Добавил:
Опубликованный материал нарушает ваши авторские права? Сообщите нам.
Вуз: Предмет: Файл:

Bioregenerative Engineering Principles and Applications - Shu Q. Liu

..pdf
Скачиваний:
119
Добавлен:
10.08.2013
Размер:
26.55 Mб
Скачать

716 VASCULAR REGENERATIVE ENGINEERING

Bloch K, Eddy RL, Shows TB, Quertermous T: cDNA cloning and chromosomal assignment of the gene encoding endothelin 3, J Biol Chem 264:18156–61, 1989.

Bolk S, Angrist M, Xie J, Yanagisawa M, Silvestri JM et al: Endothelin-3 frameshift mutation in congenital central hypoventilation syndrome, Nature Genet 13:395–6, 1996.

Dupin E, Glavieux C, Vaigot P, Le Douarin NM: Endothelin 3 induces the reversion of melanocytes to glia through a neural crest-derived glial-melanocytic progenitor, Proc Natl Acad Sci USA 97:7882–7, 2000.

Gopal Rao VVN, Loffler C, Hansmann I: The gene for the novel vasoactive peptide endothelin 3 (EDN3) is localized to human chromosome 20q13.2-qter, Genomics 10:840–1, 1991.

Lee S, Lin M, Mele A, Cao Y, Farmar J et al: Proteolytic processing of big endothelin-3 by the Kell blood group protein, Blood 94:1440–50, 1999.

Endothelin Receptor A

Bourgeois C, Robert B, Rebourcet R, Mondon F, Mignot TM et al: Endothelin-1 and ET(A) receptor expression in vascular smooth muscle cells from human placenta: A new ET(A) receptor messenger ribonucleic acid is generated by alternative splicing of exon 3, J Clin Endocr Metab 82:3116–23, 1997.

Campia U, Cardillo C, Panza JA: Ethnic differences in the vasoconstrictor activity of endogenous endothelin-1 in hypertensive patients, Circulation 109:3191–5, 2004.

Hosoda K, Nakao K, Tamura N, Arai H, Ogawa Y et al: Organization structure chromosomal assignment and expression of the gene encoding the human endothelin-A receptor, J Biol Chem 267:18797–804, 1992.

Maggi M, Barni T, Fantoni G, Mancina R, Pupilli C et al: Expression and biological effects of endothelin-1 in human gonadotropin-releasing hormone-secreting neurons, J Clin Endocr Metab 85:1658–65, 2000.

Maurer M, Wedemeyer J, Metz M, Piliponsky AM, Weller K et al: Mast cells promote homeostasis by limiting endothelin-1-induced toxicity, Nature 432:512–6, 2004.

Tzourio C, El Amrani M, Poirier O, Nicaud V, Bousser MG et al: Association between migraine and endothelin type A receptor (ETA -231 A/G) gene polymorphism, Neurology 56:1273–7, 2001.

Endothtelin Receptor B

Arai H, Nakao K, Takaya K, Hosoda K, Ogawa Y et al: The human endothelin-B receptor gene: Structural organization and chromosomal assignment, J Biol Chem 268:3463–70, 1993.

Baynash AG, Hosoda K, Giaid A, Richardson JA, Emoto N et al: Interaction of endothelin-3 with endothelin-B receptor is essential for development of epidermal melanocytes and enteric neurons, Cell 79:1277–85, 1994.

Carpenter T, Schomberg S, Steudel W, Ozimek J, Colvin K et al: Endothelin B receptor deficiency predisposes to pulmonary edema formation via increased lung vascular endothelial cell growth factor expression, Circ Res 93:456–63, 2003.

Ceccherini I, Zhang AL, Matera I, Yang G, Devoto M et al: Interstitial deletion of the endothelin-B receptor gene in the spotting lethal (sl) rat. Hum Mol Genet 4:2089–96, 1995.

Elshourbagy NA, Adamou JE, Gagnon AW, Wu HL, Pullen M et al: Molecular characterization of a novel human endothelin receptor splice variant, J Biol Chem 271:25300–7, 1996.

