jdm

Journal of Diabetes & Metabolism

ISSN - 2155-6156

Review Article - (2014) Volume 5, Issue 3

Hedgehog Signaling Pathway is Linked with Age-Related Diseases

Monireh Dashty*
Department of Cell Biology, University Medical Center Groningen, University of Groningen, The Netherlands
*Corresponding Author: Monireh Dashty, Department of Cell Biology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, Tel: +31 (0)50 3632536, Fax: +31 (0)50 3632522 Email:

Abstract

Disorders related to chronic inflammation and ageing such as metabolic syndrome, atherosclerosis, osteoporosis and cancers appear to have different pathogenesis. However, by studying the pathways related to the stem cell (SC) signaling network, like Hedgehog signaling and in particular its influence on the adipogenesis - osteogenesis interaction, we could find a close link between the pathogenesis of these disorders. Hedgehog signaling is an active pathway during embryogenesis that seems to be silent in adults. However, it strongly affects the maintenance of adult tissue homeostasis and the highly proliferative and differentiated cells (e.g., SCs, adipocytes and osteocytes). Malfunction of these cells is considered as the basic etiology of a group of age-related diseases such as atherosclerosis, osteoporosis and cancer.

Keywords: Hedgehog signaling pathway, Age-related diseases, Stem cell signaling network, Adipogenesis-osteogenesis interaction

Introduction

Proliferation, differentiation and development of zygote cell for creation of a living entity are one of the astonishing events in medical science, which still remain to be uncovered. From the early stages of development, each cell of embryo mass gains specific characteristics and gene expression properties that are different from others [1,2]. This diversity in the property of cells leads to compartmentation of a group of cells with similar properties in different segments known as organs till the end of development [3]. Hedgehog signaling is one of the highly conserved and key pathways in modulation of differentiation and determination of cell fate. Therefore, it is the regulator of developmental cell fate [4] and it remains to be active till the late stages of embryogenesis (morphogenesis) [5]. Concentration of Hedgehog proteins among embryo segments are different [6]. Hedgehog protein functions together with other oncogenetic and embryogenesis pathways such as transforming growth factor beta (TGF-β), Wnt/ Wingless, Notch and Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathways and receptor tyrosine kinases such as epidermal growth factor (EGF) and fibroblast growth factor (FGF) for modulation of embryonic development [3,7]. After birth, the highly proliferative, differentiative and plasticity capacities of embryonic cells remain and are represented in multipotent cells such as mesenchymal SCs (MSCs) and preadipocytes [8,9]. These cells are able to differentiate into a variety of different cells including adipocyte and osteoblast lineages [10]. The possibility for SCs to follow either the adipogenesis or osteogenesis processes [8] might be the main etiology in the pathogenesis of age-related disorders such as atherosclerosis and osteoporosis. Hedgehog signaling regulates maintenance of integrity and function of tissues/organs and proper regeneration of SCs [11,12] for proper proliferation and differentiation of tissue cells and tissue repair. Therefore, it can considered as a anti-geriatric factor [13]. Malfunction of SC signaling networks [14] increase susceptibility of the body to cancers [7,15]. Moreover, Hedgehog signaling pathway regulates the immune system. Hyperactivity of Hedgehog signaling during embryonic development and immune impairment states such as cancer shows an intense correlation between immune system activity and Hedgehog signaling activity [7,11,15].

Antithetical Effect of Ageing and Diabetes on Hedgehog Signaling

There are different conditions, which function as a bilateral switching of the multipotent cells, like MSCs, to either the adipocyte or osteocyte lineages. Sonic Hedgehog (Shh) has an anti-adipogenicosteoblastic effect on MSCs. Osteogenesis capacity of Shh is accelerated due to upregulation of osteogenic transcription factors [16]. In diabetic patients, hyperglycemia interferes with Shh signaling and Shh-induced bone regeneration; therefore, it stimulates osteoporosis. Hyperglycemia also inhibits osteogenesis through osteogenesis-related genes such as BMP4, Runx2 and osteopontin (OPN), which results in increase in the activity of alkaline phosphatase (ALP) and matrix mineralization in BMSCs [17].

Hedgehog signaling also regulates β-cells-insulin production [18]. During ageing, impairment of Hedgehog signaling function could be the etiology of atrophy, fatty infiltration and fibrosis in pancreas [19] and insulin production. Furthermore, in muscle satellite cells and bone marrow (BM) [20] there is an increased tendency toward adipogenesis at the cost of osteogenesis [21], which leads to unresponsiveness in peripheral tissues and hyperglycemia. In this regulation, the fate of adipocytes depends on PPARγ activation, which has an adipogenic and anti-osteoblastic capacity [22]. Moreover, low level of Shh also correlates to impaired osteogenesis and consequently osteoporosis during ageing [23]. In ageing and diabetic conditions the adipogenesis property of the bone marrow is increased, however the brown adipose tissue (BAT) characteristic of bone marrow is attenuated [24].

Morphogens, Regulatory Proteins of Embryonic Development, Osteogenesis and Adipogenesis

In addition to Hedgehog signaling pathway other morphogens also function in the differentiation of osteoblasts and adipocytes [25]. Morphogens are regulatory proteins like Hedgehog molecules, Wnts [26], fibroblast growth factors (FGF) [27] and bone morphogenetic proteins (BMPs) that integrate together to regulate embryonic development. Metabologens are morphogen proteins that initiate, promote and maintain metabolism and homeostasis [28] and function in the development of brown adipose tissue (BAT adipogenesis), energy metabolism and iron metabolism [29] and regulate embryonic patterning [30].

BMPs metabologens belong to the TGF superfamily members [31]. TGF regulatory ligands, which operate via SMADs, mainly regulate proliferation, differentiation, development, tissue homeostasis and immune system. They inhibit adipogenesis [32] and osteoblast differentiation via inhibition of Core-binding factor alpha-1 (Cbfa1)/ Runx2 [33] and stimulate chondrogenesis [34]. FGF and Wnt signaling also influences osteogenesis, chondrogenesis and adipogenesis [1,35]. Wnt proteins, as in Hedgehog signaling, have anti-adipogenesis and osteoblast differentiation properties [36]. FGF functions directly or via regulation of BMP and Wnt [37]. Activation of Wnt and BMP pathways are involved in tissue repair and triggered by skeletal injury [20]. Dysfunction of these morphogenetic pathways accounts for the susceptibility to a group of cancers [7,35].

