Skip to main content
Cardiovascular Research logoLink to Cardiovascular Research
. 2020 Oct 18;118(1):84–96. doi: 10.1093/cvr/cvaa299

TNAP as a therapeutic target for cardiovascular calcification: a discussion of its pleiotropic functions in the body

Claudia Goettsch 1, Agnieszka Strzelecka-Kiliszek 2, Laurence Bessueille 3, Thibaut Quillard 4, Laura Mechtouff 5,6, Slawomir Pikula 7, Emmanuelle Canet-Soulas 8, Millan Jose Luis 9, Caroline Fonta 10, David Magne 11,
PMCID: PMC8752354  PMID: 33070177

Abstract

Cardiovascular calcification (CVC) is associated with increased morbidity and mortality. It develops in several diseases and locations, such as in the tunica intima in atherosclerosis plaques, in the tunica media in type 2 diabetes and chronic kidney disease, and in aortic valves. In spite of the wide occurrence of CVC and its detrimental effects on cardiovascular diseases (CVD), no treatment is yet available. Most of CVC involve mechanisms similar to those occurring during endochondral and/or intramembranous ossification. Logically, since tissue-nonspecific alkaline phosphatase (TNAP) is the key-enzyme responsible for skeletal/dental mineralization, it is a promising target to limit CVC. Tools have recently been developed to inhibit its activity and preclinical studies conducted in animal models of vascular calcification already provided promising results. Nevertheless, as its name indicates, TNAP is ubiquitous and recent data indicate that it dephosphorylates different substrates in vivo to participate in other important physiological functions besides mineralization. For instance, TNAP is involved in the metabolism of pyridoxal phosphate and the production of neurotransmitters. TNAP has also been described as an anti-inflammatory enzyme able to dephosphorylate adenosine nucleotides and lipopolysaccharide. A better understanding of the full spectrum of TNAP’s functions is needed to better characterize the effects of TNAP inhibition in diseases associated with CVC. In this review, after a brief description of the different types of CVC, we describe the newly uncovered additional functions of TNAP and discuss the expected consequences of its systemic inhibition in vivo.

Keywords: Tissue non-specific alkaline phosphatase, Cardiovascular calcification, Therapeutic target, Inhibition, Inflammation

Graphical Abstract

graphic file with name cvaa299f4.jpg

1. Introduction

Cardiovascular calcification (CVC) is an independent risk factor for cardiovascular morbidity and mortality. CVC is a common process across ethnicities in the general population and increases with age.1 It is essential to distinguish between three main types of CVC. Two types of CVC with common risk factors and pathogenesis may occur in most ageing adults: intimal atherosclerotic plaque calcification and aortic valve calcification.1,2 The third type of CVC affects the tunica media, in individuals with chronic kidney disease (CKD) or with type 2 diabetes. Media calcification indeed causes arterial stiffness, increased pulse pressure, and left ventricular hypertrophy.3 In CKD, calcification is very likely responsible for the high cardiovascular mortality,4 patients with end-stage renal disease have a 30 times higher risk of death compared to the general population.5 In type 2 diabetes, media calcification first appears in the feet and develops proximally, worsening the risk of lower limb amputation.6,7 Consequences of atherosclerotic intimal plaque calcification are less clear,3 although it is virtually present in every adult.1 Since the calcium score is positively associated with cardiovascular mortality risk,8 it was commonly thought that advanced plaques with heavy calcifications are unstable. This view has been challenged, particularly by the fact that statins increase the calcium score,9 and a consensus has emerged that strongly calcified plaques could be in fact more stable.10 More recently, microcalcifications have been evidenced in early plaques,11–13 where they may destabilize plaques by exerting pro-inflammatory effects and generating mechanical stress within the fibrous cap.14–17 Finally, although aortic valve calcification share many features with intimal calcification, it undoubtedly has a harmful biomechanical and clinical impact.2 Until recently, lack of therapeutic targets and/or of pharmacological inhibitors have prevented CVC inhibition. The identification of tissue-nonspecific alkaline phosphatase (TNAP) as a central player in physiology as well as several types of CVC, and the development of approaches to inhibit TNAP have opened new possibilities.

2. TNAP, a therapeutic target to block cardiovascular calcification

All types of CVC develop at least in part through mechanisms mimicking endochondral (through a cartilage template) and/or intramembranous (without a cartilage template) ossification. Both ossification types have been evidenced in the tunica media of arteries in both humans and rodents with CKD18,19 and diabetes,20,21 in human aortic valves,22,23 and in mouse and human atherosclerotic plaques.10,24–26 In humans, mineralization in endochondral and intramembranous ossification relies on TNAP activity. Indeed, the most severe genetic TNAP deficiencies (hypophosphatasia, HPP) lead to the perinatal death of foetuses devoid of minerals in their whole skeleton.27 Increasing evidence indicates that TNAP plays a role in most types of CVC. TNAP appears as a central player in valve calcification, based on ex vivo experimental models.28,29 In Apolipoprotein (Apo)E-deficient mice, TNAP activity precedes calcification in atherosclerotic plaques.30 In rats, nephrectomized to mimic CKD, TNAP was also detected slightly before vascular calcification (VC) initiation.31 Importantly, local TNAP activation in arteries may be sufficient to trigger VC since its overexpression in vascular smooth muscle cells (VSMCs) or in endothelial cells is sufficient to induce massive and lethal arterial calcification in mice.32–34 Besides its local action on CVC, TNAP may accelerate CVC from the circulation. Indeed, circulating TNAP activity is an independent predictor of mortality in the general population and in individuals with metabolic syndrome (MetS), who have increased serum TNAP levels.35–40 Serum TNAP activity is associated with coronary artery calcium score.41 Moreover, circulating TNAP is associated with increased risk of cardiovascular death in patients with CKD.42–44 In the light of these results, TNAP has emerged as a promising therapeutic target to block CVC, but until recently this target lacked tools to be efficiently inhibited in vivo. Two approaches, based on inhibition of expression or activity, have recently been developed and several preclinical studies have been conducted with encouraging results. These advances open the way to clinical studies aiming to prevent or treat CVC. On the other hand, increasing evidence indicates that TNAP is not only the key player in mineralization but also a ubiquitous enzyme that exerts various and necessary functions in different organs. It is therefore mandatory to thoroughly analyse these functions.

3. TNAP, a ubiquitous enzyme with broad substrate specificity

3.1. TNAP is a membrane-anchored homodimeric enzyme

Humans have in their genome four different loci expressing four distinct alkaline phosphatase (AP) isoenzyme.45 Three of these genes, ALPI, ALPP, and ALPPL2, are tissue-specific in their expression pattern restricted to the intestine, placenta, and germ cells, respectively. The fourth gene, ALPL, is designated as TNAP, since it is expressed in bone, liver, kidney, brain, among others. The 3D structure of mammalian TNAP has not been elucidated so far and can only be assessed based on its 57% identity and 74% homology to the human ALPP and Escherichia coli AP for which crystal structures are known. Both function physiologically as homodimers46 (Figure 1). Structural features of APs involve the monomer–monomer interface the active site with divalent cation-binding sites, the crown domain, the N-terminal arm, a glycosylphosphatidylinositol (GPI) anchor, and N-linked glycosylation sites. The monomer–monomer interface exhibits a strong hydrophobic character demonstrating that <30% of the amino acid residues are involved in two hydrogen-bonding interactions.48 This feature is crucial for stability and enzymatic function. The active site contains three metal-binding sites surrounding the catalytic serine residue that are essential for TNAP enzymatic activity.49 The metal-binding site M1 and M2 are occupied by Zn2+ and M3 is occupied by Mg2+. An additional metal-binding site—M4—is suggested for the binding of Ca2+ that does not alter TNAP catalytic activity.50 The N-terminal α-helix domain together with the crown domain, which is a flexible loop, stabilize the dimeric structure, and determine allosteric properties (Figure 1). In all mammalian AP homodimers, the N-terminal α-helix (residues 9–25) of one monomeric subunit encircle the contralateral subunit reaching towards its active site. These N-terminal α-helical folding is crucial for the structural stability of the second monomer in TNAP and is largely responsible for the allosteric behaviour of mammalian APs.51 Mutation of Arg374 (R374A) and deletion of 5 and 9N-terminal amino acids cause structural and functional disruption.52 In addition, TNAP contains five putative N-linked glycosylation sites (N123, N213, N254, N286, and N413), which are important for catalytic activity.53,54 Another crucial structural element is the GPI anchor enabling the binding of mammalian APs to the surface of the plasma membrane. Plasma membrane-localized phospholipases can enzymatically cleave the anchor, releasing TNAP in the circulation. Both anchored and free APs are active.

Figure 1.

Figure 1

Structural features of TNAP. AMP, adenosine monophosphate; ATP, adenosine triphosphate; GPI, glycosyl phosphatidyl inositol; LPS, lipopolysaccharide; OPN, osteopontin; PPi, inorganic pyrophosphate; PLP, pyridoxal phosphate; TNAP, tissue-nonspecific alkaline phosphatase. Modified from Ref.47

3.2. TNAP has weak substrate specificity in vitro

APs catalyse the hydrolysis of a broad range of phosphate monoesters to form inorganic phosphate (Pi) and alcohol (or phenol) (Figure 1). For example, purified TNAP is able to hydrolyze adenosine triphosphate (ATP), ADP, AMP, inorganic pyrophosphate (PPi), glucose-6-phosphate, β-glycerophosphate, or p-nitrophenyl-phosphate in vitro.55 The reaction mechanisms have been investigated in AP from E. coli, which has an active site with strong homology to mammalian APs, with differences at only three positions,56 suggesting similar reaction mechanisms.57 The reaction generates a serine-phosphate intermediate to produce Pi and an alcohol or phenol. Pi then fills the entire volume of the active site pocket and acts as a strong competitive inhibitor of the enzyme, in a negative feedback loop.58 The two Zn2+ ions play a crucial role in the reaction by coordinating the hydroxyl group of serine within the active site, preparing it for nucleophilic attack of the phosphate monoester substrate and binding the substrate/product.59 Additionally, an arginine residue is important in the initial binding of the substrate and in the release of Pi.56 Finally, the presence of the Mg2+ ion is required for deprotonation of the serine residue.49 The presence of the two Zn2+ ions but not that of the Mg2+ ion in the nucleotide pyrophosphatase phosphodiesterase (NPP) enzymes suggests that the Mg2+ ion could play a fundamental role in the discrimination between the binding and hydrolysis of phosphate monoester and diester.60,61 Indeed, in addition to their phosphomonoesterase activity, APs may have a weaker but significant phosphodiesterase activity,60,61 including TNAP that is able to hydrolyze phosphodiesters in vitro.62 Whether it is present in vivo remains speculative, since all in vivo demonstrated TNAP substrates are monoesters.

3.3. TNAP has a small but growing number of pathophysiological substrates

The mineralizing function of TNAP relies on inorganic pyrophosphate (PPi) hydrolysis rather than on Pi generation. In 1962, Fleisch and Bisaz63 proposed that PPi in the plasma reaches tissues to prevent collagen mineralization, which is triggered by the local hydrolysis of PPi through TNAP. This hypothesis was confirmed by Russell et al. who measured increased PPi excretion in patients with HPP.64 In the 2000s, the group of JL Millan crossed TNAP-deficient mice (Alpl-deficient mice) with mice deficient in genes involved in the generation of extracellular PPi. One of these genes is Ank, the ortholog of the human ANKH gene, encoding an exporter of PPi in the extracellular compartment, whose mutations result in craniometaphyseal dysplasia.65 The other one is Enpp1 (ectonucleotidase pyrophosphatase phosphodiesterase 1) encoding an enzyme that generates PPi from extracellular ATP, whose deficiency in humans is associated with generalized arterial calcification of infancy.66 Crossing Alpl-deficient mice with Ank mice or Enpp1-deficient mice not only reduced the ectopic calcification phenotype of PPi-deficient mice but also prevented the mineralization defects of Alpl-deficient mice.67,68 Finally, Murshed et al.69 demonstrated that TNAP induces mineralization only in tissues containing a fibrillar collagen, which serves as a template for crystal deposition, explaining why only bones and teeth are physiologically mineralized (Figure 2A). However, TNAP is not involved in crystal nucleation but in crystal growth. Bones from HPP patients and TNAP-deficient mice still contain small crystals in matrix vesicles [the so-called extracellular vesicles (EVs) released by mineralizing bone cells70] that have failed to growth and multiply within collagen fibrils.47 This paradigm is strengthened by the fact that PPi ions may not act by inhibiting crystal nucleation, but by binding to the hydration shell of apatite crystals, and hindering Pi binding.71 The molecular mechanisms involved in crystal nucleation remain obscure. Intravesicular dephosphorylation of phosphocholine by phospho1 and calcium ion import by annexins have been proposed as mechanisms to increase ion concentration and induce crystal nucleation.70 Alternatively, poly-ADP ribose released in EVs by senescent bone cells and VSMCs has been proposed to participate to crystal nucleation in bone and the vasculature.72,73 Recent evidence also suggest that phosphatidylserine-mediated nucleation could be the predominant mechanism to produce the very first mineral nuclei during MV-mediated bone/cartilage mineralization.74

Figure 2.