Gariepy CE, Cass DT, Yanagisawa M: Null mutation of endothelin receptor type B gene in spotting lethal rats causes aganglionic megacolon and white coat color, Proc Natl Acad Sci USA 93:867– 72, 1996.

Gariepy CE, Ohuchi T, Williams SC, Richardson JA, Yanagisawa M: Salt-sensitive hypertension in endothelin-B receptor-deficient rats, J Clin Invest 105:925–33, 2000.

BIBLIOGRAPHY 717

Gariepy CE, Williams SC, Richardson JA, Hammer RE, Yanagisawa M: Transgenic expression of the endothelin-B receptor prevents congenital intestinal aganglionosis in a rat model of Hirschsprung disease, J Clin Invest 102:1092–101, 1998.

Hosoda K, Hammer RE, Richardson JA, Baynash AG, Cheung JC et al: Targeted and natural (piebald-lethal) mutations of endothelin-B receptor gene produce megacolon associated with spotted coat color in mice, Cell 79:1267–76, 1994.

Iwashita T, Kruger GM, Pardal R, Kiel MJ, Morrison SJ: Hirschsprung disease is linked to defects in neural crest stem cell function, Science 301:972–6, 2003.

Khodorova A, Navarro B, Jouaville LS, Murphy JE, Rice FI et al: Endothelin-B receptor activation triggers an endogenous analgesic cascade at sites of peripheral injury, Nature Med 9:1055–61, 2003.

Shin MK, Levorse JM, Ingram RS, Tilghman SM: The temporal requirement for endothelin receptor-B signalling during neural crest development, Nature 402:496–501, 1999.

Shin MK, Russell LB, Tilghman SM: Molecular characterization of four induced alleles at the Ednrb locus, Proc Natl Acad Sci USA 94:13105–10, 1997.

Nishiyama M, Masaki T, Yanagisawa M, Sekiya S, Kimura S: Novel mutations of the endothelin B receptor gene in patients with Hirschsprung’s disease and their characterization, J Biol Chem 273:11378–83, 1998.

Yang LL, Gros R, Kabir MG, Sadi A, Gotlieb AI et al: Conditional cardiac overexpression of endothe- lin-1 induces inflammation and dilated cardiomyopathy in mice, Circulation 109:255–61, 2004.

Zhu L, Lee HO, Jordan CS, Cantrell VA, Southard-Smith EM et al: Spatiotemporal regulation of endothelin receptor-B by SOX10 in neural crest-derived enteric neuron precursors, Nature Genet 36:732–7, 2004.

Human protein reference data base, Johns Hopkins University and the Institute of Bioinformatics, at http://www.hprd.org/protein.

15.4. Regulation of Leukocytes and Platelet Adhesion

MCP1

Ambati J, Anand A, Fernandez S, Sakurai E, Lynn BC et al: An animal model of age-related macular degeneration in senescent Ccl-2- or Ccr-2-deficient mice, Nature Med 9:1390–7, 2003.

Corrigall VM, Arastu M, Khan S, Shah C, Fife M et al: Functional IL-2 receptor beta (CD122) and gamma (CD132) chains are expressed by fibroblast-like synoviocytes: Activation by IL-2 stimulates monocyte chemoattractant protein-1 production, J Immun 166:4141–7, 2001.

Galindo M, Santiago B, Rivero M, Rullas J, Alcami J et al: Chemokine expression by systemic sclerosis fibroblasts: Abnormal regulation of monocyte chemoattractant protein 1 expression, Arthritis Rheum 44:1382–6, 2001.

Gosling J, Slaymaker S, Gu L, Tseng S, Zlot CH et al: MCP-1 deficiency reduces susceptibility to atherosclerosis in mice that overexpress human apolipoprotein B, J Clin Invest 103:773–8, 1999.

Gu L, Tseng S, Horner RM, Tam C, Loda M et al: Control of TH2 polarization by the chemokine monocyte chemoattractant protein-1, Nature 404:407–11, 2000.

Lu B, Rutledge BJ, Gu L, Fiorillo J, Lukacs NW et al: Abnormalities in monocyte recruitment and cytokine expression in monocyte chemoattractant protein 1-deficient mice, J Exp Med 187:601– 8, 1998.

