当前位置: 首页 > 医学版 > 期刊论文 > 内科学 > 糖尿病学杂志 > 2005年 > 第9期 > 正文
编号:11256720
Type 2 Diabetes Causes Remodeling of Cerebrovasculature via Differential Regulation of Matrix Metalloproteinases and Collagen Synthesis
     1 Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, Georgia

    2 Department of Biostatistics, Medical College of Georgia, Augusta, Georgia

    3 Department of Physiology, Medical College of Georgia, Augusta, Georgia

    4 Vascular Biology Center, Medical College of Georgia, Augusta, Georgia

    ABSTRACT

    The risk of cerebrovascular disease is four- to sixfold higher in patients with diabetes. Vascular remodeling, characterized by extracellular matrix deposition and an increased media-to-lumen ratio, occurs in diabetes and contributes to the development of complications. However, diabetes-induced changes in the cerebrovascular structure remain unknown. Endothelin-1 (ET-1), a potent vasoconstrictor with profibrotic properties, is chronically elevated in diabetes. To determine diabetes-mediated changes in the cerebrovasculature and the role of ET-1 in this process, type 2 diabetic Goto-Kakizaki (GK) rats were administered an ETA receptor antagonist for 4 weeks. Middle cerebral arteries were harvested and studies were performed to determine vascular structure. Tissue and plasma ET-1 levels were increased in GK rats compared with controls. Significant medial hypertrophy and collagen deposition resulted in an increased wall-to-lumen ratio in diabetic rats that was reduced by ETA receptor antagonism. Vascular matrix metalloproteinase (MMP)-2 activity was higher, but MMP-1 levels were significantly reduced in GK rats, and MMP levels were restored to control levels by ETA receptor antagonism. We conclude that ET-1 promotes cerebrovascular remodeling in type 2 diabetes through differential regulation of MMPs. Augmented cerebrovascular remodeling may contribute to an increased risk of stroke in diabetes, and ETA receptor antagonism may offer a novel therapeutic target.

    Type 2 diabetes, a disease that affects more than 17 million Americans, holds a two- to sixfold increased risk for cerebrovascular disease and stroke (1,2), and 70% of patients with a recent stroke have overt diabetes or pre-diabetes characterized by impaired fasting glucose or impaired glucose tolerance (3). However, the underlying basis of this predisposition remains unclear. Diabetic vascular complications are associated with remodeling of the vessel wall in the retinal, renal, and mesenteric circulations. However, diabetes-induced structural changes in the cerebral microvessels are unknown.

    The endothelium is an early target in diabetes, and dysfunction of vascular endothelial cells has a role in the diabetic vascular disease process (4). For example, the release of vasodilator and antiproliferative mediators such as nitric oxide and prostaglandin-I2 is decreased, whereas production of endothelin-1 (ET-1) is increased (5). A significant correlation has been observed between plasma ET-1 levels and diabetes complications. In addition to being vasoconstrictive, ET-1 is also mitogenic. In streptozotocin-induced diabetes, nonselective ET receptor antagonism prevents extracellular matrix deposition in the retina as well as in the mesenteric arteries, providing evidence for a causal relationship (6,7). Nonselective blockade of ET receptors also prevents increased myogenic tone of cerebral vessels in diabetes (8), but the effect on vascular structure and the underlying mechanisms remain to be identified.

    The matrix metalloproteinases (MMPs) are a family of zinc-dependent proteases that are responsible for extracellular matrix turnover (9). In pathological states such as cardiovascular disease, the MMPs may become deleterious because of dysregulation and can result in tissue injury and inflammation. In experimental hypertension and atherosclerosis models, for example, increased MMP activity contributes to cardiac and vascular complications (10eC13). Although it has been shown that diabetes enhances aortic MMP activity, mesangial cells grown under high-glucose conditions have been shown to exhibit decreased MMP activity, which contributes to diabetic nephropathy via matrix accumulation. Clearly, the regulation of MMPs and their relative contribution to pathological processes varies between tissues; however, the regulation of MMP expression and activity in the cerebrovasculature and especially in diabetes is not known. To identify diabetes-induced pathological changes in the cerebrovasculature that may contribute to the development of cerebrovascular complications in diabetes, the current study had three objectives: 1) to assess the diabetes-induced structural changes in cerebral vessels, 2) to determine the expression and activity of the MMP system in these vessels, and 3) to determine to what extent ET-1 regulates cerebrovascular MMPs and remodeling. The central hypothesis was that increased MMP activity would be associated with hypertrophic remodeling of the cerebral vessels in the diabetic state and that blockade of the ET system would ameliorate this process.