Figure 2

TNAP’s involvement in PPi dephosphorylation leading to the mineralization of collagen-rich tissues (A) and in AMP dephosphorylation leading to resolution of inflammation (B). Ado, adenosine; AMP, adenosine monophosphate; ANKH, progressive ankylosis protein homolog; ATP, adenosine triphosphate; ENPP1, ectonucleotidase pyrophosphatase phosphodiesterase 1; Pi, inorganic phosphate; PPi, inorganic pyrophosphate; TNAP, tissue-nonspecific alkaline phosphatase; VSMC, vascular smooth muscle cell.

Another in vivo substrate of TNAP is pyridoxal phosphate (PLP), the major circulating form of vitamin B6. PLP dephosphorylation in the blood is necessary to allow pyridoxal uptake by cells, where it is phosphorylated again to participate as a cofactor in many reactions.47 In particular, PLP is required for the synthesis of the inhibitory neurotransmitter γ-aminobutyric acid (GABA) in the central nervous system, explaining in part why severe HPP is associated with epileptic seizures.47 Moreover, Alpl-deficient mice have increased levels of cystathionine in their brain, a molecule of the transsulfuration pathway that is controlled by PLP-dependent enzymes.75 Since the transsulfuration pathway is responsible for the synthesis of important anti-oxidant molecules, such as glutathione and hydrogen sulphide, TNAP may indirectly participate in anti-oxidant capacity.

Increasing evidence suggests that adenosine nucleotides are in vivo TNAP substrates. In addition to its intracellular energetic role, ATP is released by all cells, during necrosis, apoptosis, or in response to inflammatory or mechanical stresses.76 Extracellular ATP binds to P2 purinergic receptors in neighbouring cells and activates multiple intracellular pathways. These effects are restrained by CD39 that dephosphorylates ATP into AMP removing two Pi, and CD73 that removes the third Pi to generate adenosine.76 To date, convincing data indicate that TNAP participates at least in AMP dephosphorylation (Figure 2B). Alpl-deficient mice have less adenosine in their brain than control mice,75 suggesting that other tissues also exhibit lack of adenosine. In the blood, TNAP might be a significant contributor to AMP hydrolysis.77 In addition, human neutrophils, which are the most abundant leucocytes in the blood, have very low expression of CD73 and high levels of TNAP,77,78 which is responsible for AMP dephosphorylation77 (Figure 2B). A contribution of TNAP to the dephosphorylation of high AMP levels has also been reported in bronchial epithelial cells.79,80 Given the well-known anti-inflammatory role of adenosine, these data identify TNAP as a possible anti-inflammatory ectonucleotidase.78 The contribution of TNAP to AMP hydrolysis increases with increasing AMP concentrations, suggesting that TNAP is particularly involved in intense pro-inflammatory responses. TNAP’s function in AMP dephosphorylation may significantly interfere with CVC. Patients with arterial calcification due to deficiency of CD73 (ACDC) present with extensive medial calcification.81 The group of St Hilaire demonstrated that decreased adenosine production in absence of CD73 stimulates the expression of TNAP to compensate for deficient AMP dephosphorylation, leading to parallel PPi hydrolysis and calcification.82 Importantly, stimulated TNAP expression due to CD73 deficiency relies on the transcription factor FOXO1 in cells from ACDC patients, and calcified femoropopliteal arteries form non-ACDC patients exhibit increased FOXO1 levels as compared from non-calcified arteries.83 Considered together with the fact that the pro-inflammatory cytokines TNF-α and IL-1β stimulate TNAP expression and calcification in human mesenchymal stem cells (MSCs) while they decrease RUNX2 levels and osteoblast differentiation,84 these results indicate that dysregulated expression of TNAP in pro-inflammatory conditions might be sufficient to trigger CVC. Finally, TNAP dephosphorylates ATP in culture of neurons,85 hypertrophic chondrocytes,78 and MSCs,86 but the in vivo relevance of these findings and whether they are associated with TNAP involvement in inflammation or calcification remains uncertain and deserve further investigation.

Lipopolysaccharide (LPS) is a well-known pro-inflammatory compound synthesized by Gram-negative bacteria. The pro-inflammatory effects of LPS relies on the phosphate groups in the lipid A region.87 IAP dephosphorylates LPS in the gut and prevents the development MetS resulting from repeated LPS absorption accompanying high fat diets.88 LPS is indeed absorbed during fatty meals,89 and contributes to postprandial inflammation and metabolism.90 IAP would thus limit postprandial endotoxemia and keep postprandial inflammation in physiological limits.88 It is likely that in the blood TNAP participates in the dephosphorylation of LPS that has escaped from IAP in the gut. Increasing evidence indicates that human TNAP has the ability to dephosphorylate LPS in vitro and in vivo.91–96 Whether LPS dephosphorylation by TNAP also participates in the control of postprandial inflammation and metabolism is yet only speculative but deserves investigation.

Finally, TNAP may dephosphorylate proteins in vivo. To our knowledge, the first protein that has been suggested to be a TNAP substrate is osteopontin (OPN). OPN is a highly phosphorylated glycoprotein expressed in multiple cell types and may exert multiple functions. OPN can act as an inflammatory cytokine,97 an inducible inhibitor of ectopic calcification,98 and a promoter of regression of ectopic calcification99 and bone resorption.100 Importantly, phosphorylated but not unphosphorylated OPN has been proposed to inhibit ectopic calcification.101  Alpl-deficient mice have increased levels of OPN, and crossing Alpl-deficient mice with Opn-deficient mice partly corrects their mineralization defects.102 TNAP likely dephosphorylates OPN on residues from two distinct regions to stimulate mineralization.103 These effects of TNAP on mineralization mediated by OPN dephosphorylation seem to counter-regulate those relying on PPi hydrolysis since the extracellular Pi/PPi ratio and OPN appear to regulate the expression of the same genes involved in the control of mineralization.102,103 Among other proteins that have been proposed to be TNAP substrates, Tau protein will be discussed in chapter 3.4.

4. Physiological TNAP’s functions and potential consequences of its inhibition

4.1. Inhibition of TNAP expression and/or activity to prevent CVC

Until recently, the most potent available TNAP inhibitor was levamisole (or tetramisole, the racemic mixture of levamisole and its enantiomer dexamisole). However, this inhibitor has TNAP-independent effects,104 for instance, on voltage-dependent sodium channels and/or noradrenaline uptake.104 The first potent and selective TNAP inhibitor is arylsulfonamide 2,5-dimethoxy-N-(quinolin-3-yl) benzenesulfonamide, also known as MLS-0038949.105 MLS-0038949 specifically inhibits TNAP with no effect on IAP. It is however unknown whether MLS-0038949 differently inhibits liver and bone TNAP, which have the same amino acid sequence but differ by their glycosylation pattern.53 MLS-0038949 reduced calcification in culture of VSMCs.106,107 However, MLS-0038949 has only modest pharmacokinetics properties, and had to be modified for translational approaches. In 2018, the group of JL Millan developed 5-((5-chloro-2-methoxyphenyl)sulfonamido)nicotinamide, also known as SBI-425, as a potent and highly selective TNAP inhibitor adapted for in vivo experiments. This inhibitor was first validated in mice overexpressing TNAP under the control of the Tagln promoter active in VSMCs.32 These mice develop extensive arterial calcification, left ventricular hypertrophy and fibrosis, higher systolic blood pressure, and early and sudden mortality, consistent with pressure overload-induced hypertrophy and progression to heart failure. Mice that orally or intravenously received a daily single dose of 10 mg/kg of SBI-425 showed a complete inhibition of plasma TNAP activity, and reduced arterial calcification and prolonged lifespan in TNAP-overexpressing mice.32 SBI-425 also prevented VC in a mouse model of pseudoxanthoma elasticum, a genetic disease associated with impaired PPi generation,108 and in mice with CKD induced by adenine and phosphorus-rich diet.109 Treatment with SBI-425 at 10 or 30 mg/kg/day did not affect kidney function, but fully prevented early death.109 This was associated with the prevention of aorta calcification but not of heart calcification, likely explained by the low penetration of SBI-425 in tissues.110 Nevertheless, these results indicate that TNAP inhibition in general, and SBI-425 in particular, may represent a very promising strategy to treat patients with CKD. On the other hand, patients with CKD also have atherosclerotic intimal plaque calcification that has a very different impact on the vasculature than the specific medial calcification developed by these patients.3 To date, one published article reported the effects of SBI-425 on atherosclerotic plaque calcification in a mouse model of familial hypercholesterolemia, but these mice were crossed with mice overexpressing TNAP in endothelial cells,34 making results difficult to interpret. Oral supplementation of SBI-425 30 mg/kg/day in the wicked high cholesterol (WHC) mouse model induced by a point mutation in the low-density lipoprotein receptor, decreased coronary calcium accumulation, and left ventricular hypertrophy.34 These results indicate that SBI-425 may inhibit intimal atherosclerosis plaque calcification in addition to inhibiting medial calcification associated with CKD. More studies are needed to have a clearer vision of the effects of SBI-425 on cardiovascular mortality.

Besides SBI-425, potential novel inhibitors were tested chemically and putative binding modes were suggested by molecular docking simulations. The isonicotinohydrazone derivative (E)-N'-(4-hydroxy-3-methoxybenzylidene)isonicotinohydrazide was identified as the most potent TNAP inhibitor of the tested derivatives.111 Further, derivatives of 4-quinolones, hybrid compounds from chalcone and 1,2-benzothiazine pharmacophores and trinary benzocoumarin-thiazoles-azomethine derivatives were suggested as selective inhibitors of AP isoenzymes.112–114 Whether these compounds are suitable for in vitro and/or in vivo experiments remains to be determined.

Another approach to diminish TNAP activity is to reduce its protein levels. Recently, the epigenetic regulator apabetalone, an orally available bromodomain and extraterminal (BET) protein inhibitor which is in clinical development for cardiovascular disease treatment, was identified as a TNAP inhibitor acting on expression level. Apabetalone diminished the induction of ALPL mRNA, TNAP protein and enzyme activity in primary human hepatocytes and VSMCs via the transcriptional regulator BRD4 that caused reduction of VC.115 Additionally, apabetalone prevented induction of inflammatory cytokines in vitro and reduced multiple mediators of lipid metabolism and chronic vascular inflammation in the plasma of CVD patients.116 Potential beneficial cardiovascular effects of apabetalone are under investigation. While apabetalone treatment reduced cardiovascular risk and improved kidney function in CKD patients, patients with recent acute coronary syndrome, type 2 diabetes, and low high-density lipoprotein cholesterol levels, did not demonstrate beneficial cardiovascular effects.117–119 In CKD and CVD patients, apabetalone also reduced circulating TNAP levels.119 However, whether the potential favourable cardiovascular effects of apabetalone are causally linked to TNAP reduction remains to be demonstrated.

Therefore, tools now exist to inhibit TNAP and block CVC. A tight evaluation of TNAP inhibitors is however needed in vivo, since as its name indicates, TNAP is ubiquitous, as perfectly illustrated by its pattern of activity during development (Figure 3).120 During vertebrate development and growth, TNAP is active in bone, liver, kidney, nervous, and immune systems, where it exerts known or unknown effects. Its activity changes during ageing, being progressively weaker in bone, but higher in other tissues depending on the presence or absence of different age-related diseases. The next subchapters review these known TNAP functions.

Figure 3.

Figure 3

TNAP activity detected by histochemistry in E15 mouse embryo (bar 500 µm). Taken with permission from Ref.120

4.2. TNAP as an anti-inflammatory enzyme

TNAP is present in the blood as a soluble (anchorless) isoform, originating primarily from bone and liver. Bone is the main source of blood TNAP during growth, when the skeleton is being built, whereas liver progressively becomes the main source of blood TNAP in ageing adults when bone formation slows down.121 Distinction between bone and liver TNAPs in the blood is possible but technically challenging. Bone TNAP levels can be quantified by ELISA, although cross-reactivity as high as 18% has been reported with liver TNAP.122 Unfortunately, there is to date no available ELISA that specifically measures liver TNAP in the blood, which can only be assessed through total TNAP activity. Nevertheless, since increased bone TNAP levels are relatively uncommon in adults, increased TNAP activity in the blood often reflects liver disorders, in particular, during impaired bile flow or cholestasis in primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC) but also non-alcoholic fatty liver disease (NAFLD),123 and more generally MetS.35–40 Circulating TNAP may generate adenosine from AMP to exert anti-inflammatory effects,77 and/or dephosphorylate LPS to detoxify it, in a context of sepsis and/or to regulate postprandial endotoxaemia.92 TNAP is also present in the blood anchored at the membrane of neutrophils,124 the most abundant leukocytes in human blood, where it exerts anti-inflammatory functions.77,78,125 Neutrophil TNAP may support soluble TNAP to hydrolyze PLP, AMP, and LPS, but its presence at the membrane more likely participates in the control of the autocrine effects of adenosine and its nucleotides, in particular on neutrophil survival,126 migration,127 or secretion of IL-1β.78 Finally, TNAP is also present in the blood attached to the membrane of endothelial cells in arterioles and capillaries.34,128,129 Its specific function in endothelial cells is unknown. It might also be related to nucleotide and/or LPS dephosphorylation, or to transport from blood to the endothelium.

