabolism, decreased hepatic steatosis, and are protected from endothelial dysfunction and adipose inflammation [68–70]. Upon analysis, these mice showed a reduction in leukocyte accumulation and expression of pro-inflammatory genes with a concomitant reduction in adipocyte hypertrophy and free fatty acid levels [69]. Importantly, compared to transgenic mice expressing the entire human SELP gene responsible for coding P-selectin, P-selectin-deficient mice have significantly reduced atherogenesis [71].
More translationally, clinical studies have been conducted to better understand the role of PSGL-1 and P-selectin in obesity-induced disease. Through the study of gene expression levels, the SELP gene has been found to be upregulated 1.5 fold in the omentum of morbidly obese diabetic patients [72]. When stratified further, those patients with hyperlipidemia were noted to have a 2.9 fold increase in gene expression relative to those with normal lipid panel [72]. Accordingly, P-selectin is upregulated after consumption of a single high-fat meal [73] and elevated lipid and triglyceride levels are associated with increased concentrations of P-selectin at follow-up [74]. Moreover, in a study of obese women without other cardiovascular risk factors, visceral body fat was found to correlate with endothelial dysfunction and elevated circulating P-selectin levels [75]. In this group, reduction of body weight (of at least 10%) was associated with a decrease in cytokine levels reflecting a reduction in endothelial activation [75]. Lastly, in evaluation of patients with insulin resistance and metabolic syndrome, platelet expression of P-selectin, platelet-leukocyte aggregates, and plasma P-selectin were all found to be increased [76–83], including a cohort of 2,570 participants in the Framingham Heart Study who, in spite of having metabolic syndrome, were free of both overt diabetes and cardiovascular disease [84].
Taken together, the evidence in both animal models and human studies demonstrating the critical role of chronic inflammation in obesity-induced insulin resistance and metabolic syndrome suggest that novel therapies for preventing and treating resultant disease are possible through specific targeting of PSGL-1/P-selectin interactions.
APPROACHES TO BLOCKADE OF PSGL-1/P-SELECTIN INTERACTIONS
Current therapeutic approaches targeted at interfering with PSGL-1/P-selectin interactions have led to the development of PSGL-1 and/or P-selectin antibodies, the creation of recombinant immunoglobulins to competitively bind P-selectin (rPSGL-Ig), and the design of molecular therapies (Figures 2A and 2B). In this section, results from prior literature focusing on each of these approaches in various disease models will be discussed, along with foreseeable challenges with applying these approaches to the development of new treatments for obesity-induced metabolic syndrome.
Figure 2.
Approaches targeted at interfering with PSGL-1/P-selectin interactions act on either (A) P-selectin or (B) PSGL-1. Different therapies include the following: 1. anti-P-selectin antibodies, 2. P-selectin aptamers, 3. glycomimetics, 4. recombinant immunoglobulins that competitively bind P-selectin (rPSGL-Ig), 5. glycosyltransferase inhibitors, and 6. anti-PSGL-1 antibodies. PSGL-1: P-selectin glycoprotein ligand-1; EGF: epidermal growth factor domain; SCR: short consensus repeat.
Antibody-mediated therapies
Antibody-mediated therapies targeted at PSGL-1 and P-selectin have been developed not only to aid in understanding their role in cellular interactions, but also as potential treatments for clinical disease. This therapeutic modality makes use of an antibody specific to either PSGL-1 or P-selectin that after binding renders them inactive, and in turn impacts underlying disease that relies on PSGL-1/P-selectin interaction for initiation and/or progression [85]. In studies relevant to metabolic syndrome, for instance, mice with diet-induced obesity and resultant increase in mesenteric perivascular adipose tissue macrophage content, as well as vascular oxidative stress, were administered weekly injections of PSGL-1 blocking antibody [70, 86]. In these investigations, this antibody was noted to be effective in preventing endothelial dysfunction and reducing visceral adipose inflammation [70, 86]. Furthermore, through chronic inflammation and adipokine production abrogation, PSGL-1 blockade maintained local nitric oxide (NO) levels and preserved the vessel wall in a non-activated, quiescent state [70, 86]. As endothelial dysfunction is predictive of later atherosclerosis and cardiovascular complications [87–90], it is encouraging that PSGL-1 blocking antibody has been shown to inhibit visceral fat-induced atherosclerosis [91]. Further efforts investigating antibody-mediated therapies targeting PSGL-1 are summarized in Table 1.
Table 1.
Summary of animal studies evaluating anti-PSGL-1 based antibody-mediated therapies that target PSGL-1/P-Selectin interactions.
Disease | Model | Effect | References | |
---|---|---|---|---|
Inflammation | Pancreatitis | Mouse – Caerulin induced acute pancreatitis |
Reduction in leukocyte rolling | [1] |
Uveitis | Mouse – Uveitis induced by systemic injection of lipopolysaccaride |
Reduction of retinal leukostasis and severe uveitis manifestations |
[2] | |
Encephalomyelitis (EAE) |
Mouse – Myelin basic protein injection |
Reduction in EAE incidence and severity |
[3] | |
Peritonitis | Mouse – Intraperitoneal thioglycollate bouillon |
Decreased neutrophil accumulation | [4] | |
Chronic colitis | Mouse – Oral dextran sodium sulfate | Reduced disease activity index and mucosal injury |
[5] | |
Crohn’s disease | Mouse – Spontaneous ileitis | Improvement in ileitis | [6] | |
Infection | Sepsis | Mouse – Cecal ligation and puncture | Decreased leukocyte accumulation and pulmonary edema, and improved pulmonary function |
[7, 8] |
T-cell mediated |
Graft versus host | Mouse – Bone marrow transplant | Induced apoptosis of abnormally activated T-cells |
[9] |
Type I diabetes | Mouse – Non-obese diabetic | |||
Neurological | Epilepsy | Mouse – Systemic pilocarpine injection |
Prevented of disease development | [10] |
Ischemia- Reperfusion |
Intestinal | Mouse – Superior mesenteric artery clamping |
Reduced leukocyte and platelet adhesion |
[11, 12] |
Cerebral | Mouse – Bilateral common carotid artery occlusion |
Decreased leukocyte-endothelial- platelet cell interactions |
[13] | |
Malignancy | Gastric cancer | Mouse – Human gastric carcinoma tissue implant |
Decreased metastatic rate of tumor cells |
[14] |
Multiple Myeloma (MM) | Mouse – Intravenous injection of MM cell line |
Decreased proliferation and adhesion of tumor cells; increased sensitivity to chemotherapeutics |
[15] | |
Vascular disease |
Thrombosis | Mouse – Inferior vena cava ligation Mouse – Systemic IL-1β administration and carotid photochemical injury Baboon – Inferior vena cava balloon occlusion |
Decreased thrombus mass and vein wall inflammation |
[16–18] |
Neointimal Formation | Mouse – Carotid artery wire injury | Reduced neointimal formation and plaque macrophage content |
[19] | |
Hepatic | Acute obstructive cholestasis |
Mouse – Bile duct ligation | Reduced elevation in liver enzymes and decreased leukocyte rolling |
[20] |
Transplantation | Rat – Liver | Reduced leukocyte infiltration and improved hepatic function |
[21] |
REFERENCES
Abdulla A, Awla D, Hartman H, Weiber H, Jeppsson B, Regner S, et al. Platelets regulate P-selectin expression and leukocyte rolling in inflamed venules of the pancreas. Eur J Pharmacol. 2012;682:153–60.
Almulki L, Noda K, Amini R, Schering A, Garland RC, Nakao S, et al. Surprising up-regulation of P-selectin glycoprotein ligand-1 (PSGL-1) in endotoxin-induced uveitis. FASEB J. 2009;23:929–39.
Deshpande P, King IL, Segal BM. IL-12 driven upregulation of P-selectin ligand on myelin-specific T cells is a critical step in an animal model of autoimmune demyelination. J Neuroimmunol. 2006;173:35–44.
Borges E, Eytner R, Moll T, Steegmaier M, Campbell MA, Ley K, et al. The P-selectin glycoprotein ligand-1 is important for recruitment of neutrophils into inflamed mouse peritoneum. Blood. 1997;90:1934–42.
Rijcken EM, Laukoetter MG, Anthoni C, Meier S, Mennigen R, Spiegel HU, et al. Immunoblockade of PSGL-1 attenuates established experimental murine colitis by reduction of leukocyte rolling. Am J Physiol Gastrointest Liver Physiol. 2004;287:G115–24.
Inoue T, Tsuzuki Y, Matsuzaki K, Matsunaga H, Miyazaki J, Hokari R, et al. Blockade of PSGL-1 attenuates CD14+ monocytic cell recruitment in intestinal mucosa and ameliorates ileitis in SAMP1/Yit mice. J Leukoc Biol. 2005;77:287–95.
Roller J, Wang Y, Rahman M, Schramm R, Laschke MW, Menger MD, et al. Direct in vivo observations of P-selectin glycoprotein ligand-1-mediated leukocyte-endothelial cell interactions in the pulmonary microvasculature in abdominal sepsis in mice. Inflamm Res. 2013;62:275–82.
Asaduzzaman M, Rahman M, Jeppsson B, Thorlacius H. P-selectin glycoprotein-ligand-1 regulates pulmonary recruitment of neutrophils in a platelet-independent manner in abdominal sepsis. Br J Pharmacol. 2009;156:307–15.
Huang CC, Lu YF, Wen SN, Hsieh WC, Lin YC, Liu MR, et al. A novel apoptosis-inducing anti-PSGL-1 antibody for T cell-mediated diseases. Eur J Immunol. 2005;35:2239–49.
Fabene PF, Navarro Mora G, Martinello M, Rossi B, Merigo F, Ottoboni L, et al. A role for leukocyte-endothelial adhesion mechanisms in epilepsy. Nat Med. 2008;14:1377–83.
Cooper D, Chitman KD, Williams MC, Granger DN. Time-dependent platelet-vessel wall interactions induced by intestinal ischemia-reperfusion. Am J Physiol Gastrointest Liver Physiol. 2003;284:G1027–33.