    RESEARCH DESIGN AND METHODS

    The Medical College of Georgia institutional animal care and use committee approved all protocols. Male Wistar and Goto-Kakizaki (GK) rats were obtained from Taconic (Germantown, NY) at 8 weeks of age. All animals were individually housed at the Medical College of Georgia’s animal care facility, were allowed access to food and water ad libitum, and were maintained on a 12-h light/dark cycle. During housing, drinking water, weight, and blood glucose measurements were performed twice weekly. Glucose measurements were taken from the tail vein and measured on a commercially available glucose meter (AccuChek; Roche Diagnostics, Indianapolis, IN). At 12 weeks of age, when all GK animals became overtly diabetic, telemetry transmitters for blood pressure measurements were implanted as previously reported (14). After a 2-week recovery period, control and diabetic animals were administered the ETA selective antagonist ABT-627 (5 mg · kgeC1 · dayeC1) or vehicle only (15,16). The drug was dissolved in drinking water at a concentration based on the animal’s weight and daily water consumption. Treatment was maintained until the time the animals were killed at 18 weeks of age. Animals were anesthetized with sodium pentobarbital and exsanguinated via the abdominal aorta. On death, the brain was removed and placed in ice-cold physiological saline solution. For immunohistochemistry, middle cerebral arteries were perfused with Histogel (Richard Allen Scientific, Kalamazoo, MI) and then excised and embedded in the same matrix. On gelling of the matrix, the embedded vessel was placed in 10% formalin for storage. For protein studies, vessels were excised, snap-frozen in liquid nitrogen, and stored at eC80°C.

    Plasma measurements.

    Plasma ET-1 and insulin were measured by specific enzyme-linked immunosorbent assay kits from Alpco Diagnostics (Windham, NH). Plasma triglycerides and cholesterol were measured using commercially available kits (Wako, Richmond, VA).

    Glucose clearance experiments.

    Additional diabetic and control animals were restrained awake, and baseline glucose was checked via the tail vein. A glucose bolus (0.6 mg/kg) was injected via the dorsal tail vein, and glucose readings were taken at 5-min intervals for a total duration of 1 h.

    Tissue homogenization and zymography.

    Snap-frozen middle cerebral arteries were placed in modified radioimmunoprecipitation assay buffer (50 mmol/l Tris-HCl, 1% Nonidet P-40, 0.25% Na-deoxycholate, 150 mmol/l NaCl, 1 mmol/l phenylmethylsulfonyl fluoride, 1 e/ml aprotinin, 1 e/ml leupeptin, 1 e/ml pepstatin, 1 mmol/l sodium orthovanadate, and 1 mmol/l sodium fluoride) and sonicated at room temperature for 8- to 10-s bursts. Samples were placed on ice between sonications. Total protein was measured using the Bradford method (BioRad, Richmond, CA) in a 96-well format and read on a Quant spectrophotometer. BSA was used as a standard. On the day of the experiment, samples (20 e protein/sample) were loaded onto 10% gelatin zymogram gels (BioRad) and separated by molecular weight under nonreducing conditions. The gels were then rinsed twice in 2.5% Triton X-100 and incubated for 24 h in substrate buffer containing 21 mmol/l Tris-HCl, 10 mmol/l CaCl2, and 0.04% NaN3. Gels were then stained by Coomassie blue R-250 followed by destaining in 55% methanol and 7% acetic acid. Lytic activity was viewed as clear bands on a dark blue background and was quantitated by densitometric analysis (Gel-Pro version 3.1; Media Cybernetics, Carlsbad, CA). Kaleidoscope protein standards (Bio-Rad) and recombinant MMP-2 and -9 proteins (Calbiochem, San Diego, CA) were run in parallel with all samples. Tissue inhibitor of metalloproteinase-2 (TIMP-2) levels were measured by enzyme-linked immunosorbent assay (Amersham Biosciences, Piscataway, NJ).