If indeed TNAP in the blood exerts important anti-inflammatory effects, its inhibition may have detrimental effects in several age-related diseases. Ageing is associated with a low-grade systemic inflammation, characterized by increased levels of C-reactive protein (CRP), and of cytokines, such as TNF-α and IL-1β.130–132 Exacerbation of this systemic age-associated inflammation may therefore adversely impact health during ageing. Moreover, TNAP inhibition may not only impact systemic inflammation but also tissue inflammation in several diseases, since neutrophils are often the first cells to be recruited in inflamed tissues, e.g. in atherosclerotic plaques, where neutrophils are the first cells to enter and stimulate inflammation.133,134 In plaques, TNAP inhibition may therefore have more complex consequences than only blocking calcification.

4.3. TNAP’S crucial role in skeletal/dental mineralization

The most severe form of HPP leads to the perinatal death of foetuses completely devoid of bone mineral,135 highlighting the requirement for TNAP to mineralize the skeleton and teeth. Bone formation proceeds either by endochondral ossification, in particular, in long bones, or by intramembranous ossification in flat bones. In the former process, mesenchymal cells differentiate under the control of the transcription factor SOX9 into proliferative chondrocytes, which then maturate into mineralizing hypertrophic cells expressing the transcription factor RUNX2.136 During this process, TNAP is expressed in proliferative chondrocytes, but its expression is strongly stimulated by RUNX2 in hypertrophic chondrocytes, where TNAP hydrolyzes extracellular PPi allowing growth plate mineralization.137–140 During endochondral ossification, osteoblasts form bone on this calcified cartilage plate, whereas during endomembranous ossification, they produce bone without the need for a cartilage template. In both cases, osteoblasts express TNAP under the control of RUNX2 and anchor it to their cell membrane to hydrolyze PPi.140 In the teeth, TNAP is expressed in odontoblasts and ameloblasts, the cells responsible for dentin and enamel mineralization, respectively.140,141 This explains why HPP in humans and lack of TNAP in mice result not only in bone hypomineralization but also in decreased dentin and enamel mineralization,141 and lack of acellular cementum, a tissue highly sensitive to extracellular PPi concentration.142,143 In addition to its pro-mineralizing role relying on PPi hydrolysis in bone and tooth cells, TNAP may exert functions in MSCs. TNAP has indeed been identified as a marker of human MSCs,144 where its stimulation may lead to ATP hydrolysis leading to the promotion of osteoblastogenesis at the expense of adipogenesis.86 In this context, it is logical to anticipate that TNAP inhibition will slow down bone formation and exacerbate age-related osteoporosis. These side effects of TNAP inhibition are a concern for patients with CKD, who are prone to develop mineral bone disorder (MBD). They are also relevant for post-menopausal women, who may be at increased risk of developing osteoporosis with a severity inversely associated to the calcium score,145 and have a high risk of cardiovascular mortality.146 The first reports of TNAP inhibition using SBI-425, in a mouse model of pseudoxanthoma elasticum108 and in a mouse model of CKD-MBD109 did not report significant effects on bone architecture. These reports in mice with daily administration of SBI-425 for several weeks were reassuring and paved the way to subsequent studies. On the other hand, one article reported that in addition to preventing the warfarin-induced VC in rats, administration of SBI-425 for 7 weeks decreased bone formation rate and mineral apposition rate, and increased osteoid maturation time, effects that did not yet impact bone architecture as determined by µCT.147 Collectively, these studies indicate that TNAP inhibition with SBI-425 for several weeks has no harmful effects on bone architecture, but that in case of long treatments with high doses, bone homoeostasis has to be carefully controlled. Moreover, if indeed TNAP is an anti-inflammatory enzyme acting by dephosphorylating adenosine nucleotides and LPS, its inhibition on the long term may adversely impact bone homoeostasis through enhanced inflammaging. It is indeed well-known that inflammaging drives the decline in bone mass associated with ageing.148 TNF-α in particular strongly inhibits the expression of RUNX2 in osteoblasts149 and induces its degradation by the proteasome, resulting in impaired osteoblastogenesis.150 Inflammaging-associated reduction of bone TNAP expression may render bones more susceptible to inflammation in a vicious cycle. Alpl+/− mice have increased levels of IL-1β and IL-6 in their bones, which is probably due to TNAP’s nucleotidase activity.78 As mentioned above, SBI-425 does not seem to have dramatic effects on bone homoeostasis but most of the experiments were not realized in aged animals with osteoporosis, which deserve further consideration.

4.4. Emerging functions of TNAP in the central nervous system

TNAP is strongly active in the brain during development (Figure 3), where it is expressed by endothelial and neuronal cells. In endothelial cells, TNAP is active in both the luminal and abluminal sides of the cell membrane, particularly in the arterial part of the microvasculature, but not the venous system.151,152 In human brain vessels, TNAP activity can be detected from gestational ages,153,154 while in mouse brain vessels TNAP is active several days after birth.151,155 This subcellular distribution suggests a role in active transport across capillary endothelial cells, and in blood brain barrier (BBB) permeability.156,157 This role likely includes PLP dephosphorylation in the blood and pyridoxal transport across the BBB. Lack of TNAP in both Alpl-deficient mice and newborns with HPP results in higher PLP levels in blood, reduced GABA synthesis, and epileptic seizures. In addition to endothelial TNAP, neuronal TNAP is likely crucial for normal brain development and functions. During development, neuronal TNAP activity is observed in both grey matter and white matter.158 Interestingly, the association of TNAP with neurogenesis in embryonic rodent brain is maintained in adulthood, with elevated TNAP activity in neurogenic niches.159 Multiple neuroanatomical abnormalities have been observed by MRI in HPP infants, such as hypodensity of the white matter, dilated ventricles, multicystic encephalopathy, parenchymal lesions,160–164 while delay in myelination and synaptogenesis, and abnormalities in the spinal nerve roots were reported in Alpl-deficient mice.139,165 Several mechanisms have been proposed to explain TNAP contribution in brain development: dephosphorylation or interaction with extracellular matrix proteins, such as laminin and collagen,166–168 a role in signal transduction through interaction with PrPc, a plasma membrane GPI anchored protein in lipid rafts highly expressed in the central nervous system,166 regulation of PLP-dependent enzymes, impacting synthesis of GABA, serotonin and dopamine,75,169,170 and a ectonucleotidase function involved in axon formation and growth,85 neurotransmission,171,172 and contributing to the epileptic phenotype of TNAP-deficient pups.173

SBI-425 administration in mice does not trigger epileptic seizures, suggesting that the remaining TNAP activity is sufficient to dephosphorylate PLP in the blood and allow GABA synthesis in the brain. It is however possible that TNAP inhibition may trigger seizures in some patients. Indeed, epileptic seizures have been reported in children treated for paediatric nephrotic syndrome with levamisole.104 In this case, pyridoxine supplementation could prevent these seizures as it does in the infantile and perinatal forms of hypophosphatasia.160,174,175 In addition, SBI-425 does not cross the BBB in healthy mice,176 indicating that it should not alter TNAP functions in brain cells in animals and patients with a normal BBB. However, SBI-425 may cross the BBB whose integrity is compromised by systemic and local inflammation,176 which might be the case in ageing adults with MetS and neurodegenerative disorders, such as Alzheimer’s disease (AD). Analyses of post-mortem AD brains showed an increase in TNAP protein, activity, or expression in hippocampus and temporal gyrus, which are cerebral regions targeted by tau pathology.177,178 Patients with AD also seem to have increased serum levels of TNAP, which are associated with cognitive dysfunction.177,179–181 Activation of TNAP in AD may have both beneficial and harmful effects. First, TNAP may exert protective effects on neuroinflammation by its ability to dephosphorylate ATP into adenosine.75,85,182,183 Second, TNAP may also protect from AD development helping to maintain a functional BBB.176 On the other hand, a scenario based on in vitro approaches, proposed that TNAP participates to AD progression by dephosphorylating extracellular hyperphosphorylated Tau protein.178 Unphosphorylated Tau interacting with muscarinic receptors induces an increase in intracellular calcium that affects calcium homoeostasis triggering neuronal cell death184; it promotes phosphorylation of intracellular Tau which has neurotoxic effects, and it increases TNAP expression.178,185 Thus, a positive feedback loop would result in a link between increases in TNAP and Tau levels, increase in neuronal loss and decline in brain functions. In this complex context, exploring the effects of TNAP inhibition in models of AD and neuroinflammation deserve consideration.

Finally, TNAP inhibition may participate in brain damages induced by ischaemic stroke. Ischaemic strokes account for 85% of strokes and are due to large vessel atherosclerosis in 15–20% of cases.186 Serum TNAP activity is increased in patients with acute ischaemic stroke and is associated with a poor functional outcome, stroke recurrence, and mortality.187–193 TNAP may not only be a biomarker but also an active player in post-ischaemic reperfusion injuries. TNAP may play a protective role in post-ischaemic BBB leakage.176,194 Moreover, TNAP may also reduce neuroinflammation following stroke. Stroke leads to cell necrosis, which triggers the release of ATP and a subsequent inflammation that is resolved by the dephosphorylation of ATP into adenosine.195 The reported involvement of TNAP in ATP hydrolysis and/or adenosine generation in neurons85,182,183 and in neutrophils,77,78 which are recruited by ATP release,196 and play multiple pro-inflammatory roles during stroke,197–199 may suggest that TNAP inhibition might worsen post-ischaemic neuroinflammation. Therefore, the effects of TNAP inhibition on brain damages and functional outcome after stroke deserves investigation.

4.5. TNAP’S functions in liver and kidneys

TNAP’s former name was liver/bone/kidney (L/B/K) AP. The expression of TNAP in liver and kidney is known for a long time, but its function in these tissues remains elusive. In the liver, TNAP is localized at the canalicular membrane of hepatocytes and the apical area of the cytoplasm of bile duct epithelial cells.140,200,201 Cholangitis is associated with increased release of liver TNAP into the blood,123 suggesting that TNAP participates in bile excretion. TNAP may dephosphorylate ATP at the cholangiocyte surface to participate in the regulation of bile pH in response to the secretion of bile salts.202 Additionally, liver TNAP may participate in LPS detoxification and excretion in the bile.91–94,203 Whether this regulation takes place under pathological conditions during bacterial infection, or physiologically to regulate postprandial endotoxemia is worth exploring. Finally, it is possible that liver expresses TNAP mainly to release it in the blood in adults to control systemic inflammation when bone formation is reduced and bone TNAP release in the blood is decreased. To our knowledge, no effect of TNAP inhibition on the liver has been reported to date, but most studies were conducted in mice which, in contrast to humans and rats, have very weak expression of liver TNAP.201

In the kidney, TNAP is expressed at the brush borders of proximal renal tubules.140,204 This renal expression of TNAP may appear surprising since prevention of mineralization in urine is supposed to rely on the presence of PPi, the mineralization inhibitor inactivated by TNAP.205 However, the main production site of PPi may be the distal nephron where TNAP is absent,205 which may explain why TNAP does not induce urinary tract calcification. Instead, in the kidney brush border, TNAP induced by LPS may be able to dephosphorylate and detoxify it.206,207 Whether this function occurs in physiological and/or in pathological situations remains obscure, but it is worth exploring.

5. Conclusion

An increasing number of articles now indicate that TNAP is responsible, or at least involved, in CVC, and tools to inhibit its expression and activity are now available. Several preclinical studies showed that TNAP inhibition strongly reduces VC.108,109,147 Since TNAP is ubiquitous and likely has several important functions in addition to its well-established role in the control of the Pi/PPi ratio during physiological and ectopic mineralization, there is now a compelling need to perform studies to better understand these additional functions, identifying its associated substrates, and elucidating these new metabolic pathways.

Conflict of interest: J.L.M. is a co-inventor on a patent covering SBI-425 (PCT WO 2013126608).

Funding

C.G., A.S.K., L.B., S.P., and D.M. received financial support from European Research Area Network on Cardiovascular Diseases (ERA-NET CVD, Microexploration project 2018–21) to work on TNAP and atherosclerotic plaque calcification; C.G. received funding from the German Research Foundation (GO1801/5-1 SFB/TRR219); D.M. received support from Fondation de France (2019–20, project 00086497) to work on TNAP, inflammation, and plaque calcification. J.L.M. is funded by grant DE12889 from the National Institute of Dental and Craniofacial Research, NIH, USA.

Contributor Information

Claudia Goettsch, Department of Internal Medicine I, Cardiology, Medical Faculty, RWTH Aachen University, Aachen, Germany.

Agnieszka Strzelecka-Kiliszek, Laboratory of Biochemistry of Lipids, Nencki Institute of Experimental Biology, 3 Pasteur Street, 02-093 Warsaw, Poland.

