用户名: 密码: 验证码:
HTG和糖尿病复合地鼠模型的建立及诱发肾损伤机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
现已证实高甘油三酯血症是动脉粥样硬化等多种心脑血管疾病的独立危险因子,目前富含甘油三酯的脂蛋白对肾脏的致病性也逐渐得到重视,本研究首次建立糖尿病高甘油三酯血症金黄地鼠模型,并探讨了高甘油三酯对肾脏的损伤机制。采用链脲佐菌素注射联合高脂高胆固醇饮食饲喂的方法,诱导地鼠肝脏甘油三酯分泌增加,清除减慢,血浆甘油三酯显著升高,建立了饮食诱导的非转基因高甘油三酯血症动物模型。通过对此模型血液流变学、肾功能、肾脏病理组织学变化等的检测,发现动物出现蛋白尿、肾脏肥大、肾小球系膜基质扩张、糖原沉积等肾脏病理改变,表明高甘油三酯在诱发早期糖尿病肾损伤中发挥着重要的作用。进一步研究发现高甘油三酯致肾损伤的机制为脂代谢关键基因SREBP-1c表达上调、TGF-β通路活化及氧化应激增加,导致高甘油三酯血症引发肾脏损伤。SREBP-1c表达的上调引起地鼠脂肪酸合成增加,脂质沉积于肝脏、肾脏,启动了肾损害的关键步骤,同时致纤维化生长因子TGF-β、CTGF、PAI-1及炎性因子IL-6、TNF-α上调,证实高甘油三酯血症通过激活TGF-β通路导致肾小球病变,另外,高甘油三酯血症加速了高糖所致的氧化应激,形成丙二醛等脂质过氧化物加速肾脏损伤。
Diabetic nephropathy (DN), a serious complication of diabetes, is the changes of renal structure and function that caused by diabetic microangiopathy. It is also the major cause of morbidity in diabetes. DN patients often have lipid metabolic disorder and abnormal lipoprotein composition. The reserch between dyslipidmia and renal lesion has become a hot topic. The current study has confirmed the pathogenicity of dyslipidemia to the illed kidney, however, the present research mainly focused on the effect of hypercholesterinemia, especially small/dense LDL and ox-LDL in kidney disease. Since hypertriglyceridemia has been confirmed to be the independent risk factor in artherosclerosis and coronary arteriosclerotic heart disease, triglyceride rich lipoprotein, such as very low density lipoprotein, intermediated-density lipoprotein received attention in the pathogenesis of kidney disease. People realized that triglyceride, as well as cholesterol may be also an independent risk factor, and plays an important role in the development and prognosis in kidney disease. The mechanism on how hypertriglyceridemia works in the development of diabetic nephropathy have not yet been fully elucidated. This is partly because of a lack of proper hypertriglyceridemia model. Lipid metabolism of mice and rat are quite different from that in human being when treated with high calory diet, plasma triglyceride and cholesterol level can’t reach high and also can’t maintain for a long time, so mice and rat are not suitable for lipid study. Genetically modified animals presented hypertriglyceridemia, however, the survival rate is low and they are not easily available. So it is very important to establish an appropriate animal model for further studies of mechanism on triglyceride toxicity in kidney.
     In the present study, Syrian golden hamsters were used as an animal model to study diabetic dyslipidemia regarding their hepatic lipid metabolism is similar to that found in human. Through streptozocin injection and high fat/cholesterol diet fed, we established a diabetic hypertriglyceridemia animal model mimicking diabetic dyslipidemia patients in a short period of time, with the advantage of shorter time-consuming, higher plasma lipid concentration and easier operation. Further, we confirmed the toxicity of HTG on kidney. Changes in haemodynamics, lipid metabolism, inflammation reaction, oxidative stress were studied to explain the mechanism of hypertriglyceridemia in the development of diabetic nephropathy.
     In this study, diet-induced/non-genetically modified HTG animal model was established by injection of streptozotocin and fed with a diet enriched with fat and cholesterol. Plasma triglyceride and cholesterol levels reached to 1000mg/dl in hamsters fed with high fat/cholesterol diet for 4 weeks, however, in diabetic hamsters fed with high fat/high cholesterol diet (D-HFHC), plasma triglyceride concentration reached to 20000mg/dl, plasma cholesterol concentration reached to 4500mg/dl, which indicates that diabetes and cholesterol in diet plays an important role in inducing hypertriglyceridemia, and it depends on saturated fatty acid added in diet. Enlargement and lipid deposition in liver were found in D-HFHC group, whilst HDL-C, MDA levels increased, indicates a high risk for artherosclerosis and other cardial-cerebrovascular disease. The main reason of high calorie diet induced hypertriglyceridemia in diabetic hamster is that the clearance of triglyceride decreased 33 times, and liver triglyceride secretion increased about 6 times. Meanwhile, liver fatty acid synthesis increased, which induced lipid deposition in liver and kidney. Hemorrheology mainly studies the blood flow and blood cell deformation, vascular system diseases may occure when heart, blood vessels and blood volume under normal circumstances, just because abnormity in hemorheological characteristics. The present study showed that hypertriglyceridemia induced changes in hemorheological characteristics, such as high blood viscosity, decreased erythrocyte deformability, increased erythrocyte osmotic fragility, decreased electronegativity et al, all these hemorheological changes and HTG, low HDL-C raised susceptibility of diabetic microangiopathy including diabetic nephropathy.
