Open access

Biological Effects of Skeletal Renin-Angiotensin System in Osteoporosis

Written By

Yan Zhang and Liang Wang

Submitted: 01 September 2014 Published: 04 March 2015

DOI: 10.5772/59299

From the Edited Volume

Advances in Osteoporosis

Edited by Yannis Dionyssiotis

Chapter metrics overview

1,787 Chapter Downloads

View Full Metrics

1. Introduction

1.1. Classical renin-angiotensin system

The renin–angiotensin system (RAS) is an endocrine system that governs body fluid, electrolyte balance and blood pressure. Within the classical RAS, angiotensinogen (AGT) secreted by liver is enzymatically cleaved to angiotensin (ANG) I by kidney-derived renin. ANG I is, hereafter, cleaved by angiotensin-converting enzyme (ACE) to generate the effector hormone ANG II, which exerts various biological actions through its receptors, ANG II type 1 receptor (AT1) and ANG II type 2 receptor (AT2). The initial reaction between the enzyme renin and the substrate AGT is the rate-limiting step of the RAS [1].

As the RAS is a hormonal cascade that is thought to act as a master controller of blood pressure and fluid balance within the body [2], the systematic RAS has been an important target of antihypertensive medications. There are several groups of drugs in this category that affect different parts of the RAS axis, including ACE inhibitors (ACEI) and angiotensin receptor blockers (ARB), both of which are widely used for anti-hypertension treatment [3]. Additionally, Aliskiren, the first orally active direct renin inhibitor approved for clinical use, is a small molecule competitive inhibitor that specifically inhibits the enzymatic activity of renin [4, 5], consequently it could effectively suppress the rate-limiting step within RAS cascade to reduce the production of ANG II. The recent evidences have shown the effective blood pressure control of Aliskiren, generally well tolerated as monotherapy or in combination with other antihypertensive drugs [3, 6].

Advertisement

2. Tissue renin–angiotensin system

From an evolutionary point of view, it is cost-effective to have a common system to potentiate and effect the actions of the regulating hormones. Interestingly, the RAS is also found in primitive animals without a closed circulatory system, which indicates that the system is far more than a mediator of vasoconstriction [7]. It is now evident that the components of RAS, in addition to the classical pathway, are produced and acting locally in multiple tissues, a concept known as tissue RAS [7]. The local effects of tissue RAS are diverse and depend on the specific tissues involved.

The functional tissue RAS is postulated to participate in various physiological and pathological processes such as insulin secretion [8], glomerular sclerosis [9], renal inflammation [10], atherosclerosis [11], cardiac hypertrophy [12], brain ischemia [13] and follicular development and endometrial cancer in female reproductive tract [14]. A growing body of studies has demonstrated that the diabetic complications, such as cardiovascular disease [15], nephropathy [16] and retinopathy [17], are caused by the high activity of tissue RAS and the increased production of ANG II in local tissues, and the clinical practice has revealed that these pathological alterations associated with diabetes were significantly improved in response to the treatment with RAS inhibitors [15-17]. Additionally, hyperglycemia, obesity, hypertension, and cortisol, well-known risk factors of metabolic disease, are all stimulators on tissue RAS, whereas glucagon-like peptide-1, vitamin D, and aerobic exercise could, to some extent, prevent metabolic disease through inhibiting tissue RAS [7]. Thus, the factors and drugs suppressing tissue RAS activity have potential in improving RAS-involved tissue injuries.

Recent in vivo studies showed that the components of RAS, such as renin, ACE, and ANG II receptors, were expressed in the local milieu of bone [1, 18-20], and in vitro study identified the expression of ANG II receptors in primary osteoblasts derived from newborn mouse calvaria [1], indicating the components of RAS are expressed locally in bone microenvironment. Our further animal studies demonstrated that the local RAS in bone was involved in age-related osteoporosis of aging mice [21], and bone deteriorations of mice with either obstructive nephropathy [22] or type 1 diabetes [23]. Other groups elucidated the involvement of skeletal RAS in the process of fracture healing in a mouse femur fracture model [24], and the steroid-induced osteonecrosis in rabbits [20] as well as the development of postmenopausal osteoporosis in ovariectomized (OVX) animal models [25, 26] and glucocorticoid-induced osteoporosis [19]. Therefore, it concludes that the local RAS exists in bone tissue and plays an important role in local bone metabolism.

Advertisement

3. Action of angiotensin II on bone

ANG II has been postulated to be able to act upon the cells involved in bone metabolism through receptors located in osteoblasts and osteoclasts or regulate blood flow in bone marrow capillaries. At the end of last century, the studies showed that ANG II stimulated DNA and collagen synthesis and decreased alkaline phosphatase (ALP) activity in bone cell populations derived from calvariae of fetal rat [27] and newborn rat [28]. Similar effects of ANG II were observed in osteoblastic ROS17/2.8 cells [29] and human adult bone cells obtained by collagenase digestion from trabecular bone [27]. The clonal cell analysis, autoradiographic studies, and receptor subtype analysis suggested that ANG II might be intimately involved in the proliferation of the osteoblast-rich populations of cells through the AT1 receptor [27, 28], which also plays one of the essential roles in bone metabolism as a mechanoreceptor of osteoblasts [30].