Laurence Bessueille, Institute of Molecular and Supramolecular Chemistry and Biochemistry (ICBMS), UMR CNRS 5246, Université Claude Bernard Lyon 1, Bâtiment Raulin, 43 Bd du 11 novembre 1918, Lyon 69622 Villeurbanne Cedex, France.

Thibaut Quillard, PHY-OS Laboratory, UMR 1238 INSERM, Université de Nantes, CHU de Nantes, France.

Laura Mechtouff, Stroke Department, Hospices Civils de Lyon, France; CREATIS Laboratory, CNRS UMR 5220, Inserm U1044, Université Claude Bernard Lyon 1, Lyon, France.

Slawomir Pikula, Laboratory of Biochemistry of Lipids, Nencki Institute of Experimental Biology, 3 Pasteur Street, 02-093 Warsaw, Poland.

Emmanuelle Canet-Soulas, CarMeN Laboratory, Univ Lyon, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Lyon, France.

Millan Jose Luis, Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.

Caroline Fonta, Brain and Cognition Research Center CerCo, CNRS UMR5549, Université de Toulouse, France.

David Magne, Institute of Molecular and Supramolecular Chemistry and Biochemistry (ICBMS), UMR CNRS 5246, Université Claude Bernard Lyon 1, Bâtiment Raulin, 43 Bd du 11 novembre 1918, Lyon 69622 Villeurbanne Cedex, France.