     In order to explain the effects of hypertriglyceridemia in diabetic renal injury, normal or diabetic hamsters were fed with high fat diet (HF), high cholesterol diet (HC), and high fat diet/high cholesterol diet (HFHC). 2 weeks after diet treatment, diabetic hamsters exhibited increased kidney weight/body weight ratio and albumin in urine. Although plasma TC levels are similar between diabetic hamsters fed with high cholesterol diet and diabetic hamsters fed with high fat/high cholesterol diet, the latter has an evident renal pathology change of diabetic nephropathy, eg. renal hypertrophy, lipid deposition, proliferation of glomerular mesangial cell, extracellular matrix(ECM)deposition, fibrosis etal. So it is confirmed that hypertriglyceridemia plays an important role in the early development of diabetic nephropathy. mRNA expression of cytokines indicated that the increase of SREBP-1 upregulated TGF-β, TGF-βwas secreted by mesangial cell, it can promote cell hypertrophy, increase the production of extracellular matrix, it can also reduce the degradation of matrix by increasing the activity of matrix catabolic enzymes inhibitor, so it is thought to be the key factor of kidney lesion. Besides, PAI-1 and CTGF, which are the main factors in kidney fibrosis and deposition of ECM also increased, promoted renal fibrosis. Increased TNF-αand IL-6 also plays an important role. Upregulation of NADPH oxidase subunit p47phox, p467phox and Nox4 indicated that oxidative stress was also involved. Renal oxidative stress can induce apoptosis, dysfunction of endothelial cell, white blood cell infiltration and hypercoagulabale state. In conclusion, deposition of lipid in glomerular caused monocyte-macrophages aggregation and activation. Changes of renal hemodynamics, activation of cytokines and vasoactive substances, inhibition of ECM degradation and increase of oxidative stress are the reason that hypertriglyceridemia caused diabetic nephropathy lesion.
     Disorder of lipid metabolism is a common clinical situation in primary and secondary renal disease; it is also the independent pathogenic factor. Body reaction of dyslipidemia is complex, the present study provided experimental evidence and laid the foundation for study the relationship among diet induced hypertriglyceridemia, kidney lesion, fatty liver, fat and insulin resistence.
引文
[1]李玲.糖尿病肾病的治疗及其进展[J].实用药物与临床, 2005(8):3
    [2] Savage S, Nagel NJ, Estacio RO, et al. Clinical factors associated with urinary albumin excretion in type II diabetes[J]. Am J Kidney Dis, 1995(25):836-844
    [3] Nicollerat JA. Implications of the United Kingdom Prospective Diabetes Study (UKPDS) results on patient management[J]. Diabetes Educ, 2000(26 Suppl):8-10
    [4] Youssef S, Nguyen DT, Soulis T, et al. Effect of diabetes and aminoguanidine therapy on renal advanced glycation end-product binding[J]. Kidney Int, 1999(55):907-916
    [5] Vlassara H,Bucala R. Recent progress in advanced glycation and diabetic vascular disease: role of advanced glycation end product receptors[J]. Diabetes, 1996(45 Suppl 3):S65-66
    [6] Soulis T, Cooper ME, Vranes D, et al. Effects of aminoguanidine in preventing experimental diabetic nephropathy are related to the duration of treatment[J]. Kidney Int, 1996(50):627-634
    [7] Vay D, Vidali M, Allochis G, et al. Antibodies against advanced glycation end product Nepsilon-(carboxymethyl)lysine in healthy controls and diabetic patients[J]. Diabetologia, 2000(43):1385-1388
    [8] Cooper ME, Gilbert RE,Epstein M. Pathophysiology of diabetic nephropathy[J]. Metabolism, 1998(47):3-6
    [9] Makino H, Shikata K, Kushiro M, et al. Roles of advanced glycation end-products in the progression of diabetic nephropathy[J]. Nephrol Dial Transplant, 1996(11 Suppl 5):76-80
    [10] Horiuchi S, Higashi T, Ikeda K, et al. Advanced glycation end products and their recognition by macrophage and macrophage-derived cells[J]. Diabetes, 1996(45 Suppl 3):S73-76