Santen S, Schramm R, Menger MD, Wang Y, Jeppsson B, Thorlacius H. P-selectin glycoprotein ligand-1 regulates chemokine-dependent leukocyte recruitment in colonic ischemia-reperfusion. Inflamm Res. 2007;56:452–8.
Ishikawa M, Sekizuka E, Yamaguchi N, Nakadate H, Terao S, Granger DN, et al. Angiotensin II type 1 receptor signaling contributes to platelet-leukocyte-endothelial cell interactions in the cerebral microvasculature. Am J Physiol Heart Circ Physiol. 2007;292:H2306–15.
Chen JL, Chen WX, Zhu JS, Chen NW, Zhou T, Yao M, et al. Effect of P-selectin monoclonal antibody on metastasis of gastric cancer and immune function. World J Gastroenterol. 2003;9:1607–10.
Muz B, Azab F, de la Puente P, Rollins S, Alvarez R, Kawar Z, et al. Inhibition of P-Selectin and PSGL-1 Using Humanized Monoclonal Antibodies Increases the Sensitivity of Multiple Myeloma Cells to Bortezomib. Biomed Res Int. 2015;2015:417586.
Myers DD, Hawley AE, Farris DM, Wrobleski SK, Thanaporn P, Schaub RG, et al. P-selectin and leukocyte microparticles are associated with venous thrombogenesis. J Vasc Surg. 2003;38:1075–89.
Downing LJ, Wakefield TW, Strieter RM, Prince MR, Londy FJ, Fowlkes JB, et al. Anti-P-selectin antibody decreases inflammation and thrombus formation in venous thrombosis. J Vasc Surg. 1997;25:816–27.
Wang H, Kleiman K, Wang J, Luo W, Guo C, Eitzman DT. Deficiency of P-selectin glycoprotein ligand-1 is protective against the prothrombotic effects of interleukin-1beta. J Thromb Haemost. 2015;13:2273–6.
Phillips JW, Barringhaus KG, Sanders JM, Hesselbacher SE, Czarnik AC, Manka D, et al. Single injection of P-selectin or P-selectin glycoprotein ligand-1 monoclonal antibody blocks neointima formation after arterial injury in apolipoprotein E-deficient mice. Circulation. 2003;107:2244–9.
Dold S, Laschke MW, Zhau Y, Schilling M, Menger MD, Jeppsson B, et al. P-selectin glycoprotein ligand-1-mediated leukocyte recruitment regulates hepatocellular damage in acute obstructive cholestasis in mice. Inflamm Res. 2010;59:291–8.
Tsuchihashi S, Fondevila C, Shaw GD, Lorenz M, Marquette K, Benard S, et al. Molecular characterization of rat leukocyte P-selectin glycoprotein ligand-1 and effect of its blockade: protection from ischemia-reperfusion injury in liver transplantation. J Immunol. 2006;176:616–24.
Although the breadth of experience in the development of PSGL-1 and P-selectin blocking antibodies is extensive, major challenges have been described for antibody-mediated therapies. These drawbacks include high cost, the effectiveness of monoclonal agents as clinical therapies, limited shelf-life [92], and the potential development and consequences of acquired immunogenicity given the likely need for repeated administration [93–99].
Recombinant therapies
Recombinant strategies have also been explored to suppress PSGL-1/P-selectin interactions. This approach uses synthetic biology to develop competitive inhibitors of P-selectin [100]. To this end, researchers have expressed a soluble form of PSGL-1 through the co-transfection of Chinese hamster ovary (CHO) cells with PSGL-1 complementary DNA (cDNA) and a sialyl Lewis X (SLeX)-forming fucosyltransferase [101], yielding a functional PSGL-1 mimetic that specifically binds to P-selectin [100]. Further modifications to increase half-life and binding were performed through the addition of recombinant immunoglobulin to ultimately create rPSGL-Ig [102]. Despite reports that have found inhibitory antibodies to PSGL-1 to be more effective than rPSGL-Ig, a discrepancy attributed to dosage effect, improper timing of drug delivery, or lack of species specificity given that rPSGL-Ig is structurally based on human rather than rodent PSGL-1 [103], a number of encouraging studies in animal models using recombinant therapy have been reported (Table 2).
Table 2.
Summary of animal studies evaluating recombinant PSGL-1 (rPSGL-Ig) that targets PSGL-1/P-Selectin interactions.
Disease | Model | Effect | References | |
---|---|---|---|---|
Inflammation | Traumatic | Rat – Noble-Collip drum shock | Attenuated pathophysiological consequences and decreased leukocyte rolling |
[1] |
Pulmonary | Rat – Acid aspiration | Reduced brochoalveolar lavage neutrophil counts and pulmonary vascular permeability index |
[2] | |
Mouse – Lipopolysaccride induced lung injury |
Decreased recruitment of inflammatory cells and improved lung repair |
[3] | ||
Arthritis | Mouse – Collagen-induced arthritis | Suppressed disease progression | [4] | |
Ischemia- Reperfusion |
Intestinal | Rat – Superior mesenteric artery occlusion |
Attenuated neutrophil-endothelial cell adherence |
[5] |
Renal | Rat –Warm and in-situ perfused cold ischemia |
Decreased leukocyte infiltration and renal dysfunction |
[6] | |
Cardiac | Cat – Reversible left anterior descending artery occlusion |
Preserved vascular endothelial function and decreased myocardial injury |
[7] | |
Canine – Coronary artery balloon occlusion |
Reduced neutrophil infiltration, myeloperoxidase activity, and myocardial injury |
[8] | ||
Pig – Reversible left anterior descending artery occlusion |
Decreased infarct size and improved myocardial perfusion |
[9] | ||
Vascular Disease |
Thrombosis | Pig – Iliac artery | Accelerated thrombolysis through inhibition of leukocyte accumulation |
[10] |
Neointimal Formation | Pig – Coronary artery balloon injury | Decrease neointimal hyperplasia | [11] | |
Pig – Double common carotid artery balloon injury |
Decreased platelet and leukocyte adhesion and reduced restenosis |
[12] | ||
Transplantation | Rat – Liver | Decreased hepatocyte injury as well as neutrophil adhesion and migration |
[13] | |
Rat – Kidney | Decreased tubular cell damage and reduced amount of maintenance immunosuppression required |
[14, 15] | ||
Rat – Cardiac | Reduced infiltration of mononuclear cells and decreased myocardial infarction and apoptosis |
[16] |
REFERENCES
Scalia R, Hayward R, Armstead VE, Minchenko AG, Lefer AM. Effect of recombinant soluble P-selectin glycoprotein ligand-1 on leukocyte-endothelium interaction in vivo. Role in rat traumatic shock. Circ Res. 1999;84:93–102.
Kyriakides C, Austen W, Jr., Wang Y, Favuzza J, Moore FD, Jr., Hechtman HB. Endothelial selectin blockade attenuates lung permeability of experimental acid aspiration. Surgery. 2000;128:327–31.
Shao HZ, Qin BY. rPSGL-1-Ig, a recombinant PSGL-1-Ig fusion protein, ameliorates LPS-induced acute lung injury in mice by inhibiting neutrophil migration. Cell Mol Biol (Noisy-le-grand). 2015;61:1–6.
Sumariwalla PF, Malfait AM, Feldmann M. P-selectin glycoprotein ligand 1 therapy ameliorates established collagen-induced arthritis in DBA/1 mice partly through the suppression of tumour necrosis factor. Clin Exp Immunol. 2004;136:67–75.
Hayward R, Lefer AM. Acute mesenteric ischemia and reperfusion: protective effects of recombinant soluble P-selectin glycoprotein ligand-1. Shock. 1999;12:201–7.
Takada M, Nadeau KC, Shaw GD, Marquette KA, Tilney NL. The cytokine-adhesion molecule cascade in ischemia/reperfusion injury of the rat kidney. Inhibition by a soluble P-selectin ligand. J Clin Invest. 1997;99:2682–90.
Hayward R, Campbell B, Shin YK, Scalia R, Lefer AM. Recombinant soluble P-selectin glycoprotein ligand-1 protects against myocardial ischemic reperfusion injury in cats. Cardiovascular research. 1999;41:65–76.
Wang K, Zhou X, Zhou Z, Tarakji K, Qin JX, Sitges M, et al. Recombinant soluble P-selectin glycoprotein ligand-Ig (rPSGL-Ig) attenuates infarct size and myeloperoxidase activity in a canine model of ischemia-reperfusion. Thrombosis and Haemostasis. 2002;88:149–54.
Hansen A, Kumar A, Wolf D, Frankenbergerova K, Filusch A, Gross ML, et al. Evaluation of cardioprotective effects of recombinant soluble P-selectin glycoprotein ligand-immunoglobulin in myocardial ischemia-reperfusion injury by real-time myocardial contrast echocardiography. J Am Coll Cardiol. 2004;44:887–91.
Kumar A, Villani MP, Patel UK, Keith JC, Jr., Schaub RG. Recombinant soluble form of PSGL-1 accelerates thrombolysis and prevents reocclusion in a porcine model. Circulation. 1999;99:1363–9.
Wang K, Zhou Z, Zhou X, Tarakji K, Topol EJ, Lincoff AM. Prevention of intimal hyperplasia with recombinant soluble P-selectin glycoprotein ligand-immunoglobulin in the porcine coronary artery balloon injury model. J Am Coll Cardiol. 2001;38:577–82.
Bienvenu J-G, Tanguay J-F, Theoret J-F, Kumar A, Schaub RG, Merhi Y. Recombinant soluble P-selectin glycoprotein ligand-1-Ig reduces restenosis through inhibition of platelet-neutrophil adhesion after double angioplasty in swine. Circulation. 2001;103:1128–34.
Dulkanchainun TS, Goss JA, Imagawa DK, Shaw GD, Anselmo DM, Kaldas F, et al. Reduction of hepatic ischemia/reperfusion injury by a soluble P-selectin glycoprotein ligand-1. Ann Surg. 1998;227:832–40.
Gasser M, Waaga-Gasser AM, Grimm MW, Grimm MR, Lenhard MS, Kist-van Holthe JE, et al. Selectin blockade plus therapy with low-dose sirolimus and cyclosporin A prevent brain death-induced renal allograft dysfunction. Am J Transplantation. 2005;5:662–70.