Role of Hedgehog Signaling in Maintenance of Cell Functions and Tissue Repair

Hedgehog signaling seems to be a silent pathway during adult life; however, during ischemia and tissue repair, it (particularly Shh) becomes re-activated. Shh regulates maintenance of vasculature and stimulates angiogenesis including vascular development and capillary morphogenesis through phosphoinositide 3-kinase (PI3K) [38]. Impairment of Shh signaling by PI3-K inhibitor (e.g. LY294002), cyclopamine (Shh receptor inhibitor) and diabetic hyperglycemia delays wound healing through suppression of cutaneous constitutive nitric oxide synthase (NOS) function [39] and explains the etiology of diabetic foot syndrome.

In diabetic state, via activation of Shh and consequently induction of NO release, endothelial cells are protected against injury and it improves delayed wound healing. Shh-induced neovascularization occurs via controlling of anti-apoptotic cytokines, production of proangiogenic agents and recruitment of BM-derived SCs. Control of endothelium-bone interaction by Hedgehog signaling plays a crucial role in this repair [40,41]. This interaction has a signigicant role in the pathogenesis of hypertension and atherosclerosis [42]. Furthermore, Shh proteins have a productive effect on cardiac injuries [43] via recruitment of endothelial progenitor cells and fibroblast proliferation. Intramyocardial gene therapy with Shh is used for treatment of myocardial infarction, which carried out through different ways including (i) expression of Shh on fibroblasts and cardiomyocytes (ii) direct activation of myocardial neovascularization (iii) stimulation of endothelial progenitor cells of BM in neovascularization and (iv) morphogenic role of Shh in embryogenesis and tissue repair [44]. However, using Hedgehog signaling as one of the therapeutic ways for treatment of myocardial infarctions or ischemia induced-cerebral injuries is controversial because abnormal Hedgehog signaling activity and its angiogenesis induction could account for the development of cancers [7,43].

Role of Hedgehog Signaling in Energy Metabolism

Energy metabolism is a chemical process for the regulation of energy retour in the body via its components; lipoproteins and metabolic tissues mainly adipose tissue. Hedgehog signaling affects all components of lipid retour including carriers and adipose tissue cells. In humans, Indian Hedgehog (Ihh) is presented on VLDL particle. Transport of VLDL-Ihh to endothelium is essential for maintaining of primary endothelial cells (ECs). The similarity between lipid compositions of insect lipophorins and LDL [4] and existence of Indian Hedgehog on VLDL [45] represents the conserved property of carrying Hedgehog proteins by lipoproteins. Furthermore, adipose tissue consist of macrophages, ECs, fibroblasts, SCs and adipocytes that have metabolic and immunological roles [46]. Adipocytes are one of the sites of Ihh synthesis [8,45]. In rodent adipocytes, Hedgehog signaling (mainly Shh) interferes with adipocytes differentiation. One speculation in this regard is that in adipogenic conditions, Shh functions upstream of peroxisome proliferator-activated receptor gamma (PPARγ); induce GATA2 and 3 anti-adipogenic transcription factors. Another argument would be due to the inhibitory effect of Hedgehog signaling on pro-adipogenic genes via influence on transcription and/or activity of key adipogenic factors like CCAAT/enhancer binding protein alpha (CEBPα), Sterol Regulatory Element Binding Protein (SREBP)-1c and PPARγ. The third possibility could be through its influence on TAZ (transcriptional cofactors with PDZ-binding motif) or retinoblastoma protein because these cofactors activate osteogenic genes and inhibit adipogenic genes. Hedgehog signaling also plays a role downstream of the anti-adipogenic proteins like Wnt proteins, preadipocyte factor (pref)-1 and plasminogen activator inhibitor 1 (PAI-1). Therefore, reduction in the function of Hedgehog signaling is essential for induction of adipocyte differentiation [8]. It is shown that there is a crosstalk between Hedgehog and Wnt pathways, whereby inhibition of β-catenin occurs by direct interaction with Gli3 repressor. Therefore, Hedgehog signaling activation via prevention of Gli3R protein and Wnt signaling activation, inhibits adipocyte differentiation [47].

During adipocyte differentiation, activity of Hedgehog signaling is decreased. However, inhibition of Hedgehog signaling alone does not stimulate adipocyte differentiation and it merely alters adipocyte morphology and insulin sensitivity [10]. Wnt signaling has an important influence on muscle insulin sensitivity, and this is mediated by effect on myogenic and adipogenic systems [21]. Hedgehog signaling controls fat storage via blocking the level of white adipose tissue (WAT), but has no effect on BAT. Cyclic adenosine monophosphate (cAMP) and glucocorticoid induce obesity through blocking of Hedgehog signaling [48]. In metabolism, there is a close interaction between the bone and adipose tissue [49,50] (Figure 1).

diabetes-metabolism-Hedgehog-signaling

Figure 1: Hedgehog signaling pathway and age-related diseases. Hedgehog signaling pathway is a morphogen, which regulates the embryonic cells during development and the multipotent cells in adults for tissue remodelling and maintenance of the function of tissues. The anti-adipogenesis and osteogenesis properties of Hedgehog signaling pathway regulate bone – adipose tissue interaction and consequently metabolic system function. Therefore, it has a direct effect in prevention of the age-related diseases.

Hedgehog Signaling Pathway and Cancer

Fibroblasts are the main cells of the connective tissue that are involved in the synthesis of the extracellular matrix and collagen in the wound healing process. They also have a close similarity with SCs [51]. These two properties influence the function of the fibroblasts on cancer induction. Fibroblasts are produced by epithelial- tomesenchymal transition (EMT) mechanism, which has a role in many developmental processes [52]. Tissue damage enhances fibroblast proliferation. Hedgehog signaling has an anti-adipogenic, pro- EMT capacity. In liver fibrosis, stellate cells, which have epithelial and adipocyte phenotypes, differentiate to myofibroblastic-hepatic stellate cells (MF-HSC) through activation of Hedgehog signaling. Leptin, through activation of Hedgehog signaling and inhibition of adiposity, stimulates differentiation toward EMT; therefore, leptin is modulator of fibrogenesis. Leptin also induces JAK/STAT pathway for gene expression of MSCs. Thus, leptin-related Hedgehog signaling activation is important for expression of genes that mediate cells fate and have an influence on different EMT-related processes including liver fibrosis and cancer metastasis during obesity [53].