    [11] Schmidt AM, Hori O, Cao R, et al. RAGE: a novel cellular receptor for advanced glycation end products[J]. Diabetes, 1996(45 Suppl 3):S77-80
    [12] Wendt T, Tanji N, Guo J, et al. Glucose, glycation, and RAGE: implications for amplification of cellular dysfunction in diabetic nephropathy[J]. J Am Soc Nephrol, 2003(14):1383-1395
    [13] Moskovitz J, Yim MB,Chock PB. Free radicals and disease[J]. Arch Biochem Biophys, 2002(397):354-359
    [14] Yamamoto T, Sato T, Mori T, et al. Clinical efficacy of insulin-like growth factor-1 in a patient with autoantibodies to insulin receptors: a case report[J]. Diabetes Res Clin Pract, 2000(49):65-69
    [15] Brownlee M. Biochemistry and molecular cell biology of diabetic complications[J]. Nature, 2001(414):813-820
    [16] Negrean V, Suciu I, Sampelean D, et al. Rheological changes in diabetic microangiopathy[J]. Rom J Intern Med, 2004(42):407-413
    [17] Le Devehat C, Vimeux M,Khodabandehlou T. Blood rheology in patients with diabetes mellitus[J]. Clin Hemorheol Microcirc, 2004(30):297-300
    [18]陈世金,唐春蓉,冯成全.糖尿病肾病患者血液流变学改变及其临床意义[J].四川医学, 2007(28):1236-1237
    [19] Buyukkocak S, Ozturk HS, Tamer MN, et al. Erythrocyte oxidant/antioxidant status of diabetic patients[J]. J Endocrinol Invest, 2000(23):228-230
    [20] Riedle B,. DK. Reactive oxygen species cause direct damage of Engelbreth-Holm-Swarm matrix[J]. Am. J. Pathol., 1997(151):215-231
    [21] Hartmut Scheuer, Wilfried Gwinner, Jan Hohbach, et al. Oxidant stress in hyperlipidemia-induced renal damage[J]. Am J Physiol Renal Physiol, 2000(278):63-74
    [22] Takebayashi K, Matsumoto S, Aso Y, et al. Aldosterone blockade attenuates urinary monocyte chemoattractant protein-1 and oxidative stress in patients with type 2 diabetes complicated by diabetic nephropathy[J]. J Clin Endocrinol Metab, 2006(91):2214-2217
    [23] Haidara MA, Mikhailidis DP, Rateb MA, et al. Evaluation of the effect of oxidative stress and vitamin E supplementation on renal function in rats with streptozotocin-induced Type 1 diabetes[J]. J Diabetes Complications, 2009(23):130-136
    [24] Nam SM, Lee MY, Koh JH, et al. Effects of NADPH oxidase inhibitor on diabetic nephropathy in OLETF rats: the role of reducing oxidative stress in its protective property[J]. Diabetes Res Clin Pract, 2009(83):176-182
    [25] Zou AP, Li N,Cowley AW, Jr. Production and actions of superoxide in the renal medulla[J]. Hypertension, 2001(37):547-553
    [26] Rueckschloss U, Galle J, Holtz J, et al. Induction of NAD(P)H oxidase by oxidizedlow-density lipoprotein in human endothelial cells: antioxidative potential of hydroxymethylglutaryl coenzyme A reductase inhibitor therapy[J]. Circulation, 2001(104):1767-1772
    [27] Zou AP,Cowley AW, Jr. Reactive oxygen species and molecular regulation of renal oxygenation[J]. Acta Physiol Scand, 2003(179):233-241
    [28] Kitada M, Koya D, Sugimoto T, et al. Translocation of glomerular p47phox and p67phox by protein kinase C-beta activation is required for oxidative stress in diabetic nephropathy[J]. Diabetes, 2003(52):2603-2614
    [29] Susztak K, Raff AC, Schiffer M, et al. Glucose-induced reactive oxygen species cause apoptosis of podocytes and podocyte depletion at the onset of diabetic nephropathy[J]. Diabetes, 2006(55):225-233
    [30] Baricos WH, Cortez SL, el-Dahr SS, et al. ECM degradation by cultured human mesangial cells is mediated by a PA/plasmin/MMP-2 cascade[J]. Kidney Int, 1995(47):1039-1047
    [31] Schnaper HW, Kopp JB, Poncelet AC, et al. Increased expression of extracellular matrix proteins and decreased expression of matrix proteases after serial passage of glomerular mesangial cells[J]. J Cell Sci, 1996(109 ( Pt 10)):2521-2528
    [32] Wilmer WA, Dixon CL,Hebert C. Chronic exposure of human mesangial cells to high glucose environments activates the p38 MAPK pathway[J]. Kidney Int, 2001(60):858-871