    Immunohistochemistry and vessel morphometry.

    Middle cerebral artery segments were fixed in 10% formalin, embedded in paraffin, sectioned at 4 e, and mounted on treated slides. Slides were then deparaffinized, blocked (Super Block; Biogenex Labs, San Ramon, CA), and placed in PBS for 5 min. Slides were then incubated with ET-1 or CD68 primary antibody obtained from Peninsula Laboratories (San Carlos, CA) and Dako (Carpinteria, CA), respectively, at room temperature; washed; and then incubated in secondary antibody (LSAB2-HRP kit; Dako), followed by incubation with streptavidineChorseradish peroxidase. Bound antibody was detected with 3,3'-diaminobenzidine substrate kit. Additional slides were incubated with only the secondary antibody to determine nonspecific staining. Slides were viewed using an Axiovert microscope (Zeiss, Thornwood, NY) and captured using Spot software. For morphometric studies, 4-e vessel segments were subjected to Verhoeff van Gieson’s elastic staining or picro-polychrome staining. Images were captured and analyzed using Spot software, and wall-to-lumen ratios were calculated.

    Western blotting.

    Protein levels of MMPs (MMP-1 and -2) were determined by immunoblotting, as previously described (15). All blots were restained with anti-actin antibody for equal protein loading.

    Statistical analysis.

    The profile of blood glucose changes over time was analyzed for group differences (control versus diabetic) using a repeated-measures ANOVA in which the interaction of group by time was the test of interest. A Tukey adjustment was used for the post hoc comparison of the groups at each time point. A rank transformation (17) was applied to all other data before analysis to address issues of nonnormality. A 2 x 2 ANOVA was used to investigate the main effects of disease (control versus diabetic) and drug (vehicle versus ABT-627) and the interaction between disease and drug. Effects were considered statistically significant at P < 0.05. SAS version 8.2 was used for all analyses. Results are expressed as the means ± SE.

    RESULTS

    Animal data.

    Metabolic parameters for control and diabetic (GK) animals are summarized in Table 1. Diabetic animals were significantly smaller than controls, and ETA antagonism did not affect animal weight. GK animals displayed mildly elevated blood glucose and blood pressure without hyperlipidemia and hyperinsulinemia. Experiments were performed in diabetic and control animals to assess glucose clearance capacity. Figure 1 indicates that the maximal response to glucose challenge was significantly higher in untreated diabetic animals versus control (259% of baseline vs. 168%, P < 0.05). Additionally, after 30 min diabetic animals exhibited a decreased ability to clear glucose and return to normal levels, providing evidence for impaired glucose tolerance (P < 0.05).

    Local and systemic ET-1 levels.

    Plasma ET-1 levels were elevated approximately threefold in diabetic animals (P = 0.001) (Fig. 2A). Tissue ET-1 was assessed by immunohistochemistry and, as shown in Fig. 2B, there was enhanced endothelial and adventitial staining in the middle cerebral arteries from diabetic animals compared with controls. Staining in the absence of the primary ET-1 antibody was determined to eliminate nonspecific staining by the secondary antibody.

    Cerebrovascular MMP expression and activity.

    MMP activity in middle cerebral arteries from treated and untreated diabetic GK and control Wistar rats was assessed using gelatin zymography. A representative zymogram is shown in Fig. 3A. Lytic activity, corresponding with the molecular weight of active MMP-2, was detected in all samples. Densitometric analysis demonstrated that MMP-2 activity was increased in diabetes (P < 0.0001) and that ETA receptor antagonism prevented this increase in activation (P = 0.001 vs. untreated GK) (Fig. 3A). Because TIMP-2 is the endogenous inhibitor of MMP-2, TIMP-2 levels were measured to determine whether the increase in MMP-2 activity arises from a decrease in its inhibitor. Although there was no significant difference between the control and diabetic groups, there was a trend for increased TIMP-2 levels with ETA antagonism.