When investigating the direct effects of ANG II on matured osteoblasts, the results revealed that ANG II inhibited the expression of mRNA for osteocalcin, which is a protein that is specifically expressed during maturation of osteoblastic cells, decreased the activity of ALP, the number and the total area of mineralized nodules as well as reduced the accumulation of calcium in cells and the matrix layer [31]. Besides, the ANG II-involved impairment of bone formation may be attributed that it altered the expression of Cbfa1 by activating the cAMP signaling pathway and subsequently reduced osteoblast number and osteoblastic function [32]. SOST, which encodes sclerostin, is a secretory product of osteocytes that counters Wnt signaling, thereby negatively regulates bone formation [33]. The AT1-involved inhibition on bone formation was highly correlated with its regulation on downstream factor SOST, as the decreased SOST expression in osteocytes was observed in AT1-deficient mice [34]. Furthermore, the treatment with ANG II strikingly increased the expressions of matrix metalloproteinase (MMP)-3 and-13 through MAPK signaling pathways via the AT1 in osteoblastic ROS17/2.8 cells, suggesting that ANG II stimulated the degradation process that occurs during extracellular matrix (ECM) turnover in osteoid by increasing the production of MMP-3 and-13 in osteoblasts [29]. Additionally, ANG II induced mitochondrial dysfunction and promoted apoptosis via JNK signaling pathway in primary mouse calvaria osteoblast [35]. Taken together, the target genes including Cbfa1, SOST, MMP-3 and MMP-13, and the signaling pathways like MAPK and JNK are involved in the mediation of ANG II on osteoblastic function and bone formation.

Osteoblast modulates osteoclast differentiation by producing both positive and negative regulators, most notably receptor activator of NF-κB ligand (RANKL) and osteoprotegerin (OPG), respectively [36]. Of note, it has been recently found that ANG II could induce the differentiation of bone marrow mononuclear cells to multinuclear cells and the number of multinuclear cells in osteoclasts as well as increase tartrate-resistant acid phosphatase (TRAP)-positive multinuclear osteoclasts due to its stimulation on the expression of osteoclastogenesis-supporting cytokine, RANKL in osteoblasts, leading to the activation of osteoclasts [1, 25], whereas these effects were completely blocked by either ANG II type 1 receptor blockade (olmesartan) or mitogen-activated protein kinase kinase inhibitors (U0126) [25]. Importantly, ANG II itself had no capacity to induce osteoclast differentiation and did not potentiate osteoclast formation triggered by RANKL, while it stimulated the formation of osteoclasts in the co-culture system of primary osteoblasts and bone marrow macrophages in a dose-dependent manner [1, 32]. Taken together, these results suggested that ANG II stimulates osteoclastogenesis by acting on osteoblastic cells (i.e., ‘the soil cells’), but not through a direct action on hematopoietic ‘seed cells’ [1].

It was found that the TRAP activity and the TRAP-positive stained area were significantly increased in the tibia of OVX rats with systemic administration of ANG II at a subpresser dose (200 ng/kg/min), and the treatment with ANG II significantly induced the ovariectomy-induced increase in urinary level of deoxypyridinoline [25]. The ratio of ALP to TRAP was significantly decreased in the tibia of OVX rats upon to ANG II treatment. These results suggested that ANG II accelerated the turnover of bone metabolism, which is similar to the typical pattern in elderly postmenopausal women who are at high risk for osteoporosis [25]. Of importance, the bone density as assessed by double energy X ray absorptiometry (DEXA) was significantly decreased in the tibia of OVX rats by ANG II. These results suggested that ANG II directly accelerated estrogen deficiency-induced osteoporosis.

Advertisement

4. Interaction of AT1 and AT2 on bone

Previous studies have focused on the ANG II–AT1 interactions since these are the best described and considered the most important. However, the system is complex and several other components probably play significant roles as well [7]. Several publications raise the possibility that AT1 and AT2 carry out negative cross-talk within fibroblasts and vascular endothelial cells with respect to each other’s signaling pathways and responses [37]. This may be of particular importance when the AT1 are pharmacologically blocked.

