当前位置: 首页 > 期刊 > 《动脉硬化血栓血管生物学》 > 2004年第7期 > 正文
编号:11330580
Vascular Calcification
http://www.100md.com 《动脉硬化血栓血管生物学》
     Moeen Abedin; Yin Tintut; Linda L. Demer

    From the Departments of Medicine (M.A., Y.T., L.L.D.) and Physiology (L.L.D.), The David Geffen School of Medicine at University of California, Los Angeles, Calif.

    ABSTRACT

    Vascular calcification, long thought to result from passive degeneration, involves a complex, regulated process of biomineralization resembling osteogenesis. Evidence indicates that proteins controlling bone mineralization are also involved in the regulation of vascular calcification. Artery wall cells grown in culture are induced to become osteogenic by inflammatory and atherogenic stimuli. Furthermore, osteoclast-like cells are found in calcified atherosclerotic plaques, and active resorption of ectopic vascular calcification has been demonstrated. In general, soft tissue calcification arises in areas of chronic inflammation, possibly functioning as a barrier limiting the spread of the inflammatory stimulus. Atherosclerotic calcification may be one example of this process, in which oxidized lipids are the inflammatory stimulus. Calcification is widely used as a clinical indicator of atherosclerosis. It progresses nonlinearly with time, following a sigmoid-shaped curve. The relationship between calcification and clinical events likely relates to mechanical instability introduced by calcified plaque at its interface with softer, noncalcified plaque. In general, as calcification proceeds, interface surface area increases initially, but eventually decreases as plaques coalesce. This phenomenon may account for reports of less calcification in unstable plaque. Vascular calcification is exacerbated in certain clinical entities, including diabetes, menopause, and osteoporosis. Mechanisms linking them must be considered in clinical decisions. For example, treatments for osteoporosis may have unanticipated effects on vascular calcification; the converse also applies. Further understanding of processes governing vascular calcification may yield new therapeutic options for vascular disease.

    Vascular calcification involves a complex biomineralization process that resembles osteogenesis. It is regulated by bone proteins and osteoblast-like cells found in the artery wall. Understanding its mechanism has implications for management of vascular disease, particularly in the context of bone disease.

    Key Words: calcification ? atherosclerosis ? inflammation ? bone ? vascular

    Introduction

    Vascular calcification is an important manifestation of atherosclerosis. For more than half a century, it has been associated with a poor prognosis attributable to vascular disease.1 Its presence is a strong indicator of chronic inflammatory disease, usually atherosclerosis, and its extent directly relates to the overall burden of atherosclerotic disease.2 Despite its clinical relevance, research on the mechanism of mineral deposition in arteries has been limited and remains at an early scientific stage relative to research on other aspects of atherosclerosis, such as lipoprotein biochemistry and inflammation.

    Mechanisms of Vascular Calcification

    Vascular calcification recapitulates embryonic osteogenesis. Pathologists in the 19th century recognized the presence of bone-like tissue within atherosclerotic arteries, with lamellar structure, osteoblast-like cells, and hematopoietic elements.3 Yet, for most of the 20th century, vascular calcification has been regarded as a passive, unregulated, degenerative process occurring within advanced atherosclerotic plaques. The concept of regulated ossification as the mechanism behind vascular calcification has re-emerged only in the past decade.4,5 Ossification has been identified histologically in 60% of restenotic aortic valves after balloon valvuloplasty.6 Approximately 15% of carotid atherosclerotic plaque specimens7 and calcified cardiac valve tissue8 have ossification. Vascular calcification may include both osteogenic and chondrogenic differentiation. In humans, it is primarily osteogenic with bone tissue formation, whereas in mice, it is primarily chondrogenic with cartilage formation. Although osteoblasts and chondroblasts are distinct cell types, they have substantial overlap in mineralization mechanism and gene expression, including alkaline phosphatase, Cbfa-1, and osteopontin. Many key regulators of bone formation and bone structural proteins are expressed in atherosclerotic plaques, including bone morphogenetic protein-2 (BMP-2),9 osteopontin,10–12 matrix -carboxyglutamic acid protein (MGP),13 and osteoprotegerin (OPG).14 These factors are discussed in this review. Many other bone-related factors are likely to have important roles but are beyond the scope of this review.

    Development of Mineral Formation

    Some of the earliest evidence that vascular calcification is regulated came from Anderson, who found matrix vesicles (structures involved in osteogenesis) in human calcified aortas.15 Further evidence of an ossification process within the vasculature came from in vitro studies of vascular medial cells. Giachelli et al brought vascular calcification research to the molecular level by showing expression of a bone matrix protein in a special population of vascular smooth muscle cells.16 Subsequently, we and others found expression of osteogenic factors, such as bone morphogenetic protein, osteopontin, and matrix GLA protein in cultured vascular cells.9,13 In vitro vascular cell calcification has been demonstrated by several groups. Primary cultures of vascular smooth muscle cells express bone proteins and mineralize when treated with ?-glycerophosphate,17 which serves as an inorganic phosphate donor in the presence of alkaline phosphatase. This enzyme is expressed by these cells and is used as a marker for osteogenic and chondrogenic differentiation. Primary vascular smooth muscle cells (VSMCs) include subpopulations of cells with different phenotypes. As VSMC cultures undergo osteogenic differentiation, they also lose expression of smooth muscle-specific markers,18 indicating de-differentiation. A subpopulation of VSMC, isolated by dilutional cloning, spontaneously expresses bone proteins and produces a mineralized matrix in culture, similar to that of osteoblasts.9,19 This subpopulation, "calcifying vascular cells," retains the ability to differentiate into other mesenchymal lineages besides osteoblasts,20 similar to cells of the clonal line C3H10T1/2. Towler et al have shown that developmental homeobox-related transcription factors regulate lineage determination in mesenchymal progenitor cells in vitro, and vascular calcification in vivo.21,22 In stem cell lines in general, cultures become a mix of differentiated and undifferentiated cells, because the parent cells give rise to daughter cells, some of which are stem cells and some of which are differentiated. Overall, the evidence suggests that mineralized matrix appears to be produced by the osteoblastic daughter cells of a multipotent vascular cell. These early observations establishing in vitro models of vascular calcification have permitted a wide array of investigation into its cellular and molecular mechanisms.

    Mineral Resorption

    In most regulated biological systems, anabolic processes are accompanied by counteracting catabolic processes. In the case of vascular calcification, the catabolic process would be mineral resorption. Histologically, the bone-like structures within atherosclerotic lesions have sculpted features similar to resorptive and remodeling sites in trabecular bone. In addition, recent molecular and cellular evidence point to an active resorption process and regression in ectopic valvular calcification.23 In bone, mineral is resorbed by osteoclasts, which are large, multinucleated cells with a characteristic ruffled border. They form an actin ring, where the cell tightly apposes to mineralized matrix, allowing a protease-rich, acidic microenvironment necessary for controlled resorption.24 Functional osteoclasts express tartrate-resistant acid phosphatase, cathepsin K, calcitonin receptors, H+ATPase, and carbonic anhydrase II; however, none of these proteins is specific to osteoclasts. The definitive identifying characteristic is formation of resorption pits on mineralized surfaces. In atherosclerotic lesions, multinucleated tartrate-resistant acid phosphatase-positive cells have been observed histologically at the edges of bone-like structures.25 Osteoclasts derive from the monocytic line of hematopoietic cells, and macrophages exposed to particulate calcium mineral have been reported to undergo osteoclastic differentiation.26 Thus, atherosclerotic lesions, being rich in monocytes and macrophages,27 have an abundant source of preosteoclasts. Maturation of preosteoclasts (such as peripheral blood mononuclear cells) to osteoclasts requires 2 cytokines: monocyte colony-stimulating factor and the ligand for receptor activator of NF-kappaB (RANKL).28 Both are also present in atherosclerotic lesions: monocyte colony-stimulating factor is a well-established component of atherosclerotic plaque,29,30 and RANKL was recently demonstrated in the vasculature.14,31 At the same time that they participate in resorption, macrophages may contribute to mineral formation indirectly by producing inflammatory cytokines and producing lipid oxidation products that promote vascular cell mineralization. The presence of osteoclasts in the artery wall is controversial. Although cells with osteoclastic features have been demonstrated, functional activity of these cells is difficult to demonstrate or exclude.

