Next Article in Journal
Expression of Vitamin D Receptor (VDR) Positively Correlates with Survival of Urothelial Bladder Cancer Patients
Next Article in Special Issue
Interleukins 6 and 15 Levels Are Higher in Subcutaneous Adipose Tissue, but Obesity Is Associated with Their Increased Content in Visceral Fat Depots
Previous Article in Journal
Quantitative Proteomics Analysis of Herbaceous Peony in Response to Paclobutrazol Inhibition of Lateral Branching
Previous Article in Special Issue
Expression of PD-1 Molecule on Regulatory T Lymphocytes in Patients with Insulin-Dependent Diabetes Mellitus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Pathways Regulating Macrovascular Pathology and Vascular Smooth Muscle Cells Phenotype in Type 2 Diabetes

Department of Biomedicine and Prevention, Institute of Anatomic Pathology, Tor Vergata University of Rome, Rome 00133, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2015, 16(10), 24353-24368; https://doi.org/10.3390/ijms161024353
Submission received: 5 August 2015 / Revised: 21 September 2015 / Accepted: 8 October 2015 / Published: 14 October 2015
(This article belongs to the Special Issue Molecular Research on Obesity and Diabetes)

Abstract

:
Type 2 diabetes mellitus (T2DM) is a disease reaching a pandemic proportion in developed countries and a major risk factor for almost all cardiovascular diseases and their adverse clinical manifestations. T2DM leads to several macrovascular and microvascular alterations that influence the progression of cardiovascular diseases. Vascular smooth muscle cells (VSMCs) are fundamental players in macrovascular alterations of T2DM patients. VSMCs display phenotypic and functional alterations that reflect an altered intracellular biomolecular scenario of great vessels of T2DM patients. Hyperglycemia itself and through intraparietal accumulation of advanced glycation-end products (AGEs) activate different pathways, in particular nuclear factor-κB and MAPKs, while insulin and insulin growth-factor receptors (IGFR) are implicated in the activation of Akt and extracellular-signal-regulated kinases (ERK) 1/2. Nuclear factor-κB is also responsible of increased susceptibility of VSMCs to pro-apoptotic stimuli. Down-regulation of insulin growth-factor 1 receptors (IGFR-1R) activity in diabetic vessels also influences negatively miR-133a levels, so increasing apoptotic susceptibility of VSMCs. Alterations of those bimolecular pathways and related genes associate to the prevalence of a synthetic phenotype of VSMCs induces extracellular matrix alterations of great vessels. A better knowledge of those biomolecular pathways and related genes in VSMCs will help to understand the mechanisms leading to macrovascular alterations in T2DM patients and to suggest new targeted therapies.

1. Introduction

Diabetes mellitus is a human metabolic disease with an economic impact on medical care. Around 10% of European population is affected by type-2 diabetes mellitus (T2DM), which develops from a complex interplay between eating habits, excess body fat, genetics, and other common disorders, such as obstructive sleep apnea. In parallel with T2DM, the incidence of insulin resistance and metabolic syndrome also increases because of the epidemic diffusion of obesity among populations of the industrialized world [1,2]. Hyperglycemia defines a full-blown T2DM. T2DM-induced pathologic changes involve diffusely the vasculature, and hyperglycemia is considered a major risk factor in promoting pathological arterial remodeling and clinically-adverse events, such as stroke and acute coronary syndrome [3]. Given those complications, it is surprising that less or minimal attention has been given to T2DM-related macrovascular disease and the role of vascular smooth muscle cells (VSMCs), the predominant cell population in the arterial wall. This review will highlight the biomolecular pathways leading to VSMC phenotypic alterations and how they contribute to pathological macrovascular remodeling in T2DM patients.

2. Heterogeneity of Vascular Smooth Muscle Cells and T2DM

VSMCs in the healthy tunica media are phenotypically heterogeneous and characterized by a variable switch from a “contractile” to a dedifferentiated phenotype [4,5,6]. Contractile or differentiated phenotypes, typical of VSMCs of the normal vessels, exhibits a highly organized cytoskeleton, with defined F-actin filaments that maintain contractile function, and high levels of α-smooth muscle actin, smooth muscle myosin heavy chain and h1-calponin [7]. During the cardiovascular disorders, VSMC cytoskeletal reorganization leads to the prevalence of a synthetic phenotype. Synthetic VSMCs display alterations in organelle distribution, aberrant matrix metabolism, increased proliferation and migration [8], and expression of specific glycoproteins [9]. Key characteristics of dedifferentiation of VSMCs are nuclear enlargement, increased ribosomal content, and enlarged Golgi apparatus [10,11]. Changes of VSMC behavior are phenotypically-regulated and lead to receptor activation that regulates proliferation, migration, and survival. Functionally, VSMC dedifferentiation impairs proliferative and migratory capacity through an increased sensitivity to growth factor stimulation and mitogenic factors [10,12]. Changes in VSMC phenotype also critically influence the response to pharmacologically-active substances [13,14,15,16]. Differences in VSMC phenotype can be easily identified in vitro. Two cell populations can be isolated from post-injury rat aorta and carotid artery: a spindle-shaped phenotype with the classic “hill-and-valley” growth pattern, typical of cultured VSMCs, and an epithelioid phenotype growing in a monolayer with cobblestone morphology that are isolated from the neointimal tissue after balloon injury [17,18]. Epithelioid VSMCs display a different response to growth factors [18,19]. The differentiated and synthetic VSMCs phenotypes are not stable and they can switch in culture, under particular conditions, such as growth factors (i.e., platelet-derived growth factors-BB (PDGF-BB), fibroblast growth factor-2 (FGF-2), transforming growth factor-β (TGF-β)) [19,20,21], or matrix molecules as collagen [22]. The metabolic milieu of insulin resistance and T2DM can progress for decades before clinical diagnosis [23]. During this period, hyperglycemia and hyperinsulinemia exert a direct effect on vascular cells, potentially causing detrimental changes in their phenotype and function (Figure 1). Increased susceptibility to cardiovascular diseases in T2DM patients suggests that a pathological phenotype of VSMCs is worthy of detailed study [24]. VSMCs from primary cultures from T2DM vessels were morphologically distinct from non-diabetic VSMCs. In particular, arterial and venous VSMCs from T2DM patients lose the typical “hill-and-valley” spindle-shaped appearance and adopt a more rhomboid phenotype [25]. VSMCs from diabetic patients show a significant increase of proliferation, adhesion and contact inhibition, associated with an increase of atheromatous process and restenosis [25]. Intimal hyperplasia appears closely linked to the synthetic phenotype of VSMCs and must be considered in order to counteract atherosclerosis and restenosis in T2DM patients [25,26].
Figure 1. Aortopathy and diabetes: microscopic aspects. (A) Haematoxylin-Eosin-stained section of aortic aneurism of diabetic patient shows reduced cellularity and increased extracellular matrix accumulation; Immunohistochemical staining with 3,3-diaminobenzidine for (B) α-smooth actin and (C) smooth muscle myosin show variability of expression and shape of normal aortic VSMCs; (D) Ultrathin sections were counterstained with uranyl acetate and lead citrate and photographed by using a Hitachi 7100 transmission electron microscope, showing medial VSMCs with irregular nucleus surrounded from abundant extracellular matrix. Original magnification: (AC), 400×; and (D), 4500×.
Figure 1. Aortopathy and diabetes: microscopic aspects. (A) Haematoxylin-Eosin-stained section of aortic aneurism of diabetic patient shows reduced cellularity and increased extracellular matrix accumulation; Immunohistochemical staining with 3,3-diaminobenzidine for (B) α-smooth actin and (C) smooth muscle myosin show variability of expression and shape of normal aortic VSMCs; (D) Ultrathin sections were counterstained with uranyl acetate and lead citrate and photographed by using a Hitachi 7100 transmission electron microscope, showing medial VSMCs with irregular nucleus surrounded from abundant extracellular matrix. Original magnification: (AC), 400×; and (D), 4500×.
Ijms 16 24353 g001