Asaba et al. determined the relative contribution of the two receptors for transducing the osteoclastogenesis-supporting function of ANG II in osteoblasts by knocking down the expression of each of the receptors with siRNA in primary osteoblasts in culture [1]. In AT1-knockdown osteoblasts, the stimulatory effect of ANG II on osteoclast formation was somewhat enhanced. In AT2-knockdown osteoblasts, in contrast, the osteoclastogenic potential was markedly attenuated [1], which was consistent with that AT2 deficiency increased bone mass of distal metaphyseal regions of femoral in mice as well as the treatment with AT2 blocker PD123319 suppressed ANG II-induced increase in the number of osteoclasts in organ cultures of bone [18]. Asaba et al.’s study suggested that the action of ANG II on osteoblasts in terms of stimulating osteoclastogenesis was mainly mediated through the AT2 and AT1 might exert an inhibitory effect on AT2. These findings in osteoblasts are consistent with the notion that the functions of AT1 and AT2 are in many cases counter-regulatory to each other [7]. However, they are contrary to the conclusion that AT2 is the protective arm of RAS and counterbalances pathological processes and enable recovery from disease [38]. Thus, further studies are needed to dissect the signaling pathways downstream of each receptor in osteoblasts.

Advertisement

5. ACE inhibitors and osteoporosis

Osteoporosis, hypertension, diabetes are major chronic diseases in older subjects and the latter two are well known to be high risk factors for osteoporosis. As ACE inhibitors are usually prescribed for hypertension, cardiac failure, and diabetic nephropathy [23, 39], it is important to know the prospective effects of ACEI on bones of these patients taking ACEI treatment.

Previously, most of clinical studies demonstrated that patients treated with ACEI showed an increased bone mineral density (BMD) and a reduced fracture risk [40-45]. The menopausal and hypertensive women who followed treatment with ACEI fosinopril did not present the physiological loss of bone mass that affected to menopausal women without treatment [43]. A large case–control analysis carried out in the UK, suggested a possibly decreased fracture risk associated with longer-term use of ACE inhibitors [40], and in an open prospective study including 134 patients with low to moderate hypertension and stable BMD, the plasma calcium and 25-hydroxyvitamin D levels were both increased in patients treated with the ACEI quinapril [41]. It also significantly increased BMD of lumbar spine in female subjects with ACE DD genotype, which could induce a higher level of ANG II [41]. The research group from Hong Kong performed two large scale cohort studies which investigated the risk factors for osteoporotic fractures in Hong Kong-dwelling elderly Chinese, and their data concluded that male ACEI users had higher BMD at the total hip, female neck and lumbar spine than non-users. Likewise, female ACEI users also had higher BMD than non-users, although only significant at the femoral neck [42].

While, in the contrary to the above mentioned beneficial effects of ACEI on bone health, the recent emerging evidences indicated that ACEI use did not change the rate and risk of fracture [46], and even led to greater bone loss [39, 47, 48]. The same research group from Hong Kong recently also stated that female continuous users of ACEI had increased bone loss both in total hip and femur neck [47]. A large sample size study in American men also supported this theory by showing that ACEI use was associated with increased bone loss [39], moreover, another prospective study-a cohort study of atomic bomb survivors in Japan, demonstrated that ACEI use was associated with increased bone loss of femoral neck in older Japanese [48].

Similarly in animal studies, the use of ACEI enalapril (10-20 mg/kg, i.g.) did not show positive effects on bone function of OVX mice [49] or OVX spontaneously hypertensive rats (SHR) [50], and the administration of enalapril (0.4 mg/kg, i.p.) in a dose recommended for the treatment of hypertension did not cause significant changes in bone density, the ash and mineral content or morphometric parameters of the femur in female Wistar rats [51]. Another ACEI moexipril, when given alone at oral dose of 10 mg/kg, had no effect on the cancellous bone site in either OVX or sham-operated rats and did not hamper the osteoprotective effects of 17beta-estradiol [52]. Even though the treatment of Tsukuba hypertensive mouse with enalapril improved osteoporosis [1] as well as the OVX rats in response to the treatment with ACEI captopril (1 or 5 mg/kg) showed the increased trabecular area of lumbar vertebrae (L4) and the improved biomechanical properties by increasing L5 break stress and elastic modulus [26], our recent published article elucidated that the treatment with captopril (10 mg/kg, i.g.) significantly elevated serum level of TRAP 5b, and had a trend to decrease BMD of trabecular bone and damage micro-architecture of proximal tibial head and distal femoral end in type 1 diabetic mice [23].

Based on the facts that ANG II locally in bone tissue has detrimental effects to bone function and ACE is the major enzyme producing ANG II, it is surprising that ACEI could not improve even accelerate bone loss in both humans and animals. Since the modest changes in ACE level affect the levels of its substrates much more than its products, indicating that relatively small changes in the levels of ACE affect kinin level more than ANG II level [53], a possible reason comes from the regulation of ACE on kinin-kallikrein system within which bradykinin can stimulate bone resorption and reduce BMD [47]. Another possible explanation we should consider is that although short-term ACEI therapy was associated with decreased ANG II level, there were some evidences that long-term ACE inhibition resulted in a return of ANG II towards baseline level, so-called ‘ACE escape’ [47]. The complete mechanisms have not yet been fully demonstrated. More research needs to be carried out to clarify the influence of the ACEI treatment on bone health as this might be of clinical relevance when antihypertensive therapy is initiated, particularly in hypertensive women who typically suffer from a concomitant rapid onset of osteoporosis after menopause [52].