    Nonatherosclerotic Vascular Calcification

    Not all vascular calcification occurs in the presence of atherosclerosis. Metabolic disorders, such as uremia, hyperparathyroidism, and diabetes are associated with development of medial calcification that occurs even in areas without adjacent atherosclerosis. The distinction is based on the fact that medial calcification appears to be amorphous rather than organized bone tissue. However, areas of atherosclerosis-associated calcification can appear amorphous, particularly in early stages. The same conditions associated with medial calcification are also associated with accelerated atherosclerosis, so that many patients with medial calcification also have atherosclerosis. Just as they overlap clinically, they may also overlap in some aspects of mechanism.

    Molecular Determinants

    Matrix Gla Protein

    MGP, first identified as a bone matrix protein,32 is unusual because of its small size and the post-translational modification of its glutamic acid residues by -carboxylation. This modification appears to be important in its function.33 The role of MGP in calcified atherosclerotic plaque is unclear. MGP is expressed in bone at relatively constant levels through early development and adulthood.34 It is also found in normal artery wall and at increased levels in atherosclerotic plaques.13 In rat aortic smooth muscle cells, MGP expression increases with increasing extracellular calcium concentration.35 These findings suggest that MGP participates in a homeostatic regulatory mechanism to control and limit mineralization, but it is ultimately overwhelmed.

    Interestingly, the MGP knockout mouse has extensive aortic calcification in which the wall of the aorta is replaced with calcified cartilage.36 A simple conclusion from this model is that MGP directly inhibits calcium mineral formation. However, these mice also have a bone phenotype manifest as osteopenia, fractures, short stature, and inappropriate calcification of the growth plate. The simple model would not explain osteopenia and cartilaginous metaplasia of the aorta in the MGP null mice. A more satisfactory explanation would be that MGP regulates mineralization indirectly through effects on differentiation. Recent evidence indicates that MGP inhibits mesenchymal cell differentiation to the osteogenic lineage by sequestering the potent osteogenic and chondrogenic differentiation factor, BMP-2, thus preventing its interaction with receptors.37–39 Adding further complexity, the effect of MGP on BMP-2 depends on the degree of MGP -carboxylation38 and the ratio of concentrations of the 2 molecules.40 Lack of function (from insufficient -carboxylation), rather than the amount of MGP may be the factor that increases risk of calcification. Indeed, MGP isolated from calcified atherosclerotic plaque from rats has incomplete -carboxylation.41

    MGP -carboxylation is performed by the vitamin K-dependent enzyme, -carboxylase. Conditions causing a relative deficiency of functional vitamin K may increase vascular calcification because of incomplete -carboxylation and reduced function of MGP. In postmenopausal women, the presence of aortic calcification is associated with lower overall vitamin K intake.42 Because the medication warfarin interferes with the availability of bioactive vitamin K, it may also interfere with MGP function. Important questions have been raised about its use in hemodialysis patients who are subject to extensive vascular calcification and cardiovascular risk.43

    Osteopontin

    Osteopontin is a matrix protein containing an RGD motif that binds to cells via the -v, ?-3 integrin, which functions as an important calcification inhibitor. Giachelli et al showed that OPN is associated with clinical atherosclerosis, colocalizing with calcified atherosclerotic plaques.10 This group went on to show that it is expressed by macrophages as well as smooth muscle and endothelial cells within plaque.44 Plasma OPN levels are significantly greater in patients with coronary artery disease (CAD), increasing with disease severity, independent of conventional risk factors.45 OPN directly inhibits calcification of cultured bovine aortic smooth muscle cells46 and inhibits aortic valve calcification in vivo,23 most likely through self-aggregation and adhesion to apatite crystals through specific amino acid moieties.47 This ability of osteopontin to block growth of crystals may have important physiological and pathological significance.

    OPN may also affect calcification by stimulating resorption. OPN binding to -v, ?-3 integrin on osteoclasts leads to a decrease in cytosolic calcium, a change associated with osteoclast activation to a resorptive phenotype.48 Exciting new evidence from Steitz et al suggest that this binding also promotes resorption of ectopic calcification by inducing expression of carbonic anhydrase II, which is key in creating the acidic environment required for resorption. In wild-type mice, there is regression of calcification, concurrent with accumulation of OPN and osteoclast-like cells within transplanted cardiovascular tissue.23 Recombinant OPN induces resorption of ectopic bone implanted in muscle.49

    Mice deficient in both OPN and MGP have accelerated aortic calcification compared with mice deficient only in MGP, consistent with the concept that OPN inhibits mineralization.50 The phenotype of double knockout, OPN–/–, apo E–/– mice varies with gender. The male mice develop increased vascular calcification compared with wild-type and with apoE–/– mice, but no difference in overall aortic atherosclerosis. In contrast, the female double knockout mice have decreased overall aortic atherosclerosis. These findings provide in vivo evidence that atherosclerosis and vascular calcification may have at least some independent regulatory mechanisms.51

    Osteoprotegerin

    The recent discovery of OPG, a member of the tumor necrosis factor- receptor superfamily,52 introduced another link between bone and vascular metabolism. OPG is an indirect inhibitor of osteoclastogenesis. It functions as a soluble "decoy" receptor that binds and inhibits RANKL. RANKL activation of its receptor, RANK, is essential for maturation of osteoclast progenitors.53 OPG is expressed in cultured coronary artery smooth muscle cells,54 and OPG and RANKL are found in normal arteries.14 In the rat model, OPG treatment prevents warfarin-induced vascular calcification.55 OPG transgenic mice lack functional osteoclasts, leading to inadequately trabecularized (osteopetrotic) bone.52 In contrast, OPG–/– mice have profound osteoporosis with multiple fractures. Interestingly, these mice also have vascular calcification in some arteries.56 The physiological role of OPG may be dependent on its levels relative to RANKL. RANKL, a tumor necrosis factor superfamily member, has now been shown to promote calcification of cardiac valve myofibroblasts.57 Unopposed RANKL activity may in part explain the medial calcification found in OPG null mice.

