Next Article in Journal
Metabolomic Insights of the Effects of Bacterial Algicide IRI-160AA on Dinoflagellate Karlodinium veneficum
Next Article in Special Issue
Influence of Genetic West African Ancestry on Metabolomics among Hypertensive Patients
Previous Article in Journal
The MicroRNA miR-277 Controls Physiology and Pathology of the Adult Drosophila Midgut by Regulating the Expression of Fatty Acid β-Oxidation-Related Genes in Intestinal Stem Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Glycobiology of Pulmonary Arterial Hypertension

Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Metabolites 2022, 12(4), 316; https://doi.org/10.3390/metabo12040316
Submission received: 1 March 2022 / Revised: 23 March 2022 / Accepted: 28 March 2022 / Published: 1 April 2022

Abstract

:
Pulmonary arterial hypertension (PAH) is a progressive pulmonary vascular disease of complex etiology. Cases of PAH that do not receive therapy after diagnosis have a low survival rate. Multiple reports have shown that idiopathic PAH, or IPAH, is associated with metabolic dysregulation including altered bioavailability of nitric oxide (NO) and dysregulated glucose metabolism. Multiple processes such as increased proliferation of pulmonary vascular cells, angiogenesis, apoptotic resistance, and vasoconstriction may be regulated by the metabolic changes demonstrated in PAH. Recent reports have underscored similarities between metabolic abnormalities in cancer and IPAH. In particular, increased glucose uptake and altered glucose utilization have been documented and have been linked to the aforementioned processes. We were the first to report a link between altered glucose metabolism and changes in glycosylation. Subsequent reports have highlighted similar findings, including a potential role for altered metabolism and aberrant glycosylation in IPAH pathogenesis. This review will detail research findings that demonstrate metabolic dysregulation in PAH with an emphasis on glycobiology. Furthermore, this report will illustrate the similarities in the pathobiology of PAH and cancer and highlight the novel findings that researchers have explored in the field.

1. Introduction

Pulmonary vascular diseases (PVDs) are a group of pulmonary ailments resulting from precapillary or postcapillary hypertension and include pulmonary embolism, chronic thromboembolic disease, pulmonary veno-occlusive disease, arteriovenous malformations, pulmonary edema, and pulmonary hypertension (PH) [1,2]. Several clinical studies have estimated that PVDs occur in 20–25 million persons worldwide; however, the occurrence as well as the pathogenesis still remain unknown. Currently, PH is the most complex and widely studied PVD and is therefore the focus of this review.
In 2009, PH was defined by a mean pulmonary artery pressure (mPAP) of ≥25 mm Hg at rest that is measured during right heart catheterization (RHC) [3,4]. It was recently recommended to lower the mPAP threshold to ≥20 mm Hg in 2019 based on scientific studies [5,6]. PH is often associated with other PVDs or diseases such as congenital heart disease, coronary artery disease, liver cirrhosis, and COPD, as well as personal and environmental factors such as tobacco use, drug use, and toxin stimuli. Therefore, PH has been classified by the World Health Organization (WHO) as a five-group system according to etiology: Group 1, PAH; Group 2, PH due to left heart disease; Group 3, PH due to chronic lung disease or hypoxia; Group 4, Chronic Thromboembolic PH (CTEPH); and Group 5, PH due to unclear multifactorial mechanisms [5].
Pulmonary Arterial Hypertension (PAH), the Group 1 subcategory of PH, is characterized hemodynamically by the presence of pre-capillary PH with an end-expiratory pulmonary artery wedge pressure (PAWP) ≤ 15 mm Hg and a pulmonary vascular resistance >3 Wood units [7], which frequently leads to right ventricular (RV) hypertrophy/failure and early mortality. RHC remains essential for a diagnosis of IPAH [8]. From the time of diagnosis, PAH has an average survival of 2–3 years without treatment and affects mostly women; however, men diagnosed with PAH have lower survival rates [9,10]. The pathobiology of PAH is complex; features include vasoconstriction, inflammation, genetics/epigenetics, and/or environmental cues [11,12,13,14,15,16] as well as increased cell proliferation, vascular remodeling, and angiogenesis [17,18,19,20,21,22]. Furthermore, metabolic dysfunction that includes deficits in nitric oxide (NO) production [23,24,25,26,27], insulin resistance [28,29,30,31], leptin dysregulation [32,33,34], dyslipidemia, and increased lipid oxidation [35,36,37,38,39,40,41,42] have been established in IPAH and may affect disease progression. Currently, the primary therapies for PAH are those that target the mediators of vasoconstriction (e.g., NO, prostacyclin, or endothelin) [23,43,44,45], while other pathomechanisms including cell proliferation, increased angiogenesis, inflammation, and plexiform lesions (a hallmark of PAH) have no known treatment options. This review will discuss metabolic dysregulation with an emphasis on novel findings in glycobiology observed in Group 1 PAH, where the most of the investigations have been documented.

1.1. Metabolism Studies in PAH

Over the years, evidence for the role of metabolites in PAH has been documented and determined to affect the many processes mentioned above. Other reports have shown that metabolites including glucose [30,46,47,48,49], ammonia [50], arginine [51,52], glutamine [53,54,55], and fatty acids are dysregulated [35,37,41,56]. Interestingly, altered glucose influx into cells can lead to glucose intolerance in PAH, and studies have shown that glycated hemoglobin levels correlate with idiopathic PAH (IPAH) diagnosis [30,57]. This suggests that chronic hyperglycemia may perpetuate glucose influx and insulin resistance in IPAH patients.
Several reports have also emphasized the similarities between cancer and PAH [58,59,60,61]. For example, cancer cells, such as PAH, display increased cellular glucose uptake and altered glucose metabolism [62,63,64,65]. Highly proliferative cancer cells require immense energy and utilize excess metabolites, a characteristic that has become a primary research focus in PAH. Highly proliferative cells have been postulated to alter their glycolytic metabolism rates and to shift from glucose oxidation and CO2 production to aerobic glycolysis despite normal oxygen levels (also known as the Warburg effect) [66,67]. Rapidly growing cells benefit from aerobic glycolysis because it improves their survival rate when total cell numbers increase, and the oxygen supply is drastically reduced. Highly proliferative cells, which switch to aerobic glycolysis, yield less ATP than those that utilize mitochondrial respiration (oxidative phosphorylation). In turn, these cells take up glucose more rapidly when levels are abundantly available.
Similar findings of vascular cell proliferation and changes in glucose metabolism have been determined in IPAH. In particular, (18F)-Fluoro-deoxyglucose-PET (FDG-PET) scans have shown that the lungs of patients with IPAH have increased uptake of glucose and have a higher glycolytic rate than healthy individuals [48,68]. In addition, IPAH endothelial cells display three-fold more glycolysis than normal cells, which contributes to an increased rate of growth and proliferation [68]. Other studies have shown that pulmonary vascular cells from several PH animal models and IPAH human tissue have hyperpolarized mitochondria and suppressed glucose oxidation and respiration. Glycolysis in PAH is enhanced, and mitochondrial dysfunction with reduced oxidative phosphorylation has also been observed in PAH (and cancer) [69,70,71,72,73,74]. Multiple reports have shown that a key enzyme, pyruvate dehydrogenase (PDH), which regulates pyruvate influx (a product of glycolysis) into the mitochondria, is inhibited and results in the suppression of glucose oxidation [72,75]. The role of PDH in PAH (and cancer) has been demonstrated through pharmacological inhibition with dichloroacetate (DCA), a known activator of PDH, and knockout of fatty acid metabolic enzymes [41,76,77,78,79]. Both of these interventions alter the ‘glycolytic shift’, augment glucose oxidation, and reverse PAH phenotypes in several models of PAH. In the PAH pulmonary vasculature, cells have been postulated to adapt to these metabolic alterations (nutrient stress conditions) by reprograming their metabolism and protein homeostasis, resulting in increased survival and proliferation.

1.2. Carbohydrate Metabolism and Glycosylation

Most of the proteins produced by the human body contain linked sugar chains or glycans, and all multicellular organisms utilize these molecules as biosignals in normal physiology [80,81,82]. Glycans are formed as secondary gene products by the concerted action of glycosyltransferases, glycosidases, and high energy sugar nucleotides (e.g., UDP-GlcNAc, UDP-GalNAc, and CMP-sialic acid). Therefore, the biosynthesis of glycans is not controlled by an interventional template, and their structures are much less rigidly defined than those of proteins and nucleic acids. In addition, carbohydrates and glycans are dynamic and altered by the cellular microenvironment. This is exemplified by malignant cells in which metabolic dysregulation has been shown to alter glycan structures [83,84,85]. An investigation of glycosylation changes driven by disease has already produced some novel insights. In particular, changes in the molar proteoglycan (PG) ratios have been demonstrated in patients with aging skin and with arthritis [86,87]. In addition, altered glycan patterns of glycolipids have been linked to aging [88,89,90], and glycan changes on glycoproteins have also been shown in patients with Alzheimer’s disease [91,92,93,94,95,96,97] as well as cardiovascular disease [98,99,100], COPD [101,102,103,104,105,106], and other pulmonary diseases [107,108,109,110,111,112,113,114,115]. Studying glycans, carbohydrate precursors/pathways, and/or the glycan machinery (i.e., glycosyltransferases and glycosylhydrolases) may hold essential information that is critical for uncovering mechanism(s) in these metabolic diseases, including PAH, where these post-translational modifications have not been widely studied.

1.3. Hexosamine Biosynthetic Pathway

The hexosamine biosynthetic pathway (HBP) has been documented as a cellular sensor for nutrient uptake [116,117,118,119]. Indeed, cells during proliferation use excess metabolites, namely glucose, glutamine, acetyl-CoA, and uridine triphosphate (UTP) to fuel their demanding energy requirements, all of which are also funneled into the HBP. This pathway is critical for the synthesis of the highly ‘energy’ charged sugar nucleotide, UDP-GlcNAc, which is one of the most fundamentally important building blocks for all glycosylation events. UDP-GlcNAc is also a precursor to other sugar nucleotides such as UDP-GalNAc and CMP-sialic acid that are essential for generating oligosaccharides, which make up all glycoconjugates (e.g., glycoproteins and proteoglycans) and glycosaminoglycans (GAGs).

1.4. Intracellular Glycosylation (O-GlcNAc) in PAH

UDP-GlcNAc biosynthesis has been most studied for its role as the substrate/precursor for the O-linked N-Acetylglucosamine (O-GlcNAc) post-translational protein modification. The ‘GlcNAc’ moiety from UDP-GlcNAc is transferred and covalently attached to proteins on serine/threonine residues [120,121] through catalytic activity of the O-GlcNAc transferase (OGT). Conversely, the removal of ‘GlcNAc’ is performed by O-GlcNAc hydrolase (OGA) [122,123,124]. The O-GlcNAc modification is found throughout the cell including the cytoplasm, nucleus, and mitochondria [125,126,127]. O-GlcNAc has been demonstrated to have an antagonistic role on phosphorylation [128,129,130]. Since its discovery, more than 1900 articles have characterized protein O-GlcNAcylation with over 5000 human proteins identified to bear O-GlcNAc [131].
The addition/removal of O-GlcNAc is a dynamic process under the influence of the cellular environment (i.e., stress, hormones, and nutrient flux) [132,133,134,135,136,137,138]. For this reason, the O-GlcNAc modification has been well documented as a nutrient sensor and flux mediator [123,125,133,137,139]. Alterations in glucose uptake, similar to those found in IPAH, can regulate protein O-GlcNAc levels and are likely to influence protein activity/function. On the other hand, an imbalance in the OGT, OGA, and O-GlcNAc has been documented to impact glucose utilization in cancer metabolism [140,141,142,143,144,145]. The crux of O-GlcNAc modification is regulated by the concentration of UDP-GlcNAc sugar pools, which is governed by flux into the HBP, suggesting that the intrinsic mechanisms and regulators of the pathway may be controlled by O-GlcNAc.
The O-GlcNAc modification of proteins in vascular disease has been described [100,135,146,147,148,149]. However, the exact role of the O-GlcNAc modification in vascular disease is still poorly understood. In particular, the role of this modification in PAH is just beginning to be recognized. We recently showed that alterations in glucose uptake and flux into the HBP led to augmented OGT expression/activity, increased proliferation in IPAH pulmonary artery smooth muscle cells (PASMCs), and faster time to clinical worsening in PAH (as defined by hospitalization, lung transplantation, or death; n = 86 PAH patients) [139]. In addition, we later demonstrated that reductions in O-GlcNAc levels contributed to impaired IPAH vascular sprouting and de novo vascularization [150] as well as alterations in eNOS activity and NO production [151]. Other studies followed our initial reports showing a connection between increased O-GlcNAc, RV glucose uptake, RV metabolic derangements, and RV function in PAH [152,153]. These findings indicate a direct role for the O-GlcNAc modification in PAH disease pathology.