This epithelial stromal reaction also has a crucial role in pathogenesis of a group of diseases including breast cancer. Normal breast tissue is composed of stromal and epithelial cells that communicate with each other through extracellular matrix (ECM). Normal crosstalk between these two parts is important for prevention of breast cancer. Epithelial cells also react with human adipose fibroblasts (HAFs) [54]. In carcinomatous breast cancer, a layer of these undifferentiated fibroblasts (HAFs) surround malignant epithelial cells. Epithelial cells secrete tumor necrosis factor (TNF), which via binding to its receptor, TNF receptor (TNFR)-1, inhibits adipogenesis. Fibroblasts, via over-expression of aromatase, which produce estrogen, influence the growth and progression of malignant epithelial cells. Estrogen selectively modulates TNFRs synthesis in HAF, whereby it stimulates TNFR1 expression and inhibits TNFR2 expression. TNFR1 inhibits adipogenesis and TNFR2 stimulates adipogenesis. Therefore, estrogen enhances anti-adipogenic property of TNF through TNFR1 stimulation. In breast cancer, cholesterol derivatives such as specific oxysterol (LXR-independently) and estrogen [55] have anti antiadipogenic- osteoblastic effect via influence on Hedgehog signaling. Estrogen, via stimulation of estrogen receptor alpha (ERα), can induce Shh expression [56,57] and promote motility and invasion of breast cancer cells. This characteristic of estrogen is applied through upregulation of Shh signaling in ERα cells. Reduction of estrogen leads to down-regulation of Shh gene expression [57]. Treatment of cells with estradiol (E2) enhances osteogenesis and inhibits adipogenesis via ER. Another factor that enhances the invasive properties of breast cancer is EGF receptor (EGFR), which enhances tissue factor (TF) expression. TF is a coagulation factor which is secreted by cancer cells and ECs after injury. Alteration of EMT-like features (controlled by Hedgehog signaling) in breast cancer cells modulate EGFR-mediated expression of TF-FVIIa, which consequently stimulate coagulopathy, angiogenesis and metastasis properties of cancer cells [58].

The influence of Hedgehog signaling on fibroblasts and cancer progression is also proved on pancreatic cancer [59,60], ovarian cancer [61], basal cell carcinoma [62], lung malignancy [63] and hepatocarcinogenesis [64]. Hence, Hedgehog pathway blockers [62], together with tyrosine kinase inhibitors [65], proteasome inhibitors, mTOR inhibitors, PI3K inhibitors [66], histone deacetylase inhibitors [67], growth factor inhibitors [68], 3-hydroxy-3-methylglutaryl coenzyme-A reductase (HMG-CoAR) inhibitors [69], angiogenesis inhibitors and agents targeting matrix metalloproteinases, cyclooxygenase-2 (COX-2) [70] are considered as the cancer growth blockers.

Hedgehog Signaling Pathway and Atherosclerosis

Atherosclerosis is a complex process, which is caused through disturbance of SMC proliferation, EC dysfunction, coagulation and immune system [71]. In the process of atherosclerosis, plaque calcification has a close relation to the anti-adipogenesis osteogenesis properties of Hedgehog signaling pathway. Calcification of plaque is an active process and is composed of hydroxyapatite and regulatory proteins including osteopontin, TGFβ, BMPs and RANKL. These proteins, together with oxidized cholesterol, stimulate muscle segment homeobox 2 (Msx2) transcription factor (activator of Wnt signaling) and osteogenic pathway. They also stimulate vascular smooth muscle cells (vSMCs) for OPN expression and upregulation of chondroosteogenic pathway in fibroblasts. Hyperphosphatemia of end-stage renal diseases, via stimulation of Cbfa1/Runx2 transcription factor, can stimulate vascular calcification. Other stimulators of vascular calcification are hyperglycemia, estrogen, Vitamin D toxicity, PTH/ PTHrP receptors [72] and Wnt/LDL receptor-related protein 5 (Lrp5)/ β-catenin pathway [73].

A group of conditions such as T2D, ageing, abnormal valve functions, hypercholesterolemia and chronic renal failure [72] are involved in pathogenesis of atherosclerosis. The pathogenesis related to these metabolic disorders including chronic inflammation induction via recruitment of immune cells (macrophages, lymphocytes) and their cytokines (TNFα, IL-6, IL-1β), retention of lipoproteins, oxidative modifications and renin-angiotensin system (RAS) activation in particular angiotensin converting enzyme (ACE), AngII and Angiotensin II type-1 receptor (AT1-R). Accumulation of extracellular matrix proteoglycans and its interaction with apolipoprotein B (ApoB) of LDL mediates deposition of atherogenic lipoproteins such as LDL, which contains ACE and Hedgehog molecules [13,73] (Figure 2). LDLACE, together with chymase of mast cells, converts Ang I to Ang II, which via activation of AT1-R, promotes fibrosis [74], proteoglycans synthesis and expression of enzyme generating oxidants (e.g., reactive oxygen species (ROS)), which further oxidizes lipoproteins. AT1-R is expressed pathologically in fibroblasts. Oxidation of LDL-cholesterol stimulates fibroblasts toward osteoprogenitor activity and calcification. Oxidized LDL (ox-LDL) recruits monocytes from circulation to initiate inflammation via induction of adhesion and chemotactic molecules on ECs. Monocytes are converted to macrophages to infiltrate toward and phagocytize ox-LDLs to form foam cells [75]. Activated macrophages secrete RANKLs, IL-1β and TNFs and express ROS. Foam cells increase oxidant stress in cells, resulting in impairment of fibrinolysis and increased expression of proteoglycan in myofibroblast and lipoprotein retention. In summary, atherosclerotic calcification appear to be induced via Ang II and lipids oxidation by foam cells cytokines through influence on nuclear factor-kappa B (NFκB) and mitogen-activated protein kinase (MAPK) pathways. In addition, expression of matrix metalloproteinases (MMPs) is increased with a role in regulation of vascular calcification, degradation of extracellular matrix and plaque instability.