    Recently, Pritzker et al showed a novel role of OPG in microvascular endothelial cell survival by binding TRAIL and preventing its interaction with death-inducing TRAIL receptors.58 Recent studies have shown an association between OPG levels and CAD in humans. In patients with cerebrovascular disease, OPG levels are independently associated with cardiovascular mortality but not with bone mineral density.59 In patients with stable angina, OPG levels are associated with significant coronary artery narrowing.60,61 Interestingly, estrogen therapy is associated with increased OPG levels.62

    Inorganic Phosphate

    Phosphate levels were long thought to influence mineralization only through physico-chemical means. However, new evidence indicates that phosphate regulates and coordinates cell signaling and gene expression by dynamic transport processes. Treatment of VSMCs with supplemental inorganic phosphate (Pi) increases expression of bone regulatory proteins including Cbfa-1 and osteocalcin and, simultaneously, downregulates smooth muscle lineage markers.18 Extracellular phosphate is taken up by VSMC via a sodium-dependent phosphate transporter, Pit-1, and increased intracellular phosphate in VSMC induces mineralization-related genes.63

    Exogenous addition of proteins to cells in culture, in general, may nonspecifically affect mineralization. By virtue of the charge they carry, added protein may change the free concentration of inorganic phosphate in the media. This, in turn, can alter intracellular phosphate transport, resulting in a change in expression of mineralization-related genes.64

    The positive effects of inorganic phosphate on calcification are potently inhibited by inorganic pyrophosphate. Extracellular inorganic pyrophosphate, which inhibits hydroxyapatite crystal deposition, is generated by nucleotide pyrophosphate/phosphodiesterase (NPP). NPP1 knockout mice or mice expressing a truncated, inactive isozyme spontaneously develop calcification.65 Familial idiopathic infantile arterial calcification, a clinical syndrome associated with recessive mutations in the NPP1 gene, is characterized by extensive fibrointimal hyperplasia and premature calcification of the aorta, valvular dysfunction, and cardiomyopathy. This syndrome also includes periarticular calcification similar to that seen in NPP1-deficient mice.66

    Teleology of Vascular Osteogenesis

    The concept that vascular calcification is related to osteogenesis is not unexpected, given the interaction of vascular and bone cells in normal embryonic skeletogenesis. Bone formation relies on an underlying vascular architecture and scaffold. In embryonal development, endochondral ossification follows invasion of neoangiogenic vessels into calcified cartilage matrix. Once the vasculature is established, pre-osteoblasts, originating from the angiogenic pericytes surrounding the vasculature or from blood-borne mesenchymal cells, differentiate and initiate mineralization.67 The requirement for angiogenesis holds true for bone formation in fracture healing in adults as well as in skeletogenesis of the embryo.

    Just as vascular calcification is closely associated with chronic inflammatory atherosclerosis, soft tissue calcification is also associated with sites of chronic inflammation. Examples include tuberculosis, hepatic echinococcal infection, foreign body retention, scleroderma, sarcoidosis, pancreatitis, and cancer. The mechanism underlying this association between ectopic ossification and chronic inflammation is not clear. One possibility is that activated immune cells recruited to sites of chronic inflammation typically release cytokines and free radicals, which oxidize lipids present in these sites.68 These factors may in turn modulate osteogenic regulatory genes, affecting differentiation of putative mesenchymal progenitor cells. For example, inflammatory cytokines69 as well as modified lipids and lipoproteins70 induce osteoblastic differentiation and mineralization of calcifying vascular cells, which were recently shown to be multipotential.20,71 In this way, soft tissue and atherosclerotic calcification may be driven by cytokines and modified lipids resulting from chronic inflammation (Figure 1). Genetic models that exhibit vascular calcification in the absence of atherosclerosis remain relevant because they may be caused by direct changes in osteogenic regulatory gene function downstream of the primary effect of atherosclerosis itself.

    Figure 1. Schematic diagram depicting parallel mechanisms in soft tissue versus atherosclerotic and nonatherosclerotic vascular calcification. Rectangles represent mycobacteria or other chronic inflammatory stimuli such as foreign bodies, abscesses, bacteria, parasites, neoplastic cells, or autoimmune antigens. In soft tissue calcification, these stimuli activate an immune response that results in cytokine and oxidized lipid production (hatch marks), which modify osteogenic regulatory genes to promote osteogenesis. Atherosclerosis activates a similar cascade of events. The stimuli for nonatherosclerotic vascular calcification are less well-established but may include hyperphosphatemia, AGEs, and other metabolic factors. The mechanism for vascular calcification in many animal models may involve primary changes in the osteogenic regulatory genes or their expression.

    Clinical Significance

    Histopathologically, more extensive calcification is associated with more significant coronary stenosis.72 Radiographically detected vascular calcification is generally accepted as a sensitive marker for atherosclerosis. The association with clinical outcome was first made with aortic calcification detected on plain radiographs, a relatively insensitive technique. Coronary artery calcification (CAC) can be detected in a more sensitive manner using electron beam and multidetector computed tomography.73 Current technology allows completion of a high-resolution scan in a few seconds during a single breath-hold. ECG gating can provide similar scans from conventional computed tomography imaging. These commercially available scans provide a total score indicating the extent of calcium detected, measured as "mass" or a calcium "score." The presence of CAC is sensitive and specific for detection of clinically significant CAD74 and for identifying patients at risk for adverse cardiac events.75 The scores correlate with number of artery segments having angiographically significant disease,76 although not with angiographic stenosis severity. In general, they correlate well with overall atherosclerotic plaque burden.2

    The American Heart Association and American College of Cardiology recently published guidelines for the use of coronary artery calcium score as a diagnostic tool for clinical decision-making. The guidelines affirm the sensitivity of coronary calcium for the presence of coronary atherosclerosis. Patients with no detectable coronary calcium have a relatively low risk for subsequent cardiovascular events. Therefore, it has been recommended that CAC be used as a tool to identify asymptomatic patients at low to intermediate risk, particularly the elderly, who may benefit from more aggressive risk factor modification. The guidelines emphasize that CAC should not be used for the diagnosis of obstructive CAD given the poor specificity of CAC for significant flow-limiting coronary plaques.77

    CAC may have a different significance than other types of vascular calcification (eg, valve calcification, medial artery calcification). For example, aortic valve calcification, another type of vascular calcification, is a strong independent determinant of clinical progression of aortic stenosis,78 a relationship that holds even in the setting of end-stage renal disease79 in which vascular calcification is often severe. Although not widely appreciated, CAC does not uniformly correlate with mitral valve and aortic valve calcification.80 Thus, clinical significance of vascular calcification depends in part on location.

    Controversy exists over the prognostic significance of CAC. In patients with multiple cardiovascular risk factors, CAC adds modest predictive value over traditional risk factors for predicting acute coronary events.81 A consensus has developed that coronary calcification is associated with chronic symptomatic CAD rather than with acute coronary events, which are attributed to plaque rupture and coronary thrombosis. This notion is based on intravascular ultrasound studies, which have shown less extensive coronary calcification in patients with acute coronary syndromes compared with patients with chronic stable angina.82,83 In patients presenting with acute coronary syndromes, intravascular ultrasound studies show less calcification in the event-related coronary lesion than in stable plaques in other vessels.84 These findings have been interpreted as evidence that vascular calcification is protective against acute events. However, its effects on plaque stability may not depend exclusively on the amount of calcium within the plaque.