1.5. The Role of Extracellular Matrix (ECM) Glycosaminoglycans (GAGs) in PAH

ECM is a dynamic scaffold with a fundamental role in regulating tissue and cellular function. The ECM is made up of proteins and PGs that greatly influence ECM development, stem cell differentiation, cellular morphology, and cell signaling [154]. In disease pathology, dysregulated ECM remodeling has been shown to contribute to dysfunctional cellular processes such as cell signaling, migration, proliferation, and adhesion. In particular, PGs are known to facilitate the aforementioned processes. In addition, PGs are proteins glycosylated with anionic GAGs [155,156,157,158,159,160,161]. GAG chains are typically long and unbranched with alternating disaccharide units of either D-glucuronic or L-iduronic acid and either GlcNAc or GalNAc. Often, one or both sugars contain sulfate moieties with the exception of hyaluronic acid (HA) that does not bear sulfation and is not covalently linked to a core protein. PGs are categorized by their GAG chain and fall into groups containing either heparan sulfate (HS), chondroitin sulfate (CS), dermatan sulfate (DS), or keratan sulfate (KS) [162]. Based on the vascular abnormalities (e.g., remodeling, intimal thickening, and plexiform lesion formation) observed in PAH, it is clear that altered PG biosynthesis/turnover may contribute to the pathogenesis of the disease.
In PAH, remodeling of the pulmonary vasculature and neovascularization are known consequences of disruptive changes to the ECM of vascular cells/tissue. Interestingly enough, hypoxia has been shown to potentiate GAG synthesis in human primary lung fibroblasts from normal lungs [163,164] and may offer similar regulation of GAG synthesis/deposition in PH. In PH animal models, ECM remodeling has been suggested to occur during early hypoxia exposure, which precedes increases in RV systolic pressure (RVSP) and RV hypertrophy [165,166]. Furthermore, hypoxia, vascular dysfunction, and dysregulated glucose metabolism, all of which are contributors to IPAH pathogenesis, have been suggested to modulate PG synthesis/turnover [167,168,169,170,171,172,173,174]. Several groups have identified specific PGs and their potential role in IPAH including HA, perlecan, versican, aggrecan, and syndecan.

1.6. Hyaluronan (HA) in PAH

HA is a large GAG composed of an alternate repeat of two sugars, GlcNAc and glucuronic acid (GlcUA), and is found in the pericellular matrix and ECM [175]. HA also exists in the synovial fluid, dermis, and vitreous body [176]. HA is formed at the cell membrane by any of three HA synthase enzymes (HAS1, HAS2, or HAS3) by alternately adding GlcUA and GlcNAc to the reducing end of the nascent polysaccharide, which is extruded through the cell membrane into the extracellular space [177,178]. Conversely, the turnover/degradation of HA differs from tissue to tissue and is performed by the hyaluronidases (Hyal1 and Hyal2). Recent reports have shown that HA synthesis is controlled through cytosolic UDP-GlcNAc levels generated from the HBP [179,180]. Through the regulatory production of UDP-GlcNAc sugar pools, the HBP controls not only the intracellular energy state of the cell but also the extracellular integrity surrounding the cell through HA production.
Multiple studies over the past few decades have contributed to our understanding of HA and its role in pulmonary health and disease [181,182,183,184]. Once thought to be only a scaffolding molecule of the ECM, research has shown that HA is an important active regulator of inflammation, airway hyperresponsiveness [185,186,187], lung injury [188,189,190], and fibrosis in the lung [112,113,188,191]. The presence of HA in IPAH has also been described. Elevated levels of HA in IPAH were first demonstrated by Aytekin et al. in 2008 [192]. They showed increased plasma HA levels in IPAH patients as well as abnormal levels in the plexogenic lesions and PASMCs. In addition, Lauer, Aytekin, et al. showed increased binding of inflammatory cells to a pathological form of HA (covalently modified with heavy chains from inter-alpha-inhibitor) in IPAH, suggesting that there may be a role for HA in regarding the processes of remodeling and inflammation in the disease [193]. Others showed similar findings, where HA content was elevated in IPAH compared to control donor lungs [194], as well as a potential role for endothelin-1 in HA synthesis and THP-1 monocyte adhesion [195].
Even though the role of HA has been recognized in PAH, its regulation and precise functional role in the disease still remains unknown. Cell culture models may hold the key to understanding the regulatory mechanisms associated with HA synthesis. In culture models, HA production can be induced by inhibitors of protein synthesis as well as the viral mimetic polyinosinic acid:polycytidylic (poly I:C) [196]. Similarly, the addition of high glucose to cell cultures can stimulate HA synthesis in cultured cell models [197]. A possible mechanism for the increase in HA levels in PASMCs may be due to the vast uptake of glucose [49,139], similar to the uptake described in PAECs [68]. HA, along with other GAGs, in PAH may be functionally involved in the remodeling process as well as the metabolic abnormalities that contribute to the overall disease pathology including angiogenesis, proliferation, and inflammation.

1.7. Perlecan in PAH

Heparan sulfate proteoglycans (HSPGs) contain a core protein linked to HS GAG chains [154]. Examples of HSPGs are perlecan, agrin, type XVIII collagen α1, and glypican. HSPGs are located in the ECM and provide structural support as well as promote cell-to-cell and cell-to-matrix interactions, and cell motility and migration [198]. In addition, the HSPGs interact with cytokines, chemokines, morphogens, and growth factors while facilitating spatial/temporal cell signaling and morphogen gradient formation between cells and in tissues [155,157,199,200]. Perlecan, a major HSPG in the vasculature, is important in vasculogenesis and maintaining vascular tone [201,202]. Interestingly, both perlecan and agrin were found to be higher in PAH patients [203]. Most of the research that has been carried out has been conducted on perlecan, which has been observed in high abundance in junctions between PAH PAECs and PASMCs [204,205,206]. In addition, the role of perlecan in PAH may be linked to its involvement in EC barrier function, cell-to-cell interactions between PAECs and PASMCs, and inhibition of SMC proliferation [204], all of which are known to be dysregulated in the disease pathogenesis.

1.8. Versican and Aggrecan in PAH

Versican and aggrecan both belong to a PG family called lecticans. These chondroitin sulfate proteoglycans (CSPGs) also contain DS and KS, are found in the ECM, and are the largest and most highly glycosylated PGs. Aggrecan is the best studied member of this group due to its abundance in cartilage and brain and has the ability to withstand compressive forces as well as changes in water composition (desorption/resorption) [154]. Versican is predominantly found in the pericellular and interstitial ECM of connective tissues, blood vessels, brain, and leukocytes [154] and has been documented for its role in cell adhesion, migration, and proliferation [207,208]. Interestingly enough, both versican and aggrecan can form super molecules with HA in the ECM. Recently, levels of CSPGs were found to be high in PAH PASMCs. In particular, versican was shown to be increased in medial thickened regions, the neointima, and in the plexiform lesions as well as overall lung tissues of patients with IPAH [209]. Aggrecan was also found in the intima, media, and adventitia of both small and large IPAH vessels [210]. These data suggest that CSPGs may have a pathogenic role in PAH and may be involved in vascular remodeling and cell proliferation.

1.9. Other PGs in PAH

Syndecan is a PG that contains both HS and CS GAGs and is made up of four members (Syndecan 1–4). These PGs are membrane bound and are found in most nucleated cells [154]. Syndecan has been shown to be dysregulated in several cancers and facilitates cell adhesion, migration, and actin cytoskeletal organization as well as to interact with multiple growth factors and matrix components [211,212,213]. Syndecan can be cleaved, shedding an active ectodomain containing GAGs, which retain potent biological activity [214,215,216]. It was recently reported that while plasma levels of Syndecan-1 (along with other GAGs/PGs) increased in monocrotaline (MCT)-induced PAH rats, they decreased in the pulmonary arteries compared to wild-type controls [217], suggesting destruction of the pulmonary vascular glycocalyx. Other reports have shown that prolargin, which interacts with PGs (and does not contain GAGs), has some potential in differentiating PH subtypes based on the level of pulmonary vascular remodeling [218], indirectly suggesting that PGs are involved in the disease remodeling process. Overall, these data suggest that Syndecan, as well as other PG interacting proteins, may regulate vascular remodeling and contribute to overall PAH disease severity.

1.10. Galectins (Carbohydrate Lectins) in PAH

Galectins (Gal) are β-galactoside-binding lectins expressed in several cells types and tissues [219]. They are involved in immune responses such as inflammation, metabolism, wound healing, autophagy, signaling, and angiogenesis [220,221,222,223,224]. Currently, there are 15 different galectins that have been identified. They have been classified based on their biochemical structure into three groups: (1) Gals-1, -2, -5, -7, -10, -11, -13, -14, and -15 exists as monomers or dimers and display a single carbohydrate-recognition domain (CRD); (2) Gals-4, -6, -8, -9, and -12 form tandem-repeats and have two CRDs with a linker peptide; and (3) Gal-3 (chimera-type) CRD and an non-lectin domain with the ability to oligomerize [225].
Gal-1 and -3 have been studied in cancer, fibrosis, infection, and auto-immune disease and are known to interact with signaling molecules, transcriptional regulators, lysosomal proteins, cell surface receptors, and ECM proteins through glycans on the surface of these proteins [224,225,226,227]. They have also been shown to promote angiogenesis through interaction with a specific glycoform of vascular endothelial growth factor receptor (VEGFR) [228,229] and have been studied in cardiovascular disease [230,231].
In PAH, the majority of the investigations involve the functional consequences of increased Gal-3 in the disease [232,233,234,235]. This may be due to the uniqueness of Gal-3 and its single classification, limiting redundancy from other isoforms during the investigation. Nevertheless, Gal-1 has also been studied in PAH [236] though there is no literature exploring the other galectin isoforms in the disease. Interestingly, direct investigation of carbohydrate-galectin interactions has not been studied in PAH. It is worth postulating that these carbohydrate ligands may be affected by the same environmental cues (e.g., nutrient levels, ROS, etc.) that modulate metabolic pathways in PAH. In particular, the alterations in glycan machinery (glycosyltransferases and glycosidases), levels of sugar nucleotide substrates, and overall glycan composition could be similar to our findings with OGT, UDP-GlcNAc, and O-GlcNAc in IPAH [150]. Therefore, strategies to determine the changes in glycan as well as the glycosyltransferases/hydrolases involved are likely to be of interest.

1.11. Sialylation in PAH

Not much is known about the role of sialylation in PAH. Recently, Morrow et al. reported in a FASEB journal abstract that IgG sialylation/glycosylation was increased in PAH patients, affecting IgG binding to endothelial cells [237]. Free sialic acid, which is a signature found in cancer and cardiovascular disease, was also suggested to be increased in the serum of PAH patients compared to controls in another abstract by Morrow et al. [238]. Other studies showed that sialylation of the von Willebrand factor, a known marker for worse survival rates in patients with PAH [239], was reduced in PAH (or precapillary PH) [240]. These data suggest that altered sialic acid levels in PAH may have a role in the disease pathogenesis; specifically, terminal sialylation of glycoproteins can influence the binding and function galectins as well as other carbohydrate lectins.

2. Conclusions

Pulmonary hypertension has key features of progressive thickening and remodeling of the vasculature caused by endothelial dysfunction and aberrant pulmonary vascular cell proliferation. Tremendous effort has been put into investigating the pathobiology of PAH and key features that could reverse/slow the disease process. In particular, there is a need to explore more therapeutic options other than vasodilation in PAH. Our knowledge of metabolic dysfunction in PAH has advanced over the years; however, there are still important molecular details regulated by metabolic changes that we do not fully understand, and investigating glycans and their role in PAH may hold key details.
It is clear from past reports in cancer as well as recent reports in PAH that aberrant glycosylation can cause or perpetuate disease processes (Figure 1). Intracellular glycosylation such as the O-GlcNAc modification is highly dynamic, and its tight regulation by the cell is important in PAH. Targeting the O-GlcNAc transferase or the O-GlcNAc hydrolase may help to slow or reverse the metabolic changes. Indeed, the O-GlcNAc modification is important in the regulation of specific PAH cellular phenotypes and more studies are needed to determine specific proteins that may be under the control of the O-GlcNAc modification in the disease.
PGs/GAGs (HA) and galectins are increased in PAH. In addition, sialylation may be altered. However, there specific roles and how they contribute to vascular remodeling and regulation of vascular function as well as cell proliferation, cell signaling, and cell differentiation/morphology have not been defined. Are the changes observed in these glycans/glycoconjugates a consequence of the metabolic derangements observed in PAH? Or do these glycoconjugates, when altered, have a direct role in the PAH pathogenesis? Studying the specific role of glycans and/or glycoconjugates is critical to fully understanding the molecular mechanisms associated with dysregulated metabolism in PAH. These efforts cannot move forward without some technical challenges, which are contingent on the availability and accessibility of better tools for quantifying and precisely determining the roles of glycans and glycosylation events in cell biology. Targeted approaches to regulating glycosyltransferases/hydrolases, sugar nucleotides, and/or specific glycoconjugates/glycoforms are needed to advance our knowledge of pulmonary hypertension as well as other diseases defined by metabolic dysregulation.