3. T2DM-Induced Macrovascular Disease: Different Players

Hyperglycemia is a major risk factor for arterial diseases and leads to vascular pathology through multiple processes. High glucose concentrations activate nuclear factor-κB (NF-κB) [27,28]. The latter modulates several pro-inflammatory and pro-atherosclerotic target genes in VSMCs, endothelial cells (ECs) and macrophages. NF-κB activation is essential for early events during the progression of atherosclerotic lesions, in particular the adhesion of circulating monocytes to ECs and the transmigration in the subendothelial space. In fact, hyperglycemia-induced monocyte adhesion to cultured aortic ECs in vitro by NF-κB activation increased the expression of pro-inflammatory genes and endothelial adhesion molecules, in particular vascular cell adhesion molecule 1 (VCAM-1) and monocyte chemoattractant protein-1 (MCP-1) [29,30]. MCP-1 is an important chemotactic factor for monocytes, mainly secreted by ECs, macrophages and adipocytes [31]. MCP-1 and its receptor CC chemokine receptors2 (CCR2) are important players in the monocyte recruitment. MCP-1 and CCR2 are likewise important for the onset of cardiovascular pathology and the negative effect of these circulating factors induces EC dysfunction, adhesion of monocytes and subsequent vascular remodeling with foam cells accumulation and plaque development. Expression of adhesion molecules and MCP-1 characterizes the inflammatory response of ECs in vitro [32,33] and appears up-regulated in aged great vessels [34]. After migration into the subendothelial space, monocytes differentiate into macrophages that are increased in the artery wall of diabetic patients [35]. Experimental data indicated that in response to hyperglycemia, macrophages can also locally proliferate [36]. Increased NF-κB activity also characterizes VSMC exhibiting a synthetic phenotype [16] and NF-κB inhibition promotes VSMC apoptosis [37]. NF-κB also regulates the pro-apoptotic response of intimal VSMCs to neuromediators, as pro-nerve growth factor [38]. Mitochondrial nicotinamide adenine dinucleotide phosphate oxidase (NADPH) activity, in particular the Nox4 isoform, is a dominant mechanism for endothelial reactive oxygen species (ROS) production [32,39]. Endothelial cells under high levels of glucose showed an increase expression of Nox2 and Nox4 isoform, and greater Rac-1 activity; the latter is a small G protein, important for intracellular NADPH oxidase assembly and activation [40]. T2DM induced Rac-1 activity through its translocation from cytoplasm to plasma membrane, promoting NADPH oxidase subunit assembly [41]. The latter is a necessary mechanism to trigger the ROS-generating NADPH oxidase enzymatic system. In diabetic mice vessels it was found an increased Rac-1 and NADPH oxidase activity, demonstrating that molecular processes leading to NADPH oxidase-dependent generation of ROS in T2DM implicate Rac-1 activation [41]. So, oxidative stress-induced vascular endothelial dysfunction and Rac-1 activity depend on hyperglycemic conditions of diabetic patients [41]. The selective inhibition of Rac-1, blocking a key step in oxidative stress generation, could reduce hyperglycemia-induced vascular damage and consequently the cardiovascular risk in T2DM [41]. Together, hyperglycemia and ROS accumulation inhibit the endothelial production of nitric oxide, leading to endothelial-dependent relaxation dysfunction that characterizes vascular pathology [42,43]. In diabetic patients, ROS accumulation contributes to LDL modification and oxidation [44]. Epidemiologic data indicate that in human subjects, LDL are the most atherogenic lipoproteins, particularly susceptible to oxidative modifications from other atherogenic risk, such as cigarette smoke, diabetes, and insulin resistance [45]. In diabetic patients, insulin resistance also promotes the progression of plaque to vulnerable lesions [46]. Lipid accumulation further stimulates recruitment of macrophages and other inflammatory cells [47], contributing to a state of vascular inflammation. The latter induces intimal VSMC accumulation, which display macrophagic activity and increased synthetic activity with extracellular collagen deposition [48] in a sort of accelerated vascular aging [49,50]. Diabetes-induced vascular alterations share same aspects with age-related macroangiopathy, and both represent a risk factor for atherosclerosis [49,51]. Aging potentiates other atherogenic risk factors, including diabetes [52] and VSMCs, also drive the age-related vascular changes [53]. The age-related diffuse intimal thickening in great vessels the principal site for the progression of atherosclerotic lesions [49,51]. VSMCs from old donors show increased expression of stem markers [54]. Vascular expression of flt-1+ and c-kit+ influences VSMC properties [55]. In particular, flt-1 signaling regulates NF-κB-mediated cell survival [56], in line with the hypothesis that VSMCs with a stem phenotype promote arterial remodeling [54,57]. Since it has been hypothesized that circulatory precursors can be beneficial in the stabilization of plaques [58]. It is suggestive to hypothesize that circulating and resident cells with stem phenotype play a different role in aortic remodeling observed in T2DM patients [59].

4. VSMCs and T2DM-Related Atherogenic Risk Factors

Similar to that observed during the development of atherosclerotic lesions, VSMCs undergo a variety of T2DM changes and display a different phenotype in great vessels. A better knowledge of factors regulating VSMC phenotype is crucial to suggest new targeted therapies to prevent aortopathy in T2DM patients.

4.1. Hyperglycemia

High glucose concentrations induce VSMC proliferation and play a pivotal role in the progression of diabetic vascular alterations [60]. It appears relevant to investigate the molecular mediators in response to increased blood high glucose levels as a potential therapeutic target. Interferon regulatory factor-1 (IRF-1) is a molecular mediator of vascular diseases. Several studies provided evidences that IRF-1 reduces vascular cell growth under normal glucose conditions, whereas at high concentrations IRF-1 promotes VSMC proliferation [61]. Other studies indicated that ROS accumulation influences proliferation by activating cyclin/CDK [62]. ROS accumulation also induces endothelial dysfunction [32,39]. High glucose-induced stimulation increases intracellular ROS and stimulates the activation of ERK 1/2, a mitogen-activated protein kinase essential for VSMC growth [63]. IRF-1 over-expression promotes ERK-1 activation under high-glucose condition. It is likely that the IRF-1-mediated pathway sustains hyperglycemia-dependent VSMC proliferation [63]. Elevated glucose concentration also stimulate extracellular matrix synthesis and accumulation [64], likely mediated by the activity of TGF-β [65] and its downstream mediator the connective tissue growth factor, that regulates vascular fibrosis [65]. High glucose levels increased connective tissue growth factor protein and mRNA in VSMCs and its deletion by siRNA inhibited the high glucose-induced VSMC proliferation [66]. The involvement of connective tissue growth factor in high glucose-induced proliferation suggests that its down-regulation may prevent abnormal VSMC growth in the vessels of diabetic patients.