    Plaque Stability

    It is more likely that risk of plaque rupture caused by calcification is biphasic and dose-dependent, based on mechanical stress considerations. In addition to fluid stresses, the artery wall is subject to pulsatile and steady solid mechanical stresses. Stresses generated by blood pressure are pulsatile. Steady solid stresses are generated by intrinsic and extrinsic tension; arteries are usually stretched beyond rest length in their normal anatomic configuration, generating longitudinal tension even in the absence of blood pressure. Both kinds of stress can be transmitted in all 3 axes, longitudinal, circumferential, and radial. In a theoretical analysis based on anatomic configuration, plaques that had ruptured were predicted to have higher circumferential stress than normal artery segments.85 However, rupture is best predicted by failure analysis that incorporates all 3 components, circumferential, longitudinal, and radial stresses. Failure stress, known as von Mises or maximal principal stress, tends to occur at interfaces between materials of different stiffness. Because calcified atherosclerotic plaque is at least 4- to 5-times more stiff than cellular plaque,86 failure stress would be expected to concentrate at interfaces between calcified and noncalcified regions within an artery.87

    As the degree of calcification increases, the number of interfaces between rigid and distensible plaque initially would increase until the point at which the rigid plaques coalesce. Further calcification would result in decreased interface area. Because failure stress concentrates at interface areas, it follows that risk of rupture increases with the degree of calcification to the point when plaques coalesce (Figure 2). Calcification beyond this point may be associated with decreasing risk of plaque rupture. Ultimately, the most valuable prognostic parameter may be total surface area of mineral deposits rather than calcium score or mass. This hypothesis may be difficult to test epidemiologically because interface area is difficult to measure, and, like the calcium score itself, may not be independent of atherosclerosis severity.

    Figure 2. Relationship between plaque interface area versus total calcium mass. Biomechanical principles suggest the risk of plaque rupture should correlate with interface area, which eventually decreases as calcified plaques begin to coalesce. The corresponding figure insets illustrate how the interface (the circumference around the black area) eventually decreases as calcified areas continue to form and grow.

    Assessing CAC Progression as a Clinical Endpoint

    The dynamics of progression of arterial calcification should be considered in clinical trials in which coronary artery calcium score progression is regarded as a surrogate end point for overall atherosclerotic disease progression. The problem lies in annualization of the CAC progression rate from studies with a short or variable time period between scans (eg, 2 months). Such annualization requires the assumption that calcium mass increases linearly with time. Evidence from Yoon et al suggests the process is nonlinear, and probably sigmoidal (Figure 3). In their study of 217 subjects who had at least 2 electron beam computed tomography scans separated by a mean interval of 25 months, the rate of progression of calcium volume score (calcification pixel area x slice thickness) increased with increasing baseline calcium score.88

    Figure 3. Coronary artery calcium progression. CAC progression rate increases as a function of baseline CAC volume score. The curve is extrapolated with permission from data published by Yoon et al.88 The dotted portion of the curve was added to illustrate that the curve is probably sigmoid-shaped, because there is a theoretical upper limit for CAC accumulation.

    Studies using coronary calcium measurements must also address interscan reproducibility. Depending on whether the technique includes interslice gaps, difficulty with reproducibility of positioning can introduce variability as high as 18%, even when scans are repeated immediately.89 Error caused by interscan variability essentially introduces noise into the data, making studies prone to beta error. By using comparable interscan time periods and comparable baseline scores between treated and control groups, trials can prevent bias introduced by nonlinear progression. The recent ACC/American Heart Association guidelines recommend against use of serial coronary calcium measurements to assess progression or regression of coronary atherosclerosis for these reasons.77

    Links With Other Clinical Factors

    Many clinical entities influence biomineralization. Because vascular calcification can include osteogenesis, effects on bone and vessel mineralization must be taken into account when considering treatment that affects mineralization (Table).

    Summary of Apparent Effects of Clinical Factors on Bone and Artery Mineralization

    Diabetes

    Diabetes is clinically associated with vascular calcification and osteopenia.90,91 The presence of radiologically detectable arterial calcification is a strong marker of future cardiovascular events in diabetic patients.92 Although this was initially attributed to medial (rather than intimal) calcification, the distinction was based on continuous versus interrupted x-ray density pattern, which may not be sensitive enough to distinguish the 2 histological types in cases of extensive vascular calcification. At the cellular level, advanced glycation end products associated with diabetes promote mineralization in cultures of microvascular pericytes.93 As noted earlier, Towler et al showed that in genetically modified mice, induction of type II diabetes activates expression of developmental osteogenic transcription factors and proteins in the ascending aorta. Bidder et al showed that glucose also regulates osteopontin gene expression at the transcriptional level.94 Although these mice were hyperlipidemic, diabetes in the absence of hyperlipidemia does induce vascular calcification in a hamster model.95 CAC progression appears to correlate with glycemic control in type I diabetes,96 and peripheral vascular calcification relates to the degree of glycemic control in type II diabetes.97 Interestingly, however, the degree of hyperglycemia does not clearly correlate with the degree of coronary calcification in patients with familial diabetes type II,98 corresponding with the limited relationship between glycemic control and cardiovascular outcomes.99

    Menopause

    Controversy surrounds estrogen and hormone replacement therapy and their effects on cardiovascular disease and vascular calcification. Observational studies suggested estrogen use reduced cardiovascular disease in general and coronary calcification in particular.100 However, in vitro studies using cultured vascular smooth muscle cells showed that estradiol promoted vascular calcification.101 This in vitro finding was challenged initially based on the prevailing view that estrogen conferred cardiovascular protection. However, this view was reversed once randomized controlled trials were performed in large numbers, pointing to the hazard of reliance on observational studies,102,103 and opening the possibility that vascular calcification contributes to adverse effects. Some clinical studies now confirm higher calcium content in coronary plaques of women using hormone replacement therapy (HRT),104,105 whereas other observational studies report reduction in coronary calcification with HRT.106 Conclusions about the relationship between estrogen and coronary calcification cannot be reliably determined from observational studies. Unfortunately, the only data available now regarding estrogen and CAC are observational. Clinical decision-making with respect to HRT should continue to be guided by results of randomized, prospective clinical trials.

    Osteoporosis

    Osteoporosis, like atherosclerosis, is associated with aging, diabetes, smoking, inactivity, and elevated cholesterol.107 Osteoporosis and atherosclerosis tend to occur in the same patients and correlate in severity, even after adjustment for age. In a 10-year longitudinal study of 236 women who experienced menopause during follow-up, those with progression of aortic calcification also had significantly greater decreases in bone mineral density compared with those without progression of vascular calcification.108 The question has been raised whether calcium is "transferred" from bone to artery. However, calcium and phosphate levels in the serum are normally under such tight regulatory control that such a mechanism is unlikely. If transient hypercalcemia were to occur, one would expect diffuse calcium deposition in other tissues, not just the vasculature, as in hypercalcemic metastatic calcification. Moreover, passive deposition alone could not explain the presence of complete bone tissue found in vascular calcification.

    Interestingly, bisphosphonate therapy for osteoporosis may affect vascular calcification. These compounds accumulate in the aorta, especially in animals with diet-induced atherosclerosis,109 reflecting their lipophilic nature and/or their affinity for hydroxyapatite mineral.110 While enhancing mineral density in skeletal bone, bisphosphonates reduce atherosclerosis in animal models.111,112 Bisphosphonates also reduce rat vascular smooth muscle cell proliferation113 and reduce neointimal formation after carotid balloon injury in rabbits.114 In a rat model using vitamin D and warfarin treatment to induce vascular calcification, bisphosphonates also inhibit mineralization.115 It has also been reported that patients receiving bisphosphonate therapy have no significant difference in progression of coronary calcification based on electron beam tomographic imaging.116

    Summary

    Vascular calcification is a clinical marker for atherosclerosis and may represent a special example of the general phenomenon of soft tissue calcification surrounding chronic inflammatory foci. Subintimal lipid deposition induces a type of "atheroscleritis," a chronic inflammatory response that contributes to the development of mature atheromata. The chronic inflammatory foci, atherosclerotic plaques, contain modified lipids, lipoproteins, and inflammatory cytokines that can regulate osteogenic differentiation of vascular cells. Evidence from clinical observations, animal models, and molecular studies suggest factors that regulate bone cell differentiation and mineralization, including BMP-2, MGP, OPN, OPG, and inorganic pyrophosphate also regulate vascular calcification.