diabetes-metabolism-atherosclerotic-plaque

Figure 2: Pathogenesis of atherosclerotic plaque. Normal arteriole is composed of endothelium and the subendothelium or BM. An intact endothelium produces PGI2 and NO, which retain hemostatic components in an inactive form and inhibits monocyte and platelet aggregation. An injured endothelium releases Vwf and TF to start coagulation and exposes BM to the circulation to stimulate circulatory platelets and monocytes to the place of injury. This forms a plaque for repairing the injury and prevention of blood loss. BM composed of a collagen and elastic fibers as well as SMCs that provide support around vessels. IEL and EEL line two sides of tunica intima and separate SMCs from the media and adventitial layers. In normal state, intact IEL prevents migration of SMCs into media space, however in atherosclerotic state, the intima layer growth and macrophage-cathepsis K degradates IEL. This leads to migration of SMCs from intima to media for release of collagen and plaque stabilization. Moreover, pericyte myofibroblasts in atherosclerotic plaque is able to differentiate into osteoblasts and chondrocytes, adventitial myofibrolasts, calcifying vascular cells, valvular interstitial cells and foam cell macrophages, which release cytokines, chemokines and transcription factors. They, as well as fibroblasts, express enzymes generating oxidants (ROS). The processe of plaque formaton and inflammation starts by trapping of LDL containing ACE and Hedgehog (Hh) molecules and binding of them to the proteoglycan (lipid core). Hedgehog molecules function for T cell activation and cytokine production and SMC proliferation and migration into plaque. It functions in regenerative processes including chemotaxis, revascularization, anti-adipogenesis and osteogenesis that are crucial in plaque calcification. Thereafter, LDL follows the oxidative modification processes and forms ox-LDL, which is taken up by macrophages and form foam cells. IL-6 and Ang2 produced by ox-LDL activate foam cell. Ox-LDL also enhances expression of the adhesion and chemoattractant molecules (VCAM-1, ICAM-1, MCP-1) by endothelial cells, which lead to leukocyte recruitment to the plaque. ACE of LDL converts Ang I to Ang II, which activates (AT1-R) on fibroblasts and produce both proteoglycans and oxidants by fibrobelast/myofibroblasts. Hh particle is assumed to be present on the monocytes, LDL, fibroblasts and SMCs. TF and factor V are assumed to have a role in plaque calcification (Dashty, et al, BioEssays, 2012).
BM: Basement Membrane; PGI2: Prostacyclin; NO: Nitric Oxide; vWF: vonwilebrand Factor; TF: Tissue Factor; SMC: Smooth Muscle Cells; IEL: Internal Elastic Laminas; EEL: External Elastic Laminas; ROS: Reactive Oxygen Species; Ox-LDL: Oxidized-Low Density Lipoprotein; IL: Interleukin; Ang: Angiopoietin; VCAM: Vascular Cell Adhesion Protein; ICAM: Intercellular Adhesion Molecule; MCP: Monocyte Chemoattractant Protein; ECs: Endothelial Cells. ACE: Angiotensin-Converting Enzyme; AT1-R: Angiotensin II Type I Receptors.

Besides, Hedgehog signaling, together with FGF, plays an important role in vascular remodeling. This property is also important in tissue engineering for bone regeneration and plaque calcification. During wound healing, Shh signaling stimulates angiogenesis using FGF. Among cells of blood vessels, fibroblasts, periosteal cells and SCs derived from perivascular adipose tissue express Shh gene. During atherosclerosis, Shh and FGF enhance induction of angiopoietins (Ang I or Ang II) and probably vascular endothelial growth factor (VEGF) in fibroblasts [40,76]. In vascular calcification state, pericytemyofibroblasts are differentiated to osteogenic lineage [77]. Pericytes also regulate angiogenesis and thereby link neovascularization with calcification. One assumption in this regard is that EC cytokines induce differentiation and calcification of osteoprogenitor cells. Moreover, some angiogenic factors such as FGF-2 and VEGF-C have also osteogenic and chondrogenic effects. Progenitor cells of both ECs and perivascular cells including pericytes or SMCs are similar. Pericytes are derived from different origins e.g., SMCs, fibroblasts, ECs, BM cells and can differentiate into many different cell types including osteoblasts, chondrocytes, SMCs, adipocytes, fibroblasts and Leydig cells. Therefore, they act as sources of progenitor cells in inflammatory states to repair the disease state [78] (Figure 3).

diabetes-metabolism-Adipogenesis-versus

Figure 3: Adipogenesis versus osteogenesis in ectopic calcification. Induction ofcommon vascular progenitor cells lead to differentiation of cells to either endothelial, pericytes or smooth muscle cells. Pricytes myofibroblasts are also derived from different progenitor cells and fibroblasts. Myofibroblasts have the ability to follow either the adipogenesis or the osteogenesis pathways.

Fibroblasts can also modulate atherosclerosis progression [79] via regulation of EMT, stimulation of fibrosis and synthesis of proteoglycan (regulated by coagulation factors) [80]. Coagulation system in close association with inflammation is also important in atherothrombosis. Rupture of endothelium releases coagulation factors and exposes the subendothelial layer to circulation to attract circulatory platelets and monocytes to the place of injury. Shh is one of these chemoattractants [2], which is expressed on monocytes and plaque cells including fibroblasts and SMCs [81]. Release of TF from ECs [82] enhances angiogenesis, which has a negative effect on plaques [58]. Another coagulation factor which could have an interaction with Hedgehog signaling and have a role in atherosclerosis is factor V (FV), as it may be involved in early mammalian development [83].

Role of Cholesterol in Hedgehog Signaling and Atherosclerosis

There is a direct association between hypercholesterolemia with initiation and exacerbation of atherosclerotic plaque formation. Sterol metabolites are one of the mediators of Hedgehog signaling pathway. In Hedgehog producing cells, cholesterol joins to the produced Hedgehog molecules and functions for destination of Hedgehog molecules to target cells. In Hedgehog receiving cells, sterol secretion induces the inhibitory function of Ptch receptor on Smo receptor in the absence of Hedgehog molecules [84]. Thus, sterol disorders affect Hedgehog pathway and development either by improper sterolation of Hedgehog protein or by a reduced responsiveness of cells to Hedgehog proteins. Transport of Hedgehog molecules occurs via lipophorin in Drosophila and via VLDL in mammals, which is loaded by Hedgehog molecules in adipocytes. VLDL has a crucial role in atherosclerosis pathogenesis; which would be due to the effect of Hedgehog signaling. Existence of Hedgehog molecules on LDL is also speculated [4,85,86]. After trapping of lipoproteins in plaques, Hedgehog molecules stimulate different events in plaques including the regenerative processes (e.g., chemotaxis, revascularization of surrounding tissue, anti-adipogenesis and osteogenesis), proliferation and migration of SMCs [87,88] and T cell activation. T cell activation secretes receptor activator of nuclear factor kappa-B ligand (RANKLs) that in addition to its role in the immune system, plays a role in bone formation and calcification within atherosclerotic plaques and breast cancer [89]. This represents the existence of a close cross-talk between bone metabolism and the immune system [90,91]. RANKLs via TRAF6 adaptor protein stimulate different inflammatory pathways [92]. Therefore, Hedgehog signaling affects bone formation via RANKL (and PTHrP) expression [93,94]. Moreover, Ihh and Shh signaling play an important role in vascular remodeling through induction of VEGF and angiopoietin in fibroblasts [76]. In atherosclerosis, this property may enhance the calcification process [95] in plaques because stimulation of angiogenesis is one of the requirements of the ectopic calcification [78]. Another parameter that affects osteogenesis during atherosclerosis is cathepsin K enzyme that is produced by BM-derived macrophages and stimulates fragmentation of the elastin layer and SMC migration to plaques (Figure 2) [13,96]. Hedgehog signaling stimulates osteoclastic activity via upregulation of Cathepsin K [56].