Author Contributions

Conceptualization, J.W.B. and S.K.; writing—original draft preparation, S.V., P.C., S.K. and J.W.B.; writing—review and editing, S.V., P.C., J.S.D., S.K. and J.W.B. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Institutes of Health (R01HL152246 to J.W.B. and R01HL160911 to S.K.).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mélot, C.; Naeije, R. Pulmonary vascular diseases. Compr. Physiol. 2011, 1, 593–619. [Google Scholar] [PubMed]
  2. Barnes, J.W.; Tonelli, A.R.; Heresi, G.A.; Newman, J.E.; Mellor, N.E.; Grove, D.E.; Dweik, R.A. Novel Methods in Pulmonary Hypertension Phenotyping in the Age of Precision Medicine (2015 Grover Conference Series). Pulm. Circ. 2016, 6, 439–447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Badesch, D.B.; Champion, H.C.; Sanchez, M.A.G.; Hoeper, M.M.; Loyd, J.E.; Manes, A.; McGoon, M.; Naeije, R.; Olschewski, H.; Oudiz, R.J.; et al. Diagnosis and Assessment of Pulmonary Arterial Hypertension. J. Am. Coll. Cardiol. 2009, 54, S55–S66. [Google Scholar] [CrossRef] [PubMed]
  4. Galiè, N.; Hoeper, M.M.; Humbert, M.; Torbicki, A.; Vachiery, J.L.; Barbera, J.A.; Beghetti, M.; Corris, P.; Gaine, S.; Gibbs, J.S.; et al. Guidelines for the diagnosis and treatment of pulmonary hypertension: The Task Force for the Diagnosis and Treatment of Pulmonary Hypertension of the European Society of Cardiology (ESC) and the European Respiratory Society (ERS), endorsed by the International Society of Heart and Lung Transplantation (ISHLT). Eur. Heart J. 2009, 30, 2493–2537. [Google Scholar]
  5. Simonneau, G.; Montani, D.; Celermajer, D.S.; Denton, C.P.; Gatzoulis, M.A.; Krowka, M.; Williams, P.G.; Souza, R. Haemodynamic definitions and updated clinical classification of pulmonary hypertension. Eur. Respir. J. 2019, 53, 1801913. [Google Scholar] [CrossRef]
  6. Simonneau, G.; Hoeper, M.M. The revised definition of pulmonary hypertension: Exploring the impact on patient management. Eur. Heart J. 2019, 21, K4–K8. [Google Scholar] [CrossRef] [Green Version]
  7. Hoeper, M.M.; Bogaard, H.J.; Condliffe, R.; Frantz, R.; Khanna, D.; Kurzyna, M.; Langleben, D.; Manes, A.; Satoh, T.; Torres, F.; et al. Definitions and diagnosis of pulmonary hypertension. J. Am. Coll. Cardiol. 2013, 62, D42–D50. [Google Scholar] [CrossRef] [Green Version]
  8. Galie, N.; Humbert, M.; Vachiery, J.L.; Gibbs, S.; Lang, I.; Torbicki, A.; Simonneau, G.; Peacock, A.; Vonk Noordegraaf, A.; Beghetti, M.; et al. 2015 ESC/ERS Guidelines for the Diagnosis and Treatment of Pulmonary Hypertension. Rev. Esp. Cardiol. (Engl. Ed.) 2016, 69, 177. [Google Scholar]
  9. Memon, H.A.; Park, M.H. Pulmonary Arterial Hypertension in Women. Methodist Debakey Cardiovasc. J. 2017, 13, 224–237. [Google Scholar] [CrossRef]
  10. D’Alonzo, G.E.; Barst, R.J.; Ayres, S.M.; Bergofsky, E.H.; Brundage, B.H.; Detre, K.M.; Fishman, A.P.; Goldring, R.M.; Groves, B.M.; Kernis, J.T.; et al. Survival in patients with primary pulmonary hypertension. Results from a national prospective registry. Ann. Intern. Med. 1991, 115, 343–349. [Google Scholar] [CrossRef]
  11. Hassoun, P.M. Pulmonary Arterial Hypertension. N. Engl. J. Med. 2021, 385, 2361–2376. [Google Scholar] [CrossRef] [PubMed]
  12. Yan, Y.; He, Y.Y.; Jiang, X.; Wang, Y.; Chen, J.W.; Zhao, J.H.; Ye, J.; Lian, T.Y.; Zhang, X.; Zhang, R.J.; et al. DNA methyltransferase 3B deficiency unveils a new pathological mechanism of pulmonary hypertension. Sci. Adv. 2020, 6, eaba2470. [Google Scholar] [CrossRef]
  13. Wang, X.-J.; Lian, T.-Y.; Jiang, X.; Liu, S.-F.; Li, S.-Q.; Jiang, R.; Wu, W.-H.; Ye, J.; Cheng, C.-Y.; Du, Y.; et al. Germline BMP9 mutation causes idiopathic pulmonary arterial hypertension. Eur. Respir. J. 2019, 53, 1801609. [Google Scholar] [CrossRef] [PubMed]
  14. Evans, J.D.; Girerd, B.; Montani, D.; Wang, X.J.; Galie, N.; Austin, E.D.; Elliott, G.; Asano, K.; Grunig, E.; Yan, Y.; et al. BMPR2 mutations and survival in pulmonary arterial hypertension: An individual participant data meta-analysis. Lancet Respir. Med. 2016, 4, 129–137. [Google Scholar] [CrossRef] [Green Version]
  15. Wang, X.-J.; Xu, X.-Q.; Sun, K.; Liu, K.-Q.; Li, S.-Q.; Jiang, X.; Zhao, Q.-H.; Wang, L.; Peng, F.-H.; Ye, J.; et al. Association of Rare PTGIS Variants With Susceptibility and Pulmonary Vascular Response in Patients With Idiopathic Pulmonary Arterial Hypertension. JAMA Cardiol. 2020, 5, 677–684. [Google Scholar] [CrossRef] [Green Version]
  16. Klouda, T.; Yuan, K. Inflammation in Pulmonary Arterial Hypertension. Adv. Exp. Med. Biol. 2021, 1303, 351–372. [Google Scholar]
  17. Voelkel, N.F.; Gomez-Arroyo, J.; Abbate, A.; Bogaard, H.J.; Nicolls, M.R. Pathobiology of pulmonary arterial hypertension and right ventricular failure. Eur. Respir. J. 2012, 40, 1555–1565. [Google Scholar] [CrossRef] [Green Version]
  18. Rabinovitch, M.; Guignabert, C.; Humbert, M.; Nicolls, M.R. Inflammation and immunity in the pathogenesis of pulmonary arterial hypertension. Circ. Res. 2014, 115, 165–175. [Google Scholar] [CrossRef]
  19. Meloche, J.; Renard, S.; Provencher, S.; Bonnet, S. Anti-inflammatory and immunosuppressive agents in PAH. Handb. Exp. Pharmacol. 2013, 218, 437–476. [Google Scholar]
  20. Humbert, M.; Guignabert, C.; Bonnet, S.; Dorfmuller, P.; Klinger, J.R.; Nicolls, M.R.; Olschewski, A.J.; Pullamsetti, S.S.; Schermuly, R.T.; Stenmark, K.R.; et al. Pathology and pathobiology of pulmonary hypertension: State of the art and research perspectives. Eur. Respir. J. 2019, 53, 1801887. [Google Scholar] [CrossRef] [Green Version]
  21. Tuder, R.M.; Chacon, M.; Alger, L.; Wang, J.; Taraseviciene-Stewart, L.; Kasahara, Y.; Cool, C.D.; Bishop, A.E.; Geraci, M.; Semenza, G.L.; et al. Expression of angiogenesis-related molecules in plexiform lesions in severe pulmonary hypertension: Evidence for a process of disordered angiogenesis. J. Pathol. 2001, 195, 367–374. [Google Scholar] [CrossRef] [PubMed]
  22. Jeffery, T.K.; Morrell, N.W. Molecular and cellular basis of pulmonary vascular remodeling in pulmonary hypertension. Prog. Cardiovasc. Dis. 2002, 45, 173–202. [Google Scholar] [CrossRef]
  23. Koress, C.; Swan, K.; Kadowitz, P. Soluble Guanylate Cyclase Stimulators and Activators: Novel Therapies for Pulmonary Vascular Disease or a Different Method of Increasing cGMP? Curr. Hypertens. Rep. 2016, 18, 42. [Google Scholar] [CrossRef] [PubMed]
  24. Xu, W.; Kaneko, F.T.; Zheng, S.; Comhair, S.A.; Janocha, A.J.; Goggans, T.; Thunnissen, F.B.; Farver, C.; Hazen, S.L.; Jennings, C.; et al. Increased arginase II and decreased NO synthesis in endothelial cells of patients with pulmonary arterial hypertension. FASEB J. 2004, 18, 1746–1748. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Kaneko, F.T.; Arroliga, A.C.; Dweik, R.A.; Comhair, S.A.; Laskowski, D.; Oppedisano, R.; Thomassen, M.J.; Erzurum, S.C. Biochemical reaction products of nitric oxide as quantitative markers of primary pulmonary hypertension. Am. J. Respir. Crit. Care Med. 1998, 158, 917–923. [Google Scholar] [CrossRef]
  26. Giaid, A.; Saleh, D. Reduced expression of endothelial nitric oxide synthase in the lungs of patients with pulmonary hypertension. N. Engl. J. Med. 1995, 333, 214–221. [Google Scholar] [CrossRef]
  27. Rubin, L.J. Primary pulmonary hypertension. N. Engl. J. Med. 1997, 336, 111–117. [Google Scholar] [CrossRef] [Green Version]
  28. Brunner, N.W.; Skhiri, M.; Fortenko, O.; Hsi, A.; Haddad, F.; Khazeni, N.; Zamanian, R.T. Impact of insulin resistance on ventricular function in pulmonary arterial hypertension. J. Heart Lung Transplant. 2014, 33, 721–726. [Google Scholar] [CrossRef]
  29. Hansmann, G.; Wagner, R.A.; Schellong, S.; Perez, V.A.; Urashima, T.; Wang, L.; Sheikh, A.Y.; Suen, R.S.; Stewart, D.J.; Rabinovitch, M. Pulmonary arterial hypertension is linked to insulin resistance and reversed by peroxisome proliferator-activated receptor-gamma activation. Circulation 2007, 115, 1275–1284. [Google Scholar] [CrossRef]
  30. Pugh, M.E.; Robbins, I.M.; Rice, T.W.; West, J.; Newman, J.H.; Hemnes, A.R. Unrecognized glucose intolerance is common in pulmonary arterial hypertension. J. Heart Lung Transplant. 2011, 30, 904–911. [Google Scholar] [CrossRef]
  31. Zamanian, R.T.; Hansmann, G.; Snook, S.; Lilienfeld, D.; Rappaport, K.M.; Reaven, G.M.; Rabinovitch, M.; Doyle, R.L. Insulin resistance in pulmonary arterial hypertension. Eur. Respir. J. 2009, 33, 318–324. [Google Scholar] [CrossRef]
  32. Aytekin, M.; Tonelli, A.R.; Farver, C.F.; Feldstein, A.E.; Dweik, R.A. Leptin deficiency recapitulates the histological features of pulmonary arterial hypertension in mice. Int. J. Clin. Exp. Pathol. 2014, 7, 1935–1946. [Google Scholar]
  33. Santos, M.; Reis, A.; Goncalves, F.; Ferreira-Pinto, M.J.; Cabral, S.; Torres, S.; Leite-Moreira, A.F.; Henriques-Coelho, T. Adiponectin levels are elevated in patients with pulmonary arterial hypertension. Clin. Cardiol. 2014, 37, 21–25. [Google Scholar] [CrossRef]
  34. Tonelli, A.R.; Aytekin, M.; Feldstein, A.E.; Dweik, R.A. Leptin Levels Predict Survival in Pulmonary Arterial Hypertension. Pulm. Circ. 2012, 2, 214–219. [Google Scholar] [CrossRef] [Green Version]
  35. Talati, M.; Hemnes, A. Fatty acid metabolism in pulmonary arterial hypertension: Role in right ventricular dysfunction and hypertrophy. Pulm. Circ. 2015, 5, 269–278. [Google Scholar] [CrossRef] [Green Version]
  36. Cracowski, J.L.; Cracowski, C.; Bessard, G.; Pepin, J.L.; Bessard, J.; Schwebel, C.; Stanke-Labesque, F.; Pison, C. Increased lipid peroxidation in patients with pulmonary hypertension. Am. J. Respir. Crit. Care Med. 2001, 164, 1038–1042. [Google Scholar] [CrossRef]
  37. Brittain, E.L.; Talati, M.; Fessel, J.P.; Zhu, H.; Penner, N.; Calcutt, M.W.; West, J.D.; Funke, M.; Lewis, G.D.; Gerszten, R.E.; et al. Fatty Acid Metabolic Defects and Right Ventricular Lipotoxicity in Human Pulmonary Arterial Hypertension. Circulation 2016, 133, 1936–1944. [Google Scholar] [CrossRef]
  38. Cathey, S.S. Breath analysis in pulmonary arterial hypertension. Eur. J. Hum. Genet. EJHG 2014, 145, 551–558. [Google Scholar]
  39. Chen, J.; Tang, H.; Sysol, J.R.; Moreno-Vinasco, L.; Shioura, K.M.; Chen, T.; Gorshkova, I.; Wang, L.; Huang, L.S.; Usatyuk, P.V.; et al. The sphingosine kinase 1/sphingosine-1-phosphate pathway in pulmonary arterial hypertension. Am. J. Respir. Crit. Care Med. 2014, 190, 1032–1043. [Google Scholar] [CrossRef]
  40. Ross, D.J.; Hough, G.; Hama, S.; Aboulhosn, J.; Belperio, J.A.; Saggar, R.; Van Lenten, B.J.; Ardehali, A.; Eghbali, M.; Reddy, S.; et al. Proinflammatory high-density lipoprotein results from oxidized lipid mediators in the pathogenesis of both idiopathic and associated types of pulmonary arterial hypertension. Pulm. Circ. 2015, 5, 640–648. [Google Scholar] [CrossRef] [Green Version]