Sartipy P, Loskutoff DJ: Monocyte chemoattractant protein 1 in obesity and insulin resistance, Proc Natl Acad Sci USA 100:7265–70, 2003.

15.5. Smooth Muscle Cells

Arner A, Malmqvist U: Cross-bridge cycling in smooth muscle: A short review, Acta Physiol Scand 164(4):363–72, 1998.

718 VASCULAR REGENERATIVE ENGINEERING

Kohama K, Ye LH, Hayakawa K, Okagaki T: Myosin light chain kinase: An actin-binding protein that regulates an ATP-dependent interaction with myosin, Trends Pharmacol Sci 8:284–7, 1996.

Winder SJ, Allen BG, Clement-Chomienne O, Walsh MP: Regulation of smooth muscle actinmyosin interaction and force by calponin, Acta Physiol Scand 164(4):415–26, 1998.

Liu SQ, Fung YC: Relationship between hypertension hypertrophy and opening angle of zero-stress state of arteries following aortic constriction, J Biomech Eng 111:325–35, 1989.

Liu SQ, Fung YC: Influence of STZ-diabetes on zero-stress state of rat pulmonary and systemic arteries, Diabetes 41:136–46, 1992.

Liu SQ, Fung YC: Material coefficients of the strain energy function of pulmonary arteries in normal and cigarette smoke-exposed rats, J Biomech 26:1261–9, 1993.

Liu SQ, Fung YC: Changes in the structure and mechanical properties of pulmonary arteries of rats exposed to cigarette smoke, Am Rev Resp Dis 148:768–77, 1993.

Fung YC, Liu SQ: Determination of the mechanical properties of the different layers of blood vessels in vivo, Proc Natl Acad Sci USA 92:2169–73, 1995.

Liu SQ, Fung YC: Indicial functions of arterial remodeling in response to locally altered mechanical stress, Am J Physiol 270: H1323–33, 1996.

Liu SQ: Alterations in structure of elastic laminae of rat pulmonary arteries in hypoxic hypertension, J Appl Physiol 81:2147–55, 1996.

Liu SQ: Regression of hypoxic hypertension-induced changes in the elastic laminae of rat pulmonary arteries, J Appl Physiol 82:1677–84, 1997.

Liu SQ: Influence of tensile strain on smooth muscle cell orientation in rat blood vessels, J Biomech Eng 120:313–20, 1998.

Liu SQ: Biomechanical basis of vascular tissue engineering, Cri Rev Biomed Eng 27:75–148, 1999.

Liu SQ, Moore MM, Glucksberg MR, Mockros LF, Grotberg JB et al: Partial prevention of monocyte and granulocyte activation in experimental vein grafts using a biomechanical engineering approach, J Biomech 32:1165–75, 1999.

Liu SQ, Moore MM, Yap C: Prevention of tensile stress-induced endothelial and smooth muscle cell injury in experimental vein grafts, J Biomech Eng 122:31–8, 2000.

Moore MM, Goldman J, Patel A, Chien S, Liu SQ: Role of mechanical stretch in the induction of vascular cell death, J Biomech 34:289–97, 2001.

Goldman J, Zhong L, Liu SQ: Degradation of α actin filaments in vascular smooth muscle cells in response to mechanical stretch, Am J Physiol 284:H1839–47, 2003.

15.6. Regulation by the Renin–Angiotensin–Aldosterone System

Renin

Caron KMI, James LR, Kim HS, Knowles J, Uhlir R et al: Cardiac hypertrophy and sudden death in mice with a genetically clamped renin transgene, Proc Natl Acad Sci USA 101:3106–11, 2004.

Frossard PM, Lestringant GG, Malloy MJ, Kane JP: Human renin gene BglI dimorphism associated with hypertension in two independent populations, Clin Genet 56:428–33, 1999.

Gribouval O, Gonzales M, Neuhaus T, Aziza J, Bieth E et al: Mutations in genes in the reninangiotensin system are associated with autosomal recessive renal tubular dysgenesis, Nature Genet 37:964–8, 2005.