Advertisement

6. Angiotensin receptor blockers and osteoporosis

The clinical profiles of users of ACEI and ARB were very similar. In the USA, ARB was usually prescribed when ACEI was not tolerated, thus explaining the smaller number of ARB users [39] and limited human studies of ARB and BMD or fracture risk in the literatures [46]. The completed studies on the affections of ARB on bone function in human and animals have shown contradictory results.

The recent population-based, retrospective cohort study with propensity score-matching using administrative databases in Ontario, Canada to examine the risk of osteoporosis-related fractures in hypertensive elderly patients treated with ARBs versus ACEIs, showed that there was no significant difference between the effects of ARBs and ACE inhibitors on hip and other osteoporotic fractures [54]. A large cohort study on Medicare beneficiaries with a diagnosis of hypertension initiating single-drug therapy for anti-hypertension treatment suggested the increasingly protective effect of ARB on relative fracture risk over time [46]. While, the study with large sample size of community-dwelling older adults from six different geographic regions demonstrated that the use of ARBs did not have any significant overall effect on bone loss in older men [39].

The contradictory results about the actions of ARBs on bone metabolism were also shown among animal studies. The treatment with telmisartan, olmesartan, and losartan, could reduce bone loss of OVX mice [49], attenuate the ovariectomy-induced decrease in BMD [25], and increase bone strength, mass and trabecular connections of OVX rats femur [55, 56], respectively. Moreover, telmisartan partially protected from thiazolidinedione-induced bone loss by actively blocking thiazolidinedione-induced anti-osteoblastic activity via maintaining PPARγ serine 112 phosphorylation [57], and promoted fracture healing in a mice model [58]. However, some studies reported ARBs did not cause significant changes of bone properties in normal female rats [51], type 2 diabetic mice [57], OVX rats [59] or orchiectomized rats [60]. Importantly, it was noted that in some animal models ARBs may lead to more bone injuries [1, 61]. The treatment of transgenic Tsukuba hypertensive mouse with losartan resulted in exacerbation of the low bone mass phenotype [1]. The study in our group demonstrated a trend of losartan to promote the loss of bone mass and the deteriorations of trabecular bone micro-architecture in type 1 diabetic mice due to the compensatory stimulation of bone RAS activation as shown by the up-regulation of renin and ANG II expression in bone tissue [61].

Advertisement

7. Perspective

It has been argued that neither ACEIs nor ARBs completely block the RAS cascade due to the disruption of the feedback inhibition of renin production [62]. The increase in renin activity stimulates the conversion of ANG I and ultimately ANG II, which largely limits the efficacy of RAS inhibition [63]. The increased renin can also act through the prorenin/renin receptor, which may cause tissue damages independent of ANG II [64]. Thus, as compared to single treatment with RAS inhibitors, whether combining renin inhibitor, like Aliskiren, with ARB or ACEI could generate better therapeutic effects on tissue injuries, such as osteoporosis, should be further clarified.

As discussed in this chapter, RAS locally plays a key role in the modulation of bone metabolism. However, over the past 10 years, several studies have presented evidences for the existence of a new arm of the RAS, namely the ACE2/ANG-(1–7)/Mas axis [65]. The identification of the ACE homolog, ACE2 as a key ANG-(1–7)-forming enzyme, unravels the existence of a distinct enzymatic pathway for the production of ANG-(1-7), which has a broad range of effects in different organs and tissues that goes beyond its initially described cardiovascular and renal actions [66]. This heptapeptide exerts its actions through binding to a G protein-coupled receptor Mas, distinct from AT1 and AT2 [67]. It is now accepted that the ACE2/ANG-(1–7)/Mas axis is able to counteract most of the deleterious actions of the ACE/ANG II/AT1 axis, especially in pathological conditions [68] such as cardiac dysfunction, increased blood pressure, decreased baroreflex function, endothelial dysfunction, reduced reproductive function, increased thrombogenesis [66]. Thus, how the cross-talk and the interaction between the dual axis systems of RAS contribute to the maintainance of bone metabolism needs to be further investigated and elucidated for better understanding the molecular mechanism of bone metabolic diseases.

Advertisement

Acknowledgments

This work was sponsored by National Natural Science Foundation of China (No. 81202894).