Fuller TF, Sattler B, Binder L, Vetterlein F, Ringe B, Lorf T. Reduction of severe ischemia/reperfusion injury in rat kidney grafts by a soluble P-selectin glycoprotein ligand. Transplantation. 2001;72:216–22.
Coito AJ, Shaw GD, Li J, Ke B, Ma J, Busuttil RW, et al. Selectin-mediated interactions regulate cytokine networks and macrophage heme oxygenase-1 induction in cardiac allograft recipients. Lab Invest. 2002;82:61–70.
Given the positive evidence in animal studies, human clinical trials have been conducted to evaluate the effect of rPSGL-Ig in transplant and myocardial infarction patients. In a multi-center phase IIA study in renal allograft recipients, rPSGL-1 was noted to have no impact on the primary outcome measure of dialysis within the first week post-transplant [104]. In a separate single-center phase II study evaluating the role of rPSGL-Ig in deceased-donor liver transplant recipients, treatment was associated with a non-statistically significant trend towards decreased ischemia reperfusion injury and improved early liver function [105]. With regards to myocardial infarction, the P-Selectin Antagonist Limiting Myonecrosis (PSALM) phase II trial assessed patients with ST-segment elevation acute myocardial infarction who were treated with intravenous rPSGL-Ig as an adjunctive therapy to thrombolysis [106]. Due to a lack of significant benefit with regards to myocardial blood flow, ST-segment change resolution, left ventricular ejection fraction, and Thrombolysis In Myocardial Infarction (TIMI) flow grade, this trial was stopped early [106]. Taken together, trials evaluating rPSGL-Ig as a competitive inhibitor of P-selectin highlight the difficulty in translating convincing data from animal experiments to yield clinical benefit. Furthermore, additional challenges with continued development rPSGL-Ig therapies involve their failure to accurately recapitulate the native glycosylation pattern of PSGL-1 and their relatively low potency with IC50’s in the high micromolar range [102, 107].
Synthetic molecular therapies
Within the realm of synthetic molecular therapies, multiple mechanisms have been exploited to interfere with PSGL-1/P-selectin interactions, including the inhibition of enzymes required to make selectin ligands, the creation of aptamers to directly bind P-selectin, and the synthesis of glycomimetics to the N-terminus of PSGL-1 to competitively bind selectins.
Glycosyltransferase inhibitors
Structurally, ideal selectin ligands are glycoconjugates composed of protein or lipid scaffolds that contain a sLeX or sialyl Lewis A (sLeA) [108]. In order for sLeX or sLeA to be properly linked to these glycoconjuates, a series of glycosyltransferase reactions are performed by N-acetylglucosaminyl-, galactosyl-, sialyl-, and fucosyl-transferases [48]. As such, inhibitors have been developed in order to block the necessary steps of conjugating these moieties to the underlying glycan chain and thereby interfere with PSGL-1/P-selectin interactions by preventing the endogenous production of PSGL-1. Considering that these glycosyltransferase reactions take place in the endoplasmic reticulum and Golgi apparatus, along with the inherent drug design challenge of creating a molecule that is able to reach these structures, such inhibitors have been difficult to advance to in vivo studies. Nonetheless, progress has been reported by a number of investigators. Examples include the development of a cell permeable sialyltransferase (ST) inhibitor, 3Fax-Neu5Ac, which enters into the cell in a peracetylated form after which it is deacetylated by native cellular esterases and is subsequently able to inhibit the action of ST [109]. Although these inhibitors have been found to be beneficial in experimental models of cancer [109–111], their lack of selectivity is concerning as off-target effects have been noted, leading to significant morbidity such as irreversible renal injury [112]. Thus, recent efforts have focused on the encapsulation of these inhibitors in nanoparticles, such as poly(lactic-co-glycolic acid) (PLGA), displaying cancer targeting antibodies [113]. However, as this drug delivery based solution requires knowledge of a specific antigen to be targeted, further progress in the development of specific glycosyltransferase inhibitors for clinical use will require increased structural knowledge of native and pathogenic glycosyltransferases.
P-selectin aptamers
Representing a distinct synthetic approach to interfering with PSGL-1/P-selectin interactions, aptamers, or single-stranded oligonucleotides that bind target proteins, have been created with affinity for P-selectin [114]. Therapeutically, anti-mouse P-selectin aptamers have been shown to prevent adhesion of sickle red blood cells and leukocytes to endothelial cells in mice models of sickle cell disease [114]. Further, in a baboon model of venous thrombosis, anti-P-selectin aptamer (ARC5692) has been noted to promote vein recanalization, preserve valve competency, and decrease vein wall fibrosis [115]. Currently, however, such aptamers have not been used clinically, potentially due to challenges with cross-reactivity, interaction with intracellular targets, and degradation[116].
Glycomimetics
Most generally, glycomimetics represent a class of synthetic compounds that have structures similar to native carbohydrates. Given that PSGL-1 is a glyoconjugate, creating a glycomimetic of this ligand affords an opportunity to interfere with PSGL-1/selectin interactions. As one example, GMI-1070 is a pan-selectin inhibitor designed, in part, on the conformation of sLex in the carbohydrate binding region of E-selectin as delineated by nuclear magnetic resonance techniques [117–119]. This glycomimetic has been tested in mice models of multiple myeloma and has been shown to decrease tumor growth and progression, as well as restore sensitivity to chemotherapeutics such as bortezomib, which are challenged by drug resistance [120]. In patients with sickle cell disease, recent results of a prospective multicenter phase 2 study suggest that this compound may yield clinically meaningful reductions in vaso-occlusive crises with reduced use of opioid analgesia [121]. Although this pan-selectin inhibitor is potent for E-selectin, it requires relatively high concentrations to inhibit P-selectin, thus, limiting its use clinically for interfering with PSGL-1/P-selectin interactions [119, 121, 122]. Bimosiamose (TBC1269) is another pan-selectin inhibitor that has previously been shown to be effective in psoriasis [123] and has recently been used in patients with chronic obstructive pulmonary disease with evidence from a phase II trial suggesting that when administered as a nebulizer treatment along with standard bronchodilator therapies decreases pulmonary inflammatory cell infiltrate and cytokine production [124].
In addition to pan-selectin inhibitors, targeted small molecule glycomimetic inhibitors of P-selectin have also been developed, primarily for the management of thrombosis [125–131]. Examples include an orally administered, C2 benzyl substituted, quinolone salicylic acid [131]. PSI-697 was the first of two drugs in this class and in clinical trials did not show beneficial inhibition on platelet-monocyte aggregation [132]. Further modification of this compound has yielded PSI-421, which demonstrated improved pharmacokinetics and has been found to be effective in animal models of both arterial and venous injury [131]. Clinical trial data is unavailable.
While first generation glycomimetics of PSGL-1/selectin interactions have helped validate targets, they have demonstrated limited clinical benefit, which may be related to their low binding affinity to particular selectins. Specifically, the pan-selectin inhibitor GMI-1070 has been noted to have low activity against P-selectin with an IC50 of 423 µM [119], and bimosiamose (TBC1269) has similarly been noted to have an IC50 of 70 µM [133]. Likewise, even selective P-selectin inhibitors such as PSI-697 and PSI-421 have weak binding affinity with Kd’s of approximately 200 µM[131, 134].
NEXT GENERATION APPROACHES TO TARGET PSGL-1/P-SELECTIN INTEREACTIONS
Previous strategies aiming to interfere with PSGL-1/P-selectin interactions provide a compelling rationale for the development of more potent inhibitors that could be effective as disease-modifying agents for patients suffering from metabolic syndrome, in order to prevent the onset of diabetes as well as limit the induction of coronary atherosclerosis. Fundamentally, high affinity PSGL-1/P-selectin binding requires stereospecific interactions with clustered tyrosine sulfates (Tyr-SO3H), as well as a core-2 O-glycan, which has a sLex containing hexasaccharide (C2-O-sLex) [135–137]. To date, the design of most existing P-selectin inhibitors has focused on mimicking the C2 O-glycan bearing sLex moiety and in so doing, these approaches have been largely unsuccessful in accounting for the contribution of critical clustered tyrosine sulfates [119, 124, 130, 131, 138, 139]. Further synthetic challenges have included suboptimal selectivity in glycosylation [140], incompatible protecting groups for the synthesis of oligosaccharides [141], and the acid lability of tyrosine sulfates [142, 143], all of which have contributed to low yield of PSGL-1 GSP mimetics.
Recent advances have been achieved to address these limitations. Specifically, molecular dynamic simulations have been used to account for structural elements of PSGL-1/P-selectin interactions as determined by both crystallographic and experimental data, and guide the design of glycomimetics [144]. With added insight, a stereoselective scheme has been developed in order to achieve a multi-gram, preparative scale (>50g) synthesis of the C2 O-glycan [144]. Furthermore, in order to address the acid lability of tyrosine sulfate, discovery that these groups can be replaced with hydrolytically stable isoteric sulphonates (Fmoc-Phe (p-CH2SO3H) [145, 146] and Fmoc-Phe (p-SO3H)[147]) has led to an overall increase in GSP yield from < 0.5% when tyrosine O-sulfates were used to 24% when the corresponding sulfonate analogues were synthesized [144].
Through use of these approaches, a novel PSGL-1 glycomimetic model compound, GSnP-6, has been identified as a low nanomolar antagonist of PSGL-1/P-selectin interactions both in vitro and in vivo [144, 148]. GSnP-6 was the result of a screening program intended to inform the rational design of PSGL-1 mimetics in which critical, high-affinity, carbohydrate and peptide recognition epitopes were preserved [144]. Currently, GSnP-6 has the highest known binding affinity to human P-selectin with a Kd of 22 nM, comparable to that of native PSGL-1, which has a Kd of approximately 70 nM [149]. Furthermore, the compound has been noted to be stable at low pH (~5) as well as high temperature (60°C) [144]. In vivo experimentation characterizing platelet-leukocyte adhesion and platelet aggregation, confirmed that GSnP-6 inhibited early thromboinflammatory events [144].
Given the important physiologic role of PSGL-1/P-selectin interactions, potential side effects secondary to blockade will need to be considered. Nonetheless, studies in established models of bacterial sepsis have found that recombinant therapy can be given without worsening of infection, immune clearance, or increased mortality [150]. In murine models of lupus, exacerbation of nephritis has been noted [151, 152]. As treatment for metabolic syndrome would likely require chronic therapy, it will be important to assess these potential risks.