Other derivatives of cholesterol oxide in the circulation are oxysterols [23]. The levels of these molecules are modified during diseases, ageing and drug intervention states. Oxysterols have diverse roles in inflammation, differentiation, steroid production, cholesterol metabolism, lipoprotein metabolism, calcium uptake, atherosclerosis and apoptosis [97]. The natural form of oxysterols is 20s, which has both the anti-adipogenesis and pro-adipogenesis activity that is mediated by PPARγ. PPARγ is the early stage adipogenic transcription factor that stimulates adipogenesis in adipocytes together with CEBPα. 20s is also one of the regulators of bone metabolism, linking adipogenic to osteogenic pathways. 20s oxysterol has the ability to inhibit PPARγ via stimulation of Hedgehog signaling. It also stimulates both SREBP- 1c/ adipogenic differentiation of factor 1 (ADD1) and PPARγ via enhancement of liver X receptor (LXR). The inhibitory role of 20s in adipogenesis in vascular cells induces the osteoblastic activity of oxysterols and triggers calcification within atherosclerotic plaques (Figure 4) [22].

diabetes-metabolism-Bilateral-role

Figure 4: Bilateral role of oxysterols (20s) in regulation of adipogenesis. While 20s via stimulation of Hedgehog signaling has a inhibitory role in the early stage of adipogenicity, it also has a stimulatory effect on adipocyte differentiation through LXR. PPARγ has the main role in this conection. LXR: Liver X Receptor, PPARγ: Peroxisome Proliferator-Activated Receptor-γ`.

Stressor hormones like cortisol, growth hormone and norepinephrine also regulate bone remodeling. Stress hormones simulate interleukin (IL)-6 secretions that, together with other proinflammatory cytokines such as IL-1β and TNF alpha, are involved in age-related diseases such as T2D, cancer, osteoporosis and atherosclerosis. Steroidal hormones regulate expression of IL-6, which together with Ang II stimulates lipid absorption by macrophages in atherosclerotic plaques. Among these hormones, estrogen has the same properties as Hedgehog signaling in anti-adipogenic and osteogenic pathways [98]. During ageing, IL-6 mediated inflammation also plays a role in osteoporosis. Indirect inhibition of IL-6-induced inflammation by endogenous cholesterol synthesis is one of the therapeutic ways for treatment of atherosclerosis [99]. Schieffer et al. show that a balance between IL-6 and IL-10 is required in protection against atherosclerosis [100].

Conclusion

Regulation of energy metabolism is one of the most complicated events in the body and is fundamental for a healthy life. Dysregulation of the metabolic system is considered as one of the main etiology of ageing. There is a vast interaction between the energy metabolic system of the body and other systems in order to regulate its function. Boneadipose tissue interaction is considered as one of these fundamental events in regulation of metabolism. Dysregulation of this interaction is represented in the pathophysiology of age-related diseases. Hedgehog signaling pathway is the linker between the energy metabolism and bone metabolism. It is active in all cells of adipose tissue including fibroblasts, ECs, SCs and adipocytes and its anti-adipogenesis and osteogenesis property has a significant role in regulation of the energy metabolism” prevention of age-related diseases. Hedgehog signaling is considered as one of the regulators of multipotent cells. multipotent cells like SCs, fibroblasts, osteocytes and adipocytes are cells with the high capacity of proliferation, differentiation and plasticity as it is in embryonic cells. Hedgehog signaling is active during emberyogenesis and after birth it has a strong role in the maintenance of tissues. It is activated during tissue repair; therefore, it has an anti-geriatric effect. A close association between chronic inflammation as one of the main triggers of age-related diseases with an important role of Hedgehog signaling in the pathogenesis of these diseases, would be a proof of a close correlation between an active Hedgehog signaling and tolerogenic states of the immune system.