    [33] Lal MA, Brismar H, Eklof AC, et al. Role of oxidative stress in advanced glycation end product-induced mesangial cell activation[J]. Kidney Int, 2002(61):2006-2014
    [34]向红丁.糖尿病肾脏病变[J].国外医学内分泌学分册, 2004(24):125-136
    [35] Sugimoto H, Shikata K, Wada J, et al. Advanced glycation end products-cytokine-nitric oxide sequence pathway in the development of diabetic nephropathy: aminoguanidine ameliorates the overexpression of tumour necrosis factor-alpha and inducible nitric oxide synthase in diabetic rat glomeruli[J]. Diabetologia, 1999(42):878-886
    [36] Radeke HH, Meier B, Topley N, et al. Interleukin 1-alpha and tumor necrosis factor-alpha induce oxygen radical production in mesangial cells[J]. Kidney Int, 1990(37):767-775
    [37] Wolf G, Schroeder R, Ziyadeh FN, et al. High glucose stimulates expression of p27Kip1in cultured mouse mesangial cells: relationship to hypertrophy[J]. Am J Physiol, 1997(273):F348-356
    [38] Kuan CJ, al-Douahji M,Shankland SJ. The cyclin kinase inhibitor p21WAF1, CIP1 is increased in experimental diabetic nephropathy: potential role in glomerular hypertrophy[J]. J Am Soc Nephrol, 1998(9):986-993
    [39] Danilenko DM, Ring BD, Tarpley JE, et al. Growth factors in porcine full and partial thickness burn repair. Differing targets and effects of keratinocyte growth factor, platelet-derived growth factor-BB, epidermal growth factor, and neu differentiation factor[J]. Am J Pathol, 1995(147):1261-1277
    [40] Fabris B, Candido R, Carraro M, et al. Modulation of incipient glomerular lesions in experimental diabetic nephropathy by hypotensive and subhypotensive dosages of an ACE inhibitor[J]. Diabetes, 2001(50):2619-2624
    [41] D'Agord Schaan B, Lacchini S, Bertoluci MC, et al. Increased renal GLUT1 abundance and urinary TGF-beta 1 in streptozotocin-induced diabetic rats: implications for the development of nephropathy complicating diabetes[J]. Horm Metab Res, 2001(33):664-669
    [42] Clemmons DR, Moses AC, McKay MJ, et al. The combination of insulin-like growth factor I and insulin-like growth factor-binding protein-3 reduces insulin requirements in insulin-dependent type 1 diabetes: evidence for in vivo biological activity[J]. J Clin Endocrinol Metab, 2000(85):1518-1524
    [43] Flyvbjerg A, Khatir DS, Jensen LJ, et al. The involvement of growth hormone (GH), insulin-like growth factors (IGFs) and vascular endothelial growth factor (VEGF) in diabetic kidney disease[J]. Curr Pharm Des, 2004(10):3385-3394
    [44] Hirschberg R. The physiology and pathophysiology of IGF-I in the kidney[J]. Adv Exp Med Biol, 1993(343):345-366
    [45] Lupia E, Elliot SJ, Lenz O, et al. IGF-1 decreases collagen degradation in diabetic NOD mesangial cells: implications for diabetic nephropathy[J]. Diabetes, 1999(48):1638-1644
    [46] Heilig CW, Liu Y, England RL, et al. D-glucose stimulates mesangial cell GLUT1 expression and basal and IGF-I-sensitive glucose uptake in rat mesangial cells: implications for diabetic nephropathy[J]. Diabetes, 1997(46):1030-1039
    [47] Senger DR, Galli SJ, Dvorak AM, et al. Tumor cells secrete a vascular permeabilityfactor that promotes accumulation of ascites fluid[J]. Science, 1983(219):983-985
    [48] Feng D, Nagy JA, Hipp J, et al. Vesiculo-vacuolar organelles and the regulation of venule permeability to macromolecules by vascular permeability factor, histamine, and serotonin[J]. J Exp Med, 1996(183):1981-1986
    [49] Wolf G, Chen S,Ziyadeh FN. From the periphery of the glomerular capillary wall toward the center of disease: podocyte injury comes of age in diabetic nephropathy[J]. Diabetes, 2005(54):1626-1634
    [50] Amemiya T, Sasamura H, Mifune M, et al. Vascular endothelial growth factor activates MAP kinase and enhances collagen synthesis in human mesangial cells[J]. Kidney Int, 1999(56):2055-2063
    [51] Jiang T, Wang XX, Scherzer P, et al. Farnesoid X receptor modulates renal lipid metabolism, fibrosis, and diabetic nephropathy[J]. Diabetes, 2007(56):2485-2493