Although further studies are needed, it is encouraging that GSnP-6 potently blocks the adhesion of leukocyte subsets implicated in obesity-induced metabolic syndrome [144]. Most importantly, as a synthetic molecule, GSnP-6 is amenable to further modification, which provides for flexibility in drug design and delivery. Ultimately, GSnP-6 provides a unique structural scaffold for the synthesis of a range of highly potent selectin-specific antagonists, including even simpler analogues with comparable or higher selectin binding affinity.
CONCLUSIONS
Evidence in both humans and animal models have validated chronic inflammation as a promising target for the prevention and treatment of metabolic syndrome. Given the significant role of PSGL-1/P-selectin interactions in inflammation, targeting these interactions has the potential to reduce the risk of type 2 diabetes and cardiovascular disease. Although various different approaches to blocking PSGL-1/P-selectin have been the focus of prior studies, recent advancements in the synthesis of glycomimetic analogues provides a novel strategy for creating highly potent selectin-specific antagonists. As synthetic compounds can be manufactured in a cost-effective manner with a high degree of quality control and reproducibility, they warrant continued investigation as therapeutics for the increasingly prevalent, obesity related insulin resistance and metabolic syndrome.
ABBREVIATIONS
- PSGL-1
P-selectin glycoprotein ligand-1
- JNK
c-Jun N-terminal kinases
- NF-κB
nuclear factor-κB
- ACE-I
angiotensin converting enzyme inhibitors
- TLR2
toll-like receptor 2
- SPM
specialized pro-resolving mediator
- PPAR-α
peroxisome proliferator-activated receptor alpha
- MCP-1
monocyte chemoattractant protein 1
- ICAM
intracellular adhesion molecule 1
- VCAM
vascular cell adhesion protein 1
- GSP
glycosulfopeptide
- EGF
epidermal growth factor
- SCR
short consensus repeat
- EC
endothelial cell
- MPO
myeloperoxidase
- rPSGL-Ig
recombinant P-selectin glycoprotein immunoglobulin
- NO
nitric oxide
- CHO
Chinese hamster ovary
- cDNA
complementary DNA
- sLeX
sialyl Lewis X
- PSALM
P-Selectin Antagonist Limiting Myonecrosis
- TIMI
Thrombolysis In Myocardial Infarction
- sLeA
sialyl Lewis A
- ST
sialyltransferase
- PLGA
poly(lactic-co-glycolic acid)
- C2
core-2
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
REFERENCES
- 1.Grundy SM. Metabolic syndrome update. Trends Cardiovasc Med. 2016;26:364–373. doi: 10.1016/j.tcm.2015.10.004. [DOI] [PubMed] [Google Scholar]
- 2.National Cholesterol Education Program Expert Panel on Detection E, Treatment of High Blood Cholesterol in A. Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III) final report. Circulation. 2002;106:3143–3421. [PubMed] [Google Scholar]
- 3.Grundy SM. Metabolic syndrome pandemic. Arteriosclerosis, thrombosis, and vascular biology. 2008;28:629–636. doi: 10.1161/ATVBAHA.107.151092. [DOI] [PubMed] [Google Scholar]
- 4.Welty FK, Alfaddagh A, Elajami TK. Targeting inflammation in metabolic syndrome. Transl Res. 2016;167:257–280. doi: 10.1016/j.trsl.2015.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Saltiel AR, Kahn CR. Insulin signalling and the regulation of glucose and lipid metabolism. Nature. 2001;414:799–806. doi: 10.1038/414799a. [DOI] [PubMed] [Google Scholar]
- 6.Wang YC, Colditz GA, Kuntz KM. Forecasting the obesity epidemic in the aging U.S. population. Obesity. 2007;15:2855–2865. doi: 10.1038/oby.2007.339. [DOI] [PubMed] [Google Scholar]
- 7.Guilherme A, Virbasius JV, Puri V, Czech MP. Adipocyte dysfunctions linking obesity to insulin resistance and type 2 diabetes. Nat Rev Mol Cell Biol. 2008;9:367–377. doi: 10.1038/nrm2391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Olefsky JM, Glass CK. Macrophages, inflammation, and insulin resistance. Annu Rev Physiol. 2010;72:219–246. doi: 10.1146/annurev-physiol-021909-135846. [DOI] [PubMed] [Google Scholar]
- 9.Gregor MF, Hotamisligil GS. Inflammatory mechanisms in obesity. Annu Rev Immunol. 2011;29:415–445. doi: 10.1146/annurev-immunol-031210-101322. [DOI] [PubMed] [Google Scholar]
- 10.Kalupahana NS, Claycombe KJ, Moustaid-Moussa N. (n-3) Fatty acids alleviate adipose tissue inflammation and insulin resistance: Mechanistic insights. Adv Nutr. 2011;2:304–316. doi: 10.3945/an.111.000505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Osborn O, Olefsky JM. The cellular and signaling networks linking the immune system and metabolism in disease. Nat Med. 2012;18:363–374. doi: 10.1038/nm.2627. [DOI] [PubMed] [Google Scholar]
- 12.Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW., Jr Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. 2003;112:1796–1808. doi: 10.1172/JCI19246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest. 2003;112:1821–1830. doi: 10.1172/JCI19451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Gustafson B, Hammarstedt A, Andersson CX, Smith U. Inflamed adipose tissue: A culprit underlying the metabolic syndrome and atherosclerosis. Arterioscler Thromb Vasc Biol. 2007;27:2276–2283. doi: 10.1161/ATVBAHA.107.147835. [DOI] [PubMed] [Google Scholar]
- 15.Jiao P, Chen Q, Shah S, Du J, Tao B, Tzameli I, et al. Obesity-related upregulation of monocyte chemotactic factors in adipocytes: Involvement of nuclear factor-kappaB and c-Jun NH2-terminal kinase pathways. Diabetes. 2009;58:104–115. doi: 10.2337/db07-1344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Bertola A, Ciucci T, Rousseau D, Bourlier V, Duffaut C, Bonnafous S, et al. Identification of adipose tissue dendritic cells correlated with obesity-associated insulin-resistance and inducing Th17 responses in mice and patients. Diabetes. 2012;61:2238–2247. doi: 10.2337/db11-1274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Elgazar-Carmon V, Rudich A, Hadad N, Levy R. Neutrophils transiently infiltrate intra-abdominal fat early in the course of high-fat feeding. J Lipid Res. 2008;49:1894–1903. doi: 10.1194/jlr.M800132-JLR200. [DOI] [PubMed] [Google Scholar]
- 18.Talukdar S, Oh da Y, Bandyopadhyay G, Li D, Xu J, McNelis J, et al. Neutrophils mediate insulin resistance in mice fed a high-fat diet through secreted elastase. Nat Med. 2012;18:1407–1412. doi: 10.1038/nm.2885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Kintscher U, Hartge M, Hess K, Foryst-Ludwig A, Clemenz M, Wabitsch M, et al. T-lymphocyte infiltration in visceral adipose tissue: A primary event in adipose tissue inflammation and the development of obesity-mediated insulin resistance. Arterioscler Thromb Vasc Biol. 2008;28:1304–1310. doi: 10.1161/ATVBAHA.108.165100. [DOI] [PubMed] [Google Scholar]
- 20.Nishimura S, Manabe I, Nagasaki M, Eto K, Yamashita H, Ohsugi M, et al. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med. 2009;15:914–920. doi: 10.1038/nm.1964. [DOI] [PubMed] [Google Scholar]
- 21.Winer S, Chan Y, Paltser G, Truong D, Tsui H, Bahrami J, et al. Normalization of obesity-associated insulin resistance through immunotherapy. Nat Med. 2009;15:921–929. doi: 10.1038/nm.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Feuerer M, Herrero L, Cipolletta D, Naaz A, Wong J, Nayer A, et al. Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nat Med. 2009;15:930–939. doi: 10.1038/nm.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Winer DA, Winer S, Shen L, Wadia PP, Yantha J, Paltser G, et al. B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nat Med. 2011;17:610–617. doi: 10.1038/nm.2353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.DeFuria J, Belkina AC, Jagannathan-Bogdan M, Snyder-Cappione J, Carr JD, Nersesova YR, et al. B cells promote inflammation in obesity and type 2 diabetes through regulation of T-cell function and an inflammatory cytokine profile. Proc Natl Acad Sci U S A. 2013;110:5133–5138. doi: 10.1073/pnas.1215840110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Liu J, Divoux A, Sun J, Zhang J, Clement K, Glickman JN, et al. Genetic deficiency and pharmacological stabilization of mast cells reduce diet-induced obesity and diabetes in mice. Nat Med. 2009;15:940–945. doi: 10.1038/nm.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Wu D, Molofsky AB, Liang HE, Ricardo-Gonzalez RR, Jouihan HA, Bando JK, et al. Eosinophils sustain adipose alternatively activated macrophages associated with glucose homeostasis. Science. 2011;332:243–247. doi: 10.1126/science.1201475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science. 1993;259:87–91. doi: 10.1126/science.7678183. [DOI] [PubMed] [Google Scholar]
- 28.Takahashi K, Mizuarai S, Araki H, Mashiko S, Ishihara A, Kanatani A, et al. Adiposity elevates plasma MCP-1 levels leading to the increased CD11b-positive monocytes in mice. J Biol Chem. 2003;278:46654–46660. doi: 10.1074/jbc.M309895200. [DOI] [PubMed] [Google Scholar]