Nguyen G, Delarue F, Burckle C, Bouzhir L, Giller T et al: Pivotal role of the renin/prorenin receptor in angiotensin II production and cellular responses to renin, J Clin Invest 109:1417–27, 2002.

BIBLIOGRAPHY 719

Qin H, Chen YH, Yip MY, Lam-Po-Tang PRL, Morris BJ: Reassignment of human renin gene to chromosome 1q32 in studies of a (1;4)(q42;p16) translocation, Hum Hered 43:261–4, 1993.

van Hooft IMS, Grobbee DE, Derkx FHM, de Leeuw PW, Schalekamp MADH et al: Renal hemodynamics and the renin-angiotensin-aldosterone system in normotensive subjects with hypertensive and normotensive parents, New Engl J Med 324:1305–11, 1991.

Villard E, Lalau JD, van Hooft IS, Derkx FHM, Houot A-M et al: A mutant renin gene in familial elevation of prorenin, J Biol Chem 269:30307–12, 1994.

Angiotensin I

Bloem LJ, Manatunga AK, Tewksbury DA, Pratt JH: The serum angiotensinogen concentration and variants of the angiotensinogen gene in white and black children, J Clin Invest 95:948–53, 199.

Caulfield M, Lavender P, Farrall M, Munroe P, Lawson M et al: Linkage of the angiotensinogen gene to essential hypertension, New Engl J Med 330:1629–33, 1994.

Caulfield M, Lavender P, Newell-Price J, Farrall M, Kamdar S et al: Linkage of the angiotensinogen gene locus to human essential hypertension in African Caribbeans, J Clin Invest 96:687–92, 1995.

Davisson RL, Kim HS, Krege JH, Lager DJ, Smithies O et al: Complementation of reduced survival hypotension, and renal abnormalities in angiotensinogen-deficient mice by the human renin and human angiotensinogen genes, J Clin Invest 99:1258–64, 1997.

Ding Y, Stec DE, Sigmund CD: Genetic evidence that lethality in angiotensinogen-deficient mice is due to loss of systemic but not renal angiotensinogen, J Biol Chem 276:7431–6, 2001.

Gribouval O, Gonzales M, Neuhaus T, Aziza J, Bieth E et al: Mutations in genes in the reninangiotensin system are associated with autosomal recessive renal tubular dysgenesis, Nature Genet 37:964–8, 2005.

Hata A, Namikawa C, Sasaki M, Sato K, Nakamura T et al: Angiotensinogen as a risk factor for essential hypertension in Japan, J Clin Invest 93:1285–7, 1994.

Inoue I, Nakajima T, Williams CS, Quackenbush J, Puryear R et al: A nucleotide substitution in the promoter of human angiotensinogen is associated with essential hypertension and affects basal transcription in vitro, J Clin Invest 99:1786–97, 1997.

Jeunemaitre X, Soubrier F, Kotelevtsev YV, Lifton RP, Williams CS et al: Molecular basis of human hypertension: Role of angiotensinogen, Cell 71:7–20, 1992.

Katsuya T, Koike G, Yee TW, Sharpe N, Jackson R et al: Association of angiotensinogen gene T235 variant with increased risk of coronary heart disease, Lancet 345:1600–3, 1995.

Sadoshima J, Xu Y, Slayter HS, Izumo S: Autocrine release of angiotensin II mediates stretchinduced hypertrophy of cardiac myocytes in vitro, Cell 75:977–84, 1993.

Tanimoto K, Sugiyama F, Goto Y, Ishida J, Takimoto E et al: Angiotensinogen-deficient mice with hypotension, J Biol Chem 269:31334–7, 1994.

Angiotensin-Converting Enzyme

Cambien F, Poirier O, Lecerf L, Evans A, Cambou JP et al: Deletion polymorphism in the gene for angiotensin-converting enzyme is a potent risk factor for myocardial infarction, Nature 359:641–4, 1992.

Doria A, Warram JH, Krolewski AS: Genetic predisposition to diabetic nephropathy: Evidence for a role of the angiotensin I-converting enzyme gene, Diabetes 43:690–5, 1994.