  41. Sutendra, G.; Bonnet, S.; Rochefort, G.; Haromy, A.; Folmes, K.D.; Lopaschuk, G.D.; Dyck, J.R.; Michelakis, E.D. Fatty acid oxidation and malonyl-CoA decarboxylase in the vascular remodeling of pulmonary hypertension. Sci. Transl. Med. 2010, 2, 44ra58. [Google Scholar] [CrossRef]
  42. Heresi, G.A.; Aytekin, M.; Newman, J.; DiDonato, J.; Dweik, R.A. Plasma levels of high-density lipoprotein cholesterol and outcomes in pulmonary arterial hypertension. Am. J. Respir. Crit. Care Med. 2010, 182, 661–668. [Google Scholar] [CrossRef] [Green Version]
  43. Hu, J.; Xu, Q.; McTiernan, C.; Lai, Y.C.; Osei-Hwedieh, D.; Gladwin, M. Novel Targets of Drug Treatment for Pulmonary Hypertension. Am. J. Cardiovasc. Drugs 2015, 15, 225–234. [Google Scholar] [CrossRef]
  44. Ozkan, M.; Dweik, R.A.; Laskowski, D.; Arroliga, A.C.; Erzurum, S.C. High levels of nitric oxide in individuals with pulmonary hypertension receiving epoprostenol therapy. Lung 2001, 179, 233–243. [Google Scholar] [CrossRef]
  45. Giaid, A.; Yanagisawa, M.; Langleben, D.; Michel, R.P.; Levy, R.; Shennib, H.; Kimura, S.; Masaki, T.; Duguid, W.P.; Stewart, D.J. Expression of Endothelin-1 in the Lungs of Patients with Pulmonary Hypertension. N. Engl. J. Med. 1993, 328, 1732–1739. [Google Scholar] [CrossRef]
  46. Heresi, G.A.; Malin, S.K.; Barnes, J.W.; Tian, L.; Kirwan, J.P.; Dweik, R.A. Abnormal Glucose Metabolism and High-Energy Expenditure in Idiopathic Pulmonary Arterial Hypertension. Ann. Am. Thorac. Soc. 2016, 14, 190–199. [Google Scholar]
  47. Lundgrin, E.L.; Park, M.M.; Sharp, J.; Tang, W.H.; Thomas, J.D.; Asosingh, K.; Comhair, S.A.; DiFilippo, F.P.; Neumann, D.R.; Davis, L.; et al. Fasting 2-deoxy-2-[18F]fluoro-D-glucose positron emission tomography to detect metabolic changes in pulmonary arterial hypertension hearts over 1 year. Ann. Am. Thorac. Soc. 2013, 10, 1–9. [Google Scholar] [CrossRef] [Green Version]
  48. Marsboom, G.; Wietholt, C.; Haney, C.R.; Toth, P.T.; Ryan, J.J.; Morrow, E.; Thenappan, T.; Bache-Wiig, P.; Piao, L.; Paul, J.; et al. Lung (1)(8)F-fluorodeoxyglucose positron emission tomography for diagnosis and monitoring of pulmonary arterial hypertension. Am. J. Respir. Crit. Care Med. 2012, 185, 670–679. [Google Scholar] [CrossRef] [Green Version]
  49. Barnes, J.W.; Kucera, E.T.; Tian, L.; Mellor, N.E.; Dvorina, N.; Baldwin, W.W., III; Aldred, M.A.; Farver, C.F.; Comhair, S.A.; Aytekin, M.; et al. BMPR2 Mutation-independent Mechanisms of Disrupted BMP Signaling in IPAH. Am. J. Respir. Cell Mol. Biol. 2016, 55, 564–575. [Google Scholar] [CrossRef] [Green Version]
  50. Cikach, F.S., Jr.; Tonelli, A.R.; Barnes, J.; Paschke, K.; Newman, J.; Grove, D.; Dababneh, L.; Wang, S.; Dweik, R.A. Breath Analysis in Pulmonary Arterial Hypertension. Chest 2013, 145, 551–558. [Google Scholar] [CrossRef] [Green Version]
  51. Kao, C.C.; Wedes, S.H.; Hsu, J.W.; Bohren, K.M.; Comhair, S.A.A.; Jahoor, F.; Erzurum, S.C. Arginine Metabolic Endotypes in Pulmonary Arterial Hypertension. Pulm. Circ. 2015, 5, 124–134. [Google Scholar] [CrossRef] [Green Version]
  52. Demoncheaux, E.A.; Higenbottam, T.W.; Kiely, D.G.; Wong, J.M.; Wharton, S.; Varcoe, R.; Siddons, T.; Spivey, A.C.; Hall, K.; Gize, A.P. Decreased whole body endogenous nitric oxide production in patients with primary pulmonary hypertension. J. Vasc. Res. 2005, 42, 133–136. [Google Scholar] [CrossRef]
  53. Izquierdo-Garcia, J.L.; Arias, T.; Rojas, Y.; Garcia-Ruiz, V.; Santos, A.; Martin-Puig, S.; Ruiz-Cabello, J. Metabolic Reprogramming in the Heart and Lung in a Murine Model of Pulmonary Arterial Hypertension. Front. Cardiovasc. Med 2018, 5, 110. [Google Scholar] [CrossRef]
  54. Piao, L.; Fang, Y.H.; Parikh, K.; Ryan, J.J.; Toth, P.T.; Archer, S.L. Cardiac glutaminolysis: A maladaptive cancer metabolism pathway in the right ventricle in pulmonary hypertension. J. Mol. Med. 2013, 91, 1185–1197. [Google Scholar] [CrossRef] [Green Version]
  55. Egnatchik, R.A.; Brittain, E.L.; Shah, A.T.; Fares, W.H.; Ford, H.J.; Monahan, K.; Kang, C.J.; Kocurek, E.G.; Zhu, S.; Luong, T.; et al. Dysfunctional BMPR2 signaling drives an abnormal endothelial requirement for glutamine in pulmonary arterial hypertension. Pulm. Circ. 2017, 7, 186–199. [Google Scholar] [CrossRef] [Green Version]
  56. Mey, J.T.; Hari, A.; Axelrod, C.L.; Fealy, C.E.; Erickson, M.L.; Kirwan, J.P.; Dweik, R.A.; Heresi, G.A. Lipids and ketones dominate metabolism at the expense of glucose control in pulmonary arterial hypertension: A hyperglycaemic clamp and metabolomics study. Eur. Respir. J. 2020, 55, 1901700. [Google Scholar] [CrossRef]
  57. Belly, M.J.; Tiede, H.; Morty, R.E.; Schulz, R.; Voswinckel, R.; Tanislav, C.; Olschewski, H.; Ghofrani, H.A.; Seeger, W.; Reichenberger, F. HbA1c in pulmonary arterial hypertension: A marker of prognostic relevance? J. Heart Lung Transpl. 2012, 31, 1109–1114. [Google Scholar] [CrossRef]
  58. Paulin, R.; Michelakis, E.D. The metabolic theory of pulmonary arterial hypertension. Circ. Res. 2014, 115, 148–164. [Google Scholar] [CrossRef] [Green Version]
  59. Rai, P.R.; Cool, C.D.; King, J.A.; Stevens, T.; Burns, N.; Winn, R.A.; Kasper, M.; Voelkel, N.F. The cancer paradigm of severe pulmonary arterial hypertension. Am. J. Respir. Crit. Care Med. 2008, 178, 558–564. [Google Scholar] [CrossRef] [Green Version]
  60. Guignabert, C.; Tu, L.; Le Hiress, M.; Ricard, N.; Sattler, C.; Seferian, A.; Huertas, A.; Humbert, M.; Montani, D. Pathogenesis of pulmonary arterial hypertension: Lessons from cancer. Eur. Respir. Rev. Off. J. Eur. Respir. Soc. 2013, 22, 543–551. [Google Scholar] [CrossRef] [Green Version]
  61. Cool, C.D.; Kuebler, W.M.; Bogaard, H.J.; Spiekerkoetter, E.; Nicolls, M.R.; Voelkel, N.F. The hallmarks of severe pulmonary arterial hypertension: The cancer hypothesis—ten years later. Am. J. Physiol. Lung Cell. Mol. Physiol. 2020, 318, L1115–L1130. [Google Scholar] [CrossRef]
  62. Baggetto, L.G. Deviant energetic metabolism of glycolytic cancer cells. Biochimie 1992, 74, 959–974. [Google Scholar] [CrossRef]
  63. Altenberg, B.; Greulich, K.O. Genes of glycolysis are ubiquitously overexpressed in 24 cancer classes. Genomics 2004, 84, 1014–1020. [Google Scholar] [CrossRef]
  64. Bos, R.; van der Hoeven, J.J.M.; van der Wall, E.; van der Groep, P.; van Diest, P.J.; ComansUrvi Joshi, E.F.I.; Semenza, G.L.; Hoekstra, O.S.; Lammertsma, A.A.; Molthoff, C.F.M. Biologic Correlates of 18Fluorodeoxyglucose Uptake in Human Breast Cancer Measured by Positron Emission Tomography. J. Clin. Oncol. 2002, 20, 379–387. [Google Scholar] [CrossRef]
  65. Pedersen, P.L. Warburg, me and Hexokinase 2, Multiple discoveries of key molecular events underlying one of cancers’ most common phenotypes, the “Warburg Effect”, i.e., elevated glycolysis in the presence of oxygen. J. Bioenerg. Biomembr. 2007, 39, 211. [Google Scholar] [CrossRef]
  66. Warburg, O. On the origin of cancer cells. Science 1956, 123, 309–314. [Google Scholar] [CrossRef]
  67. Diaz-Ruiz, R.; Rigoulet, M.; Devin, A. The Warburg and Crabtree effects: On the origin of cancer cell energy metabolism and of yeast glucose repression. Biochim. Biophys. Acta 2011, 1807, 568–576. [Google Scholar] [CrossRef] [Green Version]
  68. Xu, W.; Koeck, T.; Lara, A.R.; Neumann, D.; DiFilippo, F.P.; Koo, M.; Janocha, A.J.; Masri, F.A.; Arroliga, A.C.; Jennings, C.; et al. Alterations of cellular bioenergetics in pulmonary artery endothelial cells. Proc. Natl. Acad. Sci. USA 2007, 104, 1342–1347. [Google Scholar] [CrossRef] [Green Version]
  69. Diebold, I.; Hennigs, J.K.; Miyagawa, K.; Li, C.G.; Nickel, N.P.; Kaschwich, M.; Cao, A.; Wang, L.; Reddy, S.; Chen, P.-I.; et al. BMPR2 Preserves Mitochondrial Function and DNA during Reoxygenation to Promote Endothelial Cell Survival and Reverse Pulmonary Hypertension. Cell Metab. 2015, 21, 596–608. [Google Scholar] [CrossRef] [Green Version]
  70. Rehman, J.; Archer, S.L. A proposed mitochondrial-metabolic mechanism for initiation and maintenance of pulmonary arterial hypertension in fawn-hooded rats: The Warburg model of pulmonary arterial hypertension. Adv. Exp. Med. Biol. 2010, 661, 171–185. [Google Scholar]
  71. Rabinovitch, M. Molecular pathogenesis of pulmonary arterial hypertension. J. Clin. Investig. 2012, 122, 4306–4313. [Google Scholar] [CrossRef]
  72. Bonnet, S.; Michelakis, E.D.; Porter, C.J.; Andrade-Navarro, M.A.; Thébaud, B.; Bonnet, S.; Haromy, A.; Harry, G.; Moudgil, R.; McMurtry, M.S.; et al. An abnormal mitochondrial-hypoxia inducible factor-1alpha-Kv channel pathway disrupts oxygen sensing and triggers pulmonary arterial hypertension in fawn hooded rats: Similarities to human pulmonary arterial hypertension. Circulation 2006, 113, 2630–2641. [Google Scholar] [CrossRef] [Green Version]
  73. Gomez-Arroyo, J.; Mizuno, S.; Szczepanek, K.; Tassell, B.V.; Natarajan, R.; dos Remedios, C.G.; Drake, J.I.; Farkas, L.; Kraskauskas, D.; Wijesinghe, D.S.; et al. Metabolic Gene Remodeling and Mitochondrial Dysfunction in Failing Right Ventricular Hypertrophy Secondary to Pulmonary Arterial Hypertension. Circ. Heart Fail. 2013, 6, 136–144. [Google Scholar] [CrossRef] [Green Version]
  74. Nagendran, J.; Gurtu, V.; Fu, D.Z.; Dyck, J.R.; Haromy, A.; Ross, D.B.; Rebeyka, I.M.; Michelakis, E.D. A dynamic and chamber-specific mitochondrial remodeling in right ventricular hypertrophy can be therapeutically targeted. J. Thorac. Cardiovasc. Surg. 2008, 136, 168–178.e163. [Google Scholar] [CrossRef] [Green Version]
  75. Michelakis, E.D.; Gurtu, V.; Webster, L.; Barnes, G.; Watson, G.; Howard, L.; Cupitt, J.; Paterson, I.; Thompson, R.B.; Chow, K.; et al. Inhibition of pyruvate dehydrogenase kinase improves pulmonary arterial hypertension in genetically susceptible patients. Sci. Transl. Med. 2017, 9, eaao4583. [Google Scholar] [CrossRef] [Green Version]
  76. Bonnet, S.; Archer, S.L.; Allalunis-Turner, J.; Haromy, A.; Beaulieu, C.; Thompson, R.; Lee, C.T.; Lopaschuk, G.D.; Puttagunta, L.; Bonnet, S.; et al. A Mitochondria-K+ Channel Axis Is Suppressed in Cancer and Its Normalization Promotes Apoptosis and Inhibits Cancer Growth. Cancer Cell 2007, 11, 37–51. [Google Scholar] [CrossRef] [Green Version]
  77. Chu, Q.S.; Sangha, R.; Spratlin, J.; Vos, L.J.; Mackey, J.R.; McEwan, A.J.; Venner, P.; Michelakis, E.D. A phase I open-labeled, single-arm, dose-escalation, study of dichloroacetate (DCA) in patients with advanced solid tumors. Investig. New Drugs 2015, 33, 603–610. [Google Scholar] [CrossRef]