- 29.Wellen KE, Hotamisligil GS. Inflammation, stress, and diabetes. J Clin Invest. 2005;115:1111–1119. doi: 10.1172/JCI25102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Cai D, Yuan M, Frantz DF, Melendez PA, Hansen L, Lee J, et al. Local and systemic insulin resistance resulting from hepatic activation of IKK-beta and NF-kappaB. Nat Med. 2005;11:183–190. doi: 10.1038/nm1166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Hirosumi J, Tuncman G, Chang L, Gorgun CZ, Uysal KT, Maeda K, et al. A central role for JNK in obesity and insulin resistance. Nature. 2002;420:333–336. doi: 10.1038/nature01137. [DOI] [PubMed] [Google Scholar]
- 32.Platten M, Youssef S, Hur EM, Ho PP, Han MH, Lanz TV, et al. Blocking angiotensin-converting enzyme induces potent regulatory T cells and modulates TH1- and TH17-mediated autoimmunity. Proc Natl Acad Sci U S A. 2009;106:14948–14953. doi: 10.1073/pnas.0903958106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Kagami S, Owada T, Kanari H, Saito Y, Suto A, Ikeda K, et al. Protein geranylgeranylation regulates the balance between Th17 cells and Foxp3+ regulatory T cells. Int Immunol. 2009;21:679–689. doi: 10.1093/intimm/dxp037. [DOI] [PubMed] [Google Scholar]
- 34.Serhan CN, Hong S, Gronert K, Colgan SP, Devchand PR, Mirick G, et al. Resolvins: a family of bioactive products of omega-3 fatty acid transformation circuits initiated by aspirin treatment that counter proinflammation signals. J Exp Med. 2002;196:1025–1037. doi: 10.1084/jem.20020760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Buckley CD, Gilroy DW, Serhan CN. Proresolving lipid mediators and mechanisms in the resolution of acute inflammation. Immunity. 2014;40:315–327. doi: 10.1016/j.immuni.2014.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Moraes LA, Piqueras L, Bishop-Bailey D. Peroxisome proliferator-activated receptors and inflammation. Pharmacol Ther. 2006;110:371–385. doi: 10.1016/j.pharmthera.2005.08.007. [DOI] [PubMed] [Google Scholar]
- 37.Robinson E, Grieve DJ. Significance of peroxisome proliferator-activated receptors in the cardiovascular system in health and disease. Pharmacol Ther. 2009;122:246–263. doi: 10.1016/j.pharmthera.2009.03.003. [DOI] [PubMed] [Google Scholar]
- 38.Fleischman A, Shoelson SE, Bernier R, Goldfine AB. Salsalate improves glycemia and inflammatory parameters in obese young adults. Diabetes Care. 2008;31:289–294. doi: 10.2337/dc07-1338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Goldfine AB, Fonseca V, Jablonski KA, Pyle L, Staten MA, Shoelson SE, et al. The effects of salsalate on glycemic control in patients with type 2 diabetes: A randomized trial. Ann Intern Med. 2010;152:346–357. doi: 10.1059/0003-4819-152-6-201003160-00004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Goldfine AB, Fonseca V, Jablonski KA, Chen YD, Tipton L, Staten MA, et al. Salicylate (salsalate) in patients with type 2 diabetes: A randomized trial. Ann Intern Med. 2013;159:1–12. doi: 10.7326/0003-4819-159-1-201307020-00003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Larsen CM, Faulenbach M, Vaag A, Ehses JA, Donath MY, Mandrup-Poulsen T. Sustained effects of interleukin-1 receptor antagonist treatment in type 2 diabetes. Diabetes Care. 2009;32:1663–1668. doi: 10.2337/dc09-0533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Moore KL, Patel KD, Bruehl RE, Li F, Johnson DA, Lichenstein HS, et al. P-selectin glycoprotein ligand-1 mediates rolling of human neutrophils on P-selectin. J Cell Biol. 1995;128:661–671. doi: 10.1083/jcb.128.4.661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Vachino G, Chang XJ, Veldman GM, Kumar R, Sako D, Fouser LA, et al. P-selectin glycoprotein ligand-1 is the major counter-receptor for P-selectin on stimulated T cells and is widely distributed in non-functional form on many lymphocytic cells. J Biol Chem. 1995;270:21966–21974. doi: 10.1074/jbc.270.37.21966. [DOI] [PubMed] [Google Scholar]
- 44.Xia L, Sperandio M, Yago T, McDaniel JM, Cummings RD, Pearson-White S, et al. P-selectin glycoprotein ligand-1-deficient mice have impaired leukocyte tethering to E-selectin under flow. J Clin Invest. 2002;109:939–950. doi: 10.1172/JCI14151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Zou X, Shinde Patil VR, Dagia NM, Smith LA, Wargo MJ, Interliggi KA, et al. PSGL-1 derived from human neutrophils is a high-efficiency ligand for endothelium-expressed E-selectin under flow. Am J Physiol Cell Physiol. 2005;289:C415–C424. doi: 10.1152/ajpcell.00289.2004. [DOI] [PubMed] [Google Scholar]
- 46.Carlow DA, Gossens K, Naus S, Veerman KM, Seo W, Ziltener HJ. PSGL-1 function in immunity and steady state homeostasis. Immunol Rev. 2009;230:75–96. doi: 10.1111/j.1600-065X.2009.00797.x. [DOI] [PubMed] [Google Scholar]
- 47.Mehta P, Cummings RD, McEver RP. Affinity and kinetic analysis of P-selectin binding to P-selectin glycoprotein ligand-1. J Biol Chem. 1998;273:32506–32513. doi: 10.1074/jbc.273.49.32506. [DOI] [PubMed] [Google Scholar]
- 48.Impellizzeri D, Cuzzocrea S. Targeting selectins for the treatment of inflammatory diseases. Expert Opin Ther Targets. 2014;18:55–67. doi: 10.1517/14728222.2013.841140. [DOI] [PubMed] [Google Scholar]
- 49.Johnston GI, Bliss GA, Newman PJ, McEver RP. Structure of the human gene encoding granule membrane protein-140, a member of the selectin family of adhesion receptors for leukocytes. J Biol Chem. 1990;265:21381–21385. [PubMed] [Google Scholar]
- 50.Chen M, Geng JG. P-selectin mediates adhesion of leukocytes, platelets, and cancer cells in inflammation, thrombosis, and cancer growth and metastasis. Arch Immunol Ther Exp (Warsz) 2006;54:75–84. doi: 10.1007/s00005-006-0010-6. [DOI] [PubMed] [Google Scholar]
- 51.Lasky LA. Lectin cell adhesion molecules (LEC-CAMs): a new family of cell adhesion proteins involved with inflammation. J Cell Biochem. 1991;45:139–146. doi: 10.1002/jcb.240450204. [DOI] [PubMed] [Google Scholar]
- 52.Blann AD, Nadar SK, Lip GY. The adhesion molecule P-selectin and cardiovascular disease. Eur Heart J. 2003;24:2166–2179. doi: 10.1016/j.ehj.2003.08.021. [DOI] [PubMed] [Google Scholar]
- 53.Blann AD, Lip GY. Hypothesis: is soluble P-selectin a new marker of platelet activation? Atherosclerosis. 1997;128:135–138. doi: 10.1016/s0021-9150(96)05980-1. [DOI] [PubMed] [Google Scholar]
- 54.Evangelista V, Manarini S, Coller BS, Smyth SS. Role of P-selectin, beta2-integrins, and Src tyrosine kinases in mouse neutrophil-platelet adhesion. J Thrombosis Haemostasis. 2003;1:1048–1054. doi: 10.1046/j.1538-7836.2003.00214.x. [DOI] [PubMed] [Google Scholar]
- 55.da Costa Martins P, van den Berk N, Ulfman LH, Koenderman L, Hordijk PL, Zwaginga JJ. Platelet-monocyte complexes support monocyte adhesion to endothelium by enhancing secondary tethering and cluster formation. Arterioscler Thromb Vasc Biol. 2004;24:193–199. doi: 10.1161/01.ATV.0000106320.40933.E5. [DOI] [PubMed] [Google Scholar]
- 56.Kim KH, Barazia A, Cho J. Real-time imaging of heterotypic platelet-neutrophil interactions on the activated endothelium during vascular inflammation and thrombus Formation in live mice. J Vis Exp. 2013 doi: 10.3791/50329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Sreeramkumar V, Adrover JM, Ballesteros I, Cuartero MI, Rossaint J, Bilbao I, et al. Neutrophils scan for activated platelets to initiate inflammation. Science. 2014;346:1234–1238. doi: 10.1126/science.1256478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Zuchtriegel G, Uhl B, Hessenauer ME, Kurz AR, Rehberg M, Lauber K, et al. Spatiotemporal expression dynamics of selectins govern the sequential extravasation of neutrophils and monocytes in the acute inflammatory response. Arterioscler Thromb Vasc Biol. 2015;35:899–910. doi: 10.1161/ATVBAHA.114.305143. [DOI] [PubMed] [Google Scholar]
- 59.Zarbock A, Ley K, McEver RP, Hidalgo A. Leukocyte ligands for endothelial selectins: specialized glycoconjugates that mediate rolling and signaling under flow. Blood. 2011;118:6743–6751. doi: 10.1182/blood-2011-07-343566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Nishimura S, Manabe I, Nagasaki M, Seo K, Yamashita H, Hosoya Y, et al. In vivo imaging in mice reveals local cell dynamics and inflammation in obese adipose tissue. J Clin Invest. 2008;118:710–721. doi: 10.1172/JCI33328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Sakai A, Kume N, Nishi E, Tanoue K, Miyasaka M, Kita T. P-selectin and vascular cell adhesion molecule-1 are focally expressed in aortas of hypercholesterolemic rabbits before intimal accumulation of macrophages and T lymphocytes. Arterioscler Thromb Vasc Biol. 1997;17:310–316. doi: 10.1161/01.atv.17.2.310. [DOI] [PubMed] [Google Scholar]