Esther CR Jr, Marino EM, Howard TE, Machaud A, Corvol P et al: The critical role of tissue angiotensin-converting enzyme as revealed by gene targeting in mice, J Clin Invest 99:2375–85, 1997.

720 VASCULAR REGENERATIVE ENGINEERING

Jeunemaitre X, Lifton RP, Hunt SC, Williams RR, Lalouel JM: Absence of linkage between the angiotensin converting enzyme locus and human essential hypertension, Nature Genet 1:72–5, 1992.

Keavney B, McKenzie C, Parish S, Palmer A, Clark S et al: Large-scale test of hypothesized associations between the angiotensin-converting-enzyme insertion/deletion polymorphism and myocardial infarction in about 5000 cases and 6000 controls, Lancet 355:434–42, 2000.

Kessler SP, deS Senanayake P, Scheidemantel TS, Gomos JB, Rowe TM et al: Maintenance of normal blood pressure and renal functions are independent effects of angiotensin-converting enzyme, J Biol Chem 278:21105–12, 2003.

Kramers C, Danilov SM, Deinum J, Balyasnikova IV, Scharenborg N et al: Point mutation in the stalk of angiotensin-converting enzyme causes a dramatic increase in serum angiotensinconverting enzyme but no cardiovascular disease, Circulation 104:1236–40, 2001.

Lindpaintner K, Lee M, Larson MG, Rao VS, Pfeffer MA et al: Absence of association or genetic linkage between the angiotensin-converting-enzyme gene and left ventricular mass, New Engl J Med 334:1023–8, 1996.

Lindpaintner K, Pfeffer MA, Kreutz R, Stampfer MJ, Grodstein F et al: A prospective evaluation of an angiotensin-converting-enzyme gene polymorphism and the risk of ischemic heart disease, New Engl J Med 332:706–11, 1995.

Marre M, Jeunemaitre X, Gallois Y, Rodier M, Chatellier G et al: Contribution of genetic polymorphism in the renin-angiotensin system to the development of renal complications in insulindependent diabetes, J Clin Invest 99:1585–95, 1997.

Montgomery HE, Clarkson P, Dollery CM, Prasad K, Losi MA et al: Association of angiotensinconverting enzyme gene I/D polymorphism with change in left ventricular mass in response to physical training, Circulation 96:741–7, 1997.

Natesh R, Schwager SLU, Sturrock ED, Acharya KR: Crystal structure of the human angiotensinconverting enzyme-lisinopril complex, Nature 421:551–4, 2003.

Ohishi M, Fujii K, Minamino T, Hagaki J, Kamitani A et al: A potent genetic risk factor for restenosis [Letter], Nature Genet 5:324–5, 1993.

Oike Y, Hata A, Ogata Y, Numata Y, Shido K et al: Angiotensin converting enzyme as a genetic risk factor for coronary artery spasm: implication in the pathogenesis of myocardial infarction, J Clin Invest 96:2975–9, 1995.

Ramaraj P, Kessler SP, Colmenares C, Sen GC: Selective restoration of male fertility in mice lacking angiotensin-converting enzymes by sperm-specific expression of the testicular isozyme, J Clin Invest 102:371–8, 1998.

AT1

AbdAlla S, Lother H, Langer A, el Faramawy Y, Quitterer U: Factor XIIIA transglutaminase crosslinks AT1 receptor dimers of monocytes at the onset of atherosclerosis, Cell 119:343–54, 2004.

AbdAlla S, Lother H, Quitterer U: AT(1)-receptor heterodimers show enhanced G-protein activation and altered receptor sequestration, Nature 407:94–8, 2000.

Bonnardeaux A, Davies E, Jeunemaitre X, Fery I, Charru A et al: Angiotensin II type 1 receptor gene polymorphisms in human essential hypertension, Hypertension 24:63–9, 1994.

Haywood GA, Gullestad L, Katsuya T, Hutchinson HG, Pratt RE et al: AT(1) and AT(2) angiotensin receptor gene expression in human heart failure, Circulation 95:1201–6, 1997.

Herzig TC, Jobe SM, Aoki H, Molkentin JD, Cowley AW Jr et al: Angiotensin II type-1a receptor gene expression in the heart: AP-1 and GATA-4 participate in the response to pressure overload,

Proc Natl Acad Sci USA 94:7543–8, 1997.