  78. McMurtry, M.S.; Bonnet, S.; Wu, X.; Dyck, J.R.; Haromy, A.; Hashimoto, K.; Michelakis, E.D. Dichloroacetate prevents and reverses pulmonary hypertension by inducing pulmonary artery smooth muscle cell apoptosis. Circ. Res. 2004, 95, 830–840. [Google Scholar] [CrossRef] [Green Version]
  79. Dyck, J.R.; Hopkins, T.A.; Bonnet, S.; Michelakis, E.D.; Young, M.E.; Watanabe, M.; Kawase, Y.; Jishage, K.; Lopaschuk, G.D. Absence of malonyl coenzyme A decarboxylase in mice increases cardiac glucose oxidation and protects the heart from ischemic injury. Circulation 2006, 114, 1721–1728. [Google Scholar] [CrossRef] [Green Version]
  80. Reily, C.; Stewart, T.J.; Renfrow, M.B.; Novak, J. Glycosylation in health and disease. Nat. Rev. Nephrol. 2019, 15, 346–366. [Google Scholar] [CrossRef]
  81. Varki, A. Biological roles of oligosaccharides: All of the theories are correct. Glycobiology 1993, 3, 97–130. [Google Scholar] [CrossRef]
  82. Varki, A. Biological roles of glycans. Glycobiology 2017, 27, 3–49. [Google Scholar] [CrossRef] [Green Version]
  83. Häuselmann, I.; Borsig, L. Altered Tumor-Cell Glycosylation Promotes Metastasis. Front. Oncol. 2014, 4, 28. [Google Scholar] [CrossRef] [Green Version]
  84. Kamigaito, T.; Okaneya, T.; Kawakubo, M.; Shimojo, H.; Nishizawa, O.; Nakayama, J. Overexpression of O-GlcNAc by prostate cancer cells is significantly associated with poor prognosis of patients. Prostate Cancer Prostatic Dis. 2014, 17, 18–22. [Google Scholar] [CrossRef] [Green Version]
  85. Kizuka, Y.; Taniguchi, N. Enzymes for N-Glycan Branching and Their Genetic and Nongenetic Regulation in Cancer. Biomolecules 2016, 6, 25. [Google Scholar] [CrossRef] [Green Version]
  86. Mathews, M.B.; Glagov, S. Acid mucopolysaccharide patterns in aging human cartilage. J. Clin. Investig. 1966, 45, 1103–1111. [Google Scholar] [CrossRef]
  87. Pal-Ghosh, S.; Tadvalkar, G.; Stepp, M.A. Alterations in Corneal Sensory Nerves During Homeostasis, Aging, and after Injury in Mice Lacking the Heparan Sulfate Proteoglycan Syndecan-1. Investig. Opthalmol. Vis. Sci. 2017, 58, 4959–4975. [Google Scholar] [CrossRef] [Green Version]
  88. Kobata, A. Glycobiology in the field of aging research—Introduction to glycogerontology. Biochimie 2003, 85, 13–24. [Google Scholar] [CrossRef]
  89. Beeley, J.G.; Blackie, R.; Baxter, A. Glycoprotein and glycolipid changes in aged erythrocytes [proceedings]. Biochem. Soc. Trans. 1977, 5, 1725–1726. [Google Scholar] [CrossRef] [Green Version]
  90. Ovsepian, L.M.; Kazarian, G.S.; Akopdzhanian, A.A.; L’Vov, M.V. Age-dependent changes in phospholipid content and neutral lipid contents in aging. Adv. Gerontol. 2012, 25, 250–254. [Google Scholar] [CrossRef]
  91. Wang, W.; Gopal, S.; Pocock, R.; Xiao, Z. Glycan Mimetics from Natural Products: New Therapeutic Opportunities for Neurodegenerative Disease. Molecules 2019, 24, 4604. [Google Scholar] [CrossRef] [Green Version]
  92. Kizuka, Y.; Kitazume, S.; Fujinawa, R.; Saito, T.; Iwata, N.; Saido, T.C.; Nakano, M.; Yamaguchi, Y.; Hashimoto, Y.; Staufenbiel, M.; et al. An aberrant sugar modification of BACE1 blocks its lysosomal targeting in Alzheimer’s disease. EMBO Mol. Med. 2015, 7, 175–189. [Google Scholar] [CrossRef]
  93. Maguire, T.M.; Gillian, A.M.; O’Mahony, D.; Coughlan, C.M.; Breen, K.C. A decrease in serum sialyltransferase levels in Alzheimer’s disease. Neurobiol. Aging 1994, 15, 99–102. [Google Scholar] [CrossRef]
  94. Fodero, L.R.; Sáez-Valero, J.; Barquero, M.S.; Marcos, A.; McLean, C.A.; Small, D.H. Wheat germ agglutinin-binding glycoproteins are decreased in Alzheimer’s disease cerebrospinal fluid. J. Neurochem. 2001, 79, 1022–1026. [Google Scholar] [CrossRef]
  95. Saito, F.; Yanagisawa, K.; Miyatake, T. Soluble derivatives of β/A4 amyloid protein precursor in human cerebrospinal fluid are both N- and O-glycosylated. Mol. Brain Res. 1993, 19, 171–174. [Google Scholar] [CrossRef]
  96. Griffith, L.S.; Mathes, M.; Schmitz, B. Beta-amyloid precursor protein is modified with O-linked N-acetylglucosamine. J. Neurosci. Res. 1995, 41, 270–278. [Google Scholar] [CrossRef]
  97. Zhu, Y.; Shan, X.; Yuzwa, S.A.; Vocadlo, D.J. The emerging link between O-GlcNAc and Alzheimer disease. J. Biol. Chem. 2014, 289, 34472–34481. [Google Scholar] [CrossRef] [Green Version]
  98. Dashti, H.; Pabon Porras, M.A.; Mora, S. Glycosylation and Cardiovascular Diseases. In The Role of Glycosylation in Health and Disease; Lauc, G., Trbojević-Akmačić, I., Eds.; Springer International Publishing: Cham, Switzerland, 2021; pp. 307–319. [Google Scholar]
  99. 99Gudelj, I.; Lauc, G. Protein N-Glycosylation in Cardiovascular Diseases and Related Risk Factors. Curr. Cardiovasc. Risk Rep. 2018, 12, 16. [Google Scholar] [CrossRef]
  100. Wright, J.N.; Collins, H.E.; Wende, A.R.; Chatham, J.C. O-GlcNAcylation and cardiovascular disease. Biochem. Soc. Trans. 2017, 45, 545–553. [Google Scholar] [CrossRef]
  101. Komaromy, A.; Reider, B.; Jarvas, G.; Guttman, A. Glycoprotein biomarkers and analysis in chronic obstructive pulmonary disease and lung cancer with special focus on serum immunoglobulin G. Clin. Chim. Acta 2020, 506, 204–213. [Google Scholar] [CrossRef]
  102. Pavić, T.; Dilber, D.; Kifer, D.; Selak, N.; Keser, T.; Ljubičić, Đ.; Vukić Dugac, A.; Lauc, G.; Rumora, L.; Gornik, O. N-glycosylation patterns of plasma proteins and immunoglobulin G in chronic obstructive pulmonary disease. J. Transl. Med. 2018, 16, 323. [Google Scholar] [CrossRef] [PubMed]
  103. Wang, X.; Inoue, S.; Gu, J.; Miyoshi, E.; Noda, K.; Li, W.; Mizuno-Horikawa, Y.; Nakano, M.; Asahi, M.; Takahashi, M.; et al. Dysregulation of TGF-β1 receptor activation leads to abnormal lung development and emphysema-like phenotype in core fucose-deficient mice. Proc. Natl. Acad. Sci. USA 2005, 102, 15791–15796. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Yamada, M.; Ishii, T.; Ikeda, S.; Naka-Mieno, M.; Tanaka, N.; Arai, T.; Kumasaka, T.; Gemma, A.; Kida, K.; Muramatsu, M.; et al. Association of fucosyltransferase 8 (FUT8) polymorphism Thr267Lys with pulmonary emphysema. J. Hum. Genet. 2011, 56, 857–860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Mészáros, B.; Járvás, G.; Farkas, A.; Szigeti, M.; Kovács, Z.; Kun, R.; Szabó, M.; Csánky, E.; Guttman, A. Comparative analysis of the human serum N-glycome in lung cancer, COPD and their comorbidity using capillary electrophoresis. J. Chromatogr. B 2019, 1137, 121913. [Google Scholar] [CrossRef] [PubMed]
  106. Krick, S.; Helton, E.S.; Easter, M.; Bollenbecker, S.; Denson, R.; Zaharias, R.; Cochran, P.; Vang, S.; Harris, E.; Wells, J.M.; et al. ST6GAL1 and α2-6 Sialylation Regulates IL-6 Expression and Secretion in Chronic Obstructive Pulmonary Disease. Front. Immunol. 2021, 12, 693149. [Google Scholar] [CrossRef]
  107. Schulz, B.; Sloane, A.J.; Robinson, L.J.; Prasad, S.S.; Lindner, R.A.; Robinson, M.; Bye, P.T.; Nielson, D.W.; Harry, J.L.; Packer, N.H.; et al. Glycosylation of sputum mucins is altered in cystic fibrosis patients. Glycobiology 2007, 17, 698–712. [Google Scholar] [CrossRef]
  108. Arnold, J.N.; Saldova, R.; Galligan, M.C.; Murphy, T.B.; Mimura-Kimura, Y.; Telford, J.E.; Godwin, A.K.; Rudd, P.M. Novel Glycan Biomarkers for the Detection of Lung Cancer. J. Proteome Res. 2011, 10, 1755–1764. [Google Scholar] [CrossRef]
  109. Xia, B.; Royall, J.A.; Damera, G.; Sachdev, G.P.; Cummings, R.D. Altered O-glycosylation and sulfation of airway mucins associated with cystic fibrosis. Glycobiology 2005, 15, 747–775. [Google Scholar] [CrossRef] [Green Version]
  110. Davril, M.; Groux-Degroote, S.; Humbert, P.; Galabert, C.; Dumur, V.; Lafitte, J.-J.; Lamblin, G.; Roussel, P. The sialylation of bronchial mucins secreted by patients suffering from cystic fibrosis or from chronic bronchitis is related to the severity of airway infection. Glycobiology 1999, 9, 311–321. [Google Scholar] [CrossRef] [Green Version]
  111. Collum, S.D.; Chen, N.; Hernandez, A.M.; Hanmandlu, A.; Sweeney, H.; Mertens, T.C.J.; Weng, T.; Luo, F.; Molina, J.G.; Davies, J.; et al. Inhibition of hyaluronan synthesis attenuates pulmonary hypertension associated with lung fibrosis. J. Cereb. Blood Flow Metab. 2017, 174, 3284–3301. [Google Scholar] [CrossRef] [Green Version]
  112. Bjermer, L.; Lundgren, R.; Hallgren, R. Hyaluronan and type III procollagen peptide concentrations in bronchoalveolar lavage fluid in idiopathic pulmonary fibrosis. Thorax 1989, 44, 126–131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Li, Y.; Jiang, D.; Liang, J.; Meltzer, E.B.; Gray, A.; Miura, R.; Wogensen, L.; Yamaguchi, Y.; Noble, P.W. Severe lung fibrosis requires an invasive fibroblast phenotype regulated by hyaluronan and CD44. J. Exp. Med. 2011, 208, 1459–1471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Dittner-Moormann, S.; Lourenco, C.M.; Reunert, J.; Nishinakamura, R.; Tanaka, S.S.; Werner, C.; Debus, V.; Zimmer, K.-P.; Wetzel, G.; Naim, H.Y.; et al. TRAPγ-CDG shows asymmetric glycosylation and an effect on processing of proteins required in higher organisms. J. Med. Genet. 2021, 58, 213–216. [Google Scholar] [CrossRef]
  115. Toppila, S.; Paavonen, T.; Laitinen, A.; Laitinen, L.A.; Renkonen, R. Endothelial Sulfated Sialyl Lewis × Glycans, Putative L-Selectin Ligands, Are Preferentially Expressed in Bronchial Asthma but Not in Other Chronic Inflammatory Lung Diseases. Am. J. Respir. Cell Mol. Biol. 2000, 23, 492–498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Darley-Usmar, V.M.; Ball, L.E.; Chatham, J.C. Protein O-linked beta-N-acetylglucosamine: A novel effector of cardiomyocyte metabolism and function. J. Mol. Cell. Cardiol. 2012, 52, 538–549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Brownlee, M. Biochemistry and molecular cell biology of diabetic complications. Nature 2001, 414, 813–820. [Google Scholar] [CrossRef]
  118. McClain, D.A. Hexosamines as mediators of nutrient sensing and regulation in diabetes. J. Diabetes Complicat. 2002, 16, 72–80. [Google Scholar] [CrossRef]
  119. Rossetti, L. Perspective: Hexosamines and nutrient sensing. Endocrinology 2000, 141, 1922–1925. [Google Scholar] [CrossRef]
  120. Kreppel, L.K.; Blomberg, M.A.; Hart, G.W. Dynamic glycosylation of nuclear and cytosolic proteins. Cloning and characterization of a unique O-GlcNAc transferase with multiple tetratricopeptide repeats. J. Biol. Chem. 1997, 272, 9308–9315. [Google Scholar] [CrossRef] [Green Version]
  121. Lubas, W.A.; Frank, D.W.; Krause, M.; Hanover, J.A. O-Linked GlcNAc transferase is a conserved nucleocytoplasmic protein containing tetratricopeptide repeats. J. Biol. Chem. 1997, 272, 9316–9324. [Google Scholar] [CrossRef] [Green Version]