References

  1. 1. Asaba Y, Ito M, Fumoto T, Watanabe K, Fukuhara R, Takeshita S, Nimura Y, Ishida J, Fukamizu A, Ikeda K. Activation of renin-angiotensin system induces osteoporosis independently of hypertension. J Bone Miner Res 2009;24(2) 241-250.
  2. 2. Namazi S, Ardeshir-Rouhani-Fard S, Abedtash H. The effect of renin angiotensin system on tamoxifen resistance. Med Hypotheses 2011;77(1) 152-155.
  3. 3. Robles NR, Cerezo I, Hernandez-Gallego R. Renin-angiotensin system blocking drugs. J Cardiovasc Pharmacol Ther 2014;19(1) 14-33.
  4. 4. Wood JM, Maibaum J, Rahuel J, Grütter MG, Cohen NC, Rasetti V, Rüger H, Göschke R, Stutz S, Fuhrer W, Schilling W, Rigollier P, Yamaguchi Y, Cumin F, Baum HP, Schnell CR, Herold P, Mah R, Jensen C, O'Brien E, Stanton A, Bedigian MP. Structure-based design of aliskiren, a novel orally effective renin inhibitor. Biochem Biophys Res Commun 2003;308(4) 698-705.
  5. 5. Zhang Y, Wang Y, Chen Y, Deb DK, Sun T, Zhao Q, Li YC. Inhibition of renin activity by aliskiren ameliorates diabetic nephropathy in type 1 diabetes mouse model. J Diabetes Mellitus 2012;2(3) 353-360.
  6. 6. Sen S, Sabırlı S, Ozyiğit T, Uresin Y. Aliskiren: review of efficacy and safety data with focus on past and recent clinical trials. Ther Adv Chronic Dis 2013;4(5) 232-241.
  7. 7. Skov J, Persson F, Frøkiær J, Christiansen JS. Tissue renin-angiotensin systems: a unifying hypothesis of metabolic disease. Front Endocrinol (Lausanne) 2014;5 23.
  8. 8. Lau T, Carlsson PO, Leung PS. Evidence for a local angiotensin-generating system and dose-dependent inhibition of glucose-stimulated insulin release by angiotensin II in isolated pancreatic islets. Diabetologia 2004;47(2) 240-248.
  9. 9. Zhang Z, Zhang Y, Ning G, Deb DK, Kong J, Li YC. Combination therapy with AT1 receptor blocker and vitamin D analog markedly ameliorates diabetic nephropathy. Proc Natl Acad Sci USA 2008;105(41) 15896-15901.
  10. 10. Zhang Y, Deb DK, Kong J, Ning G, Wang Y, Li G, Chen Y, Zhang Z, Strugnell S, Sabbagh Y, Arbeeny C, Li YC. Long-term therapeutic effect of vitamin D analog Doxercalciferol on diabetic nephropathy: strong synergism with AT1 receptor antagonist. Am J Physiol Renal Physiol 2009;297(3) F791-F801.
  11. 11. Koïtka A, Cao Z, Koh P, Watson AMD, Sourris KC, Loufrani L, Soro-Paavonen A, Walther T, Woollard KJ, Jandeleit-Dahm KA, Cooper ME, Allen TJ. Angiotensin II subtype 2 receptor blockade and deficiency attenuate the development of atherosclerosis in an apolipoprotein E-deficient mouse model of diabetes. Diabetologia 2010;53(3) 584-592.
  12. 12. Inaba S, Iwai M, Furuno M, Kanno H, Senba I, Okayama H, Mogi M, Higaki J, Horiuchi M. Role of angiotensin-converting enzyme 2 in cardiac hypertrophy induced by nitric oxide synthase inhibition. J Hypertens 2011;29(11) 2236-2245.
  13. 13. Jiang T, Gao L, Lu J, Zhang YD. ACE2-Ang-(1-7)-Mas axis in brain: A potential target for prevention and treatment of ischemic stroke. Curr Neuropharmacol 2013;11(2) 209-217.
  14. 14. Herr D, Bekes I, Wulff C. Local renin-angiotensin system in the reproductive system. Front Endocrinol (Lausanne) 2013;4 150.
  15. 15. Vijan SG. Angiotensin-converting enzyme inhibitors (ACEIs), not angiotensin receptor blockers (ARBs), are preferred and effective mode of therapy in high cardiovascular risk patients. J Indian Med Assoc 2009;107(3) 178-182.
  16. 16. Hoogwerf BJ. Renin-angiotensin system blockade and cardiovascular and renal protection. Am J Cardiol 2010;105(1 Suppl) 30A-35A.
  17. 17. Wright AD, Dodson PM. Diabetic retinopathy and blockade of the renin-angiotensin system: new data from the DIRECT study programme. Eye (Lond) 2007;24(1) 1-6.
  18. 18. Izu Y, Mizoguchi F, Kawamata A, Hayata T, Nakamoto T, Nakashima K, Inagami T, Ezura Y, Noda M. Angiotensin II type 2 receptor blockade increases bone mass. J Biol Chem 2009;284(8) 4857-4864.