BIBLIOGRAPHY 721

Kambayashi Y, Bardhan S, Takahashi K, Tsuzuki S, Inui H et al: Molecular cloning of a novel angiotensin II receptor isoform involved in phosphotyrosine phosphatase inhibition, J Biol Chem 268:24543–6, 1993.

Murphy TJ, Alexander RW, Griendling KK, Runge MS, Bernstein KE: Isolation of a cDNA encoding the vascular type-1 angiotensin II receptor, Nature 351:233–6, 1991.

Sasaki K, Murohara T, Ikeda H, Sugaya T, Shimada T et al: Evidence for the importance of angiotensin II type 1 receptor in ischemia-induced angiogenesis, J Clin Invest 109:603–11, 2002.

Sasaki K, Yamano Y, Bardhan S, Iwai N, Murray JJ et al: Cloning and expression of a complementary DNA encoding a bovine adrenal angiotensin II type-1 receptor, Nature 351:230–3, 1991.

Human protein reference data base, Johns Hopkins University and the Institute of Bioinformatics, at http://www.hprd.org/protein.

15.7. Pathogenesis, Pathology, and Clinical Features of Atherosclerosis

Ross R: Atherosclerosis—an inflammatory disease, New Engl J Med 340(2):115–26, 1999.

Ross R: The pathogenesis of atherosclerosis: A perspective for the 1990s, Nature 362(6423):801–9, 1993.

Schneider AS, Szanto PA: Pathology, 3rd ed, Lippincott Williams & Wilkins Philadelphia, 2006.

McCance KL, Huether SE: Pathophysiology: The Biologic Basis for Disease in Adults & Children, 5th ed, Elsevier Mosby, St Louis, 2006.

Porth CM: Pathophysiology: Concepts of Altered Health States, 7th ed, Lippincott Williams & Wilkins, Philadelphia, 2005.

Frazier MS, Drzymkowski JW: Essentials of Human Diseases and Conditions, 3rd ed, Elsevier Saunders, St Louis, 2004.

15.8. Animal Models of Atherosclerosis

Xu Q: Mouse models of arteriosclerosis: from arterial injuries to vascular grafts, Am J Pathol 165:1–10, 2004.

Meir KS, Leitersdorf E: Atherosclerosis in the apolipoprotein E-deficient mouse: A decade of progress, Arterioscler Thromb Vasc Biol 24:1006–14, 2004.

Leidenfrost JE, Khan MF, Boc KP, Villa BR, Collins ET et al: A model of primary atherosclerosis and post-angioplasty restenosis in mice, Am J Pathol 163(2):773–8, 2003.

15.9. Conventional Treatment of Atherosclerosis

Gershlick AH: Treating atherosclerosis: Local drug delivery from laboratory studies to clinical trials, Atherosclerosis 160:259–71, 2002.

Jordan MA, Toso RJ, Thrower D, Wilson L: Mechanism of mitotic block and inhibition of cell proliferation by taxol at low concentrations, Proc Natl Acad Sci USA 90:9552–6, 1993.

Axel DI et al: Paclitaxel inhibits arterial smooth muscle cell proliferation and migration in vitro and in vivo using local drug delivery, Circulation 96:636–45, 1997.

Liistro F et al: First clinical experience with a paclitaxel derivate-eluting polymer stent system implantation for in-stent restenosis: Immediate and long-term clinical and angiographic outcome, Circulation 105:1883–6, 2002.

Dzau VJ, Braun-Dullaeus RC, Sedding DG: Vascular proliferation and atherosclerosis: New perspectives and therapeutic strategies, Nat Med 8(11):1249–56, 2002.

Sehgal SN, Baker H, Vezina C: Rapamycin (AY-22,989), a new antifungal antibiotic. II. Fermentation, isolation and characterization, J Antibiot (Tokyo) 28:727–32, 1975.

722 VASCULAR REGENERATIVE ENGINEERING

Gregory CR, Huie P, Billingham ME, Morris RE: Rapamycin inhibits arterial intimal thickening caused by both alloimmune and mechanical injury. Its effect on cellular, growth factor, and cytokine response in injured vessels, Transplantation 55:1409–18, 1993.