4.2. Advanced Glycation End-Product

Several hypotheses aim to explain the increase of VSMC proliferation in pathological vessels. Proteins and lipids exposed to high glucose levels generate advanced glycation end-products (AGEs) as protein cross-links, flurophors, and low molecular-weight residues [42]. AGE accumulation induces the increased production of ROS. ROS accumulation plays a central role in vascular disease, including atherosclerosis and restenosis. AGE accumulation activated NF-κB in many cell types, in particular in VSMCs [67]. Moreover, AGEs also induce mitogen activated protein kinase (MAPKs) activation [68]. The role of AGE receptors (RAGEs) has been also investigated. RAGEs and galectin-3 have been linked to the progression of atherosclerosis [69,70]. VSMCs proliferation is likely mediated via galectin-3 and abnormal AGE-galectin-3 interaction has been linked to macroangiopathy in T2DM patients [71].

4.3. Hyperinsulinemia and Insulin Growth Factor

Hyperinsulinemia is an important factor for the plaque formation in T2DM patients. Insulin displayed mitogenic effects on human aortic VSMCs [72] and insulin growth factor-1 (IGF-1) activated insulin-dependent proliferation of human VSMC [73] through various pathways, including MAPK signaling [74], that in turn increases human VSMC chemotaxis [75]. IGF-1 has been also documented to influence VSMC survival [76]. IGF-1 binds to its receptor IGF-1R with high affinity, leading to the activation of IGF-1R tyrosine kinase, which in turn induces pro-survival and growth signals. Insulin resistance of VSMCs in T2DM patients is linked to angiotensin II-mediated vasculopathy [77]. In the vascular system, NO activity is able to antagonize angiotensin II. The latter is able to stimulate ROS production [77].However, the relationship between angiotensin II and insulin resistance has not been established. A prolonged oxidative stress and increased angiotensin II may cause insulin resistance thorough ROS-mediated activation of insulin receptor substrate-1 (IRS-1) of VSMCs. IRS-1 phosphorylation reduced phosphatidylinositol 3-kinase activity (PI3K) and abolished insulin-induced activation of Akt [77]. This effect induces the decrease of GLUT4 (glucose transporter type 4) translocation to the plasma membrane, which causes a reduced glucose uptake in VSMCs, demonstrating that insulin-mediated glucose uptake in VSMCs is linked to IRS-1/PI3K/Akt pathway, similarly to other insulin-sensitive tissues [78]. In addition, increased ROS and angiotensin II production are responsible for the increased VSMC proliferation, vascular inflammation and extracellular matrix deposition [79]. Persistent exposure to IGF-1 stimulates growth and migration of VSMCs. In advanced plaques, intimal VSMC hyperplasia may reduce the plaque vulnerability to rupture and it helps to stabilize atherosclerotic lesions [80].Biological effects of insulin on VSMCs are mediated by IGF-1R [81]. IGF-1R expression was down-regulated in aortas of diabetic mice, likely as the consequence an alteration in insulin responsiveness [82]. When IGF-1R is down-regulated, insulin induces Akt and ERK 1/2 phosphorylation as well as glucose uptake [82]. Akt and ERK 1/2 play a relevant regulatory role on survival and vasculogenic properties of human adipose tissue-derived stem cells [33,83]. Adventitial cells may be the source of stem cells with vasculogenic properties [17]. Enhanced insulin signaling in VSMCs results in the suppression of TNF-α-induced NF-κB activation and MCP-1 expression [82]. These results suggest that manipulation of IGF-1R activity is a therapeutic target to improve insulin sensitivity and also indicate that insulin plays a potentially anti-inflammatory role in the vasculature.

5. MicroRNAs and Diabetic Macrovascular Pathology

MicroRNAs (miRs) represent a class of new-coding small RNA species that promote mRNA cleavage and/or translational repression through base-paring to the 3ʹ untranslated region (UTR) of target mRNAs [84]. Recently miR-133a has been reported to be expressed in VSMCs and to regulate IGF-1R expression [58]. In the aorta, miR-133a was reduced in ApoE−/− mice [58]. It is expected that VSMCs may be susceptible to apoptosis when the expression of the inhibitor of apoptosis IGF-1R, is compromised, likely the consequence of low miR-133a level in VSMCs of atherosclerotic aortas [58]. MiR-133a/IGF-1R promotion of VSMC proliferation and differentiation may help to prevent plaque rupture and adverse clinical events [58]. IGF-1 increases the expression of ribonucleoprotein domain family member 6 (LARP6) in VSMCs, resulting in an increased binding of LARP6 to COL1a1 and COL1a2 mRNAs, with enhanced synthesis of collagen type I and extracellular matrix accumulation of mature collagen by promoting fibril maturation, with a more stable plaque phenotype [85]. These data indicate that the regulatory mechanisms of IGF-1 during the development of atherosclerotic plaques are quite different from those of macrovascular pathology in T2DM patients. Hyperglycemia can work in synergy with hyperinsulinemia and other factors to promote atherosclerotic plaque progression in diabetic patients. Growth-promoting effects of high glucose level on VSMCs have been described in human and rat aorta and human umbilical artery cultures [24,86,87]. These results were not confirmed in experiments using porcine SMCs, where high glucose levels did not stimulate proliferation neither via synergism with other SMC mitogens, such as PDGF [24,88].

6. Adipose Tissue and Diabetes-Related Macrovascular Dysfunction

Adipose tissue has a central role in the progression of a systemic low-grade inflammatory condition that favors vascular dysfunction in obese subjects. The local production of adipokines and chemokines by fat cells suggests a novel interplay between obesity and vascular pathology [89]. In addition, chemotactic adipokines modulate the immune cell infiltration in the adipose tissue stimulating atherosclerotic process [90]. Accumulating evidence showed that adipose tissue cells secrete adipokines and favor the development of cardiovascular complications in T2DM patients, an increase of superoxide affects cardiomyocytes and VSMC function [91]. However, adiponectin plasma level was significantly lower in adult subjects with obesity [92] and T2DM [93]. Adipose tissue-secreted adipokines promote vascular dysfunction associated with obesity and vascular pathology. Interestingly, vascular endothelial growth factor (VEGF) released from visceral adipose tissue, seemed linked to increased VSMC proliferation [94]. VEGF activity is mediated by specific receptors, VEGFR-1 and VEGFR-2 [95,96]. Increased expression of VEGFR-1 is associated to phenotype changes of VSMCs and influences biological properties and pharmacological response [54,56,97,98]. Adiponectin also induces NO production by increasing of endothelial nitric oxide synthase (eNOS) phosphorylation [99]; eNOS is critically involved in microvascular dysfunction, since it induces overproduction and release of O2 [100]. Diabetes or obesity are also associated to increased levels of free fatty acids, that increase intracellular diacylglycerol concentration in several tissues, including renal glomeruli from diabetic animals and VSMCs. Intracellular diacylglycerol promotes membrane-associated protein kinase C activity that leads to increased expression of cyclooxygenase and vasoconstrictor thromboxane A2, whereas vasodilator prostacyclin production decreases [101]. Overall, those mechanisms accelerate vascular dysfunction in people with obesity-associated insulin resistance and in diabetes.

7. Conclusions

During the last years, many studies have been focused on the investigation of molecular mechanisms regulating VSMC phenotype in pathological vessels. Hyperglycemia and hyperinsulinemia induce changes of VSMC phenotype that is critical for the progression of T2DM macrovascular pathology. Investigation of alterations of VSMCs phenotype in T2DM patients allowed the recognition of specific intracellular pathways supporting pathological macrovascular remodeling, in particular NF-κB, Akt and ERK-related genes, as summarized in Figure 2. Moreover, miR-RNAs represent a promising field of research to discover new mechanism of VSMC homeostasis in T2DM pathological vessels. The next step will be to discover therapeutic agents capable to correct the activation of T2DM-related biomolecular pathways in VSMCs in order to prevent macrovascular pathology.
Figure 2. Phenotypic switch of VSMCs in diabetic aortopathy. Schematic representation of diabetes-related intracellular signaling involved in the switch from contractile to synthetic phenotype of vascular smooth muscle cells (VSMCs) in great vessels.
Figure 2. Phenotypic switch of VSMCs in diabetic aortopathy. Schematic representation of diabetes-related intracellular signaling involved in the switch from contractile to synthetic phenotype of vascular smooth muscle cells (VSMCs) in great vessels.
Ijms 16 24353 g002

Acknowledgments

We thank Sabrina Cappelli and Antonio Volpe for the technical work.