  19. 19. Yongtao Z, Kunzheng W, Jingjing Z, Hu S, Jianqiang K, Ruiyu L, Chunsheng W. Glucocorticoids activate the local renin-angiotensin system in bone: possible mechanism for glucocorticoid-induced osteoporosis. Endocrine 2014;DOI: 10.1007/s12020-014-0196-z.
  20. 20. Zhang Y, Wang K, Song Q, Liu R, Ji W, Ji L, Wang C. Role of the local bone renin-angiotensin system in steroid-induced osteonecrosis in rabbits. Mol Med Rep 2014;9(4) 1128-1134.
  21. 21. Gu SS, Zhang Y, Li XL, Wu SY, Diao TY, Hai R, Deng H. Involvement of the skeletal renin-angiotensin system in age-related osteoporosis of ageing mice. Biosci Biotechnol Biochem 2012;76(7) 1367-1371.
  22. 22. Gu SS, Zhang Y, Wu SY, Diao TY, Gebru YA, Deng H. Early molecular responses of bone to obstructive nephropathy induced by unilateral ureteral obstruction in mice. Nephrology 2012;17(8) 767-773.
  23. 23. Diao TY, Pan H, Gu SS, Chen X, Zhang FY, Wong MS, Zhang Y. Effects of angiotensin-converting enzyme inhibitor, captopril, on bone of mice with streptozotocin-induced type 1 diabetes. J Bone Miner Metab 2014;32(3) 261-270.
  24. 24. Garcia P, Schwenzer S, Slotta JE, Scheuer C, Tami AE, Holstein JH, Histing T, Burkhardt M, Pohlemann T, Menger MD. Inhibition of angiotensin-converting enzyme stimulates fracture healing and periosteal callus formation-role of a local renin-angiotensin system. Br J Pharmacol 2010; 159(8) 1672-1680.
  25. 25. Shimizu H, Nakagami H, Osako MK, Hanayama R, Kunugiza Y, Kizawa T, Tomita T, Yoshikawa H, Ogihara T, Morishita R. Angiotensin II accelerates osteoporosis by activating osteoclasts. FASEB J 2008;22(7) 2465-2475.
  26. 26. Liu YY, Yao WM, Wu T, Xu BL, Chen F, Cui L. Captopril improves osteopenia in ovariectomized rats and promotes bone formation in osteoblasts. J Bone Miner Metab 2011;29(2) 149-158.
  27. 27. Lamparter S, Kling L, Schrader M, Ziegler R, Pfeilschifter J. Effects of angiotensin II on bone cells in vitro. J Cell Physiol 1998;175(1) 89-98.
  28. 28. Hiruma Y, Inoue A, Hirose S, Hagiwara H. Angiotensin II stimulates the proliferation of osteoblast-rich populations of cells from rat calvariae. Biochem Biophys Res Commun 1997;230(1) 176-178.
  29. 29. Nakai K, Kawato T, Morita T, Iinuma T, Kamio N, Zhao N, Maeno M. Angiotensin II induces the production of MMP-3 and MMP-13 through the MAPK signaling pathways via the AT(1) receptor in osteoblasts. Biochimie 2013;95(4) 922-933.
  30. 30. Bandow K, Nishikawa Y, Ohnishi T, Kakimoto K, Soejima K, Iwabuchi S, Kuroe K, Matsuguchi T. Low-intensity pulsed ultrasound (LIPUS) induces RANKL, MCP-1, and MIP-1beta expression in osteoblasts through the angiotensin II type 1 receptor. J Cell Physiol 2007;211(2) 392-398.
  31. 31. Hagiwara H, Hiruma Y, Inoue A, Yamaguchi A, Hirose S. Deceleration by angiotensin II of the differentiation and bone formation of rat calvarial osteoblastic cells. J Endocrinol 1998;156(3) 543-550.
  32. 32. Guan XX, Zhou Y, Li JY. Reciprocal roles of angiotensin II and Angiotensin II Receptors Blockade (ARB) in regulating Cbfa1/RANKL via cAMP signaling pathway: possible mechanism for hypertension-related osteoporosis and antagonistic effect of ARB on hypertension-related osteoporosis. Int J Mol Sci 2011;12(7) 4206-4213.
  33. 33. Paszty C, Turner CH, Robinson MK. Sclerostin: a gem from the genome leads to bone-building antibodies. J Bone Miner Res 2010;25(9) 1897-1904.
  34. 34. Kaneko K, Ito M, Fumoto T, Fukuhara R, Ishida J, Fukamizu A, Ikeda K. Physiological function of the angiotensin AT1a receptor in bone remodeling. J Bone Miner Res 2011;26(12) 2959-2966.
  35. 35. Li G, Wang M, Hao L, Loo WT, Jin L, Cheung MN, Chow LW, Ng EL. Angiotensin II induces mitochondrial dysfunction and promotes apoptosis via JNK signalling pathway in primary mouse calvaria osteoblast. Arch Oral Biol 2014;59(5) 513-523.