References

  1. Bilkovski R, Schulte DM, Oberhauser F, Gomolka M, Udelhoven M, et al. (2010) Role of WNT-5a in the determination of human mesenchymal stem cells into preadipocytes. J Biol Chem 285: 6170-6178.
  2. King PJ, Guasti L, Laufer E (2008) Hedgehog signalling in endocrine development and disease. J Endocrinol 198: 439-450.
  3. Casso DJ, Biehs B, Kornberg TB (2011) A novel interaction between hedgehog and Notch promotes proliferation at the anterior-posterior organizer of the Drosophila wing. Genetics 187: 485-499.
  4. Bijlsma MF, Spek CA, Peppelenbosch MP (2006) Hedgehog turns lipoproteins into Janus-faced particles. Trends Cardiovasc Med 16: 217-220.
  5. Ingham PW, McMahon AP (2001) Hedgehog signaling in animal development: paradigms and principles. Genes Dev 15: 3059-3087.
  6. Wilson CW, Chuang PT (2010) Mechanism and evolution of cytosolic Hedgehog signal transduction. Development 137: 2079-2094.
  7. Rubin LL, de Sauvage FJ (2006) Targeting the Hedgehog pathway in cancer. Nat Rev Drug Discov 5: 1026-1033.
  8. Cousin W, Fontaine C, Dani C, Peraldi P (2007) Hedgehog and adipogenesis: fat and fiction. Biochimie 89: 1447-1453.
  9. Gimble JM, Katz AJ, Bunnell BA (2007) Adipose-derived stem cells for regenerative medicine. Circ Res 100: 1249-1260.
  10. Fontaine C, Cousin W, Plaisant M, Dani C, Peraldi P (2008) Hedgehog signaling alters adipocyte maturation of human mesenchymal stem cells. Stem Cells 26: 1037-1046.
  11. Benson RA, Lowrey JA, Lamb JR, Howie SE (2004) The Notch and Sonic hedgehog signalling pathways in immunity. Mol Immunol 41: 715-725.
  12. Shin K, Lee J, Guo N, Kim J, Lim A, et al. (2011) Hedgehog/Wnt feedback supports regenerative proliferation of epithelial stem cells in bladder. Nature 472: 110-114.
  13. Dashti M, Peppelenbosch MP, Rezaee F (2012) Hedgehog signalling as an antagonist of ageing and its associated diseases. Bioessays 34: 849-856.
  14. Katoh M (2007) Dysregulation of stem cell signaling network due to germline mutation, SNP, Helicobacter pylori infection, epigenetic change and genetic alteration in gastric cancer. Cancer Biol Ther 6: 832-839.
  15. Tang Y, Swietlicki EA, Jiang S, Buhman KK, Davidson NO, et al. (2006) Increased apoptosis and accelerated epithelial migration following inhibition of hedgehog signaling in adaptive small bowel postresection. Am J Physiol Gastrointest Liver Physiol 290: G1280-G1288.
  16. Guan CC, Yan M, Jiang XQ, Zhang P, Zhang XL, et al. (2009) Sonic hedgehog alleviates the inhibitory effects of high glucose on the osteoblastic differentiation of bone marrow stromal cells. Bone 45: 1146-1152.
  17. Lu H, Kraut D, Gerstenfeld LC, Graves DT (2003) Diabetes interferes with the bone formation by affecting the expression of transcription factors that regulate osteoblast differentiation. Endocrinology 144: 346-352.
  18. Thomas MK, Rastalsky N, Lee JH, Habener JF (2000) Hedgehog signaling regulation of insulin production by pancreatic beta-cells. Diabetes 49: 2039-2047.
  19. Glaser J, Stienecker K (2000) Pancreas and aging: a study using ultrasonography. Gerontology 46: 93-96.
  20. James AW, Leucht P, Levi B, Carre AL, Xu Y, et al. (2010) Sonic Hedgehog influences the balance of osteogenesis and adipogenesis in mouse adipose-derived stromal cells. Tissue Eng Part A 16: 2605-2616.
  21. Abiola M, Favier M, Christodoulou-Vafeiadou E, Pichard AL, Martelly I, et al. (2009) Activation of Wnt/beta-catenin signaling increases insulin sensitivity through a reciprocal regulation of Wnt10b and SREBP-1c in skeletal muscle cells. PLoS One 4: e8509.
  22. Kim WK, Meliton V, Amantea CM, Hahn TJ, Parhami F (2007) 20(S)-hydroxycholesterol inhibits PPARgamma expression and adipogenic differentiation of bone marrow stromal cells through a hedgehog-dependent mechanism. J Bone Miner Res 22: 1711-1719.
  23. Dwyer JR, Sever N, Carlson M, Nelson SF, Beachy PA, et al. (2007) Oxysterols are novel activators of the hedgehog signaling pathway in pluripotent mesenchymal cells. J Biol Chem 282: 8959-8968.
  24. Krings A, Rahman S, Huang S, Lu Y, Czernik PJ, et al. (2012) Bone marrow fat has brown adipose tissue characteristics, which are attenuated with aging and diabetes. Bone 50: 546-552.
  25. Niemelä SM, Miettinen S, Konttinen Y, Waris T, Kellomäki M, et al. (2007) Fat tissue: views on reconstruction and exploitation. J Craniofac Surg 18: 325-335.
  26. Nusse R (2003) Wnts and Hedgehogs: lipid-modified proteins and similarities in signaling mechanisms at the cell surface. Development 130: 5297-5305.
  27. Behr B, Leucht P, Longaker MT, Quarto N (2010) Fgf-9 is required for angiogenesis and osteogenesis in long bone repair. Proc Natl Acad Sci U S A 107: 11853-11858.
  28. Reddi AH, Reddi A (2009) Bone morphogenetic proteins (BMPs): from morphogens to metabologens. Cytokine Growth Factor Rev 20: 341-342.
  29. Tseng YH, Kokkotou E, Schulz TJ, Huang TL, Winnay JN, et al. (2008) New role of bone morphogenetic protein 7 in brown adipogenesis and energy expenditure. Nature 454: 1000-1004.
  30. Cao X, Chen D (2005) The BMP signaling and in vivo bone formation. Gene 357: 1-8.
  31. Bragdon B, Moseychuk O, Saldanha S, King D, Julian J, et al. (2011) Bone morphogenetic proteins: a critical review. Cell Signal 23: 609-620.
  32. Kumar A, Ruan M, Clifton K, Syed F, Khosla S, et al. (2012) TGF-β mediates suppression of adipogenesis by estradiol through connective tissue growth factor induction. Endocrinology 153: 254-263.
  33. Kang JS, Alliston T, Delston R, Derynck R (2005) Repression of Runx2 function by TGF-beta through recruitment of class II histone deacetylases by Smad3. EMBO J 24: 2543-2555.
  34. Iwasaki M, Nakata K, Nakahara H, Nakase T, Kimura T, et al. (1993) Transforming growth factor-beta 1 stimulates chondrogenesis and inhibits osteogenesis in high density culture of periosteum-derived cells. Endocrinology 132: 1603-1608.
  35. Ng F, Boucher S, Koh S, Sastry KS, Chase L, et al. (2008) PDGF, TGF-beta, and FGF signaling is important for differentiation and growth of mesenchymal stem cells (MSCs): transcriptional profiling can identify markers and signaling pathways important in differentiation of MSCs into adipogenic, chondrogenic, and osteogenic lineages. Blood 112: 295-307.