Author Contributions

Sara Casella: writing the manuscript; Alessandra Bielli: writing the manuscript; Alessandro Mauriello: writing and revision the manuscript; Augusto Orlandi: financial support, administrative support, writing and final approval of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Yach, D.; Stucler, D.; Brownell, K.D. Epidemiologic and economic consequence of the global epidemics of obesity and diabetes. Nat. Med. 2006, 12, 62–66. [Google Scholar] [CrossRef] [PubMed]
  2. Cory, S.; Ussery-Hall, A.; Griffin-Blake, S.; Easton, A.; Vigeant, J.; Balluz, L.; Garvin, W.; Greenlund, K. Prevalence of selected risk behaviors and chronic disease and conditions—Step communities, United States, 2006–2007. MMWR Surveill. Summ. 2010, 59, 1–3. [Google Scholar] [PubMed]
  3. Orasanu, G.; Plutzky, J. The pathologic continuum of diabetic vascular disease. J. Am. Coll. Cardiol. 2009, 53, S35–S42. [Google Scholar] [CrossRef] [PubMed]
  4. Gabbiani, G.; Kocher, O.; Bloom, W.S.; Vandekerckhove, J.; Weber, K. Actin expression in smooth muscle cells of rat aortic intimal thickening, human atheromatous plaque, and cultured rat aortic media. J. Clin. Investig. 1984, 73, 148–152. [Google Scholar] [CrossRef] [PubMed]
  5. Campbell, G.R.; Campbell, J.H. Smooth muscle phenotypic changes in arterial wall homeostasis: Implications for the pathogenesis of atherosclerosis. Exp. Mol. Pathol. 1985, 42, 139–162. [Google Scholar] [CrossRef]
  6. Walker, L.N.; Bowen-Pope, D.F.; Ross, R.; Reidy, M.A. Production of platelet-derived growth factor-like molecules by cultured arterial smooth muscle cells accompanies proliferation after arterial injury. Proc. Natl. Acad. Sci. USA 1986, 83, 7311–7315. [Google Scholar] [CrossRef] [PubMed]
  7. Alexander, M.R.; Owens, G.K. Epigenetic control of smooth muscle cell differentiation and phenotypic switching in vascular development and disease. Annu. Rev. Physiol. 2012, 74, 13–40. [Google Scholar] [CrossRef] [PubMed]
  8. Owens, G.K.; Kumar, M.S.; Wamhoff, B.R. Molecular regulation of vascular smooth muscle cell differentiation in development and disease. Physiol. Rev. 2004, 84, 767–801. [Google Scholar] [CrossRef] [PubMed]
  9. Orlandi, A.; Pucci, S.; Ciucci, A.; Pichiorri, F.; Ferlosio, A.; Spagnoli, L.G. Modulation of clusterin isoforms is associated with all-trans retinoic acid-induced proliferative arrest and apoptosis of intimal smooth muscle cells. Arterioscler. Thromb. Vasc. Biol. 2005, 25, 348–353. [Google Scholar] [CrossRef] [PubMed]
  10. Thyberg, J.; Moskalewski, S. Microtubules and the organization of the Golgi complex. Exp. Cell Res. 1985, 159, 1–16. [Google Scholar] [CrossRef]
  11. Thyberg, J.; Blomgren, K.; Hedin, U.; Dryjski, M. Phenotypic modulation of smooth muscle cells during the formation of neointimal thickenings in the rat carotid artery after balloon injury: An electron-microscopic and stereological study. Cell Tissue Res. 1995, 281, 421–433. [Google Scholar] [CrossRef] [PubMed]
  12. Orlandi, A.; Calzetta, L.; Doldo, E.; Tarquini, C.; Matera, M.G.; Passeri, D. Brain natriuretic peptide modulates calcium homeostasis and epidermal growth factor receptor gene signalling in asthmatic airways smooth muscle cells. Pulm. Pharmacol. Ther. 2015, 31, 51–54. [Google Scholar] [CrossRef] [PubMed]
  13. Ferri, N.; Arnaboldi, L.; Orlandi, A.; Yokoyama, K.; Gree, R.; Granata, A.; Hachem, A.; Paoletti, R.; Gelb, M.H.; Corsini, A. Effect of S(–) perillic acid on protein prenylation and arterial smooth muscle cell proliferation. Biochem. Pharmacol. 2001, 62, 1637–1645. [Google Scholar] [CrossRef]
  14. Orlandi, A.; Marcellini, M.; Pesce, D.; Calvani, M.; Spagnoli, L.G. Propionyl-l-carnitine reduces intimal hyperplasia after injury in normocholesterolemic rabbit carotidartery by modulating proliferation and caspase 3-dependent apoptosis of vascular smooth muscle cells. Atherosclerosis 2002, 160, 81–89. [Google Scholar] [CrossRef]
  15. Orlandi, A.; Spagnoli, L.G.; Marino, B.; Mauriello, A.; de Angelis, C.; Ramacci, M.T. Propionyl-l-carnitine prevents the progression of atherosclerotic lesions in aged hyperlipemic rabbits. Atherosclerosis 1995, 114, 29–44. [Google Scholar]
  16. Orlandi, A.; Francesconi, A.; Cocchia, D.; Corsini, A.; Spagnoli, L.G. Phenotypic heterogeneity influences apoptotic susceptibility to retinoic acid and cis-platinum of rat arterial smooth muscle cells in vitro: Implications for the evolution of experimental intimal thickening. Arterioscler. Thromb. Vasc. Biol. 2001, 21, 1118–1123. [Google Scholar] [CrossRef] [PubMed]
  17. Orlandi, A.; Bennett, M. Progenitor cell-derived smooth muscle cells in vascular disease. Biochem. Pharmacol. 2010, 79, 1706–1713. [Google Scholar] [CrossRef] [PubMed]
  18. Orlandi, A.; Ehrlich, H.P.; Ropraz, P.; Spagnoli, L.G.; Gabbiani, G. Rat aortic smooth muscle cells isolated from different layers and at different times after endothelial denudation show distinct biological features in vitro. Arterioscler. Thromb. 1994, 14, 982–989. [Google Scholar] [CrossRef] [PubMed]
  19. Orlandi, A.; Ropraz, P.; Gabbiani, G. Proliferative activity and α-smooth muscle actin expression in cultured rat aortic smooth muscle cells are differently modulated by transforming growth factor-β1 and heparin. Exp. Cell Res. 1994, 214, 528–536. [Google Scholar] [CrossRef] [PubMed]
  20. Hao, H.; Ropraz, P.; Verin, V.; Camenzind, E.; Geinoz, A.; Pepper, M.S.; Gabbiani, G.; Bochaton-Piallat, M.L. Heterogeneity of smooth muscle cell populations cultured from pig coronary artery. Arterioscler. Thromb. Vasc. Biol. 2002, 22, 1093–1099. [Google Scholar] [CrossRef] [PubMed]
  21. Li, S.; Sims, S.; Jiao, Y.; Chow, L.H.; Pickering, J.G. Evidence from a novel human cell clone that adult vascular smooth muscle cells can convert reversibly between noncontractile and contractile phenotypes. Circ. Res. 1999, 85, 338–348. [Google Scholar] [CrossRef] [PubMed]