Braun-Dullaeus RC et al: Cell cycle protein expression in vascular smooth muscle cells in vitro and in vivo is regulated through phosphatidylinositol 3-kinase and mammalian target of rapamycin, Arterioscler Thromb Vasc Biol 21:1152–8, 2001.

Gallo R et al: Inhibition of intimal thickening after balloon angioplasty in porcine coronary arteries by targeting regulators of the cell cycle, Circulation 99:2164–70, 1999.

Grise MA et al: Five-year clinical follow-up after intracoronary radiation: results of a randomized clinical trial, Circulation 105:2737–40, 2002.

Waksman R et al: Intracoronary gamma-radiation therapy after angioplasty inhibits recurrence in patients with in-stent restenosis, Circulation 101:2165–71, 2000.

Scott S, O’Sullivan M, Hafizi S, Shapiro LM, Bennett MR: Human vascular smooth muscle cells from restenosis or in-stent stenosis sites demonstrate enhanced responses to p53: Implications for brachytherapy and drug treatment for restenosis, Circ Res 90:398–404, 2002.

Liu SQ: Prevention of focal intimal hyperplasia in rat vein grafts by using a tissue engineering approach, Atherosclerosis 140:365–77, 1998.

Liu SQ: Biomechanical basis of vascular tissue engineering, Crit Rev Biomed Eng 27:75–148, 1999.

Liu SQ: Focal activation of angiotensin II type 1 receptor and smooth muscle cell proliferation in the neointima of experimental vein grafts: Relation to eddy blood flow, Arterioscl Thromb Vasc Biol 19:2630–9, 1999.

15.10. Antisense Oligodeoxynucleotides for CDKs

Dzau VJ, Braun-Dullaeus RC, Sedding DG: Vascular proliferation and atherosclerosis: New perspectives and therapeutic strategies, Nat Med 8(11):1249–56, 2002.

Braun-Dullaeus RC, Mann MJ, Dzau VJ: Cell cycle progression. New therapeutic target for vascular proliferative disease, Circulation 98:82–89, 1998.

Morishita R et al: Single intraluminal delivery of antisense cdc2 kinase and proliferating-cell nuclear antigen oligonucleotides results in chronic inhibition of neointimal hyperplasia, Proc Natl Acad Sci USA 90:8474–8, 1993.

Simons M, Edelman ER, Rosenberg RD: Antisense proliferating cell nuclear antigen oligonucleotides inhibit intimal hyperplasia in a rat carotid artery injury model, J Clin Invest 93:2351–6, 1994.

Morishita R et al: A gene therapy strategy using a transcription factor decoy of the E2F binding site inhibits smooth muscle proliferation in vivo, Proc Natl Acad Sci USA 92:5855–9, 1995.

Mann MJ et al: Cell cycle inhibition preserves endothelial function in genetically engineered rabbit vein grafts, J Clin Invest 99:1295–301, 1997.

Mann MJ et al: Ex vivo gene therapy of human vascular bypass grafts with E2F decoy: The PREVENT single-centre, randomised, controlled trial, Lancet 354:1493–8, 1999.

Mangi AA, Dzau VJ: Gene therapy for human bypass grafts, Ann Med 33:153–5, 2001.

15.11. Cell Cycle-Inhibiting Genes

Smith RC et al: p21CIP1-mediated inhibition of cell proliferation by overexpression of the gax homeodomain gene, Genes Dev 11:1674–89, 1997.

BIBLIOGRAPHY 723

Yang ZY et al: Role of the p21 cyclin-dependent kinase inhibitor in limiting intimal cell proliferation in response to arterial injury, Proc Natl Acad Sci USA 93:7905–10, 1996.

Yonemitsu Y, Kaneda Y, Hataa Y, Nakashima Y, Sueishi K: Wild-type p53 gene transfer: A novel therapeutic strategy for neointimal hyperplasia after arterial injury, Ann NY Acad Sci 811:395–9, 1997.