  36. Taipaleenmäki H, Abdallah BM, AlDahmash A, Säämänen AM, Kassem M (2011) Wnt signalling mediates the cross-talk between bone marrow derived pre-adipocytic and pre-osteoblastic cell populations. Exp Cell Res 317: 745-756.
  37. Lin GL, Hankenson KD (2011) Integration of BMP, Wnt, and notch signaling pathways in osteoblast differentiation. J Cell Biochem 112: 3491-3501.
  38. Kanda S, Mochizuki Y, Suematsu T, Miyata Y, Nomata K, et al. (2003) Sonic hedgehog induces capillary morphogenesis by endothelial cells through phosphoinositide 3-kinase. J Biol Chem 278: 8244-8249.
  39. Luo JD, Hu TP, Wang L, Chen MS, Liu SM, et al. (2009) Sonic hedgehog improves delayed wound healing via enhancing cutaneous nitric oxide function in diabetes. Am J Physiol Endocrinol Metab 297: E525-E531.
  40. Fujii T, Kuwano H (2010) Regulation of the expression balance of angiopoietin-1 and angiopoietin-2 by Shh and FGF-2. In Vitro Cell Dev Biol Anim 46: 487-491.
  41. Kanczler JM, Oreffo RO (2008) Osteogenesis and angiogenesis: the potential for engineering bone. Eur Cell Mater 15: 100-114.
  42. Shao JS, Aly ZA, Lai CF, Cheng SL, Cai J, et al. (2007) Vascular Bmp Msx2 Wnt signaling and oxidative stress in arterial calcification. Ann N Y Acad Sci 1117: 40-50.
  43. Bijlsma MF, Spek CA (2010) The Hedgehog morphogen in myocardial ischemia-reperfusion injury. Exp Biol Med (Maywood) 235: 447-454.
  44. Kusano KF, Pola R, Murayama T, Curry C, Kawamoto A, et al. (2005) Sonic hedgehog myocardial gene therapy: tissue repair through transient reconstitution of embryonic signaling. Nat Med 11: 1197-1204.
  45. Queiroz KC, Tio RA, Zeebregts CJ, Bijlsma MF, Zijlstra F, et al. (2010) Human plasma very low density lipoprotein carries Indian hedgehog. J Proteome Res 9: 6052-6059.
  46. Meijer K, de Vries M, Al-Lahham S, Bruinenberg M, Weening D, et al. (2011) Human primary adipocytes exhibit immune cell function: adipocytes prime inflammation independent of macrophages. PLoS One 6: e17154.
  47. Hu H, Hilton MJ, Tu X, Yu K, Ornitz DM, et al. (2005) Sequential roles of Hedgehog and Wnt signaling in osteoblast development. Development 132: 49-60.
  48. Pospisilik JA, Schramek D, Schnidar H, Cronin SJ, Nehme NT, et al. (2010) Drosophila genome-wide obesity screen reveals hedgehog as a determinant of brown versus white adipose cell fate. Cell 140: 148-160.
  49. Nuttall ME, Shah F, Singh V, Thomas-Porch C, Frazier T, et al. (2014) Adipocytes and the regulation of bone remodeling: a balancing act. Calcif Tissue Int 94: 78-87.
  50. Rosen C, Karsenty G, MacDougald O (2012) Foreword: interactions between bone and adipose tissue and metabolism. Bone 50: 429.
  51. Schäffler A, Büchler C (2007) Concise review: adipose tissue-derived stromal cells--basic and clinical implications for novel cell-based therapies. Stem Cells 25: 818-827.
  52. Kalluri R, Neilson EG (2003) Epithelial-mesenchymal transition and its implications for fibrosis. J Clin Invest 112: 1776-1784.
  53. Choi SS, Syn WK, Karaca GF, Omenetti A, Moylan CA, et al. (2010) Leptin promotes the myofibroblastic phenotype in hepatic stellate cells by activating the hedgehog pathway. J Biol Chem 285: 36551-36560.
  54. Mimeault M, Batra SK (2007) Interplay of distinct growth factors during epithelial mesenchymal transition of cancer progenitor cells and molecular targeting as novel cancer therapies. Ann Oncol 18: 1605-1619.
  55. Deb S, Amin S, Imir AG, Yilmaz MB, Suzuki T, et al. (2004) Estrogen regulates expression of tumor necrosis factor receptors in breast adipose fibroblasts. J Clin Endocrinol Metab 89: 4018-4024.
  56. Das S, Samant RS, Shevde LA (2011) Hedgehog signaling induced by breast cancer cells promotes osteoclastogenesis and osteolysis. J Biol Chem 286: 9612-9622.
  57. Koga K, Nakamura M, Nakashima H, Akiyoshi T, Kubo M, et al. (2008) Novel link between estrogen receptor alpha and hedgehog pathway in breast cancer. Anticancer Res 28: 731-740.
  58. Milsom CC, Yu JL, Mackman N, Micallef J, Anderson GM, et al. (2008) Tissue factor regulation by epidermal growth factor receptor and epithelial-to-mesenchymal transitions: effect on tumor initiation and angiogenesis. Cancer Res 68: 10068-10076.
  59. Hwang RF, Moore TT, Hattersley MM, Scarpitti M, Yang B, et al. (2012) Inhibition of the hedgehog pathway targets the tumor-associated stroma in pancreatic cancer. Mol Cancer Res 10: 1147-1157.
  60. Spivak-Kroizman TR, Hostetter G, Posner R, Aziz M, Hu C, et al. (2013) Hypoxia triggers hedgehog-mediated tumor-stromal interactions in pancreatic cancer. Cancer Res 73: 3235-3247.
  61. Bhattacharya R, Kwon J, Ali B, Wang E, Patra S, et al. (2008) Role of hedgehog signaling in ovarian cancer. Clin Cancer Res 14: 7659-7666.
  62. Cortés J, Calvo E, Vivancos A, Perez-Garcia J, Recio JA, et al. (2014) New approach to cancer therapy based on a molecularly defined cancer classification. CA Cancer J Clin 64: 70-74.
  63. You M1, Varona-Santos J1, Singh S2, Robbins DJ2, Savaraj N3, et al. (2014) Targeting of the Hedgehog signal transduction pathway suppresses survival of malignant pleural mesothelioma cells in vitro. J Thorac Cardiovasc Surg 147: 508-516.
  64. Sicklick JK, Li YX, Jayaraman A, Kannangai R, Qi Y, et al. (2006) Dysregulation of the Hedgehog pathway in human hepatocarcinogenesis. Carcinogenesis 27: 748-757.
  65. Höpfner M, Huether A, Sutter AP, Baradari V, Schuppan D, et al. (2006) Blockade of IGF-1 receptor tyrosine kinase has antineoplastic effects in hepatocellular carcinoma cells. Biochem Pharmacol 71: 1435-1448.
  66. Höpfner M, Schuppan D, Scherübl H (2008) Treatment of gastrointestinal neuroendocrine tumors with inhibitors of growth factor receptors and their signaling pathways: recent advances and future perspectives. World J Gastroenterol 14: 2461-2473.
  67. Baradari V, Huether A, Höpfner M, Schuppan D, Scherübl H (2006) Antiproliferative and proapoptotic effects of histone deacetylase inhibitors on gastrointestinal neuroendocrine tumor cells. Endocr Relat Cancer 13: 1237-1250.