  22. Orlandi, A.; Ferlosio, A.; Gabbiani, G.; Spagnoli, L.G.; Ehrlich, P.H. Phenotypic heterogeneity influences the behavior of rat aortic smooth muscle cells in collagen lattice. Exp. Cell Res. 2005, 311, 317–327. [Google Scholar] [CrossRef] [PubMed]
  23. Forst, T.; Hohberg, C.; Pfutzner, A. Cardiovascular effects of disturbed insulin activity in metabolic syndrome and in type 2 diabetic patients. Horm. Metab. Res. 2009, 41, 123–131. [Google Scholar] [CrossRef] [PubMed]
  24. Porter, K.E.; Riches, K. The vascular smooth muscle cell: A therapeutic target in type 2 diabetes? Clin. Sci. 2013, 125, 167–182. [Google Scholar] [CrossRef] [PubMed]
  25. Faries, P.L.; Rohan, D.I.; Takahara, H.; Wyers, M.C.; Contreras, M.A.; Quist, W.C.; King, G.L.; Logerfo, F.W. Human vascular smooth muscle cells of diabetic origin exhibit increased proliferation, adhesion, and migration. J. Vasc. Surg. 2001, 33, 601–607. [Google Scholar] [CrossRef] [PubMed]
  26. Madi, H.A.; Riches, K.; Warburton, P.; O’Regan, D.J.; Turner, N.A.; Porter, K.E. Inherent differences in morphology, proliferation, and migration in saphenous vein smooth muscle cells cultured from nondiabetic and type 2 diabetic patients. Am. J. Physiol. Cell Physiol. 2009, 297, 1307–1317. [Google Scholar] [CrossRef] [PubMed]
  27. Piga, R.; Naito, Y.; Kokura, S.; Handa, O.; Yoshikawa, T. Short-term high glucose exposure induces monocyte-endothelial cells adhesion and transmigration by increasing VCAM-1 and MCP-1 expression in human aortic endothelial cells. Atherosclerosis 2007, 193, 328–334. [Google Scholar] [CrossRef] [PubMed]
  28. Yan, S.D.; Schmidt, A.M.; Anderson, G.M.; Zhang, J.; Brett, J.; Zou, Y.S.; Pinsky, D.; Stern, D. Enhanced cellular oxidant stress by the interaction of advanced glycation end products with their receptors/binding proteins. J. Biol. Chem. 1994, 269, 9889–9897. [Google Scholar] [PubMed]
  29. Otsuka, A.; Azuma, K.; Iesaki, T.; Sato, F.; Hirose, T.; Shimizu, T.; Tanaka, Y.; Daida, H.; Kawamori, R.; Watada, H. Temporary hyperglycaemia provokes monocyte adhesion to endothelial cells in rat thoracic aorta. Diabetologia 2005, 48, 2667–2674. [Google Scholar] [CrossRef] [PubMed]
  30. Scioli, M.G.; Bielli, A.; Arcuri, G.; Ferlosio, A.; Orlandi, A. Ageing and microvasculature. Vasc. Cell 2014, 6, 19. [Google Scholar] [CrossRef] [PubMed]
  31. Suzuki, M.; Tada, A.; Kanmani, P.; Watanabe, H.; Aso, H.; Suda, Y.; Nochi, T.; Miyazawa, K.; Yoda, K.; He, F.; et al. Advanced application of porcine intramuscular adipocytes for evaluating anti-adipogenic and anti-inflammatory activities of immunobiotics. PLoS ONE 2015, 10, e0119644. [Google Scholar] [CrossRef] [PubMed]
  32. Stasi, M.A.; Scioli, M.G.; Arcuri, G.; Mattera, G.G.; Lombardo, K.; Marcellini, M.; Riccioni, T.; de Falco, S.; Pisano, C.; Spagnoli, L.G.; et al. Propionyl-l-carnitine improves postischemic blood flow recovery and arteriogenetic revascularization and reduces endothelial NADPH-oxidase 4-mediated superoxide production. Arterioscler. Thromb. Vasc. Biol. 2010, 30, 426–435. [Google Scholar] [CrossRef] [PubMed]
  33. Scioli, M.G.; Cervelli, V.; Arcuri, G.; Gentile, P.; Doldo, E.; Bielli, A.; Bonanno, E.; Orlandi, A. High insulin-induced down-regulation of Erk-1/IGF-1R/FGFR-1 signaling is required for oxidative stress-mediated apoptosis of adipose-derived stem cells. J. Cell. Physiol. 2014, 229, 2077–2087. [Google Scholar] [CrossRef] [PubMed]
  34. Orlandi, A.; Francesconi, A.; Marcellini, M.; Ferlosio, A.; Spagnoli, L.G. Role of ageing and coronary atherosclerosis in the development of cardiac fibrosis in the rabbit. Cardiovasc. Res. 2004, 64, 544–552. [Google Scholar] [CrossRef] [PubMed]
  35. Venugopal, S.K.; Devaraj, S.; Yang, T.; Jialal, I. α-Tocopherol decreases superoxide anion release in human monocytes under hyperglycemic conditions via inhibition of protein kinase C-α. Diabetes 2002, 51, 3049–3054. [Google Scholar] [CrossRef] [PubMed]
  36. Spagnoli, L.G.; Orlandi, A.; Santeusanio, G. Foam cells of the rabbit atherosclerotic plaque arrested in metaphase by colchicine show a macrophage phenotype. Atherosclerosis 1991, 88, 87–92. [Google Scholar] [CrossRef]
  37. Orlandi, A.; Francesconi, A.; Marcellini, M.; di Lascio, A.; Spagnoli, L.G. Propionyl-l-carnitine reduces proliferation and potentiates Bax-related apoptosis of aortic intimal smooth muscle cells by modulating nuclear factor-κB activity. J. Biol. Chem. 2007, 282, 4932–4942. [Google Scholar] [CrossRef] [PubMed]
  38. Campagnolo, L.; Costanza, G.; Francesconi, A.; Arcuri, G.; Moscatelli, I.; Orlandi, A. Sortilin expression is essential for pro-nerve growth factor-induced apoptosis of rat vascular smooth muscle cells. PLoS ONE 2014, 9, e84969. [Google Scholar] [CrossRef] [PubMed]
  39. Scioli, M.G.; Bielli, A.; Agostinelli, S.; Tarquini, C.; Arcuri, G.; Ferlosio, A.; Costanza, G.; Doldo, E.; Orlandi, A. Antioxidant treatment prevents serum deprivation- and TNF-α-induced endothelial dysfunction through the inhibition of NADPH oxidase 4 and the restoration of β-oxidation. J. Vasc. Res. 2014, 51, 327–337. [Google Scholar] [PubMed]
  40. Abo, A.; Pick, E.; Hall, A.; Totty, N.; Teahan, C.G.; Segal, A.W. Activation of the NADPH oxidase involves the small GTP-binding protein p21rac1. Nature 1991, 353, 668–670. [Google Scholar] [CrossRef] [PubMed]
  41. Vecchione, C.; Aretini, A.; Marino, G.; Bettarini, U.; Poulet, R.; Maffei, A.; Sbroggiò, M.; Pastore, L.; Gentile, M.T.; Notte, A.; et al. Selective Rac-1 inhibition protects from diabetes-induced vascular injury. Circ. Res. 2006, 98, 218–225. [Google Scholar] [CrossRef] [PubMed]
  42. Cohen, R.A. Role of nitric oxide in diabetic complications. Am. J. Ther. 2005, 12, 499–502. [Google Scholar] [CrossRef] [PubMed]