    To determine whether increased MMP-2 activity is caused by an increase in protein levels, total MMP-2 protein in middle cerebral arteries was assessed by immunoblotting. Two bands of 72 and 67 kDa were detected corresponding to the zymogen and cleaved active forms, respectively. A representative immunoblot is shown in Fig. 4A. Densitometric analysis of both bands indicated a fourfold increase in MMP-2 protein in diabetes (P < 0.003 vs. control). However, unlike the zymography studies, ETA receptor antagonism did not cause a change in MMP-2 protein levels (Fig. 4A). Because MMP-1 is the major MMP that degrades fibrillar collagen, protein levels were determined in middle cerebral arteries by immunoblotting. There was a significant decrease in MMP-1 levels in GK rats (P = 0.034) and a trend toward restoration of MMP-1 levels to control values with ETA receptor antagonism (Fig. 4B).

    Vascular structure.

    Verhoeff van Gieson elastic staining was performed to determine vessel diameter and wall thickness (Fig. 5A). There was a twofold increase in the wall-to-lumen ratio in GK rats that was significantly reduced by ETA receptor antagonism (P = 0.042) (Fig. 5B). Compared with control rats, there was increased staining for smooth muscle fibers and collagen, as evidenced by deep purple staining by Verhoeff van Gieson elastic staining in the cross-sections from GK rats. These changes were detected primarily in the medial layer. To further evaluate the collagen deposition dynamics, picro-polychrome staining was used to differentiate the old and newly laid collagen. In untreated GK rats, there was increased blue staining for new collagen, masking the red staining for old collagen, indicating increased collagen synthesis. ETA antagonism reduced the staining pattern for new collagen. To determine whether and to what extent inflammation contributes to the increased wall thickness and collagen deposition, the same vascular cross-sections were immunostained with CD68 antibody, a marker for macrophages. There was no difference between groups (data not shown).

    DISCUSSION

    It is well established that diabetes causes hypertrophic remodeling of the peripheral vasculature, characterized by reduced lumen diameter, media enlargement, abnormalities of expression and/or localization of extracellular matrix components (such as collagen and laminin deposition in the vessel wall and accelerated formation of atherosclerotic plaques), and intimal proliferation (7,18eC21). It is also known that type 2 diabetes, a disease that affects more than 17 million Americans, holds a two- to sixfold increased risk for cerebrovascular disease and stroke. Furthermore, diabetes increases the risk of microvascular hemorrhage and poor outcome of stroke. The majority of experimental stroke models are induced by middle cerebral artery occlusion, and, although it is known that the integrity of cerebral blood vessels is very critical in the pathophysiology of stroke, diabetes-induced changes in middle cerebral artery structure have remained unknown. This study was designed to look at structural changes and potential mechanisms of altered matrix dynamics in the cerebral microvasculature in diabetes. Our findings demonstrate for the first time that mild hyperglycemia for 4eC6 weeks stimulates the local production of ET-1 and causes medial thickening in the cerebrovasculature, which are associated with increased gelatinase (MMP-2) activity and decreased collagenase (MMP-1) levels. Furthermore, MMP activation and an increased wall-to-lumen ratio can be partially prevented by the administration of an ETA receptor antagonist, providing evidence that ET-1 is in part responsible for pathological remodeling of the cerebral vessels in diabetes.

    The chemically induced streptozotocin model of type 1 diabetes, the most commonly used experimental model of diabetes, displays highly elevated glucose levels. There is still a need for spontaneous models of type 2 diabetes in which blood glucose levels are comparable to those seen in patients. GK rats generally spontaneously manifest hyperglycemia by 8eC10 weeks of age (22). It has been shown that these animals retain >40% of their total -cell mass (a number similar to human type 2 diabetes) but have impaired glucose-induced insulin release (23eC25). In addition, treatment of GK rats with nateglinide, an insulin secretagogue, reduces postprandial hyperglycemia and elicits early-phase insulin secretion, a phenomenon not observed in type 1 diabetes (26,27). Insulin resistance and hyperlipidemia often accompany type 2 diabetes, and the presence of these risk factors in GK rats has been controversial (22,28,29). Thus, we specifically assessed the presence of these risk factors in our colony. GK rats developed hyperglycemia by 12 weeks of age. Although plasma insulin levels were not elevated, impaired glucose tolerance tests demonstrated a defect in clearing glucose in GK rats when compared with normal Wistar rats, which served as control for this model. Metabolic parameters demonstrated that by 18 weeks of age (6 weeks of diabetes), this model displayed significant hyperglycemia and a slight elevation in blood pressure without hyperlipidemia and hyperinsulinemia. Recent studies identified that glucose intolerance is another risk factor for cardiovascular disease. The Hoorn Study reported increased arterial stiffness in glucose intolerance and type 2 diabetes (30). The GK rat model thus serves as a good model to study the impact of moderate changes in blood glucose on cerebrovascular complications of diabetes. Our findings are significant in that even under a relatively short duration of mildly hyperglycemic conditions without the confounding effects of hyperlipidemia and insulin resistance, there were striking pathological changes in the cerebrovascular structure.