    [52] Aucella F, Margaglione M, Vigilante M, et al. PAI-1 4G/5G and ACE I/D gene polymorphisms and the occurrence of myocardial infarction in patients on intermittent dialysis[J]. Nephrol Dial Transplant, 2003(18):1142-1146
    [53] Lee EA, Seo JY, Jiang Z, et al. Reactive oxygen species mediate high glucose-induced plasminogen activator inhibitor-1 up-regulation in mesangial cells and in diabetic kidney[J]. Kidney Int, 2005(67):1762-1771
    [54] Jiang Z, Seo JY, Ha H, et al. Reactive oxygen species mediate TGF-beta1-induced plasminogen activator inhibitor-1 upregulation in mesangial cells[J]. Biochem Biophys Res Commun, 2003(309):961-966
    [55] Neuhofer W,Pittrow D. Role of endothelin and endothelin receptor antagonists in renal disease[J]. Eur J Clin Invest, 2006(36 Suppl 3):78-88
    [56] Fukui M, Nakamura T, Ebihara I, et al. Gene expression for endothelins and their receptors in glomeruli of diabetic rats[J]. J Lab Clin Med, 1993(122):149-156
    [57] Itoh Y, Nakai A, Kakizawa H, et al. Alteration of endothelin-1 concentration in STZ-induced diabetic rat nephropathy. Effects of a PGI(2) derivative[J]. Horm Res, 2001(56):165-171
    [58] Bruno CM, Meli S, Marcinno M, et al. Plasma endothelin-1 levels and albumin excretion rate in normotensive, microalbuminuric type 2 diabetic patients[J]. J Biol Regul Homeost Agents, 2002(16):114-117
    [59] Rabelink TJ, Stroes ES, Bouter KP, et al. Endothelin blockers and renal protection: a new strategy to prevent end-organ damage in cardiovascular disease?[J]. Cardiovasc Res, 1998(39):543-549
    [60] Lee YJ, Shin SJ,Tsai JH. Increased urinary endothelin-1-like immunoreactivity excretion in NIDDM patients with albuminuria[J]. Diabetes Care, 1994(17):263-266
    [61] Morabito E, Corsico N, Serafini S, et al. Elevated urinary excretion of endothelins in streptozotocin diabetic rats[J]. Life Sci, 1994(54):PL197-200
    [62] Ohta K, Hirata Y, Shichiri M, et al. Urinary excretion of endothelin-1 in normal subjects and patients with renal disease[J]. Kidney Int, 1991(39):307-311
    [63] Looker HC, Krakoff J, Funahashi T, et al. Adiponectin concentrations are influenced by renal function and diabetes duration in Pima Indians with type 2 diabetes[J]. J Clin Endocrinol Metab, 2004(89):4010-4017
    [64] Fujita H, Morii T, Koshimura J, et al. Possible relationship between adiponectin and renal tubular injury in diabetic nephropathy[J]. Endocr J, 2006(53):745-752
    [65] Okada S, Shikata K, Matsuda M, et al. Intercellular adhesion molecule-1-deficient mice are resistant against renal injury after induction of diabetes[J]. Diabetes, 2003(52):2586-2593
    [66] Clausen P, Jacobsen P, Rossing K, et al. Plasma concentrations of VCAM-1 and ICAM-1 are elevated in patients with Type 1 diabetes mellitus with microalbuminuria and overt nephropathy[J]. Diabet Med, 2000(17):644-649
    [67] Kitagawa K, Matsumoto M, Sasaki T, et al. Involvement of ICAM-1 in the progression of atherosclerosis in APOE-knockout mice[J]. Atherosclerosis, 2002(160):305-310
    [68] Sugimoto H, Shikata K, Hirata K, et al. Increased expression of intercellular adhesion molecule-1 (ICAM-1) in diabetic rat glomeruli: glomerular hyperfiltration is a potential mechanism of ICAM-1 upregulation[J]. Diabetes, 1997(46):2075-2081
    [69] Wenzel UO,Abboud HE. Chemokines and renal disease[J]. Am J Kidney Dis, 1995(26):982-994
    [70] Rovin BH, Doe N,Tan LC. Monocyte chemoattractant protein-1 levels in patients with glomerular disease[J]. Am J Kidney Dis, 1996(27):640-646
    [71] Saitoh A, Sekizuka K, Hayashi T, et al. Detection of urinary MCP-1 in patients with diabetic nephropathy[J]. Nephron, 1998(80):99
    [72] Ito Y, Aten J, Bende RJ, et al. Expression of connective tissue growth factor in human renal fibrosis[J]. Kidney Int, 1998(53):853-861
    [73] Gilbert RE, Akdeniz A, Weitz S, et al. Urinary connective tissue growth factor excretion in patients with type 1 diabetes and nephropathy[J]. Diabetes Care, 2003(26):2632-2636