  43. De Caterina, R.; Libby, P.; Peng, H.B.; Thannickal, V.J.; Rajavashisth, T.B.; Gimbrone, M.A.; Shin, W.S.; Liao, J.K. Nitric oxide decreases cytokine-induced endothelial activation. Nitric oxide selectively reduces endothelial expression of adhesion molecules and proinflammatory cytokines. J. Clin. Investig. 1995, 96, 60–68. [Google Scholar] [CrossRef] [PubMed]
  44. Teshima, Y.; Takahashi, N.; Nishio, S.; Saito, S.; Kondo, H.; Fukui, A.; Aoki, K.; Yufu, K.; Nakagawa, M.; Saikawa, T. Production of reactive oxygen species in the diabetic heart. Roles of mitochondria and NADPH oxidase. Circ. J. 2014, 78, 300–306. [Google Scholar] [CrossRef] [PubMed]
  45. Tabas, I. Macrophage death and defective inflammation resolution in atherosclerosis. Nat. Rev. Immunol. 2010, 10, 36–46. [Google Scholar] [CrossRef] [PubMed]
  46. Tabas, I. The role of endoplasmic reticulum stress in the progression of atherosclerosis. Circ. Res. 2010, 107, 839–850. [Google Scholar] [CrossRef] [PubMed]
  47. Lundberg, B.; Suominen, L. Accumulation and mobilization of cholesteryl esters in cultured human fibroblasts exposed to free cholesterol-rich phospholipid vesicles. Atherosclerosis 1985, 56, 345–358. [Google Scholar] [CrossRef]
  48. Liu, X.; Luo, F.; Pan, K.; Wu, W.; Chen, H. High glucose upregulates connective tissue growth factor expression in human vascular smooth muscle cells. BMC Cell Biol. 2007, 8, 1. [Google Scholar] [CrossRef] [PubMed]
  49. Spagnoli, L.G.; Orlandi, A.; Mauriello, A.; Santeusanio, G.; de Angelis, C.; Lucreziotti, R.; Ramacci, M.T. Aging and atherosclerosis in the rabbit: 1. Distribution, prevalence and morphology of atherosclerotic lesions. Atherosclerosis 1991, 89, 11–24. [Google Scholar] [CrossRef]
  50. Orlandi, A.; Mauriello, A.; Marino, B.; Spagnoli, L.G. Age-related modifications of aorta and coronaries in the rabbit: A morphological and morphometrical assessment. Arch. Gerontol. Geriatr. 1993, 17, 37–53. [Google Scholar] [CrossRef]
  51. Orlandi, A.; Marcellini, M.; Spagnoli, L.G. Aging influences development and progression of early aortic atherosclerotic lesions in cholesterol-fed rabbits. Arterioscler. Thromb. Vasc. Biol. 2000, 20, 1123–1136. [Google Scholar] [CrossRef] [PubMed]
  52. Spagnoli, L.G.; Mauriello, A.; Orlandi, A.; Sangiorgi, G.; Bonanno, E. Age-related changes affecting atherosclerotic risk. Potential for pharmacological intervention. Drugs Aging 1996, 8, 275–298. [Google Scholar] [CrossRef] [PubMed]
  53. Orlandi, A.; Bochaton-Piallat, M.L.; Gabbiani, G.; Spagnoli, L.G. Aging, smooth muscle cells and vascular pathobiology: Implications for atherosclerosis. Atherosclerosis 2006, 188, 221–230. [Google Scholar] [CrossRef] [PubMed]
  54. Ferlosio, A.; Arcuri, G.; Doldo, E.; Scioli, M.G.; de Falco, S.; Spagnoli, L.G.; Orlandi, A. Age-related increase of stem marker expression influences vascular smooth muscle cell properties. Atherosclerosis 2012, 224, 51–57. [Google Scholar] [CrossRef] [PubMed]
  55. Orlandi, A.; di Lascio, A.; Francesconi, A.; Scioli, M.G.; Arcuri, G.; Ferlosio, A.; Spagnoli, L.G. Stem cell marker expression and proliferation and apoptosis of vascular smooth muscle cells. Cell Cycle 2008, 7, 3889–3897. [Google Scholar] [CrossRef] [PubMed]
  56. Orlandi, A.; Ferlosio, A.; Arcuri, G.; Scioli, M.G.; de Falco, S.; Spagnoli, L.G. Flt-1 expression influences apoptotic susceptibility of vascular smooth muscle cells through the NF-κB/IAP-1 pathway. Cardiovasc. Res. 2010, 85, 214–223. [Google Scholar] [CrossRef] [PubMed]
  57. Orlandi, A. The contribution of resident vascular stem cells to arterial pathology. J. Stem Cells 2015, 8, 9–17. [Google Scholar] [CrossRef] [PubMed]
  58. Gao, S.; Wassler, M.; Zhang, L.; Li, Y.; Wang, J.; Zhang, Y.; Shelat, H.; Williams, J.; Geng, Y.J. MicroRNA-133a regulates insulin-like growth factor-1 receptor expression and vascular smooth muscle cell proliferation in murine atherosclerosis. Atherosclerosis 2014, 232, 171–179. [Google Scholar] [CrossRef] [PubMed]
  59. Ferlosio, A.; Orlandi, A. Diabetes and aging: A different phenotypic commitment of circulating and resident stem cells? Acta Diabetol. 2012, 49, 493–494. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Chait, A.; Bornfeldt, K.E. Diabetes and atherosclerosis: Is there a role for hyperglycemia? J. Lipid Res. 2009, 50, S335–S339. [Google Scholar] [CrossRef] [PubMed]
  61. Guo, M.; Mao, X.; Ji, Q.; Lang, M.; Li, S.; Peng, Y.; Zhou, W.; Xiong, B.; Zeng, Q. Inhibition of IFN regulatory factor-1 down-regulate Th1 cell function in patients with acute coronary syndrome. J. Clin. Immunol. 2010, 30, 241–252. [Google Scholar] [CrossRef] [PubMed]
  62. Yuan, X.; Zhang, Z.; Gong, K.; Zhao, P.; Qin, J.; Liu, N. Inhibition of reactive oxygen species/extracellular signal-regulated kinases pathway by pioglitazone attenuates advanced glycation end products-induced proliferation of vascular smooth muscle cells in rats. Biol. Pharm. Bull. 2011, 34, 618–623. [Google Scholar] [CrossRef] [PubMed]
  63. Zhang, X.; Liu, L.; Chen, C.; Chi, Y.-L.; Yang, X.-Q.; Xu, Y.; Li, X.-T.; Guo, S.-L.; Xiong, S.-H.; Shen, M.R.; et al. Interferon regulatory factor-1 together with reactive oxygen species promotes the acceleration of cell cycle progression by up-regulating the cyclin E and CDK2 genes during high glucose-induced proliferation of vascular smooth muscle cells. Cardiovasc. Diabetol. 2013, 12, 147. [Google Scholar] [CrossRef] [PubMed]
  64. McGinn, S.; Poronnik, P.; Gallery, E.D.; Pollock, C.A. The effects of high glucose and atorvastatin on endothelial cell matrix production. Diabetes Med. 2004, 21, 1102–1107. [Google Scholar] [CrossRef] [PubMed]
  65. Li, J.H.; Huang, X.R.; Zhu, H.J.; Johnson, R.; Lan, H.Y. Role of TGF-β signaling in extracellular matrix production under high glucose conditions. Kidney Int. 2003, 63, 2010–2019. [Google Scholar] [CrossRef] [PubMed]