  36. 36. Zhang Y, Dong XL, Leung PC, Wong MS. Differential mRNA expression profiles in proximal tibia of aged rats in response to ovariectomy and low-Ca diet. Bone 2009;44(1) 46-52.
  37. 37. Yayama K, Okamoto H. Angiotensin II-induced vasodilation via type 2 receptor: role of bradykinin and nitric oxide. Int Immunopharmacol 2008;8(2) 312-318.
  38. 38. Namsolleck P, Recarti C, Foulquier S, Steckelings UM, Unger T. AT(2) receptor and tissue injury: therapeutic implications. Curr Hypertens Rep 2014;16(2) 416.
  39. 39. Kwok T, Leung J, Zhang YF, Bauer D, Ensrud KE, Barrett-Connor E, Leung PC; Osteoporotic Fractures inMen(MrOS) Research Group. Does the use of ACE inhibitors or angiotensin receptor blockers affect bone loss in older men? Osteoporos Int 2012;23(8) 2159-2167.
  40. 40. Schlienger RG, Kraenzlin ME, Jick SS, Meier CR. Use of beta-blockers and risk of fractures. JAMA 2004;292(11) 1326-1332.
  41. 41. Pérez-Castrillón JL, Silva J, Justo I, Sanz A, Martín-Luquero M, Igea R, Escudero P, Pueyo C, Díaz C, Hernández G, Dueñas A. Effect of quinapril, quinapril-hydrochlorothiazide, and enalapril on the bone mass of hypertensive subjects: relationship with angiotensin converting enzyme polymorphisms. Am J Hypertens 2003;16(6) 453-459.
  42. 42. Lynn H, Kwok T, Wong SY, Woo J, Leung PC. Angiotensin converting enzyme inhibitor use is associated with higher bone mineral density in elderly Chinese. Bone 2006;38(4) 584-588.
  43. 43. García-Testal A, Monzó A, Rabanaque G, González A, Romeu A. Evolution of the bone mass of hypertense menopausal women in treatment with fosinopril. Med Clin (Barc) 2006;127(18) 692-694.
  44. 44. Rejnmark L, Vestergaard P, Mosekilde L. Treatment with beta-blockers, ACE inhibitors, and calcium-channel blockers is associated with a reduced fracture risk: a nationwide case-control study. J Hypertens 2006;24(3) 581-589.
  45. 45. Ghosh M, Majumdar SR. Antihypertensive medications, bone mineral density, and fractures: a review of old cardiac drugs that provides new insights into osteoporosis. Endocrine 2014;46(3) 397-405.
  46. 46. Solomon DH, Mogun H, Garneau K, Fischer MA. Risk of fractures in older adults using antihypertensive medications. J Bone Miner Res 2011;26(7) 1561-1567.
  47. 47. Zhang YF, Qin L, Leung PC, Kwok TC. The effect of angiotensin-converting enzyme inhibitor use on bone loss in elderly Chinese. J Bone Miner Metab 2012;30(6) 666-673.
  48. 48. Masunari N, Fujiwara S, Nakata Y, Furukawa K, Kasagi F. Effect of angiotensin converting enzyme inhibitor and benzodiazepine intake on bone loss in older Japanese. Hiroshima J Med Sci 2008;57(1) 17-25.
  49. 49. Kang KY, Kang Y, Kim M, Kim Y, Yi H, Kim J, Jung HR, Park SH, Kim HY, Ju JH, Hong YS. The effects of antihypertensive drugs on bone mineral density in ovariectomized mice. J Korean Med Sci 2013;28(8) 1139-1144.
  50. 50. Ma YF, Stimpel M, Liang H, Pun S, Jee WS. Impact of antihypertensive therapy on the skeleton: effects of moexipril and hydrochlorothiazide on osteopenia in spontaneously hypertensive ovariectomized rats. J Endocrinol 1997;154(3) 467-474.
  51. 51. Broulík PD, Tesar V, Zima T, Jirsa M. Impact of antihypertensive therapy on the skeleton: effects of enalapril and AT1 receptor antagonist losartan in female rats. Physiol Res 2001;50(4) 353-358.
  52. 52. Stimpel M, Jee WS, Ma Y, Yamamoto N, Chen Y. Impact of antihypertensive therapy on postmenopausal osteoporosis: effects of the angiotensin converting enzyme inhibitor moexipril, 17beta-estradiol and their combination on the ovariectomy-induced cancellous bone loss in young rats. J Hypertens 1995;13(12 Pt 2) 1852-1856.