    [74] Nguyen TQ, Tarnow L, Andersen S, et al. Urinary connective tissue growth factor excretion correlates with clinical markers of renal disease in a large population of type 1 diabetic patients with diabetic nephropathy[J]. Diabetes Care, 2006(29):83-88
    [75] Langham RG, Kelly DJ, Gow RM, et al. Transforming growth factor-beta in human diabetic nephropathy: effects of ACE inhibition[J]. Diabetes Care, 2006(29):2670-2675
    [76] Roestenberg P, van Nieuwenhoven FA, Wieten L, et al. Connective tissue growth factor is increased in plasma of type 1 diabetic patients with nephropathy[J]. Diabetes Care, 2004(27):1164-1170
    [77] Adler SG, Kang SW, Feld S, et al. Can glomerular mRNAs in human type 1 diabetes be used to predict transition from normoalbuminuria to microalbuminuria?[J]. Am J Kidney Dis, 2002(40):184-188
    [78] Ritz E,Orth SR. Nephropathy in patients with type 2 diabetes mellitus[J]. N Engl J Med, 1999(341):1127-1133
    [79] Sun L, Halaihel N, Zhang W, et al. Role of sterol regulatory element-binding protein 1 in regulation of renal lipid metabolism and glomerulosclerosis in diabetes mellitus[J]. J Biol Chem, 2002(277):18919-18927
    [80] Abrass CK. Cellular lipid metabolism and the role of lipids in progressive renal disease[J]. Am J Nephrol, 2004(24):46-53
    [81] Ruan XZ, Varghese Z, Powis SH, et al. Dysregulation of LDL receptor under the influence of inflammatory cytokines: a new pathway for foam cell formation[J]. Kidney Int, 2001(60):1716-1725
    [82] Hussain MM, Strickland DK,Bakillah A. The mammalian low-density lipoprotein receptor family[J]. Annu Rev Nutr, 1999(19):141-172
    [83] Podrez EA, Febbraio M, Sheibani N, et al. Macrophage scavenger receptor CD36 is the major receptor for LDL modified by monocyte-generated reactive nitrogen species[J]. J Clin Invest, 2000(105):1095-1108
    [84] Han J, Hajjar DP, Tauras JM, et al. Cellular cholesterol regulates expression of themacrophage type B scavenger receptor, CD36[J]. J Lipid Res, 1999(40):830-838
    [85]赵水平.高脂血症的病因及分类[J].中国冶金工业医学杂志, 1997(14):289-295
    [86]王爱民. 2型糖尿病肾病危险因素分析[J].中国现代医学杂志, 2005(15):116
    [87]赵亮.糖尿病肾病进展与血脂异常的关系[J].河北医学, 2008(11):316
    [88] Marsh JB. Lipoprotein metabolism in experimental nephrosis[J]. J Lipid Res, 1984(25):1619-1623
    [89] Appel G. Lipid abnormalities in renal disease[J]. Kidney Int, 1991(39):169-183
    [90] Cheung AK, Parker CJ, Ren K, et al. Increased lipase inhibition in uremia: identification of pre-beta-HDL as a major inhibitor in normal and uremic plasma[J]. Kidney Int, 1996(49):1360-1371
    [91] Proctor G, Jiang T, Iwahashi M, et al. Regulation of renal fatty acid and cholesterol metabolism, inflammation, and fibrosis in Akita and OVE26 mice with type 1 diabetes[J]. Diabetes, 2006(55):2502-2509
    [92] Sohn M, Tan Y, Klein RL, et al. Evidence for low-density lipoprotein-induced expression of connective tissue growth factor in mesangial cells[J]. Kidney Int, 2005(67):1286-1296
    [93] JOLES JA, KUNTER U, JANSSEN U, et al. Early Mechanisms of Renal Injury in Hypercholesterolemic or Hypertriglyceridemic Rats[J]. Journal of the American Society of Nephrology, 2000(11):669-683
    [94] Nishida Y, Oda H,Yorioka N. Effect of lipoproteins on mesangial cell proliferation[J]. Kidney Int Suppl, 1999(71):S51-53
    [95] Sharma K, Jin Y, Guo J, et al. Neutralization of TGFbeta by anti-TGF-beta antibody attenuates kidney hypertrophy and the enhanced extracellular matrix gene expression in STZ-induced diabetic mice [J]. Diabetes, 1996 (45 ):522–530
    [96] Stevenson FT, Shearer GC,Atkinson DN. Lipoprotein-stimulated mesangial cell proliferation and gene expression are regulated by lipoprotein lipase[J]. Kidney Int, 2001(59):2062-2068
    [97] Ikezumi Y, Hurst LA, Masaki T, et al. Adoptive transfer studies demonstrate that macrophages can induce proteinuria and mesangial cell proliferation[J]. Kidney Int, 2003(63):83-95