- 62.Wu Y, Song P, Xu J, Zhang M, Zou MH. Activation of protein phosphatase 2A by palmitate inhibits AMP-activated protein kinase. J Biol Chem. 2007;282:9777–9788. doi: 10.1074/jbc.M608310200. [DOI] [PubMed] [Google Scholar]
- 63.Lindholm CR, Ertel RL, Bauwens JD, Schmuck EG, Mulligan JD, Saupe KW. A high-fat diet decreases AMPK activity in multiple tissues in the absence of hyperglycemia or systemic inflammation in rats. J Physiol Biochem. 2013;69:165–175. doi: 10.1007/s13105-012-0199-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Liu M, Liu F. Transcriptional and post-translational regulation of adiponectin. Biochem J. 2010;425:41–52. doi: 10.1042/BJ20091045. [DOI] [PubMed] [Google Scholar]
- 65.Ouedraogo R, Gong Y, Berzins B, Wu X, Mahadev K, Hough K, et al. Adiponectin deficiency increases leukocyte-endothelium interactions via upregulation of endothelial cell adhesion molecules in vivo. J Clin Invest. 2007;117:1718–1726. doi: 10.1172/JCI29623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Tsuchida A, Yamauchi T, Takekawa S, Hada Y, Ito Y, Maki T, et al. Peroxisome proliferator-activated receptor (PPAR)alpha activation increases adiponectin receptors and reduces obesity-related inflammation in adipose tissue: Comparison of activation of PPARalpha, PPARgamma, and their combination. Diabetes. 2005;54:3358–3370. doi: 10.2337/diabetes.54.12.3358. [DOI] [PubMed] [Google Scholar]
- 67.Trevaskis JL, Gawronska-Kozak B, Sutton GM, McNeil M, Stephens JM, Smith SR, et al. Role of adiponectin and inflammation in insulin resistance of Mc3r and Mc4r knockout mice. Obesity. 2007;15:2664–2672. doi: 10.1038/oby.2007.318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Russo HM, Wickenheiser KJ, Luo W, Ohman MK, Franchi L, Wright AP, et al. P-selectin glycoprotein ligand-1 regulates adhesive properties of the endothelium and leukocyte trafficking into adipose tissue. Circ Res. 2010;107:388–397. doi: 10.1161/CIRCRESAHA.110.218651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Sato C, Shikata K, Hirota D, Sasaki M, Nishishita S, Miyamoto S, et al. P-selectin glycoprotein ligand-1 deficiency is protective against obesity-related insulin resistance. Diabetes. 2011;60:189–199. doi: 10.2337/db09-1894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Wang H, Luo W, Wang J, Guo C, Wang X, Wolffe SL, et al. Obesity-induced endothelial dysfunction is prevented by deficiency of P-selectin glycoprotein ligand-1. Diabetes. 2012;61:3219–3227. doi: 10.2337/db12-0162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Zhang N, Liu Z, Yao L, Mehta-D’souza P, McEver RP. P-Selectin Expressed by a Human SELP Transgene Is Atherogenic in Apolipoprotein E-Deficient Mice. Arterioscler Thromb Vasc Biol. 2016;36:1114–1121. doi: 10.1161/ATVBAHA.116.307437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Hindle AK, Edwards C, McCaffrey T, Fu S, Brody F. Identification of cardiovascular genes in omentum from morbidly obese patients with type 2 diabetes. Int J Obes (Lond) 2010;34:1020–1027. doi: 10.1038/ijo.2010.23. [DOI] [PubMed] [Google Scholar]
- 73.Broijersen A, Karpe F, Hamsten A, Goodall AH, Hjemdahl P. Alimentary lipemia enhances the membrane expression of platelet P-selectin without affecting other markers of platelet activation. Atherosclerosis. 1998;137:107–113. doi: 10.1016/s0021-9150(97)00260-8. [DOI] [PubMed] [Google Scholar]
- 74.Fontes JD, Yamamoto JF, Larson MG, Wang N, Dallmeier D, Rienstra M, et al. Clinical correlates of change in inflammatory biomarkers: The Framingham Heart Study. Atherosclerosis. 2013;228:217–223. doi: 10.1016/j.atherosclerosis.2013.01.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Ziccardi P, Nappo F, Giugliano G, Esposito K, Marfella R, Cioffi M, et al. Reduction of inflammatory cytokine concentrations and improvement of endothelial functions in obese women after weight loss over one year. Circulation. 2002;105:804–809. doi: 10.1161/hc0702.104279. [DOI] [PubMed] [Google Scholar]
- 76.Arteaga RB, Chirinos JA, Soriano AO, Jy W, Horstman L, Jimenez JJ, et al. Endothelial microparticles and platelet and leukocyte activation in patients with the metabolic syndrome. Am J Cardiol. 2006;98:70–74. doi: 10.1016/j.amjcard.2006.01.054. [DOI] [PubMed] [Google Scholar]
- 77.Schneider DJ, Hardison RM, Lopes N, Sobel BE, Brooks MM Pro-Thrombosis Ancillary Study G. Association between increased platelet P-selectin expression and obesity in patients with type 2 diabetes: A BARI 2D (Bypass Angioplasty Revascularization Investigation 2 Diabetes) substudy. Diabetes Care. 2009;32:944–949. doi: 10.2337/dc08-1308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.De Pergola G, Pannacciulli N, Coviello M, Scarangella A, Di Roma P, Caringella M, et al. sP-selectin plasma levels in obesity: Association with insulin resistance and related metabolic and prothrombotic factors. Nutr Metab Cardiovasc Dis. 2008;18:227–232. doi: 10.1016/j.numecd.2006.09.010. [DOI] [PubMed] [Google Scholar]
- 79.Gallistl S, Sudi KM, Borkenstein M, Weinhandl G, Zotter H, Muntean W. Correlation between cholesterol, soluble P-selectin, and D-dimer in obese children and adolescents. Blood Coagul Fibrinolysis. 2000;11:755–760. doi: 10.1097/00001721-200012000-00009. [DOI] [PubMed] [Google Scholar]
- 80.Kvasnicka T, Kvasnicka J, Ceska R, Vrablik M. Increase of inflammatory state in overweight adults with combined hyperlipidemia. Nutr Metab Cardiovasc Dis. 2003;13:227–231. doi: 10.1016/s0939-4753(03)80015-7. [DOI] [PubMed] [Google Scholar]
- 81.Ingelsson E, Hulthe J, Lind L. Inflammatory markers in relation to insulin resistance and the metabolic syndrome. Eur J Clin Invest. 2008;38:502–509. doi: 10.1111/j.1365-2362.2008.01962.x. [DOI] [PubMed] [Google Scholar]
- 82.Straface E, Gambardella L, Canali E, Metere A, Gabrielli N, Arcieri R, et al. P-Selectin as a new gender associated biomarker in patients with metabolic syndrome. Int J Cardiol. 2010;145:570–571. doi: 10.1016/j.ijcard.2010.05.043. [DOI] [PubMed] [Google Scholar]
- 83.Gokulakrishnan K, Deepa R, Mohan V, Gross MD. Soluble P-selectin and CD40L levels in subjects with prediabetes, diabetes mellitus, and metabolic syndrome - The Chennai Urban Rural Epidemiology Study. Metabolism. 2006;55:237–242. doi: 10.1016/j.metabol.2005.08.019. [DOI] [PubMed] [Google Scholar]
- 84.Dallmeier D, Larson MG, Vasan RS, Keaney JF, Jr, Fontes JD, Meigs JB, et al. Metabolic syndrome and inflammatory biomarkers: A community-based cross-sectional study at the Framingham Heart Study. Diabetol Metab Syndr. 2012;4:28. doi: 10.1186/1758-5996-4-28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Huang CC, Lu YF, Wen SN, Hsieh WC, Lin YC, Liu MR, et al. A novel apoptosis-inducing anti-PSGL-1 antibody for T cell-mediated diseases. Eur J Immunol. 2005;35:2239–2249. doi: 10.1002/eji.200525849. [DOI] [PubMed] [Google Scholar]
- 86.Lee WJ, Chait A, Kim F. P-selectin glycoprotein ligand-1: A cellular link between perivascular adipose inflammation and endothelial dysfunction. Diabetes. 2012;61:3070–3071. doi: 10.2337/db12-1036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Aggoun Y, Farpour-Lambert NJ, Marchand LM, Golay E, Maggio AB, Beghetti M. Impaired endothelial and smooth muscle functions and arterial stiffness appear before puberty in obese children and are associated with elevated ambulatory blood pressure. Eur Heart J. 2008;29:792–799. doi: 10.1093/eurheartj/ehm633. [DOI] [PubMed] [Google Scholar]
- 88.Konishi M, Sugiyama S, Sato Y, Oshima S, Sugamura K, Nozaki T, et al. Pericardial fat inflammation correlates with coronary artery disease. Atherosclerosis. 2010;213:649–655. doi: 10.1016/j.atherosclerosis.2010.10.007. [DOI] [PubMed] [Google Scholar]
- 89.Alexopoulos N, McLean DS, Janik M, Arepalli CD, Stillman AE, Raggi P. Epicardial adipose tissue and coronary artery plaque characteristics. Atherosclerosis. 2010;210:150–154. doi: 10.1016/j.atherosclerosis.2009.11.020. [DOI] [PubMed] [Google Scholar]
- 90.Ueno K, Anzai T, Jinzaki M, Yamada M, Jo Y, Maekawa Y, et al. Increased epicardial fat volume quantified by 64-multidetector computed tomography is associated with coronary atherosclerosis and totally occlusive lesions. Circ J. 2009;73:1927–1933. doi: 10.1253/circj.cj-09-0266. [DOI] [PubMed] [Google Scholar]
- 91.Ohman MK, Luo W, Wang H, Guo C, Abdallah W, Russo HM, et al. Perivascular visceral adipose tissue induces atherosclerosis in apolipoprotein E deficient mice. Atherosclerosis. 2011;219:33–39. doi: 10.1016/j.atherosclerosis.2011.07.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Weiss WFt, Young TM, Roberts CJ. Principles, approaches, and challenges for predicting protein aggregation rates and shelf life. J Pharm Sci. 2009;98:1246–1277. doi: 10.1002/jps.21521. [DOI] [PubMed] [Google Scholar]