References

  • 1. Doherty TM, Fitzpatrick LA, Inoue D, Qiao JH, Fishbein MC, Detrano RC, Shah PK, Rajavashisth TB.  Molecular, endocrine, and genetic mechanisms of arterial calcification. Endocr Rev  2004;25:629–672. [DOI] [PubMed] [Google Scholar]
  • 2. Pawade TA, Newby DE, Dweck MR.  Calcification in aortic stenosis: the skeleton key. J Am Coll Cardiol  2015;66:561–577. [DOI] [PubMed] [Google Scholar]
  • 3. Vervloet M, Cozzolino M.  Vascular calcification in chronic kidney disease: different bricks in the wall?  Kidney Int  2017;91:808–817. [DOI] [PubMed] [Google Scholar]
  • 4. Schlieper G, Krüger T, Djuric Z, Damjanovic T, Markovic N, Schurgers LJ, Brandenburg VM, Westenfeld R, Dimkovic S, Ketteler M, Grootendorst DC, Dekker FW, Floege J, Dimkovic N.  Vascular access calcification predicts mortality in hemodialysis patients. Kidney Int  2008;74:1582–1587. [DOI] [PubMed] [Google Scholar]
  • 5. Foley RN, Parfrey PS, Sarnak MJ.  Clinical epidemiology of cardiovascular disease in chronic renal disease. Am J Kidney Dis  1998;32:S112–S119. [DOI] [PubMed] [Google Scholar]
  • 6. Everhart JE, Pettitt DJ, Knowler WC, Rose FA, Bennett PH.  Medial arterial calcification and its association with mortality and complications of diabetes. Diabetologia  1988;31:16–23. [DOI] [PubMed] [Google Scholar]
  • 7. Lehto S, Niskanen L, Suhonen M, RöNnemaa T, Laakso M.  Medial artery calcification. A neglected harbinger of cardiovascular complications in non-insulin-dependent diabetes mellitus. Arterioscler Thromb Vasc Biol  1996;16:978–983. [DOI] [PubMed] [Google Scholar]
  • 8. Greenland P, LaBree L, Azen SP, Doherty TM, Detrano RC.  Coronary artery calcium score combined with Framingham score for risk prediction in asymptomatic individuals. JAMA  2004;291:210–215. [DOI] [PubMed] [Google Scholar]
  • 9. Puri R, Nicholls SJ, Shao M, Kataoka Y, Uno K, Kapadia SR, Tuzcu EM, Nissen SE.  Impact of statins on serial coronary calcification during atheroma progression and regression. J Am Coll Cardiol  2015;65:1273–1282. [DOI] [PubMed] [Google Scholar]
  • 10. Mori H, Torii S, Kutyna M, Sakamoto A, Finn AV, Virmani R.  Coronary artery calcification and its progression: what does it really mean?  JACC Cardiovasc Imaging  2018;11:127–142. [DOI] [PubMed] [Google Scholar]
  • 11. Chatrou ML, Cleutjens JP, van der Vusse GJ, Roijers RB, Mutsaers PH, Schurgers LJ.  Intra-section analysis of human coronary arteries reveals a potential role for micro-calcifications in macrophage recruitment in the early stage of atherosclerosis. PLoS One  2015;10:e0142335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Roijers RB, Dutta RK, Cleutjens JP, Mutsaers PH, de Goeij JJ, van der Vusse GJ.  Early calcifications in human coronary arteries as determined with a proton microprobe. Anal Chem  2008;80:55–61. [DOI] [PubMed] [Google Scholar]
  • 13. Roijers RB, Debernardi N, Cleutjens JP, Schurgers LJ, Mutsaers PH, van der Vusse GJ.  Microcalcifications in early intimal lesions of atherosclerotic human coronary arteries. Am J Pathol  2011;178:2879–2887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Nadra I, Mason JC, Philippidis P, Florey O, Smythe CD, McCarthy GM, Landis RC, Haskard DO.  Proinflammatory activation of macrophages by basic calcium phosphate crystals via protein kinase C and MAP kinase pathways: a vicious cycle of inflammation and arterial calcification?  Circ Res  2005;96:1248–1256. [DOI] [PubMed] [Google Scholar]
  • 15. Kelly-Arnold A, Maldonado N, Laudier D, Aikawa E, Cardoso L, Weinbaum S.  Revised microcalcification hypothesis for fibrous cap rupture in human coronary arteries. Proc Natl Acad Sci USA  2013;110:10741–10746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Ehara S, Kobayashi Y, Yoshiyama M, Shimada K, Shimada Y, Fukuda D, Nakamura Y, Yamashita H, Yamagishi H, Takeuchi K, Naruko T, Haze K, Becker AE, Yoshikawa J, Ueda M.  Spotty calcification typifies the culprit plaque in patients with acute myocardial infarction: an intravascular ultrasound study. Circulation  2004;110:3424–3429. [DOI] [PubMed] [Google Scholar]
  • 17. Maldonado N, Kelly-Arnold A, Vengrenyuk Y, Laudier D, Fallon JT, Virmani R, Cardoso L, Weinbaum S.  A mechanistic analysis of the role of microcalcifications in atherosclerotic plaque stability: potential implications for plaque rupture. Am J Physiol Heart Circ Physiol  2012;303:H619–H628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Neven E, Dauwe S, De Broe ME, D'Haese PC, Persy V.  Endochondral bone formation is involved in media calcification in rats and in men. Kidney Int  2007;72:574–581. [DOI] [PubMed] [Google Scholar]
  • 19. Neven E, Persy V, Dauwe S, De Schutter T, De Broe ME, D'Haese PC.  Chondrocyte rather than osteoblast conversion of vascular cells underlies medial calcification in uremic rats. Arterioscler Thromb Vasc Biol  2010;30:1741–1750. [DOI] [PubMed] [Google Scholar]
  • 20. Shanahan CM, Cary NR, Salisbury JR, Proudfoot D, Weissberg PL, Edmonds ME.  Medial localization of mineralization-regulating proteins in association with Mönckeberg's sclerosis: evidence for smooth muscle cell-mediated vascular calcification. Circulation  1999;100:2168–2176. [DOI] [PubMed] [Google Scholar]
  • 21. Qiao JH, Mertens RB, Fishbein MC, Geller SA.  Cartilaginous metaplasia in calcified diabetic peripheral vascular disease: morphologic evidence of enchondral ossification. Hum Pathol  2003;34:402–407. [DOI] [PubMed] [Google Scholar]
  • 22. Mohler ER, Gannon F, Reynolds C, Zimmerman R, Keane MG, Kaplan FS.  Bone formation and inflammation in cardiac valves. Circulation  2001;103:1522–1528. [DOI] [PubMed] [Google Scholar]
  • 23. Torre M, Hwang DH, Padera RF, Mitchell RN, VanderLaan PA.  Osseous and chondromatous metaplasia in calcific aortic valve stenosis. Cardiovasc Pathol  2016;25:18–24. [DOI] [PubMed] [Google Scholar]
  • 24. Rattazzi M, Bennett BJ, Bea F, Kirk EA, Ricks JL, Speer M, Schwartz SM, Giachelli CM, Rosenfeld ME.  Calcification of advanced atherosclerotic lesions in the innominate arteries of ApoE-deficient mice: potential role of chondrocyte-like cells. Arterioscler Thromb Vasc Biol  2005;25:1420–1425. [DOI] [PubMed] [Google Scholar]
  • 25. Herisson F, Heymann MF, Chétiveaux M, Charrier C, Battaglia S, Pilet P, Rouillon T, Krempf M, Lemarchand P, Heymann D, Gouëffic Y.  Carotid and femoral atherosclerotic plaques show different morphology. Atherosclerosis  2011;216:348–354. [DOI] [PubMed] [Google Scholar]
  • 26. Tyson KL, Reynolds JL, McNair R, Zhang Q, Weissberg PL, Shanahan CM.  Osteo/chondrocytic transcription factors and their target genes exhibit distinct patterns of expression in human arterial calcification. Arterioscler Thromb Vasc Biol  2003;23:489–494. [DOI] [PubMed] [Google Scholar]
  • 27. Whyte MP, Fujita KP, Moseley S, Thompson DD, McAlister WH.  Validation of a novel scoring system for changes in skeletal manifestations of hypophosphatasia in newborns, infants, and children: the radiographic global impression of change scale. J Bone Miner Res  2018;33:868–874. [DOI] [PubMed] [Google Scholar]
  • 28. Rathan S, Yoganathan AP, O'Neill CW.  The role of inorganic pyrophosphate in aortic valve calcification. J Heart Valve Dis  2014;23:387–394. [PMC free article] [PubMed] [Google Scholar]
  • 29. Rattazzi M, Bertacco E, Iop L, D'Andrea S, Puato M, Buso G, Causin V, Gerosa G, Faggin E, Pauletto P.  Extracellular pyrophosphate is reduced in aortic interstitial valve cells acquiring a calcifying profile: implications for aortic valve calcification. Atherosclerosis  2014;237:568–576. [DOI] [PubMed] [Google Scholar]
  • 30. Aikawa E, Nahrendorf M, Figueiredo JL, Swirski FK, Shtatland T, Kohler RH, Jaffer FA, Aikawa M, Weissleder R.  Osteogenesis associates with inflammation in early-stage atherosclerosis evaluated by molecular imaging in vivo. Circulation  2007;116:2841–2850. [DOI] [PubMed] [Google Scholar]
  • 31. Hortells L, Sosa C, Guillén N, Lucea S, Millán Á, Sorribas V.  Identifying early pathogenic events during vascular calcification in uremic rats. Kidney Int  2017;92:1384–1394. [DOI] [PubMed] [Google Scholar]
  • 32. Sheen CR, Kuss P, Narisawa S, Yadav MC, Nigro J, Wang W, Chhea TN, Sergienko EA, Kapoor K, Jackson MR, Hoylaerts MF, Pinkerton AB, O'Neill WC, Millán JL.  Pathophysiological role of vascular smooth muscle alkaline phosphatase in medial artery calcification. J Bone Miner Res  2015;30:824–836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Savinov AY, Salehi M, Yadav MC, Radichev I, Millán JL, Savinova OV.  Transgenic overexpression of tissue-nonspecific alkaline phosphatase (TNAP) in vascular endothelium results in generalized arterial calcification. J Am Heart Assoc  2015;4:e002499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Romanelli F, Corbo A, Salehi M, Yadav MC, Salman S, Petrosian D, Rashidbaigi OJ, Chait J, Kuruvilla J, Plummer M, Radichev I, Margulies KB, Gerdes AM, Pinkerton AB, Millán JL, Savinov AY, Savinova OV.  Overexpression of tissue-nonspecific alkaline phosphatase (TNAP) in endothelial cells accelerates coronary artery disease in a mouse model of familial hypercholesterolemia. PLoS One  2017;12:e0186426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Lammers WJ, van Buuren HR, Hirschfield GM, Janssen HL, Invernizzi P, Mason AL, Ponsioen CY, Floreani A, Corpechot C, Mayo MJ, Battezzati PM, Parés A, Nevens F, Burroughs AK, Kowdley KV, Trivedi PJ, Kumagi T, Cheung A, Lleo A, Imam MH, Boonstra K, Cazzagon N, Franceschet I, Poupon R, Caballeria L, Pieri G, Kanwar PS, Lindor KD, Hansen BE, Group GPS.  Levels of alkaline phosphatase and bilirubin are surrogate end points of outcomes of patients with primary biliary cirrhosis: an international follow-up study. Gastroenterology  2014;147:1338–1349.e1335; quiz e1315. [DOI] [PubMed] [Google Scholar]
  • 36. Tonelli M, Curhan G, Pfeffer M, Sacks F, Thadhani R, Melamed ML, Wiebe N, Muntner P.  Relation between alkaline phosphatase, serum phosphate, and all-cause or cardiovascular mortality. Circulation  2009;120:1784–1792. [DOI] [PubMed] [Google Scholar]
  • 37. Abramowitz M, Muntner P, Coco M, Southern W, Lotwin I, Hostetter TH, Melamed ML.  Serum alkaline phosphatase and phosphate and risk of mortality and hospitalization. Clin J Am Soc Nephrol  2010;5:1064–1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Krishnamurthy VR, Baird BC, Wei G, Greene T, Raphael K, Beddhu S.  Associations of serum alkaline phosphatase with metabolic syndrome and mortality. Am J Med  2011;124:566.e561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Wannamethee SG, Sattar N, Papcosta O, Lennon L, Whincup PH.  Alkaline phosphatase, serum phosphate, and incident cardiovascular disease and total mortality in older men. Arterioscler Thromb Vasc Biol  2013;33:1070–1076. [DOI] [PubMed] [Google Scholar]
  • 40. Filipowicz R, Greene T, Wei G, Cheung AK, Raphael KL, Baird BC, Beddhu S.  Associations of serum skeletal alkaline phosphatase with elevated C-reactive protein and mortality. Clin J Am Soc Nephrol  2013;8:26–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Panh L, Ruidavets JB, Rousseau H, Petermann A, Bongard V, Bérard E, Taraszkiewicz D, Lairez O, Galinier M, Carrié D, Ferrières J.  Association between serum alkaline phosphatase and coronary artery calcification in a sample of primary cardiovascular prevention patients. Atherosclerosis  2017;260:81–86. [DOI] [PubMed] [Google Scholar]
  • 42. Beddhu S, Ma X, Baird B, Cheung AK, Greene T.  Serum alkaline phosphatase and mortality in African Americans with chronic kidney disease. Clin J Am Soc Nephrol  2009;4:1805–1810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Kalantar-Zadeh K, Shah A, Duong U, Hechter RC, Dukkipati R, Kovesdy CP.  Kidney bone disease and mortality in CKD: revisiting the role of vitamin D, calcimimetics, alkaline phosphatase, and minerals. Kidney Int Suppl  2010;78:S10–S21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Blayney MJ, Pisoni RL, Bragg-Gresham JL, Bommer J, Piera L, Saito A, Akiba T, Keen ML, Young EW, Port FK.  High alkaline phosphatase levels in hemodialysis patients are associated with higher risk of hospitalization and death. Kidney Int  2008;74:655–663. [DOI] [PubMed] [Google Scholar]
  • 45. Buchet R, Millán JL, Magne D.  Multisystemic functions of alkaline phosphatases. Methods Mol Biol  2013;1053:27–51. [DOI] [PubMed] [Google Scholar]
  • 46. Le Du M-H, Millán JL.  Structural evidence of functional divergence in human alkaline phosphatases. J Biol Chem  2002;277:49808–49814. [DOI] [PubMed] [Google Scholar]
  • 47. Millán JL, Whyte MP.  Alkaline phosphatase and hypophosphatasia. Calcif Tissue Int  2016;98:398–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Hummer C, Millán JL.  Gly429 is the major determinant of uncompetitive inhibition of human germ cell alkaline phosphatase by L-leucine. Biochem J  1991;274:91–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Stec B, Holtz KM, Kantrowitz ER.  A revised mechanism for the alkaline phosphatase reaction involving three metal ions. J Mol Biol  2000;299:1303–1311. [DOI] [PubMed] [Google Scholar]
  • 50. Hoylaerts MF, Van Kerckhoven S, Kiffer-Moreira T, Sheen C, Narisawa S, Millán JL.  Functional significance of calcium binding to tissue-nonspecific alkaline phosphatase. PLoS One  2015;10:e0119874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Hoylaerts MF, Manes T, Millán JL.  Mammalian alkaline phosphatases are allosteric enzymes. J Biol Chem  1997;272:22781–22787. [DOI] [PubMed] [Google Scholar]
  • 52. Hoylaerts MF, Ding L, Narisawa S, Van Kerckhoven S, Millán JL.  Mammalian alkaline phosphatase catalysis requires active site structure stabilization via the N-terminal amino acid microenvironment. Biochemistry  2006;45:9756–9766. [DOI] [PubMed] [Google Scholar]