  66. Ha, Y.M.; Lee, D.H.; Kim, M.; Kang, Y.J. High glucose induces connective tissue growth factor expression and extracellular matrix accumulation in rat aorta vascular smooth muscle cells via extracellular signal-regulated kinase 1/2. Korean J. Physiol. Pharmacol. 2013, 17, 307–314. [Google Scholar] [CrossRef] [PubMed]
  67. Bourcier, T.; Sukhova, G.; Libby, P. The nuclear factor κB signaling pathway participates in dysregulation of vascular smooth muscle cells in vitro and in human atherosclerosis. J. Biol. Chem. 1997, 272, 15817–15824. [Google Scholar] [CrossRef] [PubMed]
  68. Satoh, H.; Togo, M.; Hara, M.; Miyata, T.; Han, K.; Maekawa, H.; Ohno, M.; Hashimoto, Y.; Kurokawa, K.; Watanabe, T. Advanced glycation endproducts stimulate mitogen-activated protein kinase and proliferation in rabbit vascular smooth muscle cells. Biochem. Biophys. Res. Commun. 1997, 239, 111–115. [Google Scholar] [CrossRef] [PubMed]
  69. Park, L.; Raman, K.G.; Lee, K.J.; Lu, Y.; Ferran, L.J., Jr.; Chow, W.S.; Stern, D.; Schmidt, A.M. Suppression of accelerated diabetic atherosclerosis by the soluble receptor for advanced glycation endproducts. Nat. Med. 1998, 4, 1025–1031. [Google Scholar] [CrossRef] [PubMed]
  70. Vlassara, H.; Li, Y.M.; Imani, F.; Wojciechowicz, D.; Yang, Z.; Liu, F.T.; Cerami, A. Identification of galectin-3 as a high-affinity binding protein for advanced glycation end products (AGE): A new member of the AGE-receptor complex. Mol. Med. 1995, 1, 634–646. [Google Scholar] [PubMed]
  71. Seki, N.; Hashimoto, N.; Sano, H.; Horiuchi, S.; Yagui, K.; Makino, H.; Saito, Y. Mechanisms involved in the stimulatory effect of advanced glycation end products on growth of rat aortic smooth muscle cells. Metabolism 2003, 52, 1558–1563. [Google Scholar] [CrossRef] [PubMed]
  72. Bornfeldt, K.E.; Raines, E.W.; Nakano, T.; Graves, L.M.; Krebs, E.G.; Ross, R. Insulin-like growth factor-I and platelet-derived growth factor-BB induce directed migration of human arterial smooth muscle cells via signaling pathways that are distinct from those of proliferation. J. Clin. Investig. 1994, 93, 1266–1274. [Google Scholar] [CrossRef] [PubMed]
  73. Pfeifle, B.; Hamann, H.; Fussganger, R.; Ditschuneit, H. Insulin as a growth regulator of arterial smooth muscle cells: Effect of insulin of I.G.F.I. Diabetes Metab. 1987, 13, 326–330. [Google Scholar]
  74. Wang, C.C.; Gurevich, I.; Draznin, B. Insulin affects vascular smooth muscle cell phenotype and migration via distinct signaling pathways. Diabetes 2003, 52, 2562–2569. [Google Scholar] [CrossRef] [PubMed]
  75. Mughal, R.S.; Scragg, J.L.; Lister, P.; Warburton, P.; Riches, K.; O’Regan, D.J.; Ball, S.G.; Turner, N.A.; Porter, K.E. Cellular mechanisms by which proinsulin C-peptide prevents insulin-induced neointima formation in human saphenous vein. Diabetologia 2010, 53, 1761–1771. [Google Scholar] [CrossRef] [PubMed]
  76. Kotlyar, A.A.; Vered, Z.; Goldberg, I.; Chouraqui, P.; Nas, D.; Fridman, E.; Chen-Levy, Z.; Fytlovich, S.; Sangiorgi, G.; Spagnoli, L.G.; et al. Insulin-like growth factor I and II preserve myocardial structure in postinfarct swine. Heart 2001, 86, 693–700. [Google Scholar] [CrossRef] [PubMed]
  77. Taniyama, Y.; Hitomi, H.; Shah, A.; Alexander, R.W.; Griendling, K.K. Mechanisms of reactive oxygen species-dependent downregulation of insulin receptor substrate-1 by angiotensin II. Arterioscler. Thromb. Vasc. Biol. 2005, 25, 1142–1147. [Google Scholar] [CrossRef] [PubMed]
  78. Ketsawatsomkron, P.; Stepp, D.W.; Fulton, D.J.; Marrero, M.B. Molecular mechanism of angiotensin II-induced insulin resistance in aortic vascular smooth muscle cells: Roles of protein tyrosine phosphatase-1B. Vascul. Pharmacol. 2010, 53, 160–168. [Google Scholar] [CrossRef] [PubMed]
  79. Touyz, R.M.; Schiffrin, E.L. Signal transduction mechanisms mediating the physiological and pathophysiological actions of angiotensin II in vascular smooth muscle cells. Pharmacol. Rev. 2000, 52, 639–672. [Google Scholar] [PubMed]
  80. Von der Thüsen, J.H.; Borensztajn, K.S.; Moimas, S.; van Heiningen, S.; Teeling, P.; van Berkel, T.J.; Biessen, E.A. IGF-1 has plaque-stabilizing effects in atherosclerosis by altering vascular smooth muscle cell phenotype. Am. J. Pathol. 2011, 178, 924–934. [Google Scholar] [CrossRef] [PubMed]
  81. Johansson, G.S.; Arnqvist, H.J. Insulin and IGF-I action on insulin receptors, IGF-I receptors, and hybrid insulin/IGF-I receptors in vascular smooth muscle cells. Am. J. Physiol. Endocrinol. Metab. 2006, 291, E1124–E1130. [Google Scholar] [CrossRef] [PubMed]
  82. Engberding, N.; San Martín, A.; Martin-Garrido, A.; Koga, M.; Pounkova, L.; Lyons, E.; Lassègue, B.; Griendling, K.K. Insulin-like growth factor-1 receptor expression masks the antiinflammatory and glucose uptake capacity of insulin in vascular smooth muscle cells. Arterioscler. Thromb. Vasc. Biol. 2009, 29, 408–415. [Google Scholar] [CrossRef] [PubMed]
  83. Cervelli, V.; Scioli, M.G.; Gentile, P.; Doldo, E.; Bonanno, E.; Spagnoli, L.G.; Orlandi, A. Platelet-rich plasma greatly potentiates insulin-induced adipogenic differentiation of human adipose-derived stem cells through a serine/threonine kinase Akt-dependent mechanism and promotes clinical fat graft maintenance. Stem Cells Transl. Med. 2012, 1, 206–220. [Google Scholar] [CrossRef] [PubMed]
  84. Bartel, D.P. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 2004, 116, 281–297. [Google Scholar] [CrossRef]
  85. Blackstock, C.D.; Higashi, Y.; Sukhanov, S.; Shai, S.Y.; Stefanovic, B.; Tabony, A.M.; Yoshida, T.; Delafontaine, P. Insulin-like growth factor-1 increases synthesis of collagen type I via induction of the mRNA-binding protein LARP6 expression and binding to the 5' stem-loop of COL1a1 and COL1a2 mRNA. J. Biol. Chem. 2014, 289, 7264–7274. [Google Scholar] [CrossRef] [PubMed]