- 93.Baert F, Noman M, Vermeire S, Van Assche G, G DH, Carbonez A, et al. Influence of immunogenicity on the long-term efficacy of infliximab in Crohn’s disease. N Engl J Med. 2003;348:601–608. doi: 10.1056/NEJMoa020888. [DOI] [PubMed] [Google Scholar]
- 94.Bartelds GM, Wijbrandts CA, Nurmohamed MT, Stapel S, Lems WF, Aarden L, et al. Clinical response to adalimumab: Relationship to anti-adalimumab antibodies and serum adalimumab concentrations in rheumatoid arthritis. Ann Rheum Dis. 2007;66:921–926. doi: 10.1136/ard.2006.065615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Polman CH, O’Connor PW, Havrdova E, Hutchinson M, Kappos L, Miller DH, et al. A randomized, placebo-controlled trial of natalizumab for relapsing multiple sclerosis. N Engl J Med. 2006;354:899–910. doi: 10.1056/NEJMoa044397. [DOI] [PubMed] [Google Scholar]
- 96.Brennan PJ, Rodriguez Bouza T, Hsu FI, Sloane DE, Castells MC. Hypersensitivity reactions to mAbs: 105 desensitizations in 23 patients, from evaluation to treatment. J Allergy Clin Immunol. 2009;124:1259–1266. doi: 10.1016/j.jaci.2009.09.009. [DOI] [PubMed] [Google Scholar]
- 97.Emi Aikawa N, de Carvalho JF, Artur Almeida Silva C, Bonfa E. Immunogenicity of Anti-TNF-alpha agents in autoimmune diseases. Clin Rev Allergy Immunol. 2010;38:82–89. doi: 10.1007/s12016-009-8140-3. [DOI] [PubMed] [Google Scholar]
- 98.De Simone C, Amerio P, Amoruso G, Bardazzi F, Campanati A, Conti A, et al. Immunogenicity of anti-TNFalpha therapy in psoriasis: A clinical issue? Expert Opin Biol Ther. 2013;13:1673–1682. doi: 10.1517/14712598.2013.848194. [DOI] [PubMed] [Google Scholar]
- 99.Piaserico S, Conti A, Lo Console F, De Simone C, Prestinari F, Mazzotta A, et al. Efficacy and safety of systemic treatments for psoriasis in elderly patients. Acta Derm Venereol. 2014;94:293–297. doi: 10.2340/00015555-1719. [DOI] [PubMed] [Google Scholar]
- 100.Sako D, Comess KM, Barone KM, Camphausen RT, Cumming DA, Shaw GD. A sulfated peptide segment at the amino terminus of PSGL-1 is critical for P-selectin binding. Cell. 1995;83:323–331. doi: 10.1016/0092-8674(95)90173-6. [DOI] [PubMed] [Google Scholar]
- 101.Kumar R, Camphausen RT, Sullivan FX, Cumming DA. Core2 beta-1,6-N-acetylglucosaminyltransferase enzyme activity is critical for P-selectin glycoprotein ligand-1 binding to P-selectin. Blood. 1996;88:3872–3879. [PubMed] [Google Scholar]
- 102.Kumar A, Villani MP, Patel UK, Keith JC, Jr, Schaub RG. Recombinant soluble form of PSGL-1 accelerates thrombolysis and prevents reocclusion in a porcine model. Circulation. 1999;99:1363–1369. doi: 10.1161/01.cir.99.10.1363. [DOI] [PubMed] [Google Scholar]
- 103.Myers DD, Hawley AE, Farris DM, Wrobleski SK, Thanaporn P, Schaub RG, et al. P-selectin and leukocyte microparticles are associated with venous thrombogenesis. J Vasc Surg. 2003;38:1075–1089. doi: 10.1016/s0741-5214(03)01033-4. [DOI] [PubMed] [Google Scholar]
- 104.Gaber AO, Mulgaonkar S, Kahan BD, Woodle ES, Alloway R, Bajjoka I, et al. YSPSL (rPSGL-Ig) for improvement of early renal allograft function: A double-blind, placebo-controlled, multi-center phase IIa study. Clinical Transplantation. 2011;25:523–533. doi: 10.1111/j.1399-0012.2010.01295.x. [DOI] [PubMed] [Google Scholar]
- 105.Busuttil RW, Lipshutz GS, Kupiec-Weglinski JW, Ponthieux S, Gjertson DW, Cheadle C, et al. rPSGL-Ig for improvement of early liver allograft function: A double-blind, placebo-controlled, single-center phase II study. Am J Transplantation. 2011;11:786–797. doi: 10.1111/j.1600-6143.2011.03441.x. [DOI] [PubMed] [Google Scholar]
- 106.Mertens P, Maes A, Nuyts J, Belmans A, Desmet W, Esplugas E, et al. Recombinant P-selectin glycoprotein ligand-immunoglobulin, a P-selectin antagonist, as an adjunct to thrombolysis in acute myocardial infarction. The P-Selectin Antagonist Limiting Myonecrosis (PSALM) trial. Am Heart J. 2006;152:125, e1–e8. doi: 10.1016/j.ahj.2006.04.020. [DOI] [PubMed] [Google Scholar]
- 107.Khor SP, McCarthy K, Dupont M, Murray K, Timony G. Pharmacokinetics, pharmacodynamics, allometry, and dose selection of rPSGL-Ig for phase I trial. J Pharmacol ExpTherapeutics. 2000;293:618–624. [PubMed] [Google Scholar]
- 108.Koenig A, Jain R, Vig R, Norgard-Sumnicht KE, Matta KL, Varki A. Selectin inhibition: synthesis and evaluation of novel sialylated, sulfated and fucosylated oligosaccharides, including the major capping group of GlyCAM-1. Glycobiology. 1997;7:79–93. doi: 10.1093/glycob/7.1.79. [DOI] [PubMed] [Google Scholar]
- 109.Rillahan CD, Antonopoulos A, Lefort CT, Sonon R, Azadi P, Ley K, et al. Global metabolic inhibitors of sialyl- and fucosyltransferases remodel the glycome. Nat Chem Biol. 2012;8:661–668. doi: 10.1038/nchembio.999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Macauley MS, Arlian BM, Rillahan CD, Pang PC, Bortell N, Marcondes MC, et al. Systemic blockade of sialylation in mice with a global inhibitor of sialyltransferases. J Biol Chem. 2014;289:35149–35158. doi: 10.1074/jbc.M114.606517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Bull C, Boltje TJ, Wassink M, de Graaf AM, van Delft FL, den Brok MH, et al. Targeting aberrant sialylation in cancer cells using a fluorinated sialic acid analog impairs adhesion, migration, and in vivo tumor growth. Mol Cancer Ther. 2013;12:1935–1946. doi: 10.1158/1535-7163.MCT-13-0279. [DOI] [PubMed] [Google Scholar]
- 112.Galeano B, Klootwijk R, Manoli I, Sun M, Ciccone C, Darvish D, et al. Mutation in the key enzyme of sialic acid biosynthesis causes severe glomerular proteinuria and is rescued by N-acetylmannosamine. J Clin Invest. 2007;117:1585–1594. doi: 10.1172/JCI30954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Bull C, Boltje TJ, van Dinther EA, Peters T, de Graaf AM, Leusen JH, et al. Targeted delivery of a sialic acid-blocking glycomimetic to cancer cells inhibits metastatic spread. ACS Nano. 2015;9:733–745. doi: 10.1021/nn5061964. [DOI] [PubMed] [Google Scholar]
- 114.Gutsaeva DR, Parkerson JB, Yerigenahally SD, Kurz JC, Schaub RG, Ikuta T, et al. Inhibition of cell adhesion by anti-P-selectin aptamer: A new potential therapeutic agent for sickle cell disease. Blood. 2011;117:727–735. doi: 10.1182/blood-2010-05-285718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Diaz JA, Wrobleski SK, Alvarado CM, Hawley AE, Doornbos NK, Lester PA, et al. P-selectin inhibition therapeutically promotes thrombus resolution and prevents vein wall fibrosis better than enoxaparin and an inhibitor to von Willebrand factor. Arterioscler Thromb Vasc Biol. 2015;35:829–837. doi: 10.1161/ATVBAHA.114.304457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Lakhin AV, Tarantul VZ, Gening LV. Aptamers: problems, solutions and prospects. Acta Naturae. 2013;5:34–43. [PMC free article] [PubMed] [Google Scholar]
- 117.Rinnbauer M, Ernst B, Wagner B, Magnani J, Benie AJ, Peters T. Epitope mapping of sialyl Lewis(x) bound to E-selectin using saturation transfer difference NMR experiments. Glycobiology. 2003;13:435–443. doi: 10.1093/glycob/cwg043. [DOI] [PubMed] [Google Scholar]
- 118.Scheffler K, Brisson JR, Weisemann R, Magnani JL, Wong WT, Ernst B, et al. Application of homonuclear 3D NMR experiments and 1D analogs to study the conformation of sialyl Lewis(x) bound to E-selectin. J Biomol NMR. 1997;9:423–436. doi: 10.1023/a:1018358929268. [DOI] [PubMed] [Google Scholar]
- 119.Chang J, Patton JT, Sarkar A, Ernst B, Magnani JL, Frenette PS. GMI-1070, a novel pan-selectin antagonist, reverses acute vascular occlusions in sickle cell mice. Blood. 2010;116:1779–1786. doi: 10.1182/blood-2009-12-260513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Azab AK, Quang P, Azab F, Pitsillides C, Thompson B, Chonghaile T, et al. P-selectin glycoprotein ligand regulates the interaction of multiple myeloma cells with the bone marrow microenvironment. Blood. 2012;119:1468–1478. doi: 10.1182/blood-2011-07-368050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Telen MJ, Wun T, McCavit TL, De Castro LM, Krishnamurti L, Lanzkron S, et al. Randomized phase 2 study of GMI-1070 in SCD: reduction in time to resolution of vaso-occlusive events and decreased opioid use. Blood. 2015;125:2656–2664. doi: 10.1182/blood-2014-06-583351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Muz B, Azab F, de la Puente P, Rollins S, Alvarez R, Kawar Z, et al. Inhibition of P-Selectin and PSGL-1 Using Humanized Monoclonal Antibodies Increases the Sensitivity of Multiple Myeloma Cells to Bortezomib. Biomed Res Int. 2015;2015:417586. doi: 10.1155/2015/417586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Friedrich M, Bock D, Philipp S, Ludwig N, Sabat R, Wolk K, et al. Pan-selectin antagonism improves psoriasis manifestation in mice and man. Arch Dermatol Res. 2006;297:345–351. doi: 10.1007/s00403-005-0626-0. [DOI] [PubMed] [Google Scholar]