    Plasma ET-1 levels are elevated in type 1 and type 2 as well as experimental diabetes (31eC33). In type 1 diabetes, mixed ET receptor blockade significantly reduces the mesenteric walleCtoeClumen ratio and extracellular matrix deposition (7). Endothelial overexpression of human ET-1 was recently shown to significantly increase media-to-lumen ratio in murine mesenteric arteries (34). Nonselective blockade of ET receptors also prevented increased myogenic tone of cerebral vessels in diabetes, but the effect on vascular structure remained to be identified. Several studies have linked ET-1 to the regulation of MMPs. ETA receptor antagonism has been reported to decrease collagen accumulation and improve MMP-2 activity in kidneys from stroke-prone spontaneously hypertensive rats (35). We recently showed that environmental stress upregulates vascular MMP-2 activity via stimulation of ET-1 synthesis and ETA activation (15). Therefore, we investigated the regulation of cerebrovascular MMPs that can degrade collagen (MMP-1) and gelatin (MMP-2 and -9). Increased extracellular matrix protein synthesis, diminished MMP activity, and/or increased TIMP activity all could contribute to matrix accumulation, which is a late event in the vascular remodeling process (36). Interestingly, recent studies demonstrated that MMP activation also contributes to vascular smooth muscle cell growth and migration as well as increased collagen synthesis via several mechanisms. MMPs, especially MMP-9, degrade basement membrane and internal elastic lamina, disrupting the boundaries between vascular layers and facilitating vascular smooth muscle cell migration (37,38). Additionally, the breakdown of fibrillar collagen unmasks cryptic integrin signals buried in the extracellular matrix that serve as chemotactic stimuli for vascular smooth muscle cell migration (38,39). More importantly, MMPs activate membrane-bound proteins with growth-promoting properties via proteolytic cleavage (40eC42). For example, cleavage of heparin-binding epidermal growth factor by an MMP-dependent mechanism leads to activation of the epidermal growth factor receptor, promoting increased collagen synthesis (43). It has been demonstrated that both angiotensin-II and ET-1 enhance this transactivation process (41,43,44). In our study, we found ET-1 mediated increases in MMP-2 expression and activity. In addition, ETA receptor antagonism attenuated new collagen deposition in diabetic animals. However, it has to be recognized that with the available data, we cannot determine whether the improvement of the wall-to-lumen ratio in the ABT-627eCtreated GK group is a direct effect of receptor blockade or is attributable to the reduction of blood pressure in this group. It is conceivable that in the GK model, ET-1 promotes MMP-2 activation, which results in increased collagen synthesis. At the same time, ET-1 downregulates MMP-1 protein levels, promoting collagen deposition. These data provide evidence that ET-1 promotes matrix accumulation via differential regulation of MMP enzymes involved in collagen dynamics.

    Our results may have postischemic relevance in addition to the possible stroke-potentiating effects of MMPs in diabetes. The blood-brain barrier exists as a physical barrier to solute transport in the brain. During cerebral ischemia, the integrity of this barrier comes under attack, leading to increased permeability of the cerebral vessels (45,46). This breakdown allows for leakage of blood into the perivascular spaces, resulting in further damage to the already ischemic tissue. Several studies have demonstrated increased MMP activity and matrix degradation after focal cerebral ischemia (46eC50). Our findings of increased basal MMP activity in diabetes may account for poor stroke outcomes in hyperglycemic patients.