  • 53. Nosjean O, Koyama I, Goseki M, Roux B, Komoda T.  Human tissue non-specific alkaline phosphatases: sugar-moiety-induced enzymic and antigenic modulations and genetic aspects. Biochem J  1997;321:297–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Halling Linder C, Narisawa S, Millán JL, Magnusson P.  Glycosylation differences contribute to distinct catalytic properties among bone alkaline phosphatase isoforms. Bone  2009;45:987–993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Say JC, Ciuffi K, Furriel RP, Ciancaglini P, Leone FA.  Alkaline phosphatase from rat osseous plates: purification and biochemical characterization of a soluble form. Biochim Biophys Acta  1991;1074:256–262. [DOI] [PubMed] [Google Scholar]
  • 56. Holtz KM, Kantrowitz ER.  The mechanism of the alkaline phosphatase reaction: insights from NMR, crystallography and site-specific mutagenesis. FEBS Lett  1999;462:7–11. [DOI] [PubMed] [Google Scholar]
  • 57. Millán JL.  Alkaline phosphatases: structure, substrate specificity and functional relatedness to other members of a large superfamily of enzymes. Purinergic Signal  2006;2:335–341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Schwartz JH, Lipmann F.  Phosphate incorporation into alkaline phosphatase of E. coli. Proc Natl Acad Sci USA  1961;47:1996–2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Kim EE, Wyckoff HW.  Reaction mechanism of alkaline phosphatase based on crystal structures. Two-metal ion catalysis. J Mol Biol  1991;218:449–464. [DOI] [PubMed] [Google Scholar]
  • 60. Zalatan JG, Fenn TD, Herschlag D.  Comparative enzymology in the alkaline phosphatase superfamily to determine the catalytic role of an active-site metal ion. J Mol Biol  2008;384:1174–1189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. López-Canut V, Roca M, Bertrán J, Moliner V, Tuñón I, Promiscuity in alkaline phosphatase superfamily. Unraveling evolution through molecular simulations. J Am Chem Soc  2011;133:12050–12062. [DOI] [PubMed] [Google Scholar]
  • 62. Rezende AA, Pizauro JM, Ciancaglini P, Leone FA.  Phosphodiesterase activity is a novel property of alkaline phosphatase from osseous plate. Biochem J  1994;301:517–522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Fleisch H, Bisaz S.  Mechanism of calcification: inhibitory role of pyrophosphate. Nature  1962;195:911–911. [DOI] [PubMed] [Google Scholar]
  • 64. Russell RG.  Excretion of inorganic pyrophosphate in hypophosphatasia. Lancet  1965;286:461–464. [DOI] [PubMed] [Google Scholar]
  • 65. Nürnberg P, Thiele H, Chandler D, Höhne W, Cunningham ML, Ritter H, Leschik G, Uhlmann K, Mischung C, Harrop K, Goldblatt J, Borochowitz ZU, Kotzot D, Westermann F, Mundlos S, Braun HS, Laing N, Tinschert S.  Heterozygous mutations in ANKH, the human ortholog of the mouse progressive ankylosis gene, result in craniometaphyseal dysplasia. Nat Genet  2001;28:37–41. [DOI] [PubMed] [Google Scholar]
  • 66. Rutsch F, Ruf N, Vaingankar S, Toliat MR, Suk A, Höhne W, Schauer G, Lehmann M, Roscioli T, Schnabel D, Epplen JT, Knisely A, Superti-Furga A, McGill J, Filippone M, Sinaiko AR, Vallance H, Hinrichs B, Smith W, Ferre M, Terkeltaub R, Nürnberg P.  Mutations in ENPP1 are associated with ‘idiopathic’ infantile arterial calcification. Nat Genet  2003;34:379–381. [DOI] [PubMed] [Google Scholar]
  • 67. Hessle L, Johnson KA, Anderson HC, Narisawa S, Sali A, Goding JW, Terkeltaub R, Millan JL.  Tissue-nonspecific alkaline phosphatase and plasma cell membrane glycoprotein-1 are central antagonistic regulators of bone mineralization. Proc Natl Acad Sci USA  2002;99:9445–9449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Harmey D, Hessle L, Narisawa S, Johnson KA, Terkeltaub R, Millán JL.  Concerted regulation of inorganic pyrophosphate and osteopontin by akp2, enpp1, and ank: an integrated model of the pathogenesis of mineralization disorders. Am J Pathol  2004;164:1199–1209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Murshed M, Harmey D, Millán JL, McKee MD, Karsenty G.  Unique coexpression in osteoblasts of broadly expressed genes accounts for the spatial restriction of ECM mineralization to bone. Genes Dev  2005;19:1093–1104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Bottini M, Mebarek S, Anderson KL, Strzelecka-Kiliszek A, Bozycki L, Simão AMS, Bolean M, Ciancaglini P, Pikula JB, Pikula S, Magne D, Volkmann N, Hanein D, Millán JL, Buchet R.  Matrix vesicles from chondrocytes and osteoblasts: their biogenesis, properties, functions and biomimetic models. Biochim Biophys Acta  2018;1862:532–546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Fleisch H, Russell RG, Straumann F.  Effect of pyrophosphate on hydroxyapatite and its implications in calcium homeostasis. Nature  1966;212:901–903. [DOI] [PubMed] [Google Scholar]
  • 72. Müller KH, Hayward R, Rajan R, Whitehead M, Cobb AM, Ahmad S, Sun M, Goldberga I, Li R, Bashtanova U, Puszkarska AM, Reid DG, Brooks RA, Skepper JN, Bordoloi J, Chow WY, Oschkinat H, Groombridge A, Scherman OA, Harrison JA, Verhulst A, D’Haese PC, Neven E, Needham L-M, Lee SF, Shanahan CM, Duer MJ.  Poly(ADP-Ribose) links the DNA damage response and biomineralization. Cell Rep  2019;27:3124–3138.e3113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Ragnauth CD, Warren DT, Liu Y, McNair R, Tajsic T, Figg N, Shroff R, Skepper J, Shanahan CM.  Prelamin A acts to accelerate smooth muscle cell senescence and is a novel biomarker of human vascular aging. Circulation  2010;121:2200–2210. [DOI] [PubMed] [Google Scholar]
  • 74. Cruz MAE, Ferreira CR, Tovani CB, de Oliveira FA, Bolean M, Caseli L, Mebarek S, Luis Millán J, Buchet R, Bottini M, Ciancaglini P, Paula Ramos A.  Phosphatidylserine controls calcium phosphate nucleation and growth on lipid monolayers: a physicochemical understanding of matrix vesicles-driven biomineralization. J Struct Biol  2020;212:107607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Cruz T, Gleizes M, Balayssac S, Mornet E, Marsal G, Millán JL, Malet-Martino M, Nowak LG, Gilard V, Fonta C.  Identification of altered brain metabolites associated with TNAP activity in a mouse model of hypophosphatasia using untargeted NMR-based metabolomics analysis. J Neurochem  2017;140:919–940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Antonioli L, Pacher P, Vizi ES, Haskó G.  CD39 and CD73 in immunity and inflammation. Trends Mol Med  2013;19:355–367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Pettengill M, Robson S, Tresenriter M, Millán JL, Usheva A, Bingham T, Belderbos M, Bergelson I, Burl S, Kampmann B, Gelinas L, Kollmann T, Bont L, Levy O.  Soluble ecto-5'-nucleotidase (5'-NT), alkaline phosphatase, and adenosine deaminase (ADA1) activities in neonatal blood favor elevated extracellular adenosine. J Biol Chem  2013;288:27315–27326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Bessueille L, Briolay A, Como J, Mebarek S, Mansouri C, Gleizes M, El Jamal A, Buchet R, Dumontet C, Matera EL, Mornet E, Millan JL, Fonta C, Magne D.  Tissue-nonspecific alkaline phosphatase is an anti-inflammatory nucleotidase. Bone  2020;133:115262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Picher M, Burch LH, Hirsh AJ, Spychala J, Boucher RC.  Ecto 5'-nucleotidase and nonspecific alkaline phosphatase. Two AMP-hydrolyzing ectoenzymes with distinct roles in human airways. J Biol Chem  2003;278:13468–13479. [DOI] [PubMed] [Google Scholar]
  • 80. Woods PS, Doolittle LM, Hickman-Davis JM, Davis IC.  ATP catabolism by tissue nonspecific alkaline phosphatase contributes to development of ARDS in influenza-infected mice. Am J Physiol Lung Cell Mol Physiol  2018;314:L83–L92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. St Hilaire C, Ziegler SG, Markello TC, Brusco A, Groden C, Gill F, Carlson-Donohoe H, Lederman RJ, Chen MY, Yang D, Siegenthaler MP, Arduino C, Mancini C, Freudenthal B, Stanescu HC, Zdebik AA, Chaganti RK, Nussbaum RL, Kleta R, Gahl WA, Boehm M.  NT5E mutations and arterial calcifications. N Engl J Med  2011;364:432–442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Jin H, St Hilaire C, Huang Y, Yang D, Dmitrieva NI, Negro A, Schwartzbeck R, Liu Y, Yu Z, Walts A, Davaine JM, Lee DY, Donahue D, Hsu KS, Chen J, Cheng T, Gahl W, Chen G, Boehm M.  Increased activity of TNAP compensates for reduced adenosine production and promotes ectopic calcification in the genetic disease. Sci Signal  2016;9:ra121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Moorhead WJ, Chu CC, Cuevas RA, Callahan J, Wong R, Regan C, Boufford CK, Sur S, Liu M, Gomez D, MacTaggart JN, Kamenskiy A, Boehm M, St Hilaire C.  Dysregulation of FOXO1 (Forkhead Box O1 Protein) drives calcification in arterial calcification due to deficiency of CD73 and is present in peripheral artery disease. Arterioscler Thromb Vasc Biol  2020;40:1680–1694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Ding J, Ghali O, Lencel P, Broux O, Chauveau C, Devedjian JC, Hardouin P, Magne D.  TNF-alpha and IL-1beta inhibit RUNX2 and collagen expression but increase alkaline phosphatase activity and mineralization in human mesenchymal stem cells. Life Sci  2009;84:499–504. [DOI] [PubMed] [Google Scholar]
  • 85. Díez-Zaera M, Díaz-Hernández JI, Hernández-Álvarez E, Zimmermann H, Díaz-Hernández M, Miras-Portugal MT.  Tissue-nonspecific alkaline phosphatase promotes axonal growth of hippocampal neurons. Mol Biol Cell  2011;22:1014–1024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Liu W, Zhang L, Xuan K, Hu C, Liu S, Liao L, Li B, Jin F, Shi S, Jin Y.  prevents bone ageing sensitivity by specifically regulating senescence and differentiation in mesenchymal stem cells. Bone Res  2018;6:27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Schromm AB, Brandenburg K, Loppnow H, Zähringer U, Rietschel ET, Carroll SF, Koch MH, Kusumoto S, Seydel U.  The charge of endotoxin molecules influences their conformation and IL-6-inducing capacity. J Immunol  1998;161:5464–5471. [PubMed] [Google Scholar]
  • 88. Kaliannan K, Hamarneh SR, Economopoulos KP, Nasrin Alam S, Moaven O, Patel P, Malo NS, Ray M, Abtahi SM, Muhammad N, Raychowdhury A, Teshager A, Mohamed MM, Moss AK, Ahmed R, Hakimian S, Narisawa S, Millán JL, Hohmann E, Warren HS, Bhan AK, Malo MS, Hodin RA.  Intestinal alkaline phosphatase prevents metabolic syndrome in mice. Proc Natl Acad Sci USA  2013;110:7003–7008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Herieka M, Erridge C.  High-fat meal induced postprandial inflammation. Mol Nutr Food Res  2014;58:136–146. [DOI] [PubMed] [Google Scholar]
  • 90. Dror E, Dalmas E, Meier DT, Wueest S, Thévenet J, Thienel C, Timper K, Nordmann TM, Traub S, Schulze F, Item F, Vallois D, Pattou F, Kerr-Conte J, Lavallard V, Berney T, Thorens B, Konrad D, Böni-Schnetzler M, Donath MY.  Postprandial macrophage-derived IL-1β stimulates insulin, and both synergistically promote glucose disposal and inflammation. Nat Immunol  2017;18:283–292. [DOI] [PubMed] [Google Scholar]
  • 91. Komazin G, Maybin M, Woodard RW, Scior T, Schwudke D, Schombel U, Gisch N, Mamat U, Meredith TC.  Substrate structure-activity relationship reveals a limited lipopolysaccharide chemotype range for intestinal alkaline phosphatase. J Biol Chem  2019;294:19405–19423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Pettengill M, Matute JD, Tresenriter M, Hibbert J, Burgner D, Richmond P, Millán JL, Ozonoff A, Strunk T, Currie A, Levy O.  Human alkaline phosphatase dephosphorylates microbial products and is elevated in preterm neonates with a history of late-onset sepsis. PLoS One  2017;12:e0175936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Lei W, Ni H, Herington J, Reese J, Paria BC.  Alkaline phosphatase protects lipopolysaccharide-induced early pregnancy defects in mice. PLoS One  2015;10:e0123243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Bender B, Baranyi M, Kerekes A, Bodrogi L, Brands R, Uhrin P, Bösze Z.  Recombinant human tissue non-specific alkaline phosphatase successfully counteracts lipopolysaccharide induced sepsis in mice. Physiol Res  2015;64:731–738. [DOI] [PubMed] [Google Scholar]
  • 95. Poelstra K, Bakker WW, Klok PA, Kamps JA, Hardonk MJ, Meijer DK.  Dephosphorylation of endotoxin by alkaline phosphatase in vivo. Am J Pathol  1997;151:1163–1169. [PMC free article] [PubMed] [Google Scholar]
  • 96. Yang WH, Heithoff DM, Aziz PV, Haslund-Gourley B, Westman JS, Narisawa S, Pinkerton AB, Millán JL, Nizet V, Mahan MJ, Marth JD.  Accelerated aging and clearance of host anti-inflammatory enzymes by discrete pathogens fuels sepsis. Cell Host Microbe  2018;24:500–513.e505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Lund SA, Giachelli CM, Scatena M.  The role of osteopontin in inflammatory processes. J Cell Commun Signal  2009;3:311–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Speer MY, McKee MD, Guldberg RE, Liaw L, Yang HY, Tung E, Karsenty G, Giachelli CM.  Inactivation of the osteopontin gene enhances vascular calcification of matrix Gla protein-deficient mice: evidence for osteopontin as an inducible inhibitor of vascular calcification in vivo. J Exp Med  2002;196:1047–1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Steitz SA, Speer MY, McKee MD, Liaw L, Almeida M, Yang H, Giachelli CM.  Osteopontin inhibits mineral deposition and promotes regression of ectopic calcification. Am J Pathol  2002;161:2035–2046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Yoshitake H, Rittling SR, Denhardt DT, Noda M.  Osteopontin-deficient mice are resistant to ovariectomy-induced bone resorption. Proc Natl Acad Sci USA  1999;96:8156–8160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Jono S, Peinado C, Giachelli CM.  Phosphorylation of osteopontin is required for inhibition of vascular smooth muscle cell calcification. J Biol Chem  2000;275:20197–20203. [DOI] [PubMed] [Google Scholar]