    Teleologically, ossification that encompasses inflammatory sites, such as atherosclerotic plaque and tuberculomata, may serve to limit the spread of an infectious or noxious process and physically protect neighboring tissues from injury. However, the process reduces the vascular resilience required for hemodynamic function and introduces high mechanical failure stress in the artery wall. Findings must be interpreted cautiously in light of the complex nature of biomineralization, influenced by multiple interacting factors, many of which remain to be identified. Improved understanding of the mechanism behind vascular calcification will ultimately advance therapeutics that are likely to benefit cardiovascular health.

    References

    Chapman I. Anatomic and clinical significance of calcification of the aortic knob visualized radiographically. Am J Cardiol. 1960; 6: 281–286.

    Rumberger JA, Simons DB, Fitzpatrick LA, Sheedy PF, Schwartz RS. Coronary artery calcium area by electron-beam computed tomography and coronary atherosclerotic plaque area. A histopathologic correlative study. Circulation. 1995; 92: 2157–2162.

    Virchow R. Cellular pathology: as based upon physiological and pathological histology. (Chance F, translator; an unabridged and unaltered republication of the English translation originally published by J.B. Lippincott and Co, Philadelphia, 1863). New York: Dover Publications; 1971: 404–408.

    Demer LL, Watson KE, Bostr?m K. Mechanism of calcification in atherosclerosis. Trends Cardiovasc Med. 1994; 4: 45–49.

    Otto CM, Kuusisto J, Reichenbach DD, Gown AM, O’Brien KD. Characterization of the early lesion of ’degenerative’ valvular aortic stenosis. Histological and immunohistochemical studies. Circulation. 1994; 90: 844–853.

    Feldman T, Glagov S, Carroll JD. Restenosis following successful balloon valvuloplasty: bone formation in aortic valve leaflets. Cathet Cardiovasc Diagn. 1993; 29: 1–7.

    Hunt JL, Fairman R, Mitchell ME, Carpenter JP, Golden M, Khalapyan T, Wolfe M, Neschis D, Milner R, Scoll B, Cusack A, Mohler ER 3rd. Bone formation in carotid plaques: a clinicopathological study. Stroke. 2002; 33: 1214–1219.

    Mohler ER 3rd, Gannon F, Reynolds C, Zimmerman R, Keane MG, Kaplan FS. Bone formation and inflammation in cardiac valves. Circulation. 2001; 103: 1522–1528.

    Bostrom K, Watson KE, Horn S, Wortham C, Herman IM, Demer LL. Bone morphogenetic protein expression in human atherosclerotic lesions. J Clin Invest. 1993; 91: 1800–1809.

    Giachelli CM, Bae N, Almeida M, Denhardt DT, Alpers CE, Schwartz SM. Osteopontin is elevated during neointima formation in rat arteries and is a novel component of human atherosclerotic plaques. J Clin Invest. 1993; 92: 1686–1696.

    Ikeda T, Shirasawa T, Esaki Y, Yoshiki S, Hirokawa K. Osteopontin mRNA is expressed by smooth muscle-derived foam cells in human atherosclerotic lesions of the aorta. J Clin Invest. 1993; 92: 2814–2820.

    Hirota S, Imakita M, Kohri K, Ito A, Morii E, Adachi S, Kim HM, Kitamura Y, Yutani C, Nomura S. Expression of osteopontin messenger RNA by macrophages in atherosclerotic plaques. A possible association with calcification. Am J Pathol. 1993; 143: 1003–1008.

    Shanahan CM, Cary NR, Metcalfe JC, Weissberg PL. High expression of genes for calcification-regulating proteins in human atherosclerotic plaques. J Clin Invest. 1994; 93: 2393–2402.

    Dhore CR, Cleutjens JP, Lutgens E, Cleutjens KB, Geusens PP, Kitslaar PJ, Tordoir JH, Spronk HM, Vermeer C, Daemen MJ. Differential expression of bone matrix regulatory proteins in human atherosclerotic plaques. Arterioscler Thromb Vasc Biol. 2001; 21: 1998–2003.

    Tanimura A, McGregor DH, Anderson HC. Matrix vesicles in atherosclerotic calcification. Proc Soc Exp Biol Med. 1983; 172: 173–177.

    Giachelli C, Bae N, Lombardi D, Majesky M, Schwartz S. Molecular cloning and characterization of 2B7, a rat mRNA which distinguishes smooth muscle cell phenotypes in vitro and is identical to osteopontin (secreted phosphoprotein I, 2aR). Biochem Biophys Res Commun. 1991; 177: 867–873.

    Shioi A, Nishizawa Y, Jono S, Koyama H, Hosoi M, Morii H. ?-glycerophosphate accelerates calcification in cultured bovine vascular smooth muscle cells. Arterioscler Thromb Vasc Biol. 1995; 15: 2003–2009.

    Steitz SA, Speer MY, Curinga G, Yang HY, Haynes P, Aebersold R, Schinke T, Karsenty G, Giachelli CM. Smooth muscle cell phenotypic transition associated with calcification: upregulation of Cbfa1 and downregulation of smooth muscle lineage markers. Circ Res. 2001; 89: 1147–1154.

    Watson KE, Bostrom K, Ravindranath R, Lam T, Norton B, Demer LL. TGF-? 1 and 25-hydroxycholesterol stimulate osteoblast-like vascular cells to calcify. J Clin Invest. 1994; 93: 2106–2113.

    Tintut Y, Alfonso Z, Saini T, Radcliff K, Watson K, Bostrom K, Demer LL. Multilineage potential of cells from the artery wall. Circulation. 2003; 108: 2505–2510.

    Towler DA, Bidder M, Latifi T, Coleman T, Semenkovich CF. Diet-induced diabetes activates an osteogenic gene regulatory program in the aortas of low density lipoprotein receptor-deficient mice. J Biol Chem. 1998; 273: 30427–30434.

    Cheng SL, Shao JS, Charlton-Kachigian N, Loewy AP, Towler DA. Msx2 promotes osteogenesis and suppresses adipogenic differentiation of multipotent mesenchymal progenitors. J Biol Chem. 2003; 278: 45969–45977.

    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.

    Baron R, Neff L, Louvard D, Courtoy PJ. Cell-mediated extracellular acidification and bone resorption: evidence for a low pH in resorbing lacunae and localization of a 100-kD lysosomal membrane protein at the osteoclast ruffled border. J Cell Biol. 1985; 101: 2210–2222.

    Jeziorska M, McCollum C, Wooley DE. Observations on bone formation and remodeling in advanced atherosclerotic lesions of human carotid arteries. Virchows Arch. 1998; 433: 559–565.

    Merkel KD, Erdmann JM, McHugh KP, Abu-Amer Y, Ross FP, Teitelbaum SL. Tumor necrosis factor- mediates orthopedic implant osteolysis. Am J Pathol. 1999; 154: 203–210.

    Gerrity RG, Naito HK. Ultrastructural identification of monocyte-derived foam cells in fatty streak lesions. Artery. 1980; 8: 208–214.