    We conclude that even mild type 2 diabetes, without the confounding effects of hyperinsulinemia and hyperlipidemia, causes significant cerebrovascular remodeling via the modulation of the MMPs that are regulated by ET-1. Altered MMP activity might contribute to increased risk of stroke in diabetes, and ETA receptor blockade may provide an effective therapeutic intervention for reducing ischemic brain damage in diabetes.

    ACKNOWLEDGMENTS

    This work was supported by a grant from the National Institutes of Health (HL076236-01), an American Heart Association scientist development grant, an American Diabetes Association research grant, a Pfizer atorvastatin research award (to A.E.), and an American Heart Association southeast affiliate predoctoral fellowship award (to A.K.H.).

    The authors thank Abbott Laboratories for the ABT-627 compound.

    ET-1, endothelin-1; MMP, matrix metalloproteinase; TIMP-2, tissue inhibitor of metalloproteinase-2

    REFERENCES

    American Diabetes Association: National diabetes fact sheet,2002 [article online]. Available from http://www.diabetes.org/diabetes-statistics/national-diabetes-fact-sheet.jsp

    American Heart Association: Heart and stroke statistical update,2002

    Kernan WN, Inzucchi SE: Type 2 diabetes mellitus and insulin resistance: Stroke prevention and management. Curr Treat Options Neurol6 :443 eC450,2004

    Chakir M, Plante G: Endothelial dysfunction in diabetes mellitus. Prostaglandins Leukot Essent Fatty Acids54 :45 eC51,1996

    Klein RL, Colwell JA: Altered endothelial function in diabetes mellitus. In Contemporary Endocrinology: Endocrinology of the Vasculature. Sowers JR, Ed. Totowa, NJ, Humana Press,1996

    Evans T, Deng DX, Chen S, Chakrabarti S: Endothelin receptor blockade prevents augmented extracellular matrix component mRNA expression and capillary basement membrane thickening in the retina of diabetic and galactose-fed rats. Diabetes49 :662 eC666,2000

    Gilbert RE, Rumble JR, Cao Z, Cox AJ, van Eeden P, Allen TJ, Kelly DJ, Cooper ME: Endothelin receptor antagonism ameliorates mast cell infiltration, vascular hypertrophy, and epidermal growth factor expression in experimental diabetes. Circ Res86 :158 eC165,2000

    Dumont AS, Dumont RJ, McNeill JH, Kassell NF, Sutherland GR, Verma S: Chronic endothelin antagonism restores cerebrovascular function in diabetes. Neurosurgery52 :653 eC660,2003

    Nagase H, Woessner JF: Matrix metalloproteinases. J Biol Chem274 :21491 eC21494,1999

    Ammarguellat FZ, Gannon PO, Amiri F, Schiffrin EL: Fibrosis, matrix metalloproteinases, and inflammation in the heart of DOCA-salt hypertensive rats: role of ET(A) receptors. Hypertension39 :679 eC684,2002

    Ammarguellat F, Larouche II, Schiffrin EL: Myocardial fibrosis in DOCA-salt hypertensive rats: effect of endothelin ET(A) receptor antagonism. Circulation103 :319 eC324,2001

    Galis ZS, Sukhova G, Lark M, Libby P: Increased expression of matrix metalloproteinases and matrix degrading activity in vulnerable regions of human atherosclerotic plaques. J Clin Invest94 :2494 eC2503,1994

    Galis ZS, Khatri JJ: Matrix metalloproteinases in vascular remodeling and atherogenesis: the good, the bad, and the ugly. Circ Res90 :251 eC262,2002

    Williams JM, Pollock JS, Pollock DM: Arterial pressure response to the antioxidant tempol and ETB receptor blockade in rats on a high-salt diet. Hypertension44 :770 eC775,2004

    Ergul A, Portik-Dobos V, Giulumian AD, Molero MM, Fuchs LC: Stress upregulates arterial matrix metalloproteinase expression and activity via endothelin A receptor activation. Am J Physiol285 :H2225 eCH2232,2003

    Pollock DM, Pollock JS: Evidence for endothelin involvement in the response to high salt. Am J Physiol281 :F144 eCF150,2001

    Conover W, Iman R: Rank transformations as a bridge between parametric and nonparametric statistics. Am Stat35 :124 eC133,1981