  122. Gao, Y.; Wells, L.; Comer, F.I.; Parker, G.J.; Hart, G.W. Dynamic O-glycosylation of nuclear and cytosolic proteins: Cloning and characterization of a neutral, cytosolic beta-N-acetylglucosaminidase from human brain. J. Biol. Chem. 2001, 276, 9838–9845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Wells, L.; Gao, Y.; Mahoney, J.A.; Vosseller, K.; Chen, C.; Rosen, A.; Hart, G.W. Dynamic O-glycosylation of nuclear and cytosolic proteins: Further characterization of the nucleocytoplasmic beta-N-acetylglucosaminidase, O-GlcNAcase. J. Biol. Chem. 2002, 277, 1755–1761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Nagel, A.K.; Ball, L.E. O-GlcNAc transferase and O-GlcNAcase: Achieving target substrate specificity. Amino Acids 2014, 46, 2305–2316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Hart, G.W. Three Decades of Research on O-GlcNAcylation—A Major Nutrient Sensor That Regulates Signaling, Transcription and Cellular Metabolism. Front. Endocrinol. 2014, 5, 183. [Google Scholar] [CrossRef] [Green Version]
  126. Ma, J.; Liu, T.; Wei, A.C.; Banerjee, P.; O’Rourke, B.; Hart, G.W. O-GlcNAcomic Profiling Identifies Widespread O-Linked beta-N-Acetylglucosamine Modification (O-GlcNAcylation) in Oxidative Phosphorylation System Regulating Cardiac Mitochondrial Function. J. Biol. Chem. 2015, 290, 29141–29153. [Google Scholar] [CrossRef] [Green Version]
  127. Zachara, N.E.; O’Donnell, N.; Cheung, W.D.; Mercer, J.J.; Marth, J.D.; Hart, G.W. Dynamic O-GlcNAc modification of nucleocytoplasmic proteins in response to stress. A survival response of mammalian cells. J. Biol. Chem. 2004, 279, 30133–30142. [Google Scholar] [CrossRef] [Green Version]
  128. Kamemura, K.; Hayes, B.K.; Comer, F.I.; Hart, G.W. Dynamic interplay between O-glycosylation and O-phosphorylation of nucleocytoplasmic proteins: Alternative glycosylation/phosphorylation of THR-58, a known mutational hot spot of c-Myc in lymphomas, is regulated by mitogens. J. Biol. Chem. 2002, 277, 19229–19235. [Google Scholar] [CrossRef] [Green Version]
  129. Hart, G.W.; Slawson, C.; Ramirez-Correa, G.; Lagerlof, O. Cross talk between O-GlcNAcylation and phosphorylation: Roles in signaling, transcription, and chronic disease. Annu. Rev. Biochem. 2011, 80, 825–858. [Google Scholar] [CrossRef] [Green Version]
  130. Lefebvre, T.; Ferreira, S.; Dupont-Wallois, L.; Bussiere, T.; Dupire, M.J.; Delacourte, A.; Michalski, J.C.; Caillet-Boudin, M.L. Evidence of a balance between phosphorylation and O-GlcNAc glycosylation of Tau proteins--a role in nuclear localization. Biochim. Biophys. Acta 2003, 1619, 167–176. [Google Scholar] [CrossRef]
  131. Wulff-Fuentes, E.; Berendt, R.R.; Massman, L.; Danner, L.; Malard, F.; Vora, J.; Kahsay, R.; Olivier-Van Stichelen, S. The human O-GlcNAcome database and meta-analysis. Sci. Data 2021, 8, 25. [Google Scholar] [CrossRef]
  132. Caldwell, S.A.; Jackson, S.R.; Shahriari, K.S.; Lynch, T.P.; Sethi, G.; Walker, S.; Vosseller, K.; Reginato, M.J. Nutrient sensor O-GlcNAc transferase regulates breast cancer tumorigenesis through targeting of the oncogenic transcription factor FoxM. Oncogene 2010, 29, 2831–2842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Ferrer, C.M.; Sodi, V.L.; Reginato, M.J. O-GlcNAcylation in Cancer Biology: Linking Metabolism and Signaling. J. Mol. Biol. 2016, 428, 3282–3294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Hanover, J.A.; Krause, M.W.; Love, D.C. The hexosamine signaling pathway: O-GlcNAc cycling in feast or famine. Biochim. Biophys. Acta 2010, 1800, 80–95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Lefebvre, T.; Dehennaut, V.; Guinez, C.; Olivier, S.; Drougat, L.; Mir, A.M.; Mortuaire, M.; Vercoutter-Edouart, A.S.; Michalski, J.C. Dysregulation of the nutrient/stress sensor O-GlcNAcylation is involved in the etiology of cardiovascular disorders, type-2 diabetes and Alzheimer’s disease. Biochim. Biophys. Acta 2010, 1800, 67–79. [Google Scholar] [CrossRef]
  136. Slawson, C.; Housley, M.P.; Hart, G.W. O-GlcNAc cycling: How a single sugar post-translational modification is changing the way we think about signaling networks. J. Cell. Biochem. 2006, 97, 71–83. [Google Scholar] [CrossRef]
  137. Vaidyanathan, K.; Wells, L. Multiple tissue-specific roles for the O-GlcNAc post-translational modification in the induction of and complications arising from type II diabetes. J. Biol. Chem. 2014, 289, 34466–34471. [Google Scholar] [CrossRef] [Green Version]
  138. Wells, L.; Vosseller, K.; Hart, G.W. A role for N-acetylglucosamine as a nutrient sensor and mediator of insulin resistance. Cell. Mol. Life Sci. 2003, 60, 222–228. [Google Scholar] [CrossRef]
  139. Barnes, J.W.; Tian, L.; Heresi, G.A.; Farver, C.F.; Asosingh, K.; Comhair, S.A.; Aulak, K.S.; Dweik, R.A. O-linked beta-N-acetylglucosamine transferase directs cell proliferation in idiopathic pulmonary arterial hypertension. Circulation 2015, 131, 1260–1268. [Google Scholar] [CrossRef] [Green Version]
  140. Banerjee, P.S.; Lagerlof, O.; Hart, G.W. Roles of O-GlcNAc in chronic diseases of aging. Mol. Asp. Med. 2016, 51, 1–15. [Google Scholar] [CrossRef]
  141. Yuzwa, S.A.; Vocadlo, D.J. O-GlcNAc and neurodegeneration: Biochemical mechanisms and potential roles in Alzheimer’s disease and beyond. Chem. Soc. Rev. 2014, 43, 6839–6858. [Google Scholar] [CrossRef] [Green Version]
  142. Ferrer, C.M.; Lynch, T.P.; Sodi, V.L.; Falcone, J.N.; Schwab, L.P.; Peacock, D.L.; Vocadlo, D.J.; Seagroves, T.N.; Reginato, M.J. O-GlcNAcylation regulates cancer metabolism and survival stress signaling via regulation of the HIF-1 pathway. Mol. Cell. 2014, 54, 820–831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Yi, W.; Clark, P.M.; Mason, D.E.; Keenan, M.C.; Hill, C.; Goddard, W.A., 3rd; Peters, E.C.; Driggers, E.M.; Hsieh-Wilson, L.C. Phosphofructokinase 1 glycosylation regulates cell growth and metabolism. Science 2012, 337, 975–980. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Ma, Z.; Vosseller, K. O-GlcNAc in cancer biology. Amino Acids 2013, 45, 719–733. [Google Scholar] [CrossRef]
  145. Slawson, C.; Copeland, R.J.; Hart, G.W. O-GlcNAc signaling: A metabolic link between diabetes and cancer? Trends Biochem. Sci. 2010, 35, 547–555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Dassanayaka, S.; Jones, S.P. O-GlcNAc and the cardiovascular system. Pharmacol. Ther. 2014, 142, 62–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Zachara, N.E. The roles of O-linked beta-N-acetylglucosamine in cardiovascular physiology and disease. Am. J. Physiol. Heart Circ. Physiol. 2012, 302, H1905–H1918. [Google Scholar] [CrossRef] [Green Version]
  148. Gelinas, R.; Mailleux, F.; Dontaine, J.; Bultot, L.; Demeulder, B.; Ginion, A.; Daskalopoulos, E.P.; Esfahani, H.; Dubois-Deruy, E.; Lauzier, B.; et al. AMPK activation counteracts cardiac hypertrophy by reducing O-GlcNAcylation. Nat. Commun. 2018, 9, 374. [Google Scholar] [CrossRef] [Green Version]
  149. Hilgers, R.H.P.; Xing, D.; Gong, K.; Chen, Y.-F.; Chatham, J.C.; Oparil, S. Acute O-GlcNAcylation prevents inflammation-induced vascular dysfunction. Am. J. Physiol. Heart Circ. Physiol. 2012, 303, H513–H522. [Google Scholar] [CrossRef] [Green Version]
  150. Barnes, J.W.; Tian, L.; Krick, S.; Helton, E.S.; Denson, R.S.; Comhair, S.A.A.; Dweik, R.A. O-GlcNAc Transferase Regulates Angiogenesis in Idiopathic Pulmonary Arterial Hypertension. Int. J. Mol. Sci. 2019, 20, 6299. [Google Scholar] [CrossRef] [Green Version]
  151. Aulak, K.S.; Barnes, J.W.; Tian, L.; Mellor, N.E.; Haque, M.M.; Willard, B.; Li, L.; Comhair, S.C.; Stuehr, D.J.; Dweik, R.A. Specific O-GlcNAc modification at Ser-615 modulates eNOS function. Redox Biol. 2020, 36, 101625. [Google Scholar] [CrossRef]
  152. Prisco, S.Z.; Rose, L.; Potus, F.; Tian, L.; Wu, D.; Hartweck, L.; Al-Qazazi, R.; Neuber-Hess, M.; Eklund, M.; Hsu, S.; et al. Excess Protein O-GlcNAcylation Links Metabolic Derangements to Right Ventricular Dysfunction in Pulmonary Arterial Hypertension. Int. J. Mol. Sci. 2020, 21, 7278. [Google Scholar] [CrossRef] [PubMed]
  153. Prisco, S.Z.; Eklund, M.; Raveendran, R.; Thenappan, T.; Prins, K.W. With No Lysine Kinase 1 Promotes Metabolic Derangements and RV Dysfunction in Pulmonary Arterial Hypertension. JACC Basic Transl. Sci. 2021, 6, 834–850. [Google Scholar] [CrossRef] [PubMed]
  154. Varki, A.; Cummings, R.D.; Esko, J.D.; Freeze, H.H.; Stanley, P.; Bertozzi, C.R.; Hart, G.W.; Etzler, M.E. Essentials in Glycobiology; Cold Spring Harbor Laboratory Press: Cold Spring Harbor, NY, USA, 2009. [Google Scholar]
  155. Lin, X. Functions of heparan sulfate proteoglycans in cell signaling during development. Development 2004, 131, 6009–6021. [Google Scholar] [CrossRef] [Green Version]
  156. Chen, Q.; Sivakumar, P.; Barley, C.; Peters, D.M.; Gomes, R.R.; Farach-Carson, M.C.; Dallas, S.L. Potential role for heparan sulfate proteoglycans in regulation of transforming growth factor-beta (TGF-beta) by modulating assembly of latent TGF-beta-binding protein-1. J. Biol. Chem. 2007, 282, 26418–26430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Kraushaar, D.C.; Yamaguchi, Y.; Wang, L. Heparan sulfate is required for embryonic stem cells to exit from self-renewal. J. Biol. Chem. 2010, 285, 5907–5916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Scarpellini, A.; Germack, R.; Lortat-Jacob, H.; Muramatsu, T.; Billett, E.; Johnson, T.; Verderio, E.A. Heparan sulfate proteoglycans are receptors for the cell-surface trafficking and biological activity of transglutaminase-2. J. Biol. Chem. 2009, 284, 18411–18423. [Google Scholar] [CrossRef] [Green Version]
  159. Li, Z.; Yasuda, Y.; Li, W.; Bogyo, M.; Katz, N.; Gordon, R.E.; Fields, G.B.; Bromme, D. Regulation of collagenase activities of human cathepsins by glycosaminoglycans. J. Biol. Chem. 2004, 279, 5470–5479. [Google Scholar] [CrossRef] [Green Version]
  160. Ghabrial, A.S. A sweet spot in the FGFR signal transduction pathway. Sci. Signal 2012, 5, pe1. [Google Scholar] [CrossRef] [Green Version]
  161. Christensen, G.; Herum, K.M.; Lunde, I.G. Sweet, yet underappreciated: Proteoglycans and extracellular matrix remodeling in heart disease. Matrix Biol. 2019, 75–76, 286–299. [Google Scholar] [CrossRef] [PubMed]
  162. Iozzo, R.V.; Schaefer, L. Proteoglycan form and function: A comprehensive nomenclature of proteoglycans. Matrix Biol. 2015, 42, 11–55. [Google Scholar] [CrossRef]
  163. Papakonstantinou, E.; Karakiulakis, G.; Tamm, M.; Perruchoud, A.P.; Roth, M. Hypoxia modifies the effect of PDGF on glycosaminoglycan synthesis by primary human lung cells. Am. J. Physiol. Lung Cell. Mol. Physiol. 2000, 279, L825–L834. [Google Scholar] [CrossRef] [PubMed]