    Matsuzaki K, Udagawa N, Takahashi N, Yamaguchi K, Yasuda H, Shima N, Morinaga T, Toyama Y, Yabe Y, Higashio K, Suda T. Osteoclast differentiation factor (ODF) induces osteoclast-like cell formation in human peripheral blood mononuclear cell cultures. Biochem Biophys Res Commun. 1998; 246: 199–204.

    Rosenfeld ME, Yla-Herttuala S, Lipton BA, Ord VA, Witztum JL, Steinberg D. Macrophage colony-stimulating factor mRNA and protein in atherosclerotic lesions of rabbits and humans. Am J Pathol. 1992; 140: 291–300.

    Clinton SK, Underwood R, Hayes L, Sherman ML, Kufe DW, Libby P. Macrophage colony-stimulating factor gene expression in vascular cells and in experimental and human atherosclerosis. Am J Pathol. 1992; 140: 301–316.

    Kaden JJ, Bickelhaupt S, Grobholz R, Haase KK, Sarikoc A, Kilic R, Brueckmann M, Lang S, Zahn I, Vahl C, Hagl S, Dempfle CE, Borggrefe M. Receptor activator of nuclear factor kappaB ligand and osteoprotegerin regulate aortic valve calcification. J Mol Cell Cardiol. 2004; 36: 57–66.

    Price PA, Urist MR, Otawara Y. Matrix Gla protein, a new -carboxyglutamic acid-containing protein which is associated with the organic matrix of bone. Biochem Biophys Res Commun. 1983; 117: 765–771.

    Loeser RF, Wallin R. Cell adhesion to matrix Gla protein and its inhibition by an Arg-Gly-Asp-containing peptide. J Biol Chem. 1992; 267: 9459–9462.

    Otawara Y, Price PA. Developmental appearance of matrix GLA protein during calcification in the rat. J Biol Chem. 1986; 261: 10828–10832.

    Farzaneh-Far A, Proudfoot D, Weissberg PL, Shanahan CM. Matrix gla protein is regulated by a mechanism functionally related to the calcium-sensing receptor. Biochem Biophys Res Commun. 2000; 277: 736–740.

    Luo G, Ducy P, McKee MD, Pinero GJ, Loyer E, Behringer RR, Karsenty G. Spontaneous calcification of arteries and cartilage in mice lacking matrix GLA protein. Nature. 1997; 386: 78–81.

    Bostrom K, Tsao D, Shen S, Wang Y, Demer LL. Matrix GLA protein modulates differentiation induced by bone morphogenetic protein-2 in C3H10T1/2 cells. J Biol Chem. 2001; 276: 14044–14052.

    Wallin R, Cain D, Hutson SM, Sane DC, Loeser R. Modulation of the binding of matrix Gla protein (MGP) to bone morphogenetic protein-2 (BMP-2). Thromb Haemost. 2000; 84: 1039–1044.

    Zebboudj AF, Imura M, Bostrom K. Matrix GLA protein, a regulatory protein for bone morphogenetic protein-2. J Biol Chem. 2002; 277: 4388–4394.

    Zebboudj AF, Shin V, Bostrom K. Matrix GLA protein and BMP-2 regulate osteoinduction in calcifying vascular cells. J Cell Biochem. 2003; 90: 756–765.

    Sweatt A, Sane DC, Hutson SM, Wallin. Matrix Gla protein (MGP) and bone morphogenetic protein-2 in aortic calcified lesions of aging rats. J Thromb Haemost. 2003; 1: 178–185.

    Jie KS, Bots ML, Vermeer C, Witteman JC, Grobbee DE. Vitamin K intake and osteocalcin levels in women with and without aortic atherosclerosis: a population-based study. Atherosclerosis. 1995; 116: 117–123.

    Goodman WG. Vascular calcification in chronic renal failure. Lancet. 2001; 358: 1115–1116.

    O’Brien ER, Garvin MR, Stewart DK, Hinohara T, Simpson JB, Schwartz SM, Giachelli CM. Osteopontin is synthesized by macrophage, smooth muscle, and endothelial cells in primary and restenotic human coronary atherosclerotic plaques. Arterioscler Thromb. 1994; 14: 1648–1645.

    Ohmori R, Momiyama Y, Taniguchi H, Takahashi R, Kusuhara M, Nakamura H, Ohsuzu F. Plasma osteopontin levels are associated with the presence and extent of coronary artery disease. Atherosclerosis. 2003; 170: 333–337.

    Wada T, McKee MD, Steitz S, Giachelli CM. Calcification of vascular smooth muscle cell cultures: inhibition by osteopontin. Circ Res. 1999; 84: 166–178.

    Goldsmith HL, Labrosse JM, McIntosh FA, Maenpaa PH, Kaartinen MT, McKee MD. Homotypic interactions of soluble and immobilized osteopontin. Ann Biomed Eng. 2002; 30: 840–850.

    Miyauchi A, Alvarez J, Greenfield EM, Teti A, Grano M, Colucci S, Zambonin-Zallone A, Ross FP, Teitelbaum SL, Cheresh D, Hruska KA. Recognition of osteopontin and related peptides by an v ? 3 integrin stimulates immediate cell signals in osteoclasts. J Biol Chem. 1991; 266: 20369–20374.

    Asou Y, Rittling SR, Yoshitake H, Tsuji K, Shinomiya K, Nifuji A, Denhardt DT, Noda M. Osteopontin facilitates angiogenesis, accumulation of osteoclasts, and resorption in ectopic bone. Endocrinology. 2001; 142: 1325–1332.

    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.

    Matsui Y, Rittling SR, Okamoto H, Inobe M, Jia N, Shimizu T, Akino M, Sugawara T, Morimoto J, Kimura C, Kon S, Denhardt D, Kitabatake A, Uede T. Osteopontin deficiency attenuates atherosclerosis in female apolipoprotein e-deficient mice. Arterioscler Thromb Vasc Biol. 2003; 23: 1029–1034.

    Simonet WS, Lacey DL, Dunstan CR, Kelley M, Chang MS, Luthy R, Nguyen HQ, Wooden S, Bennett L, Boone T, Shimamoto G, DeRose M, Elliott R, Colombero A, Tan HL, Trail G, Sullivan J, Davy E, Bucay N, Renshaw-Gegg L, Hughes TM, Hill D, Pattison W, Campbell P, Boyle WJ, et al. Osteoprotegerin: a novel secreted protein involved in the regulation of bone density. Cell. 1997; 89: 309–319.

    Boyle WJ, Simonet WS, Lacey DL. Osteoclast differentiation and activation. Nature. 2003; 423: 337–342.

    Hofbauer LC, Shui C, Riggs BL, Dunstan CR, Spelsberg TC, O’Brien T, Khosla S. Effects of immunosuppressants on receptor activator of NF-kappaB ligand and osteoprotegerin production by human osteoblastic and coronary artery smooth muscle cells. Biochem Biophys Res Commun. 2001; 280: 334–339.

    Price PA, June HH, Buckley JR, Williamson MK. Osteoprotegerin inhibits artery calcification induced by warfarin and by vitamin D. Arterioscler Thromb Vasc Biol. 2001; 21: 1610–1616.

    Bucay N, Sarosi I, Dunstan CR, Morony S, Tarpley J, Capparelli C, Scully S, Tan HL, Xu W, Lacey DL, Boyle WJ, Simonet WS. Osteoprotegerin-deficient mice develop early onset osteoporosis and arterial calcification. Genes Dev. 1998; 12: 1260–1268.