  86. Cifarelli, V.; Luppi, P.; Tse, H.M.; He, J.; Piganelli, J.; Trucco, M. Human proinsulin C-peptide reduces high glucose-induced proliferation and NF-κB activation in vascular smooth muscle cells. Atherosclerosis 2008, 201, 248–257. [Google Scholar] [CrossRef] [PubMed]
  87. Nakamura, J.; Kasuya, Y.; Hamada, Y.; Nakashima, E.; Naruse, K.; Yasuda, Y.; Kato, K.; Hotta, N. Glucose-induced hyperproliferation of cultured rat aortic smooth muscle cells through polyol pathway hyperactivity. Diabetologia 2001, 44, 480–487. [Google Scholar] [CrossRef] [PubMed]
  88. Suzuki, L.A.; Poot, M.; Gerrity, R.G.; Bornfeldt, K.E. Diabetes accelerates smooth muscle accumulation in lesions of atherosclerosis: Lack of direct growth-promoting effects of high glucose levels. Diabetes 2001, 50, 851–860. [Google Scholar] [CrossRef] [PubMed]
  89. Iacobellis, G.; Gao, Y.J.; Sharma, A.M. Do cardiac and perivascular adipose tissue play a role in atherosclerosis? Curr. Diabetes Rep. 2008, 8, 20–24. [Google Scholar] [CrossRef]
  90. Henrichot, E.; Juge-Aubry, C.E.; Pernin, A.; Pache, J.C.; Velebit, V.; Dayer, J.M.; Meda, P.; Chizzolini, C.; Meier, C.A. Production of chemokines by perivascular adipose tissue: A role in the pathogenesis of atherosclerosis? Arterioscler. Thromb. Vasc. Biol. 2005, 25, 2594–2599. [Google Scholar] [CrossRef] [PubMed]
  91. Lamers, D.; Schlich, R.; Greulich, S.; Sasson, S.; Sell, H.; Eckel, J. Oleic acid and adipokines synergize in inducing proliferation and inflammatory signalling in human vascular smooth muscle cells. J. Cell Mol. Med. 2011, 15, 1177–1188. [Google Scholar] [CrossRef] [PubMed]
  92. Arita, Y.; Kihara, S.; Ouchi, N.; Takahashi, M.; Maeda, K.; Miyagawa, J.; Hotta, K.; Shimomura, I.; Nakamura, T.; Miyaoka, K.; et al. Paradoxical decrease of an adipose-specific protein, adiponectin, in obesity. Biochem. Biophys. Res. Commun. 1999, 257, 79–83. [Google Scholar] [CrossRef] [PubMed]
  93. Hotta, K.; Funahashi, T.; Arita, Y.; Takahashi, M.; Matsuda, M.; Okamoto, Y.; Iwahashi, H.; Kuriyama, H.; Ouchi, N.; Maeda, K.; et al. Plasma concentrations of a novel, adiposespecific protein, adiponectin, in type 2 diabetic patients. Arterioscler. Thromb. Vasc. Biol. 2000, 20, 1595–1599. [Google Scholar] [CrossRef] [PubMed]
  94. Schlich, R.; Willems, M.; Greulich, S.; Ruppe, F.; Knoefel, W.T.; Ouwens, D.M.; Maxhera, B.; Lichtenberg, A.; Eckel, J.; Sell, H. VEGF in the crosstalk between human adipocytes and smooth muscle cells: Depot-specific release from visceral and perivascular adipose tissue. Mediators Inflamm. 2013, 2013, 982458. [Google Scholar] [CrossRef] [PubMed]
  95. Tarallo, V.; Vesci, L.; Capasso, O.; Esposito, M.T.; Riccioni, T.; Pastore, L.; Orlandi, A.; Pisano, C.; de Falco, S. A placental growth factor variant unable to recognize vascular endothelial growth factor (VEGF) receptor-1 inhibits VEGF-dependent tumor angiogenesis via heterodimerization. Cancer Res. 2010, 70, 1804–1813. [Google Scholar] [CrossRef] [PubMed]
  96. Tarallo, V.; Lepore, L.; Marcellini, M.; dal Piaz, F.; Tudisco, L.; Ponticelli, S.; Lund, F.W.; Roepstorff, P.; Orlandi, A.; Pisano, C.; et al. The biflavonoid amentoflavone inhibits neovascularization preventing the activity of proangiogenic vascular endothelial growth factors. J. Biol. Chem. 2011, 286, 19641–19651. [Google Scholar] [CrossRef] [PubMed]
  97. Belvisi, L.; Riccioni, T.; Marcellini, M.; Vesci, L.; Chiarucci, I.; Efrati, D.; Potenza, D.; Scolastico, C.; Manzoni, L.; Lombardo, K.; et al. Biological and molecular properties of a new αvβ3/αvβ5 integrin antagonist. Mol. Cancer Ther. 2005, 4, 1670–1680. [Google Scholar] [CrossRef] [PubMed]
  98. Cicatiello, V.; Apicella, I.; Tudisco, L.; Tarallo, V.; Formisano, L.; Sandomenico, A.; Kim, Y.; Bastos-Carvalho, A.; Orlandi, A.; Ambati, J.; et al. Powerful anti-tumor and anti-angiogenic activity of a new anti-vascular endothelial growth factor receptor 1 peptide in colorectal cancer models. Oncotarget 2015, 6, 10563–10576. [Google Scholar] [CrossRef] [PubMed]
  99. Cheng, K.K.; Lam, K.S.; Wang, Y.; Huang, Y.; Carling, D.; Wu, D.; Wong, C.; Xu, A. Adiponectin-induced endothelial nitric oxide synthase activation and nitric oxide production are mediated by APPL1 in endothelial cells. Diabetes 2007, 56, 1387–1394. [Google Scholar] [CrossRef] [PubMed]
  100. Ridnour, L.A.; Thomas, D.D.; Mancardi, D.; Espey, M.G.; Miranda, K.M.; Paolocci, N.; Feelisch, M.; Fukuto, J.; Wink, D.A. The chemistry of nitrosative stress induced by nitric oxide and reactive nitrogen oxide species. Putting perspective on stressful biological situations. Biol. Chem. 2004, 385, 1–10. [Google Scholar] [CrossRef] [PubMed]
  101. Cosentino, F.; Eto, M.; de Paolis, P.; van der Loo, B.; Bachschmid, M.; Ullrich, V.; Kouroedov, A.; Delli Gatti, C.; Joch, H.; Volpe, M.; et al. High glucose causes upregulation of cyclooxygenase-2 and alters prostanoid profile in human endothelial cells: Role of protein kinase C and reactive oxygen species. Circulation 2003, 107, 1017–1023. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Casella, S.; Bielli, A.; Mauriello, A.; Orlandi, A. Molecular Pathways Regulating Macrovascular Pathology and Vascular Smooth Muscle Cells Phenotype in Type 2 Diabetes. Int. J. Mol. Sci. 2015, 16, 24353-24368. https://doi.org/10.3390/ijms161024353

AMA Style

Casella S, Bielli A, Mauriello A, Orlandi A. Molecular Pathways Regulating Macrovascular Pathology and Vascular Smooth Muscle Cells Phenotype in Type 2 Diabetes. International Journal of Molecular Sciences. 2015; 16(10):24353-24368. https://doi.org/10.3390/ijms161024353

Chicago/Turabian Style

Casella, Sara, Alessandra Bielli, Alessandro Mauriello, and Augusto Orlandi. 2015. "Molecular Pathways Regulating Macrovascular Pathology and Vascular Smooth Muscle Cells Phenotype in Type 2 Diabetes" International Journal of Molecular Sciences 16, no. 10: 24353-24368. https://doi.org/10.3390/ijms161024353

Article Metrics

Back to TopTop