    Cooper ME, Gilbert RE, Jerums G: Diabetic vascular complications. Clin Exp Pharmacol Physiol24 :770 eC775,1997

    Rumble JR, Cooper ME, Soulis T, Cox A, Wu L, Youssef S, Jasik M, Jerums G, Gilbert RE: Vascular hypertrophy in experimental diabetes: role of advanced glycation end products. J Clin Invest99 :1016 eC1027,1997

    Cooper ME, Rumble J, Komers R, Du HC, Jandeleit K, Chou ST: Diabetes-associated mesenteric vascular hypertrophy is attenuated by angiotensin-converting enzyme inhibition. Diabetes43 :1221 eC1228,1994

    Intengan HD, Schiffrin EL: Structure and mechanical properties of resistance arteries in hypertension. Hypertension36 :312 eC318,2000

    Cheng ZJ, Vaskonen T, Tikkanen I, Nurminen K, Ruskoaho H, Vapaatalo H, Muller D, Park J-K, Luft FC, Mervaala EMA: Endothelial dysfunction and salt-sensitive hypertension in spontaneously diabetic Goto-Kakizaki rats. Hypertension37 :433 eC439,2001

    Ostenson CG: The Goto-Kakizaki rat. In Animal Models of Diabetes: A Primer. Sima A, Shafrir E, Eds. Amsterdam, Harwood Academic Publishers,2001

    Seica RM, Suzuki KI, Santos RM, Do Rosario LM: [Impaired insulin secretion in isolated islets of Goto-Kakizaki rats, an animal model of non obese type 2 diabetes, is a primary event.] Acta Med Port17 :42 eC48,2004 [article in Portuguese]

    Seica RM, Martins MJ, Pessa PB, Santos RM, Rosario LM, Suzuki KI, Martins MI: [Morphological changes of islet of Langerhans in an animal model of type 2 diabetes.] Acta Med Port16 :381 eC388,2003 [article in Portuguese]

    Kitahara Y, Miura K, Takesue K, Mine T, Wada R, Uchida Y, Ito S, Yagihashi S: Decreased blood glucose excursion by nateglinide ameliorated neuropathic changes in Goto-Kakizaki rats, an animal model of non-obese type 2 diabetes. Metabolism51 :1452 eC1457,2002

    Ladriere L, Bjorkling F, Malaisse WJ: Stimulation of insulin release in hereditarily diabetic rats by mixed molecules formed of nateglinide and a succinic acid ester. Int J Mol Med5 :63 eC65,2000

    Metz SA, Meredith M, Vadakekalam J, Rabaglia ME, Kowluru A: A defect late in stimulus-secretion coupling impairs insulin secretion in Goto-Kakizaki diabetic rats. Diabetes48 :1754 eC1762,1999

    Iida KT, Kawakami Y, Suzuki M, Shimano H, Toyoshima H, Sone H, Shimada K, Iwama Y, Watanabe Y, Mokuno H, Kamata K, Yamada N: Effect of thiazolidinediones and metformin on LDL oxidation and aortic endothelium relaxation in diabetic GK rats. Am J Physiol284 :E1125 eCE1130,2003

    Schram MT, Henry RMA, van Dijk RAJM, Kostense PJ, Dekker JM, Nijpels G, Heine RJ, Bouter LM, Westerhof N, Stehouwer CDA: Increased central artery stiffness in impaired glucose metabolism and type 2 diabetes: the Hoorn Study. Hypertension43 :176 eC181,2004

    Haak T, Jungmann E, Felber A, Hillmann U, Usadel KH: Increased plasma levels of endothelin in diabetic patients with hypertension. Am J Hypertens5 :161 eC166,1992

    Collier A, Leach JP, McLellan A, Jardine A, Morton JJ, Small M: Plasma endothelin-like immunoreactivity levels in IDDM patients with microalbuminuria. Diabetes Care15 :1038 eC1040,1992

    Takahashi K, Ghatei MA, Lam HC, O’Halloran DJ, Bloom SR: Elevated plasma endothelin in patients with diabetes mellitus. Diabetologia33 :306 eC310,1990