  164. Papakonstantinou, E.; Roth, M.; Tamm, M.; Eickelberg, O.; Perruchoud, A.P.; Karakiulakis, G. Hypoxia differentially enhances the effects of transforming growth factor-beta isoforms on the synthesis and secretion of glycosaminoglycans by human lung fibroblasts. J. Pharmacol. Exp. Ther. 2002, 301, 830–837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Kerr, J.S.; Ruppert, C.L.; Tozzi, C.A.; Neubauer, J.A.; Frankel, H.M.; Yu, S.Y.; Riley, D.J. Reduction of chronic hypoxic pulmonary hypertension in the rat by an inhibitor of collagen production. Am. Rev. Respir. Dis. 1987, 135, 300–306. [Google Scholar] [PubMed]
  166. Bertero, T.; Oldham, W.M.; Cottrill, K.A.; Pisano, S.; Vanderpool, R.R.; Yu, Q.; Zhao, J.; Tai, Y.; Tang, Y.; Zhang, Y.Y.; et al. Vascular stiffness mechanoactivates YAP/TAZ-dependent glutaminolysis to drive pulmonary hypertension. J. Clin. Investig. 2016, 126, 3313–3335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Humphries, D.E.; Lee, S.-L.; Fanburg, B.L.; Silbert, J.E. Effects of hypoxia and hyperoxia on proteoglycan production by bovine pulmonary artery endothelial cells. J. Cell. Physiol. 1986, 126, 249–253. [Google Scholar] [CrossRef]
  168. Figueroa, J.E.; Tao, Z.; Sarphie, T.G.; Smart, F.W.; Glancy, D.L.; Vijayagopal, P. Effect of hypoxia and hypoxia/reoxygenation on proteoglycan metabolism by vascular smooth muscle cells. Atherosclerosis 1999, 143, 135–144. [Google Scholar] [CrossRef]
  169. Little, P.J.; Burch, M.L.; Getachew, R.; Al-Aryahi, S.; Osman, N. Endothelin-1 stimulation of proteoglycan synthesis in vascular smooth muscle is mediated by endothelin receptor transactivation of the transforming growth factor-β type I receptor. J. Cardiovasc. Pharmacol. 2010, 56, 360–368. [Google Scholar] [CrossRef]
  170. Lee, R.T.; Yamamoto, C.; Feng, Y.; Potter-Perigo, S.; Briggs, W.H.; Landschulz, K.T.; Turi, T.G.; Thompson, J.F.; Libby, P.; Wight, T.N. Mechanical strain induces specific changes in the synthesis and organization of proteoglycans by vascular smooth muscle cells. J. Biol. Chem. 2001, 276, 13847–13851. [Google Scholar] [CrossRef] [Green Version]
  171. Shimizu-Hirota, R.; Sasamura, H.; Mifune, M.; Nakaya, H.; Kuroda, M.; Hayashi, M.; Saruta, T. Regulation of Vascular Proteoglycan Synthesis by Angiotensin II Type 1 and Type 2 Receptors. J. Am. Soc. Nephrol. 2001, 12, 2609–2615. [Google Scholar] [CrossRef]
  172. Zimmer, B.M.; Barycki, J.J.; Simpson, M.A. Integration of Sugar Metabolism and Proteoglycan Synthesis by UDP-glucose Dehydrogenase. J. Histochem. Cytochem. 2021, 69, 13–23. [Google Scholar] [CrossRef]
  173. van Det, N.F.; van den Born, J.; Tamsma, J.T.; Verhagen, N.A.M.; Berden, J.H.M.; Bruijn, J.A.; Daha, M.R.; van der Woude, F.J. Effects of high glucose on the production of heparan sulfate proteoglycan by mesangial and epithelial cells. Kidney Int. 1996, 49, 1079–1089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Klein, D.J.; Cohen, R.M.; Rymaszewski, Z. Proteoglycan synthesis by bovine myocardial endothelial cells is increased by long-term exposure to high concentrations of glucose. J. Cell. Physiol. 1995, 165, 493–502. [Google Scholar] [CrossRef]
  175. Simoni, R.D.; Hill, R.L.; Vaughan, M.; Hascall, V. The discovery of hyaluronan by Karl Meyer. J. Biol. Chem. 2002, 277, e1–e2. [Google Scholar] [CrossRef]
  176. Armstrong, S.E.; Bell, D.R. Measurement of high-molecular-weight hyaluronan in solid tissue using agarose gel electrophoresis. Anal. Biochem. 2002, 308, 255–264. [Google Scholar] [CrossRef]
  177. Itano, N.; Sawai, T.; Yoshida, M.; Lenas, P.; Yamada, Y.; Imagawa, M.; Shinomura, T.; Hamaguchi, M.; Yoshida, Y.; Ohnuki, Y.; et al. Three Isoforms of Mammalian Hyaluronan Synthases Have Distinct Enzymatic Properties. J. Biol. Chem. 1999, 274, 25085–25092. [Google Scholar] [CrossRef] [Green Version]
  178. Philipson, L.H.; Schwartz, N.B. Subcellular localization of hyaluronate synthetase in oligodendroglioma cells. J. Biol. Chem. 1984, 259, 5017–5023. [Google Scholar] [CrossRef]
  179. Vigetti, D.; Deleonibus, S.; Moretto, P.; Karousou, E.; Viola, M.; Bartolini, B.; Hascall, V.C.; Tammi, M.; De Luca, G.; Passi, A. Role of UDP-N-Acetylglucosamine (GlcNAc) and O-GlcNAcylation of Hyaluronan Synthase 2 in the Control of Chondroitin Sulfate and Hyaluronan Synthesis. J. Biol. Chem. 2012, 287, 35544–35555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Jokela, T.A.; Makkonen, K.M.; Oikari, S.; Karna, R.; Koli, E.; Hart, G.W.; Tammi, R.H.; Carlberg, C.; Tammi, M.I. Cellular content of UDP-N-acetylhexosamines controls hyaluronan synthase 2 expression and correlates with O-linked N-acetylglucosamine modification of transcription factors YY1 and SP1. J. Biol. Chem. 2011, 286, 33632–33640. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Wang, A.; De La Motte, C.; Lauer, M.; Hascall, V. Hyaluronan matrices in pathobiological processes. FEBS J. 2011, 278, 1412–1418. [Google Scholar] [CrossRef] [Green Version]
  182. Dentener, M.A.; Vernooy, J.H.; Hendriks, S.; Wouters, E.F. Enhanced levels of hyaluronan in lungs of patients with COPD: Relationship with lung function and local inflammation. Thorax 2005, 60, 114–119. [Google Scholar] [CrossRef] [Green Version]
  183. Galdi, F.; Pedone, C.; McGee, C.A.; George, M.; Rice, A.B.; Hussain, S.S.; Vijaykumar, K.; Boitet, E.R.; Tearney, G.J.; McGrath, J.A.; et al. Inhaled high molecular weight hyaluronan ameliorates respiratory failure in acute COPD exacerbation: A pilot study. Respir. Res. 2021, 22, 30. [Google Scholar] [CrossRef] [PubMed]
  184. Lauer, M.E.; Dweik, R.A.; Garantziotis, S.; Aronica, M.A. The Rise and Fall of Hyaluronan in Respiratory Diseases. International. J. Cell Biol. 2015, 2015, 712507. [Google Scholar]
  185. Zhu, L.; Zhuo, L.; Kimata, K.; Yamaguchi, E.; Watanabe, H.; Aronica, M.A.; Hascall, V.C.; Baba, K. Deficiency in the Serum-Derived Hyaluronan-Associated Protein-Hyaluronan Complex Enhances Airway Hyperresponsiveness in a Murine Model of Asthma. Int. Arch. Allergy Immunol. 2010, 153, 223–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Lazrak, A.; Creighton, J.; Yu, Z.; Komarova, S.; Doran, S.F.; Aggarwal, S.; Emala, C.W.; Stober, V.P.; Trempus, C.S.; Garantziotis, S.; et al. Hyaluronan mediates airway hyperresponsiveness in oxidative lung injury. Am. J. Physiol. Cell. Mol. Physiol. 2015, 308, L891–L903. [Google Scholar] [CrossRef] [PubMed]
  187. Lamas, A.; Marshburn, J.; Stober, V.P.; Donaldson, S.H.; Garantziotis, S. Effects of inhaled high-molecular weight hyaluronan in inflammatory airway disease. Respir. Res. 2016, 17, 123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Jiang, D.; Liang, J.; Noble, P.W. Regulation of Non-Infectious Lung Injury, Inflammation, and Repair by the Extracellular Matrix Glycosaminoglycan Hyaluronan. Anat. Rec. 2010, 293, 982–985. [Google Scholar] [CrossRef] [Green Version]
  189. Singleton, P.A.; Lennon, F.E. Acute Lung Injury Regulation by Hyaluronan. J. Allergy Ther. 2011, 15 (Suppl. S4), 1–9. [Google Scholar] [CrossRef]
  190. Cui, Z.; Liao, J.; Cheong, N.; Longoria, C.; Cao, G.; DeLisser, H.M.; Savani, R.C. The Receptor for Hyaluronan-Mediated Motility (CD168) promotes inflammation and fibrosis after acute lung injury. Matrix Biol. 2018, 78–79, 255–271. [Google Scholar] [CrossRef]
  191. Liang, J.; Zhang, Y.; Xie, T.; Liu, N.; Chen, H.; Geng, Y.; Kurkciyan, A.; Mena, J.M.; Stripp, B.R.; Jiang, D.; et al. Hyaluronan and TLR4 promote surfactant-protein-C-positive alveolar progenitor cell renewal and prevent severe pulmonary fibrosis in mice. Nat. Med. 2016, 22, 1285–1293. [Google Scholar] [CrossRef]
  192. Aytekin, M.; Comhair, S.A.A.; de la Motte, C.; Bandyopadhyay, S.K.; Farver, C.F.; Hascall, V.C.; Erzurum, S.C.; Dweik, R.A. High levels of hyaluronan in idiopathic pulmonary arterial hypertension. Am. J. Physiol. Cell. Mol. Physiol. 2008, 295, L789–L799. [Google Scholar] [CrossRef] [Green Version]
  193. Lauer, M.E.; Aytekin, M.; Comhair, S.A.; Loftis, J.; Tian, L.; Farver, C.F.; Hascall, V.C.; Dweik, R.A. Modification of Hyaluronan by Heavy Chains of Inter-α-Inhibitor in Idiopathic Pulmonary Arterial Hypertension. J. Biol. Chem. 2014, 289, 6791–6798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Papakonstantinou, E.; Kouri, F.M.; Karakiulakis, G.; Klagas, I.; Eickelberg, O. Increased hyaluronic acid content in idiopathic pulmonary arterial hypertension. Eur. Respir. J. 2008, 32, 1504–1512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Yeager, M.E.; Belchenko, D.D.; Nguyen, C.M.; Colvin, K.L.; Ivy, D.D.; Stenmark, K.R. Endothelin-1, the unfolded protein response, and persistent inflammation: Role of pulmonary artery smooth muscle cells. Am. J. Respir. Cell Mol. Biol. 2012, 46, 14–22. [Google Scholar] [CrossRef] [PubMed]
  196. Hascall, V.C.; Majors, A.K.; De La Motte, C.A.; Evanko, S.P.; Wang, A.; Drazba, J.A.; Strong, S.A.; Wight, T.N. Intracellular hyaluronan: A new frontier for inflammation? Biochim. Biophys. Acta 2004, 1673, 3–12. [Google Scholar] [CrossRef]
  197. Ren, J.; Hascall, V.C.; Wang, A. Cyclin D3 mediates synthesis of a hyaluronan matrix that is adhesive for monocytes in mesangial cells stimulated to divide in hyperglycemic medium. J. Biol. Chem. 2009, 284, 16621–16632. [Google Scholar] [CrossRef] [Green Version]
  198. Sarrazin, S.; Lamanna, W.C.; Esko, J.D. Heparan sulfate proteoglycans. Cold Spring Harb. Perspect. Biol. 2011, 3, a004952. [Google Scholar] [CrossRef] [Green Version]
  199. Mii, Y.; Takada, S. Heparan Sulfate Proteoglycan Clustering in Wnt Signaling and Dispersal. Front. Cell Dev. Biol. 2020, 8, 631. [Google Scholar] [CrossRef]
  200. Yan, D.; Lin, X. Shaping morphogen gradients by proteoglycans. Cold Spring Harb. Perspect. Biol. 2009, 1, a002493. [Google Scholar] [CrossRef]
  201. Gustafsson, E.; Almonte-Becerril, M.; Bloch, W.; Costell, M. Perlecan Maintains Microvessel Integrity In Vivo and Modulates Their Formation In Vitro. PLoS ONE 2013, 8, e53715. [Google Scholar] [CrossRef] [Green Version]
  202. Segev, A.; Nili, N.; Strauss, B.H. The role of perlecan in arterial injury and angiogenesis. Cardiovasc. Res. 2004, 63, 603–610. [Google Scholar] [CrossRef] [Green Version]
  203. Abdul-Salam, V.B.; Wharton, J.; Cupitt, J.; Berryman, M.; Edwards, R.J.; Wilkins, M.R. Proteomic Analysis of Lung Tissues from Patients with Pulmonary Arterial Hypertension. Circulation 2010, 122, 2058–2067. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Chang, Y.-T.; Tseng, C.-N.; Tannenberg, P.; Eriksson, L.; Yuan, K.; Perez, V.; Lundberg, J.; Lengquist, M.; Botusan, I.; Catrina, S.-B.; et al. Perlecan Heparan Sulfate Deficiency Impairs Pulmonary Vascular Development and Attenuates Hypoxic Pulmonary Hypertension. Cardiovasc. Res. 2015, 107, 20–31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Lipke, D.W.; Arcot, S.S.; Gillespie, M.N.; Olson, J.W. Temporal alterations in specific basement membrane components in lungs from monocrotaline-treated rats. Am. J. Respir. Cell Mol. Biol. 1993, 9, 418. [Google Scholar] [CrossRef] [PubMed]