  53. 53. Kakoki M, Sullivan KA, Backus C, Hayes JM, Oh SS, Hua K, Gasim AM, Tomita H, Grant R, Nossov SB, Kim HS, Jennette JC, Feldman EL, Smithies O. Lack of both bradykinin B1 and B2 receptors enhances nephropathy, neuropathy, and bone mineral loss in Akita diabetic mice. Proc Natl Acad Sci U S A 2010;107(22) 10190-10195.
  54. 54. Butt DA, Mamdani M, Gomes T, Lix L, Lu H, Tu K; On Behalf of the Hypertension Outcome, Surveillance Team. Risk of osteoporotic fractures with angiotensin II receptor blockers versus angiotensin converting-enzyme inhibitors in hypertensive community-dwelling elderly. J Bone Miner Res 2014;DOI: 10.1002/jbmr.2271.
  55. 55. Donmez BO, Ozdemir S, Sarikanat M, Yaras N, Koc P, Demir N, Karayalcin B, Oguz N. Effect of angiotensin II type 1 receptor blocker on osteoporotic rat femurs. Pharmacol Rep 2012;64(4) 878-888.
  56. 56. Rajkumar DS, Faitelson AV, Gudyrev OS, Dubrovin GM, Pokrovski MV, Ivanov AV. Comparative evaluation of enalapril and losartan in pharmacological correction of experimental osteoporosis and fractures of its background. J Osteoporos 2013;2013 325693.
  57. 57. Kolli V, Stechschulte LA, Dowling AR, Rahman S, Czernik PJ, Lecka-Czernik B. Partial agonist, telmisartan, maintains PPARγ serine 112 phosphorylation, and does not affect osteoblast differentiation and bone mass. PLoS One 2014;9(5) e96323.
  58. 58. Zhao X, Wang JX, Feng YF, Wu ZX, Zhang Y, Shi L, Tan QC, Yan YB, Lei W. Systemic treatment with telmisartan improves femur fracture healing in mice. PLoS One 2014;9(3) e92085.
  59. 59. Li YQ, Ji H, Shen Y, Ding LJ, Zhuang P, Yang YL, Huang QJ. Chronic treatment with angiotensin AT1 receptor antagonists reduced serum but not bone TGF-beta1 levels in ovariectomized rats. Can J Physiol Pharmacol 2009;87(1) 51-55.
  60. 60. Zhang YF, Qin L, Kwok TC, Yeung BH, Li GD, Liu F. Effect of angiotensin II type I receptor blocker losartan on bone deterioration in orchiectomized male hypertensive and normotensive rats. Chin Med J (Engl) 2013;126(14) 2661-2665.
  61. 61. Zhang Y, Diao TY, Gu SS, Wu SY, Gebru YA, Chen X, Wang JY, Ran S, Wong MS. Effects of angiotensin II type 1 receptor blocker on bones in mice with type 1 diabetes induced by streptozotocin. J Renin Angiotensin Aldosterone Syst 2014;15(3) 218-227.
  62. 62. Koid SS, Ziogas J, Campbell DJ. Aliskiren reduces myocardial ischemia-reperfusion injury by a bradykinin B2 receptor-and angiotensin AT2 receptor-mediated mechanism. Hypertension 2014;63(4) 768-773.
  63. 63. Zhang Z, Zhang Y, Ning G, Kong J, Deb DK, Li YC. Combination therapy with AT1 receptor blocker and vitamin D analog markedly ameliorates diabetic nephropathy. Proc Natl Acad Sci U S A 2008;105(41) 15896-15901.
  64. 64. Zhang Y, Deb DK, Kong J, Ning G, Wang Y, Li G, Chen Y, Zhang Z, Strugnell S, Sabbagh Y, Arbeeny C, Li YC. Long-term therapeutic effect of vitamin D analog Doxercalciferol on diabetic nephropathy: strong synergism with AT1 receptor antagonist. Am J Physiol Renal Physiol 2009;297(3) F791-F801.
  65. 65. Passos-Silva DG, Verano-Braga T, Santos RA. Angiotensin-(1-7): beyond the cardio-renal actions. Clin Sci (Lond) 2013;124(7) 443-456.
  66. 66. Santos RA, Ferreira AJ, Verano-Braga T, Bader M. Angiotensin-converting enzyme 2, angiotensin-(1-7) and Mas: new players of the renin-angiotensin system. J Endocrinol 2013;216(2) R1-R17.
  67. 67. Horiuchi M, Iwanami J, Mogi M. Regulation of angiotensin II receptors beyond the classical pathway. Clin Sci (Lond) 2012;123(4) 193-203.
  68. 68. Wong TP, Ho KY, Ng EK, Debnam ES, Leung PS. Upregulation of ACE2-ANG-(1-7)-Mas axis in jejunal enterocytes of type 1 diabetic rats: implications for glucose transport. Am J Physiol Endocrinol Metab 2012;303(5) E669-E681.

Written By

Yan Zhang and Liang Wang

Submitted: 01 September 2014 Published: 04 March 2015