  68. Höpfner M, Schuppan D, Scherübl H (2008) Growth factor receptors and related signalling pathways as targets for novel treatment strategies of hepatocellular cancer. World J Gastroenterol 14: 1-14.
  69. Sutter AP, Maaser K, Höpfner M, Huether A, Schuppan D, et al. (2005) Cell cycle arrest and apoptosis induction in hepatocellular carcinoma cells by HMG-CoA reductase inhibitors. Synergistic antiproliferative action with ligands of the peripheral benzodiazepine receptor. J Hepatol 43: 808-816.
  70. Zagouri F, Papadimitriou CA, Dimopoulos MA, Pectasides D (2011) Molecularly targeted therapies in unresectable-metastatic gastric cancer: a systematic review. Cancer Treat Rev 37: 599-610.
  71. Ross R (1999) Atherosclerosis--an inflammatory disease. N Engl J Med 340: 115-126.
  72. Vattikuti R, Towler DA (2004) Osteogenic regulation of vascular calcification: an early perspective. Am J Physiol Endocrinol Metab 286: E686-696.
  73. O'Brien KD (2006) Pathogenesis of calcific aortic valve disease: a disease process comes of age (and a good deal more). Arterioscler Thromb Vasc Biol 26: 1721-1728.
  74. Lijnen P, Papparella I, Petrov V, Semplicini A, Fagard R (2006) Angiotensin II-stimulated collagen production in cardiac fibroblasts is mediated by reactive oxygen species. J Hypertens 24: 757-766.
  75. Tannock LR, King VL (2008) Proteoglycan mediated lipoprotein retention: a mechanism of diabetic atherosclerosis. Rev Endocr Metab Disord 9: 289-300.
  76. Bijlsma MF, Peppelenbosch MP, Spek CA (2006) Hedgehog morphogen in cardiovascular disease. Circulation 114: 1985-1991.
  77. Canfield AE, Doherty MJ, Wood AC, Farrington C, Ashton B, et al. (2000) Role of pericytes in vascular calcification: a review. Z Kardiol 89: 20-27.
  78. Collett GD, Canfield AE (2005) Angiogenesis and pericytes in the initiation of ectopic calcification. Circ Res 96: 930-938.
  79. Agrotis A (2008) Simvastatin, an inhibitor of epithelial-to-mesenchymal transition in experimental atherosclerotic renovascular disease? J Hypertens 26: 1553-1555.
  80. Urano A, Yamamoto C, Fujiwara Y, Kaji T (2008) [Proteoglycan as a key molecule in atherosclerosis progression: characteristics of the structure and regulation of the synthesis]. Yakugaku Zasshi 128: 365-375.
  81. Dunaeva M, Voo S, van Oosterhoud C, Waltenberger J (2010) Sonic hedgehog is a potent chemoattractant for human monocytes: diabetes mellitus inhibits Sonic hedgehog-induced monocyte chemotaxis. Basic Res Cardiol 105: 61-71.
  82. Moulton KS (2001) Plaque angiogenesis and atherosclerosis. Curr Atheroscler Rep 3: 225-233.
  83. Cui J, O'Shea KS, Purkayastha A, Saunders TL, Ginsburg D (1996) Fatal haemorrhage and incomplete block to embryogenesis in mice lacking coagulation factor V. Nature 384: 66-68.
  84. Bijlsma MF, Spek CA, Zivkovic D, van de Water S, Rezaee F, et al. (2006) Repression of smoothened by patched-dependent (pro-)vitamin D3 secretion. PLoS Biol 4: e232.
  85. Callejo A, Culi J, Guerrero I (2008) Patched, the receptor of Hedgehog, is a lipoprotein receptor. Proc Natl Acad Sci U S A 105: 912-917.
  86. Panáková D, Sprong H, Marois E, Thiele C, Eaton S (2005) Lipoprotein particles are required for Hedgehog and Wingless signalling. Nature 435: 58-65.
  87. Li F, Duman-Scheel M, Yang D, Du W, Zhang J, et al. (2010) Sonic hedgehog signaling induces vascular smooth muscle cell proliferation via induction of the G1 cyclin-retinoblastoma axis. Arterioscler Thromb Vasc Biol 30: 1787-1794.
  88. Li FH, Zhao Y, Wang XH, Shi D, Fu QN, et al. (2013) [The role of SHH pathway in PDGF-induced VSMC proliferation]. Zhonghua Yi Xue Za Zhi 93: 1490-1493.
  89. Martin TJ, Gillespie MT (2001) Receptor activator of nuclear factor kappa B ligand (RANKL): another link between breast and bone. Trends Endocrinol Metab 12: 2-4.
  90. Montecucco F, Steffens S, Mach F (2007) The immune response is involved in atherosclerotic plaque calcification: could the RANKL/RANK/OPG system be a marker of plaque instability? Clin Dev Immunol 2007: 75805.
  91. Sandberg WJ, Yndestad A, Øie E, Smith C, Ueland T, et al. (2006) Enhanced T-cell expression of RANK ligand in acute coronary syndrome: possible role in plaque destabilization. Arterioscler Thromb Vasc Biol 26: 857-863.
  92. Takayanagi H, Kim S, Koga T, Taniguchi T (2005) Stat1-mediated cytoplasmic attenuation in osteoimmunology. J Cell Biochem 94: 232-240.
  93. Mak KK, Bi Y, Wan C, Chuang PT, Clemens T, et al. (2008) Hedgehog signaling in mature osteoblasts regulates bone formation and resorption by controlling PTHrP and RANKL expression. Dev Cell 14: 674-688.
  94. Takamoto M, Tsuji K, Yamashita T, Sasaki H, Yano T, et al. (2003) Hedgehog signaling enhances core-binding factor a1 and receptor activator of nuclear factor-kappaB ligand (RANKL) gene expression in chondrocytes. J Endocrinol 177: 413-421.
  95. Doherty TM, Asotra K, Fitzpatrick LA, Qiao JH, Wilkin DJ, et al. (2003) Calcification in atherosclerosis: bone biology and chronic inflammation at the arterial crossroads. Proc Natl Acad Sci U S A 100: 11201-11206.
  96. Hofnagel O, Robenek H (2009) Cathepsin K: boon or bale for atherosclerotic plaque stability? Cardiovasc Res 81: 242-243.
  97. Colles SM, Maxson JM, Carlson SG, Chisolm GM (2001) Oxidized LDL-induced injury and apoptosis in atherosclerosis. Potential roles for oxysterols. Trends Cardiovasc Med 11: 131-138.
  98. Dang ZC, van Bezooijen RL, Karperien M, Papapoulos SE, Löwik CW (2002) Exposure of KS483 cells to estrogen enhances osteogenesis and inhibits adipogenesis. J Bone Miner Res 17: 394-405.
  99. Omoigui S (2007) The Interleukin-6 inflammation pathway from cholesterol to aging--role of statins, bisphosphonates and plant polyphenols in aging and age-related diseases. Immun Ageing 4: 1.
  100. Schieffer B, Selle T, Hilfiker A, Hilfiker-Kleiner D, Grote K, et al. (2004) Impact of interleukin-6 on plaque development and morphology in experimental atherosclerosis. Circulation 110: 3493-3500.
Citation: Dashty M (2014) Hedgehog Signaling Pathway is Linked with Age- Related Diseases. J Diabetes Metab 5:350.

Copyright: © 2014 Dashty M. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.