    [98] Sugimoto K, Isobe K, Kawakami Y, et al. The relationship between non-HDL cholesteroland other lipid parameters in Japanese subjects[J]. J Atheroscler Thromb, 2005(12):107-110
    [99] Bussolati B, Deregibus MC, Fonsato V, et al. Statins prevent oxidized LDL-induced injury of glomerular podocytes by activating the phosphatidylinositol 3-kinase/AKT-signaling pathway[J]. J Am Soc Nephrol, 2005(16):1936-1947
    [100] Bonnet F,Cooper ME. Potential influence of lipids in diabetic nephropathy: insights from experimental data and clinical studies[J]. Diabetes Metab, 2000(26):254-264
    [101] Nelson RG, Knowler WC, Pettitt DJ, et al. Incidence and determinants of elevated urinary albumin excretion in Pima Indians with NIDDM[J]. Diabetes Care, 1995(18):182-187
    [102] Ravid M, Brosh D, Ravid-Safran D, et al. Main risk factors for nephropathy in type 2 diabetes mellitus are plasma cholesterol levels, mean blood pressure, and hyperglycemia[J]. Arch Intern Med, 1998(158):998-1004
    [103] Dubois D, Chanson P, Timsit J, et al. Remission of proteinuria following correction of hyperlipidemia in NIDDM patients with nondiabetic glomerulopathy[J]. Diabetes Care, 1994(17):906-908
    [104] DeFronzo RA,Ferrannini E. Insulin resistance. A multifaceted syndrome responsible for NIDDM, obesity, hypertension, dyslipidemia, and atherosclerotic cardiovascular disease[J]. Diabetes Care, 1991(14):173-194
    [105] Bligh EG,Dyer WJ. A rapid method of total lipid extraction and purification[J]. Can J Biochem Physiol, 1959(37):911-917
    [106]马世平,章梅,王宗仁.活血通脉片对高血压患者血脂、血液流变性和细胞流变性的临床疗效观察[J].第四军医大学学报, 2001(21):292-294
    [107] Brewer HB, Jr. Hypertriglyceridemia: changes in the plasma lipoproteins associated with an increased risk of cardiovascular disease[J]. Am J Cardiol, 1999(83):3F-12F
    [108]刘雪梅,吴符火.几类高脂血症动物模型的比较[J].中西医结合学报, 2004(2):132
    [109] Wang PR, Guo Q, Ippolito M, et al. High fat fed hamster, a unique animal model for treatment of diabetic dyslipidemia with peroxisome proliferator activated receptor alpha selective agonists[J]. Eur J Pharmacol, 2001(427):285-293
    [110] Dorfman SE, Smith DE, Osgood DP, et al. Study of diet-induced changes in lipoprotein metabolism in two strains of Golden-Syrian hamsters[J]. J Nutr, 2003(133):4183-4188
    [111] Sessions VA,Salter AM. The effects of different dietary fats and cholesterol on serum lipoprotein concentrations in hamsters[J]. Biochim Biophys Acta, 1994(1211):207-214
    [112] Ebara T, Hirano T, Mamo JC, et al. Hyperlipidemia in streptozocin-diabetic hamsters as a model for human insulin-deficient diabetes: comparison to streptozocin-diabetic rats[J]. Metabolism, 1994(43):299-305
    [113] Fungwe TV, Fox JE, Cagen LM, et al. Stimulation of fatty acid biosynthesis by dietary cholesterol and of cholesterol synthesis by dietary fatty acid[J]. J Lipid Res, 1994(35):311-318
    [114] Fungwe TV, Cagen LM, Cook GA, et al. Dietary cholesterol stimulates hepatic biosynthesis of triglyceride and reduces oxidation of fatty acids in the rat[J]. J Lipid Res, 1993(34):933-941
    [115]杨文英.应重视糖尿病伴有的高甘油三酯血症的危害[J].新医学, 1998(29 ):229
    [116] Srour, Bilto YY, Juma M, et al. Exposure of human erythrocytes to oxygen radicals causes loss of deformability, increased osmotic fragility, lipid peroxidation and protein degradation[J]. Clin Hemorheol Microcirc, 2000(23):13-21
    [117] Koenig W,Ernst E. The possible role of hemorheology in atherothrombogenesis[J]. Atherosclerosis, 1992(94):93-107
    [118] Wen Z, Yao W, Xie L, et al. Influence of neuraminidase on the characteristics of microrheology of red blood cells[J]. Clin Hemorheol Microcirc, 2000(23):51-57
    [119] Wild S, Roglic G, Green A, et al. Global prevalence of diabetes: estimates for the year 2000 and projections for 2030[J]. Diabetes Care, 2004(27):1047-1053
    [120] Dominguez JH, Tang N, Xu W, et al. Studies of renal injury III: lipid-induced nephropathy in type II diabetes[J]. Kidney Int, 2000(57):92-104