  • 102. Harmey D, Johnson KA, Zelken J, Camacho NP, Hoylaerts MF, Noda M, Terkeltaub R, Millán JL.  Elevated skeletal osteopontin levels contribute to the hypophosphatasia phenotype in Akp2(-/-) mice. J Bone Miner Res  2006;21:1377–1386. [DOI] [PubMed] [Google Scholar]
  • 103. Narisawa S, Yadav MC, Millán JL.  In vivo overexpression of tissue-nonspecific alkaline phosphatase increases skeletal mineralization and affects the phosphorylation status of osteopontin. J Bone Miner Res  2013;28:1587–1598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Nowak LG, Rosay B, Czégé D, Fonta C.  Tetramisole and levamisole suppress neuronal activity independently from their inhibitory action on tissue non-specific alkaline phosphatase in mouse cortex. Subcell Biochem  2015;76:239–281. [DOI] [PubMed] [Google Scholar]
  • 105. Dahl R, Sergienko EA, Su Y, Mostofi YS, Yang L, Simao AM, Narisawa S, Brown B, Mangravita-Novo A, Vicchiarelli M, Smith LH, O’Neill WC, Millán JL, Cosford NDP.  Discovery and validation of a series of aryl sulfonamides as selective inhibitors of tissue-nonspecific alkaline phosphatase (TNAP). J Med Chem  2009;52:6919–6925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Hutcheson JD, Goettsch C, Pham T, Iwashita M, Aikawa M, Singh SA, Aikawa E.  Enrichment of calcifying extracellular vesicles using density-based ultracentrifugation protocol. J Extracell Vesicles  2014;3:25129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Kiffer-Moreira T, Yadav MC, Zhu D, Narisawa S, Sheen C, Stec B, Cosford ND, Dahl R, Farquharson C, Hoylaerts MF, Macrae VE, Millán JL.  Pharmacological inhibition of PHOSPHO1 suppresses vascular smooth muscle cell calcification. J Bone Miner Res  2013;28:81–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Li Q, Huang J, Pinkerton AB, Millan JL, van Zelst BD, Levine MA, Sundberg JP, Uitto J.  Inhibition of tissue-nonspecific alkaline phosphatase attenuates ectopic mineralization in the Abcc6. J Invest Dermatol  2019;139:360–368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Tani T, Fujiwara M, Orimo H, Shimizu A, Narisawa S, Pinkerton AB, Millán JL, Tsuruoka S.  Inhibition of tissue-nonspecific alkaline phosphatase protects against medial arterial calcification and improves survival probability in the CKD-MBD mouse model. J Pathol  2020;250:30–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Pinkerton AB, Sergienko E, Bravo Y, Dahl R, Ma CT, Sun Q, Jackson MR, Cosford NDP, Millán JL.  Discovery of 5-((5-chloro-2-methoxyphenyl)sulfonamido)nicotinamide (SBI-425), a potent and orally bioavailable tissue-nonspecific alkaline phosphatase (TNAP) inhibitor. Bioorg Med Chem Lett  2018;28:31–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Channar PA, Shah SJ, Hassan S, Nisa ZU, Lecka J, Sévigny J, Bajorath J, Saeed A, Iqbal J.  Isonicotinohydrazones as inhibitors of alkaline phosphatase and ecto-5'-nucleotidase. Chem Biol Drug Des  2017;89:365–370. [DOI] [PubMed] [Google Scholar]
  • 112. Miliutina M, Ejaz SA, Khan SU, Iaroshenko VO, Villinger A, Iqbal J, Langer P.  Synthesis, alkaline phosphatase inhibition studies and molecular docking of novel derivatives of 4-quinolones. Eur J Med Chem  2017;126:408–420. [DOI] [PubMed] [Google Scholar]
  • 113. Ashraf A, Ejaz SA, Rahman SU, Siddiqui WA, Arshad MN, Lecka J, Sévigny J, Zayed MEM, Asiri AM, Iqbal J, Hartinger CG, Hanif M.  Hybrid compounds from chalcone and 1,2-benzothiazine pharmacophores as selective inhibitors of alkaline phosphatase isozymes. Eur J Med Chem  2018;159:282–291. [DOI] [PubMed] [Google Scholar]
  • 114. Channar PA, Irum H, Mahmood A, Shabir G, Zaib S, Saeed A, Ashraf Z, Larik FA, Lecka J, Sévigny J, Iqbal J.  Design, synthesis and biological evaluation of trinary benzocoumarin-thiazoles-azomethines derivatives as effective and selective inhibitors of alkaline phosphatase. Bioorg Chem  2019;91:103137. [DOI] [PubMed] [Google Scholar]
  • 115. Gilham D, Tsujikawa LM, Sarsons CD, Halliday C, Wasiak S, Stotz SC, Jahagirdar R, Sweeney M, Johansson JO, Wong NCW, Kalantar-Zadeh K, Kulikowski E.  Apabetalone downregulates factors and pathways associated with vascular calcification. Atherosclerosis  2019;280:75–84. [DOI] [PubMed] [Google Scholar]
  • 116. Tsujikawa LM, Fu L, Das S, Halliday C, Rakai BD, Stotz SC, Sarsons CD, Gilham D, Daze E, Wasiak S, Studer D, Rinker KD, Sweeney M, Johansson JO, Wong NCW, Kulikowski E.  Apabetalone (RVX-208) reduces vascular inflammation in vitro and in CVD patients by a BET-dependent epigenetic mechanism. Clin Epigenet  2019;11:102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Nicholls SJ, Ray KK, Johansson JO, Gordon A, Sweeney M, Halliday C, Kulikowski E, Wong N, Kim SW, Schwartz GG.  Selective BET protein inhibition with apabetalone and cardiovascular events: a pooled analysis of trials in patients with coronary artery disease. Am J Cardiovasc Drugs  2018;18:109–115. [DOI] [PubMed] [Google Scholar]
  • 118. Haarhaus M, Ray KK, Nicholls SJ, Schwartz GG, Kulikowski E, Johansson JO, Sweeney M, Halliday C, Lebioda K, Wong N, Brandenburg V, Beddhu S, Tonelli M, Zoccali C, Kalantar-Zadeh K.  Apabetalone lowers serum alkaline phosphatase and improves cardiovascular risk in patients with cardiovascular disease. Atherosclerosis  2019;290:59–65. [DOI] [PubMed] [Google Scholar]
  • 119. Kulikowski E, Halliday C, Johansson J, Sweeney M, Lebioda K, Wong N, Haarhaus M, Brandenburg V, Beddhu S, Tonelli M, Zoccali C, Kalantar-Zadeh K.  Apabetalone mediated epigenetic modulation is associated with favorable kidney function and alkaline phosphatase profile in patients with chronic kidney disease. Kidney Blood Press Res  2018;43:449–457. [DOI] [PubMed] [Google Scholar]
  • 120. Brun-Heath I, Ermonval M, Chabrol E, Xiao J, Palkovits M, Lyck R, Miller F, Couraud PO, Mornet E, Fonta C.  Differential expression of the bone and the liver tissue non-specific alkaline phosphatase isoforms in brain tissues. Cell Tissue Res  2011;343:521–536. [DOI] [PubMed] [Google Scholar]
  • 121. Whyte MP, Walkenhorst DA, Fedde KN, Henthorn PS, Hill CS.  Hypophosphatasia: levels of bone alkaline phosphatase immunoreactivity in serum reflect disease severity. J Clin Endocrinol Metab  1996;81:2142–2148. [DOI] [PubMed] [Google Scholar]
  • 122. Nizet A, Cavalier E, Stenvinkel P, Haarhaus M, Magnusson P.  Bone alkaline phosphatase: an important biomarker in chronic kidney disease—mineral and bone disorder. Clin Chim Acta  2020;501:198–206. [DOI] [PubMed] [Google Scholar]
  • 123. Iluz-Freundlich D, Zhang M, Uhanova J, Minuk GY.  The relative expression of hepatocellular and cholestatic liver enzymes in adult patients with liver disease. Ann Hepatol  2020;19:204–208. [DOI] [PubMed] [Google Scholar]
  • 124. Rambaldi A, Terao M, Bettoni S, Tini ML, Bassan R, Barbui T, Garattini E.  Expression of leukocyte alkaline phosphatase gene in normal and leukemic cells: regulation of the transcript by granulocyte colony-stimulating factor. Blood  1990;76:2565–2571. [PubMed] [Google Scholar]
  • 125. Gámez-Belmonte R, Hernández-Chirlaque C, Sánchez de Medina F, Martínez-Augustin O.  Experimental acute pancreatitis is enhanced in mice with tissue nonspecific alkaline phoshatase haplodeficiency due to modulation of neutrophils and acinar cells. Biochim Biophys Acta Mol Basis Dis  2018;1864:3769–3779. [DOI] [PubMed] [Google Scholar]
  • 126. Vaughan KR, Stokes L, Prince LR, Marriott HM, Meis S, Kassack MU, Bingle CD, Sabroe I, Surprenant A, Whyte MK.  Inhibition of neutrophil apoptosis by ATP is mediated by the P2Y11 receptor. J Immunol  2007;179:8544–8553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Chen Y, Corriden R, Inoue Y, Yip L, Hashiguchi N, Zinkernagel A, Nizet V, Insel PA, Junger WG.  ATP release guides neutrophil chemotaxis via P2Y2 and A3 receptors. Science  2006;314:1792–1795. [DOI] [PubMed] [Google Scholar]
  • 128. Romanul FC, Bannister RG.  Localized areas of high alkaline phosphatase activity in endothelium of arteries. Nature  1962;195:611–612. [DOI] [PubMed] [Google Scholar]
  • 129. Romanul FC, Bannister RG.  Localized areas of high alkaline phosphatase activity in the terminal arterial tree. J Cell Biol  1962;15:73–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Ballou SP, Lozanski FB, Hodder S, Rzewnicki DL, Mion LC, Sipe JD, Ford AB, Kushner I.  Quantitative and qualitative alterations of acute-phase proteins in healthy elderly persons. Age Ageing  1996;25:224–230. [DOI] [PubMed] [Google Scholar]
  • 131. Krabbe KS, Pedersen M, Bruunsgaard H.  Inflammatory mediators in the elderly. Exp Gerontol  2004;39:687–699. [DOI] [PubMed] [Google Scholar]
  • 132. Brüünsgaard H, Pedersen BK.  Age-related inflammatory cytokines and disease. Immunol Allergy Clin North Am  2003;23:15–39. [DOI] [PubMed] [Google Scholar]
  • 133. Chèvre R, González-Granado JM, Megens RT, Sreeramkumar V, Silvestre-Roig C, Molina-Sánchez P, Weber C, Soehnlein O, Hidalgo A, Andrés V.  High-resolution imaging of intravascular atherogenic inflammation in live mice. Circ Res  2014;114:770–779. [DOI] [PubMed] [Google Scholar]
  • 134. Drechsler M, Megens RT, van Zandvoort M, Weber C, Soehnlein O.  Hyperlipidemia-triggered neutrophilia promotes early atherosclerosis. Circulation  2010;122:1837–1845. [DOI] [PubMed] [Google Scholar]
  • 135. Whyte MP.  Physiological role of alkaline phosphatase explored in hypophosphatasia. Ann N Y Acad Sci  2010;1192:190–200. [DOI] [PubMed] [Google Scholar]
  • 136. Magne D, Julien M, Vinatier C, Merhi-Soussi F, Weiss P, Guicheux J.  Cartilage formation in growth plate and arteries: from physiology to pathology. Bioessays  2005;27:708–716. [DOI] [PubMed] [Google Scholar]
  • 137. Fakhry M, Roszkowska M, Briolay A, Bougault C, Guignandon A, Diaz-Hernandez JI, Diaz-Hernandez M, Pikula S, Buchet R, Hamade E, Badran B, Bessueille L, Magne D.  TNAP stimulates vascular smooth muscle cell trans-differentiation into chondrocytes through calcium deposition and BMP-2 activation: possible implication in atherosclerotic plaque stability. Biochim Biophys Acta  2017;1863:643–653. [DOI] [PubMed] [Google Scholar]
  • 138. Liu J, Nam HK, Campbell C, Gasque KC, Millán JL, Hatch NE.  Tissue-nonspecific alkaline phosphatase deficiency causes abnormal craniofacial bone development in the Alpl(-/-) mouse model of infantile hypophosphatasia. Bone  2014;67:81–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Narisawa S, Fröhlander N, Millán JL.  Inactivation of two mouse alkaline phosphatase genes and establishment of a model of infantile hypophosphatasia. Dev Dyn  1997;208:432–446. [DOI] [PubMed] [Google Scholar]
  • 140. Hoshi K, Amizuka N, Oda K, Ikehara Y, Ozawa H.  Immunolocalization of tissue non-specific alkaline phosphatase in mice. Histochem Cell Biol  1997;107:183–191. [DOI] [PubMed] [Google Scholar]
  • 141. Yadav MC, de Oliveira RC, Foster BL, Fong H, Cory E, Narisawa S, Sah RL, Somerman M, Whyte MP, Millán JL.  Enzyme replacement prevents enamel defects in hypophosphatasia mice. J Bone Miner Res  2012;27:1722–1734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Foster BL, Nagatomo KJ, Nociti FH, Fong H, Dunn D, Tran AB, Wang W, Narisawa S, Millán JL, Somerman MJ.  Central role of pyrophosphate in acellular cementum formation. PLoS One  2012;7:e38393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Zweifler LE, Patel MK, Nociti FH, Wimer HF, Millán JL, Somerman MJ, Foster BL.  Counter-regulatory phosphatases TNAP and NPP1 temporally regulate tooth root cementogenesis. Int J Oral Sci  2015;7:27–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Estève D, Galitzky J, Bouloumié A, Fonta C, Buchet R, Magne D.  Multiple Functions of MSCA-1/TNAP in adult mesenchymal progenitor/stromal cells. Stem Cells Int  2016;2016:1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Rubin MR, Silverberg SJ.  Vascular calcification and osteoporosis–the nature of the nexus. J Clin Endocrinol Metab  2004;89:4243–4245. [DOI] [PubMed] [Google Scholar]
  • 146. van der Schouw YT, van der Graaf Y, Steyerberg EW, Eijkemans JC, Banga JD.  Age at menopause as a risk factor for cardiovascular mortality. Lancet  1996;347:714–718. [DOI] [PubMed] [Google Scholar]
  • 147. Opdebeeck B, Neven E, Millán JL, Pinkerton AB, D'Haese PC, Verhulst A.  Pharmacological TNAP inhibition efficiently inhibits arterial media calcification in a warfarin rat model but deserves careful consideration of potential physiological bone formation/mineralization impairment. Bone  2020;137:115392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Lencel P, Magne D.  Inflammaging: the driving force in osteoporosis?  Med Hypotheses  2010;75:517–521. [DOI] [PubMed] [Google Scholar]
  • 149. Gilbert L, He X, Farmer P, Rubin J, Drissi H, van Wijnen AJ, Lian JB, Stein GS, Nanes MS.  Expression of the osteoblast differentiation factor RUNX2 (Cbfa1/AML3/Pebp2alpha A) is inhibited by tumor necrosis factor-alpha. J Biol Chem  2002;277:2695–2701. [DOI] [PubMed] [Google Scholar]
  • 150. Kaneki H, Guo R, Chen D, Yao Z, Schwarz EM, Zhang YE, Boyce BF, Xing L.  Tumor necrosis factor promotes Runx2 degradation through up-regulation of Smurf1 and Smurf2 in osteoblasts. J Biol Chem  2006;281:4326–4333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Vorbrodt AW, Lossinsky AS, Wisniewski HM.  Localization of alkaline phosphatase activity in endothelia of developing and mature mouse blood-brain barrier. Dev Neurosci  1986;8:1–13. [DOI] [PubMed] [Google Scholar]
  • 152. Bell MA, Scarrow WG.  Staining for microvascular alkaline phosphatase in thick celloidin sections of nervous tissue: morphometric and pathological applications. Microvasc Res  1984;27:189–203. [DOI] [PubMed] [Google Scholar]
  • 153. Norman MG, O'Kusky JR.  The growth and development of microvasculature in human cerebral cortex. J Neuropathol Exp Neurol  1986;45:222–232. [PubMed] [Google Scholar]
  • 154. Anstrom JA, Brown WR, Moody DM, Thore CR, Challa VR, Block SM.  Temporal expression pattern of cerebrovascular endothelial cell alkaline phosphatase during human gestation. J Neuropathol Exp Neurol  2002;61:76–84. [DOI] [PubMed] [Google Scholar]