    Kaden JJ, Bickelhaupt S, Grobholz R, Haase KK, Sarikoc A, Kilic R, Brueckmann M, Lang S, Zahn I, Vahl C, Hagl S, Dempfle CE, Borggrefe M. Receptor activator of nuclear factor kappaB ligand and osteoprotegerin regulate aortic valve calcification. J Mol Cell Cardiol. 36 (1): 57–66, 2004 Jan.

    Pritzker LB, Scatena M, Giachelli CM. The role of osteoprotegerin and TNF-related apoptosis inducing ligand in human microvascular endothelial cell survival. Mol Biol Cell. 2004

    Browner WS, Lui LY, Cummings SR. Associations of serum osteoprotegerin levels with diabetes, stroke, bone density, fractures, and mortality in elderly women. J Clin Endocrinol Metab. 2001; 86: 631–637.

    Jono S, Ikari Y, Shioi A, Mori K, Miki T, Hara K, Nishizawa Y. Serum osteoprotegerin levels are associated with the presence and severity of coronary artery disease. Circulation. 2002; 106: 1192–1194.

    Schoppet M, Sattler AM, Schaefer JR, Herzum M, Maisch B, Hofbauer LC. Increased osteoprotegerin serum levels in men with coronary artery disease. J Clin Endocrinol Metab. 2003; 88: 1024–1028.

    Hofbauer LC, Schoppet M, Schuller P, Viereck V, Christ M. Effects of oral contraceptives on circulating osteoprotegerin and soluble RANK ligand serum levels in healthy young women. Clin Endocrinol (Oxf). 2004; 60: 214–219.

    Jono S, McKee MD, Murry CE, Shioi A, Nishizawa Y, Mori K, Morii H, Giachelli CM. Phosphate regulation of vascular smooth muscle cell calcification. Circ Res. 2000; 87: E10–E17.

    Block GA. Control of serum phosphorus: implications for coronary artery calcification and calcific uremic arteriolopathy (calciphylaxis). Curr Opin Nephrol Hypertens. 2001; 10: 741–747.

    Johnson K, Moffa A, Chen Y, Pritzker K, Goding J, Terkeltaub R. Matrix vesicle plasma cell membrane glycoprotein-1 regulates mineralization by murine osteoblastic MC3T3 cells. J Bone Miner Res. 1999; 14: 883–892.

    Rutsch F, Ruf N, Vaingankar S, Toliat MR, Suk A, Hohne 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, Nurnberg P. Mutations in ENPP1 are associated with ’idiopathic’ infantile arterial calcification. Nat Genet. 2003; 34: 379–381.

    Gerber HP, Ferrara N. Angiogenesis and bone growth. Trends Cardiovasc Med. 2000; 10: 223–228.

    Fiore S, Serhan CN. Formation of lipoxins and leukotrienes during receptor-mediated interactions of human platelets and recombinant human granulocyte/macrophage colony-stimulating factor-primed neutrophils. J Exp Med. 1990; 172: 1451–1457.

    Tintut Y, Patel J, Parhami F, Demer LL. Tumor necrosis factor- promotes in vitro calcification of vascular cells via the cAMP pathway. Circulation. 2000; 102: 2636–2642.

    Parhami F, Morrow AD, Balucan J, Leitinger N, Watson AD, Tintut Y, Berliner JA, Demer LL. Lipid oxidation products have opposite effects on calcifying vascular cell and bone cell differentiation. A possible explanation for the paradox of arterial calcification in osteoporotic patients. Arterioscler Thromb Vasc Biol. 1997; 17: 680–687.

    Romanov YA, Svintsitskaya VA, Smirnov VN. Searching for alternative sources of postnatal human mesenchymal stem cells: candidate MSC-like cells from umbilical cord. Stem Cells. 2003; 21: 105–110.

    Frink RJ, Achor RW, Brown AL Jr., Kincaid OW, Brandenburg RO. Significance of calcification of the coronary arteries. Am J Cardiol. 1970; 26: 241–247.

    Wong ND, Vo A, Abrahamson D, Tobis JM, Eisenberg H, Detrano RC. Detection of coronary artery calcium by ultrafast computed tomography and its relation to clinical evidence of coronary artery disease. Am J Cardiol. 1994; 73: 223–227.

    Yamamoto H, Imazu M, Hattori Y, Tadehara F, Yamakido M, Nakanishi T, Ito K. Predicting angiographic narrowing > or = 50% in diameter in each of the three major arteries by amounts of calcium detected by electron beam computed tomographic scanning in patients with chest pain. Am J Cardiol. 1998; 81: 778–780.

    Arad Y, Spadaro LA, Goodman K, Newstein D, Guerci AD. Prediction of coronary events with electron beam computed tomography. J Am Coll Cardiol. 2000; 36: 1253–1260.

    Schmermund A, Erbel R. Unstable coronary plaque and its relation to coronary calcium. Circulation. 2001; 104: 1682–1687.

    O’Rourke RA, Brundage BH, Froelicher VF, Greenland P, Grundy SM, Hachamovitch R, Pohost GM, Shaw LJ, Weintraub WS, Winters WL Jr., Forrester JS, Douglas PS, Faxon DP, Fisher JD, Gregoratos G, Hochman JS, Hutter AM Jr., Kaul S, Wolk MJ. Am College of Cardiology/Am Heart Association Expert Consensus document on electron-beam computed tomography for the diagnosis and prognosis of coronary artery disease. Circulation. 2000; 102: 126–140.

    Rosenhek R, Binder T, Porenta G, Lang I, Christ G, Schemper M, Maurer G, Baumgartner H. Predictors of outcome in severe, asymptomatic aortic stenosis. N Engl J Med. 2000; 343: 611–617.

    Wang AY, Wang M, Woo J, Lam CW, Li PK, Lui SF, Sanderson JE. Cardiac valve calcification as an important predictor for all-cause mortality and cardiovascular mortality in long-term peritoneal dialysis patients: a prospective study. J Am Soc Nephrol. 2003; 14: 159–168.

    Braun J, Oldendorf M, Moshage W, Heidler R, Zeitler E, Luft FC. Electron beam computed tomography in the evaluation of cardiac calcification in chronic dialysis patients. Am J Kidney Dis. 1996; 27: 394–401.

    Detrano RC, Wong ND, Doherty RM, Shavelle RM, Tang W, Ginzton LE, Budoff MJ and Nahara K. Coronary calcium does not accurately predict near term future coronary events in high-risk adults. Circulation. 1999; 99: 2633–2638.

    Bocksch WG, Schartl M, Beckmann SH, Dreysse S, Paeprer H. Intravascular ultrasound imaging in patients with acute myocardial infarction: comparison with chronic stable angina pectoris. Coron Artery Dis. 1994; 5: 727–735.

    Beckman JA, Ganz J, Creager MA, Ganz P, Kinlay S. Relationship of clinical presentation and calcification of culprit coronary artery stenoses. Arterioscler Thromb Vasc Biol. 2001; 21: 1618–1622.

    Ge J, Chirillo F, Schwedtmann J, Gorge G, Haude M, Baumgart D, Shah V, von Birgelen C, Sack S, Boudoulas H, Erbel R. Screening of ruptured plaques in patients with coronary artery disease by intravascular ultrasound. Heart. 1999; 81: 621–627.

    Cheng GC, Loree HM, Kamm RD, Fishbein MC, Lee R. Distribution of circumferential stress in ruptured and stable atherosclerotic lesions. A structural analysis with histopathological correlation. Circulation. 1993; 87: 1179–1187.