    [121] Vaziri ND. Dyslipidemia of chronic renal failure: the nature, mechanisms, and potential consequences[J]. Am J Physiol Renal Physiol, 2006(290):F262-272
    [122] Watts GF, Powrie JK, O'Brien SF, et al. Apolipoprotein B independently predicts progression of very-low-level albuminuria in insulin-dependent diabetes mellitus[J]. Metabolism, 1996(45):1101-1107
    [123] Orchard TJ, Chang YF, Ferrell RE, et al. Nephropathy in type 1 diabetes: a manifestation of insulin resistance and multiple genetic susceptibilities? Further evidence from the Pittsburgh Epidemiology of Diabetes Complication Study[J]. KidneyInt, 2002(62):963-970
    [124] Smulders YM, Rakic M, Stehouwer CD, et al. Determinants of progression of microalbuminuria in patients with NIDDM.A prospective study[J]. Diabetes Care, 1997(20):999-1005
    [125] Wirta OR, Pasternack AI, Mustonen JT, et al. Urinary albumin excretion rate and its determinants after 6 years in non-insulin-dependent diabetic patients[J]. Nephrol Dial Transplant, 1996(11):449-456
    [126] Oue T, Namba M, Nakajima H, et al. Risk factors for the progression of microalbuminuria in Japanese type 2 diabetic patients--a 10 year follow-up study[J]. Diabetes Res Clin Pract, 1999(46):47-55
    [127] Guijarro C, Kasiske BL, Kim Y, et al. Early glomerular changes in rats with dietary-induced hypercholesterolemia[J]. Am J Kidney Dis, 1995(26):152-161
    [128] Kasiske BL, O'Donnell MP, Schmitz PG, et al. Renal injury of diet-induced hypercholesterolemia in rats[J]. Kidney Int, 1990(37):880-891
    [129] Hattori M, Nikolic-Paterson DJ, Miyazaki K, et al. Mechanisms of glomerular macrophage infiltration in lipid-induced renal injury[J]. Kidney Int Suppl, 1999(71):S47-50
    [130] Muntner P, Coresh J, Smith JC, et al. Plasma lipids and risk of developing renal dysfunction: the atherosclerosis risk in communities study[J]. Kidney Int, 2000(58):293-301
    [131] Aligeti VR, Gandhi M, Braden R, et al. Effect of combination lipid-modifying therapy on the triglyceride lowering effect of fish oil[J]. Am J Med Sci, 2007(333):168-172
    [132] Billett MA, Bruce JS, White DA, et al. Interactive effects of dietary cholesterol and different saturated fatty acids on lipoprotein metabolism in the hamster[J]. Br J Nutr, 2000(84):439-447
    [133] McAteer MA, Grimsditch DC, Vidgeon-Hart M, et al. Dietary cholesterol reduces lipoprotein lipase activity in the atherosclerosis-susceptible Bio F(1)B hamster[J]. Br J Nutr, 2003(89):341-350
    [134] Lally S, Owens D,Tomkin GH. Genes that affect cholesterol synthesis, cholesterol absorption, and chylomicron assembly: the relationship between the liver and intestine in control and streptozotosin diabetic rats[J]. Metabolism, 2007(56):430-438
    [135] Gleeson A, Anderton K, Owens D, et al. The role of microsomal triglyceride transfer protein and dietary cholesterol in chylomicron production in diabetes[J]. Diabetologia, 1999(42):944-948
    [136] Sugano M, Yamato H, Hayashi T, et al. High-fat diet in low-dose-streptozotocin-treated heminephrectomized rats induces all features of human type 2 diabetic nephropathy: a new rat model of diabetic nephropathy[J]. Nutr Metab Cardiovasc Dis, 2006(16):477-484
    [137] Shimano H. Sterol regulatory element-binding proteins (SREBPs): transcriptional regulators of lipid synthetic genes[J]. Prog Lipid Res, 2001(40):439-452
    [138] Lindenmeyer MT, Kretzler M, Boucherot A, et al. Interstitial vascular rarefaction and reduced VEGF-A expression in human diabetic nephropathy[J]. J Am Soc Nephrol, 2007(18):1765-1776
    [139] Iglesias-De La Cruz MC, Ruiz-Torres P, Alcami J, et al. Hydrogen peroxide increases extracellular matrix mRNA through TGF-beta in human mesangial cells[J]. Kidney Int, 2001(59):87-95
    [140] Studer RK, Craven PA,DeRubertis FR. Antioxidant inhibition of protein kinase C-signaled increases in transforming growth factor-beta in mesangial cells[J]. Metabolism, 1997(46):918-925

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700