  • 155. Langer D, Ikehara Y, Takebayashi H, Hawkes R, Zimmermann H.  The ectonucleotidases alkaline phosphatase and nucleoside triphosphate diphosphohydrolase 2 are associated with subsets of progenitor cell populations in the mouse embryonic, postnatal and adult neurogenic zones. Neuroscience  2007;150:863–879. [DOI] [PubMed] [Google Scholar]
  • 156. Deracinois B, Lenfant AM, Dehouck MP, Flahaut C.  Tissue Non-specific Alkaline Phosphatase (TNAP) in Vessels of the Brain. Subcell Biochem  2015;76:125–151. [DOI] [PubMed] [Google Scholar]
  • 157. Anstrom JA, Thore CR, Moody DM, Brown WR.  Immunolocalization of tight junction proteins in blood vessels in human germinal matrix and cortex. Histochem Cell Biol  2007;127:205–213. [DOI] [PubMed] [Google Scholar]
  • 158. Fonta C, Barone P, Rodriguez Martinez L, Négyessy L.  Rediscovering TNAP in the brain: a major role in regulating the function and development of the cerebral cortex. Subcell Biochem  2015;76:85–106. [DOI] [PubMed] [Google Scholar]
  • 159. Zimmermann H, Langer D.  Tissue-nonspecific alkaline phosphatase in the developing brain and in adult neurogenesis. Subcell Biochem  2015;76:61–84. [DOI] [PubMed] [Google Scholar]
  • 160. Demirbilek H, Alanay Y, Alikaşifoğlu A, Topçu M, Mornet E, Gönç N, Özön A, Kandemir N.  Hypophosphatasia presenting with pyridoxine-responsive seizures, hypercalcemia, and pseudotumor cerebri: case report. J Clin Res Pediatr Endocrinol  2012;4:34–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Hofmann C, Liese J, Schwarz T, Kunzmann S, Wirbelauer J, Nowak J, Hamann J, Girschick H, Graser S, Dietz K, Zeck S, Jakob F, Mentrup B.  Compound heterozygosity of two functional null mutations in the ALPL gene associated with deleterious neurological outcome in an infant with hypophosphatasia. Bone  2013;55:150–157. [DOI] [PubMed] [Google Scholar]
  • 162. de Roo MGA, Abeling NGGM, Majoie CB, Bosch AM, Koelman JHTM, Cobben JM, Duran M, Poll-The BT.  Poll-The BT. Infantile hypophosphatasia without bone deformities presenting with severe pyridoxine-resistant seizures. Mol Genet Metab  2014;111:404–407. [DOI] [PubMed] [Google Scholar]
  • 163. Fukazawa M, Tezuka J, Sasazuki M, Masumoto N, Baba H, Doi T, Tsutsumi Y, Mizuno Y, Mihara F, Nakayama H.  Infantile hypophosphatasia combined with vitamin B6-responsive seizures and reticular formation lesions on magnetic resonance imaging: a case report. Brain Dev  2018;40:140–144. [DOI] [PubMed] [Google Scholar]
  • 164. Nunes ML, Mugnol F, Bica I, Fiori RM.  Pyridoxine-dependent seizures associated with hypophosphatasia in a newborn. J Child Neurol  2002;17:222–224. [DOI] [PubMed] [Google Scholar]
  • 165. Hanics J, Barna J, Xiao J, Millán JL, Fonta C, Négyessy L.  Ablation of TNAP function compromises myelination and synaptogenesis in the mouse brain. Cell Tissue Res  2012;349:459–471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Ermonval M, Baudry A, Baychelier F, Pradines E, Pietri M, Oda K, Schneider B, Mouillet-Richard S, Launay JM, Kellermann O.  The cellular prion protein interacts with the tissue non-specific alkaline phosphatase in membrane microdomains of bioaminergic neuronal cells. PLoS One  2009;4:e6497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Le Du MH, Stigbrand T, Taussig MJ, Menez A, Stura EA.  Crystal structure of alkaline phosphatase from human placenta at 1.8 A resolution. Implication for a substrate specificity. J Biol Chem  2001;276:9158–9165. [DOI] [PubMed] [Google Scholar]
  • 168. Bossi M, Hoylaerts MF, Millán JL.  Modifications in a flexible surface loop modulate the isozyme-specific properties of mammalian alkaline phosphatases. J Biol Chem  1993;268:25409–25416. [PubMed] [Google Scholar]
  • 169. Waymire KG, Mahuren JD, Jaje JM, Guilarte TR, Coburn SP, MacGregor GR.  Mice lacking tissue non-specific alkaline phosphatase die from seizures due to defective metabolism of vitamin B-6. Nat Genet  1995;11:45–51. [DOI] [PubMed] [Google Scholar]
  • 170. Balasubramaniam S, Bowling F, Carpenter K, Earl J, Chaitow J, Pitt J, Mornet E, Sillence D, Ellaway C.  Perinatal hypophosphatasia presenting as neonatal epileptic encephalopathy with abnormal neurotransmitter metabolism secondary to reduced co-factor pyridoxal-5'-phosphate availability. J Inherit Metab Dis  2010;33:25– 33. [DOI] [PubMed] [Google Scholar]
  • 171. Street SE, Kramer NJ, Walsh PL, Taylor-Blake B, Yadav MC, King IF, Vihko P, Wightman RM, Millan JL, Zylka MJ.  Tissue-nonspecific alkaline phosphatase acts redundantly with PAP and NT5E to generate adenosine in the dorsal spinal cord. J Neurosci  2013;33:11314–11322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Zhang D, Xiong W, Chu S, Sun C, Albensi BC, Parkinson FE.  Inhibition of hippocampal synaptic activity by ATP, hypoxia or oxygen-glucose deprivation does not require CD73. PLoS One  2012;7:e39772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Sebastián-Serrano Á, Engel T, de Diego-García L, Olivos-Oré LA, Arribas-Blázquez M, Martínez-Frailes C, Pérez-Díaz C, Millán JL, Artalejo AR, Miras-Portugal MT, Henshall DC, Díaz-Hernández M.  Neurodevelopmental alterations and seizures developed by mouse model of infantile hypophosphatasia are associated with purinergic signalling deregulation. Hum Mol Genet  2016;25:4143–4156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. Whyte MP.  Hypophosphatasia—aetiology, nosology, pathogenesis, diagnosis and treatment. Nat Rev Endocrinol  2016;12:233–246. [DOI] [PubMed] [Google Scholar]
  • 175. Güzel Nur B, Çelmeli G, Manguoğlu E, Soyucen E, Bircan İ, Mıhçı E.  Pyridoxine-responsive seizures in infantile hypophosphatasia and a novel homozygous mutation in ALPL gene. J Clin Res Pediatr Endocrinol  2016;8:360–364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176. Brichacek AL, Benkovic SA, Chakraborty S, Nwafor DC, Wang W, Jun S, Dakhlallah D, Geldenhuys WJ, Pinkerton AB, Millán JL, Brown CM.  Systemic inhibition of tissue-nonspecific alkaline phosphatase alters the brain-immune axis in experimental sepsis. Sci Rep  2019;9:18788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Vardy ER, Kellett KA, Cocklin SL, Hooper NM.  Alkaline phosphatase is increased in both brain and plasma in Alzheimer's disease. Neurodegenerative Dis  2012;9:31–37. [DOI] [PubMed] [Google Scholar]
  • 178. Díaz-Hernández M, Gómez-Ramos A, Rubio A, Gómez-Villafuertes R, Naranjo JR, Miras-Portugal MT, Avila J.  Tissue-nonspecific alkaline phosphatase promotes the neurotoxicity effect of extracellular tau. J Biol Chem  2010;285:32539–32548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Nho K, Kueider-Paisley A, Ahmad S, MahmoudianDehkordi S, Arnold M, Risacher SL, Louie G, Blach C, Baillie R, Han X, Kastenmüller G, Trojanowski JQ, Shaw LM, Weiner MW, Doraiswamy PM, van Duijn C, Saykin AJ, Kaddurah-Daouk R; Consortium AsDNIatADM. Association of altered liver enzymes with Alzheimer disease diagnosis, cognition, neuroimaging measures, and cerebrospinal fluid biomarkers. JAMA Netw Open  2019;2:e197978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Kellett KA, Williams J, Vardy ER, Smith AD, Hooper NM.  Plasma alkaline phosphatase is elevated in Alzheimer's disease and inversely correlates with cognitive function. Int J Mol Epidemiol Genet  2011;2:114–121. [PMC free article] [PubMed] [Google Scholar]
  • 181. Vasantharekha R, Priyanka HP, Swarnalingam T, Srinivasan AV, ThyagaRajan S.  Interrelationship between Mini-Mental State Examination scores and biochemical parameters in patients with mild cognitive impairment and Alzheimer's disease. Geriatr Gerontol Int  2017;17:1737–1745. [DOI] [PubMed] [Google Scholar]
  • 182. Ohkubo S, Kimura J, Matsuoka I.  Ecto-alkaline phosphatase in NG108-15 cells: a key enzyme mediating P1 antagonist-sensitive ATP response. Br J Pharmacol  2000;131:1667–1672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Zimmermann H, Zebisch M, Sträter N.  Cellular function and molecular structure of ecto-nucleotidases. Purinergic Signal  2012;8:437–502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Gómez-Ramos A, Díaz-Hernández M, Cuadros R, Hernández F, Avila J.  Extracellular tau is toxic to neuronal cells. FEBS Lett  2006;580:4842–4850. [DOI] [PubMed] [Google Scholar]
  • 185. Sebastián-Serrano Á, de Diego-García L, Díaz-Hernández M.  The neurotoxic role of extracellular tau protein. Int J Mol Sci  2018;19: 998–1010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Petty GW, Brown RD, Whisnant JP, Sicks JD, O’Fallon WM, Wiebers DO.  Ischemic stroke subtypes: a population-based study of incidence and risk factors. Stroke  1999;30:2513–2516. [DOI] [PubMed] [Google Scholar]
  • 187. Ryu WS, Lee SH, Kim CK, Kim BJ, Yoon BW.  Increased serum alkaline phosphatase as a predictor of long-term mortality after stroke. Neurology  2010;75:1995–2002. [DOI] [PubMed] [Google Scholar]
  • 188. Kim J, Song TJ, Song D, Lee HS, Nam CM, Nam HS, Kim YD, Heo JH.  Serum alkaline phosphatase and phosphate in cerebral atherosclerosis and functional outcomes after cerebral infarction. Stroke  2013;44:3547–3549. [DOI] [PubMed] [Google Scholar]
  • 189. Tan G, Hao Z, Lei C, Chen Y, Yuan R, Xu M, Liu M.  Subclinical change of liver function could also provide a clue on prognosis for patients with spontaneous intracerebral hemorrhage. Neurol Sci  2016;37:1693–1700. [DOI] [PubMed] [Google Scholar]
  • 190. Uehara T, Ohara T, Minematsu K, Nagatsuka K, Toyoda K.  Predictors of stroke events in patients with transient ischemic attack attributable to intracranial stenotic lesions. Intern Med  2018;57:295–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191. Zong L, Wang X, Li Z, Zhao X, Liu L, Li H, Meng X, Wang Y, Wang Y.  Alkaline phosphatase and outcomes in patients with preserved renal function: results from China National Stroke Registry. Stroke  2018;49:1176–1182. [DOI] [PubMed] [Google Scholar]
  • 192. Zhong C, You S, Chen J, Zhai G, Du H, Luo Y, Dong X, Cao Y, Liu CF, Zhang Y.  Serum alkaline phosphatase, phosphate, and in-hospital mortality in acute ischemic stroke patients. J Stroke Cerebrovasc Dis  2018;27:257–266. [DOI] [PubMed] [Google Scholar]
  • 193. Liu Y, Liang X, Xu X, Dong M, Jia S, Lu C, Wei Y.  Increased serum alkaline phosphatase in patients with acute ischemic stroke. J Stroke Cerebrovasc Dis  2019;28:21–25. [DOI] [PubMed] [Google Scholar]
  • 194. Brichacek AL, Brown CM.  Alkaline phosphatase: a potential biomarker for stroke and implications for treatment. Metab Brain Dis  2019;34:3–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195. Idzko M, Ferrari D, Eltzschig HK.  Nucleotide signalling during inflammation. Nature  2014;509:310–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. McDonald B, Pittman K, Menezes GB, Hirota SA, Slaba I, Waterhouse CC, Beck PL, Muruve DA, Kubes P.  Intravascular danger signals guide neutrophils to sites of sterile inflammation. Science  2010;330:362–366. [DOI] [PubMed] [Google Scholar]
  • 197. Jickling GC, Liu D, Ander BP, Stamova B, Zhan X, Sharp FR.  Targeting neutrophils in ischemic stroke: translational insights from experimental studies. J Cereb Blood Flow Metab  2015;35:888–901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Maestrini I, Tagzirt M, Gautier S, Dupont A, Mendyk AM, Susen S, Tailleux A, Vallez E, Staels B, Cordonnier C, Leys D, Bordet R.  MPO is partially associated with neutrophil deleterious effect in acute cerebral ischemia. Neurology  2020;95:e97–e108. [DOI] [PubMed] [Google Scholar]
  • 199. Roy-O’Reilly MA, Ahnstedt H, Spychala MS, Munshi Y, Aronowski J, Sansing LH, McCullough LD.  Aging exacerbates neutrophil pathogenicity in ischemic stroke. Aging (Albany NY)  2020;12:436–461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200. Suzuki N, Irie M, Iwata K, Nakane H, Yoshikane M, Koyama Y, Uehara Y, Takeyama Y, Kitamura Y, Sohda T, Watanabe H, Ikehara Y, Sakisaka S.  Altered expression of alkaline phosphatase (ALP) in the liver of primary biliary cirrhosis (PBC) patients. Hepatol Res  2006;35:37–44. [DOI] [PubMed] [Google Scholar]
  • 201. Halling Linder C, Englund UH, Narisawa S, Millán JL, Magnusson P.  Isozyme profile and tissue-origin of alkaline phosphatases in mouse serum. Bone  2013;53:399–408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202. Alvaro D, Benedetti A, Marucci L, Delle Monache M, Monterubbianesi R, Di Cosimo E, Perego L, Macarri G, Glaser S, Le Sage G, Alpini G.  The function of alkaline phosphatase in the liver: regulation of intrahepatic biliary epithelium secretory activities in the rat. Hepatology  2000;32:174–184. [DOI] [PubMed] [Google Scholar]
  • 203. Mimura Y, Sakisaka S, Harada M, Sata M, Tanikawa K.  Role of hepatocytes in direct clearance of lipopolysaccharide in rats. Gastroenterology  1995;109:1969–1976. [DOI] [PubMed] [Google Scholar]
  • 204. Nouwen EJ, De Broe ME.  Human intestinal versus tissue-nonspecific alkaline phosphatase as complementary urinary markers for the proximal tubule. Kidney Int Suppl  1994;47:S43–S51. [PubMed] [Google Scholar]
  • 205. Moochhala SH, Sayer JA, Carr G, Simmons NL.  Renal calcium stones: insights from the control of bone mineralization. Exp Physiol  2008;93:43–49. [DOI] [PubMed] [Google Scholar]
  • 206. Bentala H, Verweij WR, Huizinga-Van der Vlag A, van Loenen-Weemaes AM, Meijer DK, Poelstra K.  Removal of phosphate from lipid A as a strategy to detoxify lipopolysaccharide. Shock  2002;18:561–566. [DOI] [PubMed] [Google Scholar]
  • 207. Kapojos JJ, Poelstra K, Borghuis T, Van Den Berg A, Baelde HJ, Klok PA, Bakker WW.  Induction of glomerular alkaline phosphatase after challenge with lipopolysaccharide. Int J Exp Pathol  2003;84:135–144. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cardiovascular Research are provided here courtesy of Oxford University Press

RESOURCES