- 124.Watz H, Bock D, Meyer M, Schierhorn K, Vollhardt K, Woischwill C, et al. Inhaled pan-selectin antagonist Bimosiamose attenuates airway inflammation in COPD. Pulm Pharmacol Ther. 2013;26:265–270. doi: 10.1016/j.pupt.2012.12.003. [DOI] [PubMed] [Google Scholar]
- 125.Myers DD, Jr, Henke PK, Bedard PW, Wrobleski SK, Kaila N, Shaw G, et al. Treatment with an oral small molecule inhibitor of P selectin (PSI-697) decreases vein wall injury in a rat stenosis model of venous thrombosis. J Vasc Surg. 2006;44:625–632. doi: 10.1016/j.jvs.2006.05.021. [DOI] [PubMed] [Google Scholar]
- 126.Myers DD, Jr, Rectenwald JE, Bedard PW, Kaila N, Shaw GD, Schaub RG, et al. Decreased venous thrombosis with an oral inhibitor of P selectin. J Vasc Surg. 2005;42:329–336. doi: 10.1016/j.jvs.2005.04.045. [DOI] [PubMed] [Google Scholar]
- 127.Myers DD, Jr, Wrobleski SK, Longo C, Bedard PW, Kaila N, Shaw GD, et al. Resolution of venous thrombosis using a novel oral small-molecule inhibitor of P-selectin (PSI-697) without anticoagulation. Thrombosis Haemostasis. 2007;97:400–407. [PubMed] [Google Scholar]
- 128.Kaila N, Janz K, DeBernardo S, Bedard PW, Camphausen RT, Tam S, et al. Synthesis and biological evaluation of quinoline salicylic acids as P-selectin antagonists. J Med Chem. 2007;50:21–39. doi: 10.1021/jm0602256. [DOI] [PubMed] [Google Scholar]
- 129.Kaila N, Janz K, Huang A, Moretto A, DeBernardo S, Bedard PW, et al. 2-(4-Chlorobenzyl)-3-hydroxy-7,8,9,10-tetrahydrobenzo[H]quinoline-4-carboxylic acid (PSI-697): identification of a clinical candidate from the quinoline salicylic acid series of P-selectin antagonists. J Med Chem. 2007;50:40–64. doi: 10.1021/jm060631p. [DOI] [PubMed] [Google Scholar]
- 130.Meier TR, Myers DD, Jr, Wrobleski SK, Zajkowski PJ, Hawley AE, Bedard PW, et al. Prophylactic P-selectin inhibition with PSI-421 promotes resolution of venous thrombosis without anticoagulation. Thromb Haemost. 2008;99:343–351. doi: 10.1160/TH07-10-0608. [DOI] [PubMed] [Google Scholar]
- 131.Huang A, Moretto A, Janz K, Lowe M, Bedard PW, Tam S, et al. Discovery of 2-[1-(4-chlorophenyl)cyclopropyl]-3-hydroxy-8-(trifluoromethyl)quinoline-4-carbox ylic acid (PSI-421), a P-selectin inhibitor with improved pharmacokinetic properties and oral efficacy in models of vascular injury. J Med Chem. 2010;53:6003–6017. doi: 10.1021/jm9013696. [DOI] [PubMed] [Google Scholar]
- 132.Japp AG, Chelliah R, Tattersall L, Lang NN, Meng X, Weisel K, et al. Effect of PSI-697, a novel P-selectin inhibitor, on platelet-monocyte aggregate formation in humans. J Am Heart Assoc. 2013;2:e006007. doi: 10.1161/JAHA.112.006007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Beeh KM, Beier J, Meyer M, Buhl R, Zahlten R, Wolff G. Bimosiamose, an inhaled small-molecule pan-selectin antagonist, attenuates late asthmatic reactions following allergen challenge in mild asthmatics: A randomized, double-blind, placebo-controlled clinical cross-over-trial. Pulm Pharmacol Ther. 2006;19:233–241. doi: 10.1016/j.pupt.2005.07.004. [DOI] [PubMed] [Google Scholar]
- 134.Japp AGCR, Tattersall L, Lang NN, Meng X, Weisel K, Katz A, Burt D, Fox KA, Feuerstein GZ, Connolly TM, Newby DE. Effect of PSI-697, a novel P-selectin inhibitor, on platelet-monocyte aggregate formation in humans. J Am Heart Assoc. 2013;2:1–6. doi: 10.1161/JAHA.112.006007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Leppanen A, Mehta P, Ouyang YB, Ju T, Helin J, Moore KL, et al. A novel glycosulfopeptide binds to P-selectin and inhibits leukocyte adhesion to P-selectin. J Biol Chem. 1999;274:24838–24848. doi: 10.1074/jbc.274.35.24838. [DOI] [PubMed] [Google Scholar]
- 136.Leppanen A, White SP, Helin J, McEver RP, Cummings RD. Binding of glycosulfopeptides to P-selectin requires stereospecific contributions of individual tyrosine sulfate and sugar residues. J Biol Chem. 2000;275:39569–39578. doi: 10.1074/jbc.M005005200. [DOI] [PubMed] [Google Scholar]
- 137.Leppänen A, Yago TIOV, McEver RP, Cummings RD. Model glycosulfopeptides from P-selectin glycoprotein ligand-1 require tyrosine sulfation and a core 2-branched O-glycan to bind to L-selectin. J Biol Chem. 2003;278:26391–26400. doi: 10.1074/jbc.M303551200. [DOI] [PubMed] [Google Scholar]
- 138.Kaila N, Janz K, DeBernardo S, Bedard PW, Camphausen RT, Tam S, et al. Synthesis and biological evaluation of quinoline salicylic acids as P-selectin antagonists. J Med Chem. 2007;50:21–39. doi: 10.1021/jm0602256. [DOI] [PubMed] [Google Scholar]
- 139.Kranich R, Busemann AS, Bock D, Schroeter-Maas S, Beyer D, Heinemann B, et al. Rational design of novel, potent small molecule pan-selectin antagonists. J Med Chem. 2007;50:1101–1115. doi: 10.1021/jm060536g. [DOI] [PubMed] [Google Scholar]
- 140.Baumann K, Kowalczyk D, Kunz H. Total synthesis of the glycopeptide recognition domain of the P-selectin glycoprotein ligand 1. Angew Chem Int Ed. 2008;47:3445–3449. doi: 10.1002/anie.200705762. [DOI] [PubMed] [Google Scholar]
- 141.Vohra Y, Buskas T, Boons GJ. Rapid assembly of oligosaccharides: A highly convergent strategy for the assembly of a glycosylated amino acid derived from PSGL-1. J Org Chem. 2009;74:6064–6071. doi: 10.1021/jo901135k. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Huang KT, Wu BC, Lin CC, Luo SC, Chen C, Wong CH, et al. Multi-enzyme one-pot strategy for the synthesis of sialyl Lewis X-containing PSGL-1 glycopeptide. Carbohydrate Res. 2006;341:2151–2155. doi: 10.1016/j.carres.2006.04.047. [DOI] [PubMed] [Google Scholar]
- 143.Koeller KM, Smith MEB, Huang R-F, Wong C-H. Chemoenzymatic synthesis of a PSGL-1 N-terminal glycopeptide containing tyrosine sulfate and α-O-linked sialyl Lewis X. J Am Chem Soc. 2000;122:4241–4242. [Google Scholar]
- 144.Krishnamurthy VR, Sardar MY, Ying Y, Song X, Haller C, Dai E, et al. Glycopeptide analogues of PSGL-1 inhibit P-selectin in vitro and in vivo. Nat Commun. 2015;6:6387. doi: 10.1038/ncomms7387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Lam SN, Acharya P, Wyatt R, Kwong PD, Bewley CA. Tyrosine-sulfate isosteres of CCR5 N-terminus as tools for studying HIV-1 entry. Bioorg Med Chem. 2008;16:10113–10120. doi: 10.1016/j.bmc.2008.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Roosenburg S, Laverman P, Joosten L, Eek A, Oyen WJ, de Jong M, et al. Stabilized (111)in-labeled sCCK8 analogues for targeting CCK2-receptor positive tumors: Synthesis and evaluation. Bioconjug Chem. 2010;21:663–670. doi: 10.1021/bc900465y. [DOI] [PubMed] [Google Scholar]
- 147.Acharya P, Dogo-Isonagie C, LaLonde JM, Lam SN, Leslie GJ, Louder MK, et al. Structure-based identification and neutralization mechanism of tyrosine sulfate mimetics that inhibit HIV-1 entry. ACS Chem Biol. 2011;6:1069–1077. doi: 10.1021/cb200068b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Krishnamurthy VR, Dougherty A, Kamat M, Song X, Cummings RD, Chaikof EL. Synthesis of an Fmoc-threonine bearing core-2 glycan: A building block for PSGL-1 via Fmoc-assisted solid-phase peptide synthesis. Carbohydr Res. 2010;345:1541–1547. doi: 10.1016/j.carres.2010.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Ushiyama S, Laue TM, Moore KL, Erickson HP, McEver RP. Structural and functional characterization of monomeric soluble P-selectin and comparison with membrane P-selectin. J Biol Chem. 1993;268:15229–15237. [PubMed] [Google Scholar]
- 150.Opal SM, Sypek JP, Keith JC, Jr, Schaub RG, Palardy JE, Parejo NA. Evaluation of the safety of recombinant P-selectin glycoprotein ligand-immunoglobulin G fusion protein in experimental models of localized and systemic infection. Shock. 2001;15:285–290. doi: 10.1097/00024382-200115040-00006. [DOI] [PubMed] [Google Scholar]
- 151.He X, Schoeb TR, Panoskaltsis-Mortari A, Zinn KR, Kesterson RA, Zhang J, et al. Deficiency of P-selectin or P-selectin glycoprotein ligand-1 leads to accelerated development of glomerulonephritis and increased expression of CC chemokine ligand 2 in lupus-prone mice. J Immunol. 2006;177:8748–8756. doi: 10.4049/jimmunol.177.12.8748. [DOI] [PubMed] [Google Scholar]
- 152.Wang H, Knight JS, Hodgin JB, Wang J, Guo C, Kleiman K, et al. Psgl-1 Deficiency is Protective against Stroke in a Murine Model of Lupus. Sci Rep. 2016;6:28997. doi: 10.1038/srep28997. [DOI] [PMC free article] [PubMed] [Google Scholar]