  206. Vyas-Somani, A.C.; Aziz, S.M.; Arcot, S.A.; Gillespie, M.N.; Olson, J.W.; Lipke, D.W. Temporal alterations in basement membrane components in the pulmonary vasculature of the chronically hypoxic rat: Impact of hypoxia and recovery. Am. J. Med. Sci. 1996, 312, 54–67. [Google Scholar] [CrossRef]
  207. Lemire, J.M.; Merrilees, M.J.; Braun, K.R.; Wight, T.N. Overexpression of the V3 variant of versican alters arterial smooth muscle cell adhesion, migration, and proliferation in vitro. J. Cell. Physiol. 2002, 190, 38–45. [Google Scholar] [CrossRef]
  208. Sheng, W.; Wang, G.; Wang, Y.; Liang, J.; Wen, J.; Zheng, P.-S.; Wu, Y.; Lee, V.; Slingerland, J.; Dumont, D.; et al. The Roles of Versican V1 and V2 Isoforms in Cell Proliferation and Apoptosis. Mol. Biol. Cell 2005, 16, 1330–1340. [Google Scholar] [CrossRef]
  209. Chang, Y.T.; Chan, C.K.; Eriksson, I.; Johnson, P.Y.; Cao, X.; Westoo, C.; Norvik, C.; Andersson-Sjoland, A.; Westergren-Thorsson, G.; Johansson, S.; et al. Versican accumulates in vascular lesions in pulmonary arterial hypertension. Pulm. Circ. 2016, 6, 347–359. [Google Scholar] [CrossRef] [Green Version]
  210. Jandl, K.; Marsh, L.M.; Hoffmann, J.; Mutgan, A.C.; Baum, O.; Bloch, W.; Thekkekara-Puthenparampil, H.; Kolb, D.; Sinn, K.; Klepetko, W.; et al. Basement Membrane Remodeling Controls Endothelial Function in Idiopathic Pulmonary Arterial Hypertension. Am. J. Respir. Cell Mol. Biol. 2020, 63, 104–117. [Google Scholar] [CrossRef]
  211. Hassan, H.; Greve, B.; Pavao, M.S.; Kiesel, L.; Ibrahim, S.A.; Götte, M. Syndecan-1 modulates β-integrin-dependent and interleukin-6-dependent functions in breast cancer cell adhesion, migration, and resistance to irradiation. FEBS J. 2013, 280, 2216–2227. [Google Scholar] [CrossRef]
  212. Li, W.; Wang, W. Membrane tension regulates syndecan-1 expression through actin remodelling. Biochim. Biophys. Acta Gen. Subj. 2019, 1863, 129413. [Google Scholar] [CrossRef]
  213. Mochizuki, M.; Güç, E.; Park, A.J.; Julier, Z.; Briquez, P.S.; Kuhn, G.A.; Müller, R.; Swartz, M.A.; Hubbell, J.A.; Martino, M.M. Growth factors with enhanced syndecan binding generate tonic signalling and promote tissue healing. Nat. Biomed. Eng. 2020, 4, 463–475. [Google Scholar] [CrossRef] [PubMed]
  214. Wang, X.; Zuo, D.; Chen, Y.; Li, W.; Liu, R.; He, Y.; Ren, L.; Zhou, L.; Deng, T.; Ying, G.; et al. Shed Syndecan-1 is involved in chemotherapy resistance via the EGFR pathway in colorectal cancer. Br. J. Cancer 2014, 111, 1965–1976. [Google Scholar] [CrossRef] [PubMed]
  215. Su, G.; Blaine, S.A.; Qiao, D.; Friedl, A. Membrane type 1 matrix metalloproteinase-mediated stromal syndecan-1 shedding stimulates breast carcinoma cell proliferation. Cancer Res. 2008, 68, 9558–9565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Nadanaka, S.; Bai, Y.; Kitagawa, H. Cleavage of Syndecan-1 Promotes the Proliferation of the Basal-Like Breast Cancer Cell Line BT-549 Via Akt SUMOylation. Front. Cell Dev. Biol. 2021, 9, 659428. [Google Scholar] [CrossRef]
  217. Guo, J.; Yang, Z.C.; Liu, Y. Attenuating Pulmonary Hypertension by Protecting the Integrity of Glycocalyx in Rats Model of Pulmonary Artery Hypertension. Inflammation 2019, 42, 1951–1956. [Google Scholar] [CrossRef]
  218. Arvidsson, M.; Ahmed, A.; Bouzina, H.; Radegran, G. Plasma proteoglycan prolargin in diagnosis and differentiation of pulmonary arterial hypertension. ESC Heart Fail. 2021, 8, 1230–1243. [Google Scholar] [CrossRef]
  219. Johannes, L.; Jacob, R.; Leffler, H. Galectins at a glance. J. Cell Sci. 2018, 131, jcs208884. [Google Scholar] [CrossRef] [Green Version]
  220. Hughes, R.C. Galectins. In Encyclopedia of Biological Chemistry; Lennarz, W.J., Lane, M.D., Eds.; Elsevier: New York, NY, USA, 2004; pp. 171–174. [Google Scholar]
  221. Liu, F.T.; Hsu, D.K. The role of galectin-3 in promotion of the inflammatory response. Drug News Perspect. 2007, 20, 455–460. [Google Scholar] [CrossRef]
  222. Pang, J.; Rhodes, D.H.; Pini, M.; Akasheh, R.T.; Castellanos, K.J.; Cabay, R.J.; Cooper, D.; Perretti, M.; Fantuzzi, G. Increased Adiposity, Dysregulated Glucose Metabolism and Systemic Inflammation in Galectin-3 KO Mice. PLoS ONE 2013, 8, e57915. [Google Scholar] [CrossRef] [Green Version]
  223. Thurston, T.L.M.; Wandel, M.P.; von Muhlinen, N.; Foeglein, Á.; Randow, F. Galectin 8 targets damaged vesicles for autophagy to defend cells against bacterial invasion. Nature 2012, 482, 414–418. [Google Scholar] [CrossRef]
  224. Thijssen, V.L.J.L.; Postel, R.; Brandwijk, R.J.M.G.E.; Dings, R.P.M.; Nesmelova, I.; Satijn, S.; Verhofstad, N.; Nakabeppu, Y.; Baum, L.G.; Bakkers, J.; et al. Galectin-1 is essential in tumor angiogenesis and is a target for antiangiogenesis therapy. Proc. Natl. Acad. Sci. USA 2006, 103, 15975–15980. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Vasta, G.R. Chapter 8—Lectins as Innate Immune Recognition Factors: Structural, Functional, and Evolutionary Aspects. In The Evolution of the Immune System; Malagoli, D., Ed.; Academic Press: Cambridge, MA, USA, 2016; pp. 205–224. [Google Scholar]
  226. Chou, F.-C.; Chen, H.-Y.; Kuo, C.-C.; Sytwu, H.-K. Role of Galectins in Tumors and in Clinical Immunotherapy. Int. J. Mol. Sci. 2018, 19, 430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Li, L.-c.; Li, J.; Gao, J. Functions of Galectin-3 and Its Role in Fibrotic Diseases. J. Pharmacol. Exp. Ther. 2014, 351, 336–343. [Google Scholar] [CrossRef] [Green Version]
  228. Markowska, A.I.; Liu, F.T.; Panjwani, N. Galectin-3 is an important mediator of VEGF- and bFGF-mediated angiogenic response. J. Exp. Med. 2010, 207, 1981–1993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  229. Croci Diego, O.; Cerliani Juan, P.; Dalotto-Moreno, T.; Méndez-Huergo Santiago, P.; Mascanfroni Ivan, D.; Dergan-Dylon, S.; Toscano Marta, A.; Caramelo Julio, J.; García-Vallejo Juan, J.; Ouyang, J.; et al. Glycosylation-Dependent Lectin-Receptor Interactions Preserve Angiogenesis in Anti-VEGF Refractory Tumors. Cell 2014, 156, 744–758. [Google Scholar] [CrossRef] [Green Version]
  230. Zhong, X.; Qian, X.; Chen, G.; Song, X. The role of galectin-3 in heart failure and cardiovascular disease. Clin. Exp. Pharmacol. Physiol. 2019, 46, 197–203. [Google Scholar] [CrossRef] [Green Version]
  231. Chou, R.-H.; Huang, S.-S.; Kuo, C.-S.; Wang, S.-C.; Tsai, Y.-L.; Lu, Y.-W.; Chang, C.-C.; Huang, P.-H.; Lin, S.-J. Galectin-1 is associated with the severity of coronary artery disease and adverse cardiovascular events in patients undergoing coronary angiography. Sci. Rep. 2020, 10, 20683. [Google Scholar] [CrossRef]
  232. He, J.; Li, X.; Luo, H.; Li, T.; Zhao, L.; Qi, Q.; Liu, Y.; Yu, Z. Galectin-3 mediates the pulmonary arterial hypertension–induced right ventricular remodeling through interacting with NADPH oxidase. J. Am. Soc. Hypertens. 2017, 11, 275–289.e272. [Google Scholar] [CrossRef]
  233. Shen, Q.; Chen, W.; Liu, J.; Liang, Q. Galectin-3 aggravates pulmonary arterial hypertension via immunomodulation in congenital heart disease. Life Sci. 2019, 232, 116546. [Google Scholar] [CrossRef]
  234. Zhang, L.; Li, Y.M.; Zeng, X.X.; Wang, X.Y.; Chen, S.K.; Gui, L.X.; Lin, M.J. Galectin-3- Mediated Transdifferentiation of Pulmonary Artery Endothelial Cells Contributes to Hypoxic Pulmonary Vascular Remodeling. Cell. Physiol. Biochem. 2018, 51, 763–777. [Google Scholar] [CrossRef]
  235. Fulton, D.J.R.; Li, X.; Bordan, Z.; Wang, Y.; Mahboubi, K.; Rudic, R.D.; Haigh, S.; Chen, F.; Barman, S.A. Galectin-3, A Harbinger of Reactive Oxygen Species, Fibrosis, and Inflammation in Pulmonary Arterial Hypertension. Antioxid. Redox Signal 2019, 31, 1053–1069. [Google Scholar] [CrossRef] [PubMed]
  236. Case, D.; Irwin, D.; Ivester, C.; Harral, J.; Morris, K.; Imamura, M.; Roedersheimer, M.; Patterson, A.; Carr, M.; Hagen, M.; et al. Mice deficient in galectin-1 exhibit attenuated physiological responses to chronic hypoxia-induced pulmonary hypertension. Am. J. Physiol. Cell. Mol. Physiol. 2007, 292, L154–L164. [Google Scholar] [CrossRef] [PubMed]
  237. Morrow, R.; Cioffi, E.; Murphy, F.; Cioffi, D. Changes in IgG sialylation and glycosylation in pulmonary arterial hypertension (1089.3). FASEB J. 2014, 28, 1089.3. [Google Scholar] [CrossRef]
  238. Morrow, R.; Cioffi, E.; Cioffi, D. Change in Sialic Acid in Pulmonary Arterial Hypertension. FASEB J. 2013, 27, lb215. [Google Scholar] [CrossRef]
  239. Kawut, S.M.; Horn, E.M.; Berekashvili, K.K.; Widlitz, A.C.; Rosenzweig, E.B.; Barst, R.J. von Willebrand Factor Independently Predicts Long-term Survival in Patients With Pulmonary Arterial Hypertension. Chest 2005, 128, 2355–2362. [Google Scholar] [CrossRef]
  240. Lopes, A.A.; Ferraz de Souza, B.; Maeda, N.Y. Decreased sialic acid content of plasma von Willebrand factor in precapillary pulmonary hypertension. Thromb. Haemost. 2000, 83, 683–687. [Google Scholar] [PubMed]
Figure 1. The glycobiology of PAH. Changes in glycosylation have been linked to dysregulated cellular metabolism, a hallmark of both PAH and cancer. Galectins (Lectins); O-GlcNAc and sialylation (Terminal Glycosylation); as well as hyaluronan, perlecan, versican, aggrecan, and syndecan (Proteoglycans and Glycosaminoglycans) have been investigated in PAH.
Figure 1. The glycobiology of PAH. Changes in glycosylation have been linked to dysregulated cellular metabolism, a hallmark of both PAH and cancer. Galectins (Lectins); O-GlcNAc and sialylation (Terminal Glycosylation); as well as hyaluronan, perlecan, versican, aggrecan, and syndecan (Proteoglycans and Glycosaminoglycans) have been investigated in PAH.
Metabolites 12 00316 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Vang, S.; Cochran, P.; Sebastian Domingo, J.; Krick, S.; Barnes, J.W. The Glycobiology of Pulmonary Arterial Hypertension. Metabolites 2022, 12, 316. https://doi.org/10.3390/metabo12040316

AMA Style

Vang S, Cochran P, Sebastian Domingo J, Krick S, Barnes JW. The Glycobiology of Pulmonary Arterial Hypertension. Metabolites. 2022; 12(4):316. https://doi.org/10.3390/metabo12040316

Chicago/Turabian Style

Vang, Shia, Phillip Cochran, Julio Sebastian Domingo, Stefanie Krick, and Jarrod Wesley Barnes. 2022. "The Glycobiology of Pulmonary Arterial Hypertension" Metabolites 12, no. 4: 316. https://doi.org/10.3390/metabo12040316

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