    Lee RT, Grodzinsky AJ, Frank EH, Kamm RD, Schoen FJ. Structure-dependent dynamic mechanical behavior of fibrous caps from human atherosclerotic plaques. Circulation. 1993; 83: 1764–1770.

    Richardson PD, Davies MJ, Born GV. Influence of plaque configuration and stress distribution on fissuring of coronary atherosclerotic plaques. Lancet. 1989; 2: 941–944.

    Yoon HC, Emerick AM, Hill JA, Gjertson DW, Goldin JG. Calcium begets calcium: progression of coronary artery calcification in asymptomatic subjects. Radiology. 2002; 224: 236–241.

    Bielak LF, Sheedy PF 2nd, Peyser PA. Coronary artery calcification measured at electron-beam CT: agreement in dual scan runs and change over time. Radiology. 2001; 218: 224–229.

    Fuchs U, Caffier P, Schulz HG, Wieniecki P. Arterial calcification in diabetics. Virchows Arch A Pathol Anat Histopathol. 1985; 407: 431–439.

    Levin ME, Boisseau VC, Avioli LV. Effects of diabetes mellitus on bone mass in juvenile and adult-onset diabetes. N Engl J Med. 1976; 294: 241–245.

    Lehto S, Niskanen L, Suhonen M, Ronnemaa 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.

    Yamagishi S, Fujimori H, Yonekura H, Tanaka N, Yamamoto H. Advanced glycation endproducts accelerate calcification in microvascular pericytes. Biochem Biophys Res Commun. 1999; 258: 353–357.

    Bidder M, Shao JS, Charlton-Kachigian N, Loewy AP, Semenkovich CF, Towler DA. Osteopontin transcription in aortic vascular smooth muscle cells is controlled by glucose-regulated upstream stimulatory factor and activator protein-1 activities. J Biol Chem. 2002; 277: 44485–44496.

    Sima A, Popov D, Starodub O, Stancu C, Cristea C, Stern D, Simionescu M. Pathobiology of the heart in experimental diabetes: immunolocalization of lipoproteins, immunoglobulin G, and advanced glycation endproducts proteins in diabetic and/or hyperlipidemic hamster. Lab Invest. 1997; 77: 3–18.

    Snell-Bergeon JK, Hokanson JE, Jensen L, MacKenzie T, Kinney G, Dabelea D, Eckel RH, Ehrlich J, Garg S, Rewers M. Progression of coronary artery calcification in type 1 diabetes: the importance of glycemic control. Diabetes Care. 2003; 26: 2923–2928.

    Ishimura E, Okuno S, Kitatani K, Kim M, Shoji T, Nakatani T, Inaba M, Nishizawa Y. Different risk factors for peripheral vascular calcification between diabetic and non-diabetic haemodialysis patients–importance of glycaemic control. Diabetologia. 2002; 45: 1446–1448.

    Wagenknecht LE, Bowden DW, Carr JJ, Langefeld CD, Freedman BI, Rich SS. Familial aggregation of coronary artery calcium in families with type 2 diabetes. Diabetes. 2001; 50: 861–866.

    UK Prospective Diabetes Study (UKPDS) Group. Intensive blood glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). Lancet. 1998; 352: 837–853.

    McLaughlin VV, Hoff JA, Rich S. Relation between hormone replacement therapy in women and coronary artery disease estimated by electron beam tomography. Am Heart J. 1997; 134: 1115–1119.

    Balica M, Bostrom K, Shin V, Tillisch K, Demer LL. Calcifying subpopulation of bovine aortic smooth muscle cells is responsive to 17 ?-estradiol. Circulation. 1997; 95: 1954–1960.

    Hulley S, Grady D, Bush T, Furberg C, Herrington D, Riggs B, Vittinghoff E. Randomized trial of estrogen plus progestin for secondary prevention of coronary heart disease in postmenopausal women. Heart and Estrogen/progestin Replacement Study (HERS) Research Group. JAMA. 1998; 280: 605–613.

    Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C, Stefanick ML, Jackson RD, Beresford SA, Howard BV, Johnson KC, Kotchen JM, Ockene J; Writing Group for the Women’s Health Initiative Investigators. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women’s Health Initiative randomized controlled trial. JAMA. 2002; 288: 321–333.

    Christian RC, Harrington S, Edwards WD, Oberg AL, Fitzpatrick LA. Estrogen status correlates with the calcium content of coronary atherosclerotic plaques in women. J Clin Endocrinol Metab. 2002; 87: 1062–1067.

    Shemesh J, Frenkel Y, Leibovitch L, Grossman E, Pines A, Motro M. Does hormone replacement therapy inhibit coronary artery calcification? Obstet Gynecol. 1997; 89: 989–992.

    Akhrass F, Evans AT, Wang Y, Rich S, Kannan CR, Fogelfeld L, Mazzone T. Hormone replacement therapy is associated with less coronary atherosclerosis in postmenopausal women. J Clin Endocrinol Metab. 2003; 88: 5611–5614.

    Cummings SR, Melton LJ. Epidemiology and outcomes of osteoporotic fractures. Lancet. 2002; 359: 1761–1767.

    Hak AE, Pols HA, van Hemert AM, Hofman A, Witteman JC. Progression of aortic calcification is associated with metacarpal bone loss during menopause: a population-based longitudinal study. Arterioscler Thromb Vasc Biol. 2000; 20: 1926–1931.

    Ylitalo R, Monkkonen J, Urtti A, Ylitalo P. Accumulation of bisphosphonates in the aorta and some other tissues of healthy and atherosclerotic rabbits. J Lab Clin Med. 1996; 127: 200–206.

    Jarvis MF, Burns CJ, Pauls HW, Assal A, Kim JS, Cheney DL, Youssefyeh RD. Characterization of the bisphosphonate recognition site on hydroxyapatite using radioligand binding techniques with citric acid. Calcif Tissue Int. 1993; 52: 372–377.

    Kramsch DM, Chan CT. The effect of agents interfering with soft tissue calcification and cell proliferation on calcific fibrous-fatty plaques in rabbits. Circ Res. 1978; 42: 562–571.

    Hollander W, Paddock J, Nagraj S, Colombo M, Kirkpatrick B. Effects of anticalcifying and antifibrobrotic drugs on pre-established atherosclerosis in the rabbit. Atherosclerosis. 1979; 33: 111–123.

    Su JZ, Fukuda N, Kishioka H, Hu WY, Kanmatsuse K. Etidronate influences growth and phenotype of rat vascular smooth muscle cells. Pharmacol Res. 2002; 46: 7–13.

    Danenberg HD, Fishbein I, Gao J, Monkkonen J, Reich R, Gati I, Moerman E, Golomb G. Macrophage depletion by clodronate-containing liposomes reduces neointimal formation after balloon injury in rats and rabbits. Circulation. 2002; 106: 599–605.

    Price PA, Faus SA, Williamson MK. Bisphosphonates alendronate and ibandronate inhibit artery calcification at doses comparable to those that inhibit bone resorption. Arterioscler Thromb Vasc Biol. 2001; 21: 817–824.

    Hill JA, Goldin JG, Gjertson D, Emerick AM, Greaser LD, Yoon HC, Khorrami S, Aziz D, Adams JS. Progression of coronary artery calcification in patients taking alendronate for osteoporosis. Acad Radiol. 2002; 9: 1148–1152.(Mechanisms and Clinical R)