    Amiri F, Virdis A, Neves MF, Iglarz M, Seidah N, Touyz R, Reudelhuber T, Schiffrin EL: Endothelium-restricted overexpression of human endothelin-1 causes vascular remodeling and endothelial dysfunction. Circulation110 :2233 eC2240,2004

    Tostes RC, Touyz RM, He G, Ammarguellat F, Schiffrin EL: Endothelin A receptor blockade decreases expression of growth factors and collagen and improves matrix metalloproteinase-2 activity in kidneys from stroke-prone spontaneously hypertensive rats. J Cardiovasc Pharmacol39 :892 eC900,2002

    Ishikawa Y, Asuwa N, Ishii T, Ito K, Akasaka Y, Masuda T, Zhang L, Kiguchi H: Collagen alteration in vascular remodeling by hemodynamic factors. Virchows Arch437 :138 eC148,2000

    Johnson JL, van Eys GJ, Angelini GD, George SJ: Injury induces dedifferentiation of smooth muscle cells and increased matrix-degrading metalloproteinase activity in human saphenous vein. Arterioscler Thromb Vasc Biol21 :1146 eC1151,2001

    George SJ: Therapeutic potential of matrix metalloproteinase inhibitors in atherosclerosis. Expert Opin Investig Drugs9 :993 eC1007,2000

    Johnson JL, Jackson CL, Angelini GD, George SJ: Activation of matrix-degrading metalloproteinases by mast cell proteases in atherosclerotic plaques. Arterioscler Thromb Vasc Biol18 :1707 eC1715,1998

    Shah BH, Catt KJ: A central role of EGF receptor transactivation in angiotensin II-induced cardiac hypertrophy. Trends Pharmacol Sci24 :239 eC244,2003

    Saito S, Frank GD, Motley ED, Dempsey PJ, Utsunomiya H, Inagami T, Eguchi S: Metalloprotease inhibitor blocks angiotensin II-induced migration through inhibition of epidermal growth factor receptor transactivation. Biochem Biophys Res Commun294 :1023 eC1029,2002

    Konishi A, Berk BC: Epidermal growth factor receptor transactivation is regulated by glucose in vascular smooth muscle cells. J Biol Chem278 :35049 eC35056,2003

    Flamant M, Tharaux PL, Placier S, Henrion D, Coffman T, Chatziantoniou C, Dussaule JC: Epidermal growth factor receptor trans-activation mediates the tonic and fibrogenic effects of endothelin in the aortic wall of transgenic mice. FASEB J17 :327 eC329,2003

    Saito Y, Berk BC: Transactivation: a novel signaling pathway from angiotensin II to tyrosine kinase receptors. J Mol Cell Cardiol33 :3 eC7,2001

    Hamann GF, Okada Y, Fitridge R, del Zoppo GJ: Microvascular basal lamina antigens disappear during cerebral ischemia and reperfusion. Stroke26 :2120 eC2126,1995

    Rosenberg GA, Navratil M, Barone F, Feuerstein G: Proteolytic cascade enzymes increase in focal cerebral ischemia in rat. J Cereb Blood Flow Metab16 :360 eC366,1996

    Chang DI, Hosomi N, Lucero J, Heo JH, Abumiya T, Mazar AP, del Zoppo GJ: Activation systems for latent matrix metalloproteinase-2 are upregulated immediately after focal cerebral ischemia. J Cereb Blood Flow Metab23 :1408 eC1419,2003

    Magnoni S, Baker A, George SJ, Duncan WC, Kerr LE, McCulloch J, Horsburgh K: Differential alterations in the expression and activity of matrix metalloproteinases 2 and 9 after transient cerebral ischemia in mice. Neurobiol Dis17 :188 eC197,2004

    Fukuda S, Fini CA, Mabuchi T, Koziol JA, Eggleston LL Jr, del Zoppo GJ: Focal cerebral ischemia induces active proteases that degrade microvascular matrix. Stroke35 :998 eC1004,2004

    Asahi M, Asahi K, Jung JC, del Zoppo GJ, Fini ME, Lo EH: Role for matrix metalloproteinase 9 after focal cerebral ischemia: effects of gene knockout and enzyme inhibition with BB-94. J Cereb Blood Flow Metab20 :1681 eC1689,2000(Alex K. Harris, Jim R. Hu)