15.12. Nitric Oxide Synthase

Nitric Oxide Synthase 1 Gene

Guo K, Andres V, Walsh K: Nitric oxide-induced downregulation of Cdk2 activity and cyclin A gene transcription in vascular smooth muscle cells, Circulation 97:2066–72, 1998.

Ishida A, Sasaguri T, Kosaka C, Nojima H, Ogata J: Induction of the cyclin-dependent kinase inhibitor p21(Sdi1/Cip1/Waf1) by nitric oxide-generating vasodilator in vascular smooth muscle cells, J Biol Chem 272:10050–7, 1997.

von der Leyen HE et al: Gene therapy inhibiting neointimal vascular lesion: in vivo transfer of endothelial cell nitric oxide synthase gene, Proc Natl Acad Sci USA 92:1137–41, 1995.

Qian H, Neplioueva V, Shetty GA, Channon KM, George SE: Nitric oxide synthase gene therapy rapidly reduces adhesion molecule expression and inflammatory cell infiltration in carotid arteries of cholesterol-fed rabbits, Circulation 99:2979–82, 1999.

von der Leyen HE, Dzau VJ: Therapeutic potential of nitric oxide synthase gene manipulation, Circulation 103:2760–5, 2001.

Kong D, Melo LG, Mangi AA, Zhang L, Lopez-Ilasaca M et al: Enhanced inhibition of neointimal hyperplasia by genetically engineered endothelial progenitor cells, Circulation 109(14):1769–75, 2004.

Barouch LA, Harrison RW, Skaf MW, Rosas GO, Cappola TP et al: Nitric oxide regulates the heart by spatial confinement of nitric oxide synthase isoforms, Nature 416:337–40, 2002.

Bredt DS, Hwang PM, Glatt CE, Lowenstein C, Reed RR et al: Cloned and expressed nitric oxide synthase structurally resembles cytochrome P-450 reductase, Nature 351:714–8, 1991.

Brenman JE, Chao DS, Xia H, Aldape K, Bredt DS: Nitric oxide synthase complexed with dystrophin and absent from skeletal muscle sarcolemma in Duchenne muscular dystrophy, Cell 82:743–52, 1995.

Deans Z, Dawson SJ, Xie J, Young AP, Wallace D et al: Differential regulation of the two neuronal nitric-oxide synthase gene promoters by the Oct-2 transcription factor, J Biol Chem 271:32153– 8, 1996.

Burnett AL, Lowenstein CJ, Bredt DS, Chang TSK, Snyder SH: Nitric oxide: A physiologic mediator of penile erection, Science 257:401–3, 1992.

Kuo RC, Baxter GT, Thompson SH, Stricker SA, Patton C et al: NO is necessary and sufficient for egg activation at fertilization, Nature 406:633–6, 2000.

Marsden PA, Heng HHQ, Scherer SW, Stewart RJ, Hall AV et al: Structure and chromosomal localization of the human constitutive endothelial nitric oxide synthase gene, J Biol Chem 268:17478–88, 1993.

Nelson RJ, Demas GE, Huang PL, Fishman MC, Dawson VL et al: Behavioural abnormalities in male mice lacking neuronal nitric oxide synthase, Nature 378:383–6, 1995.

Newton DC, Bevan SC, Choi S, Robb GB, Millar A et al: Translational regulation of human neuronal nitric-oxide synthase by an alternatively spliced 5-prime untranslated region leader exon, J Biol Chem 278:636–44, 2003.

Sander M, Chavoshan B, Harris SA, Iannaccone ST, Stull JT et al: Functional muscle ischemia in neuronal nitric oxide synthase-deficient skeletal muscle of children with Duchenne muscular dystrophy, Proc Natl Acad Sci USA 97:13818–23, 2000.

724 VASCULAR REGENERATIVE ENGINEERING

Nitric Oxide Synthase 2A

Bloch KD, Wolfram JR, Brown DM, Roberts JD Jr, Zapol DG et al: Three members of the nitric oxide synthase II gene family (NOS2A, NOS2B, and NOS2C) colocalize to human chromosome 17, Genomics 27:526–30, 1995.

Chartrain NA, Geller DA, Koty PP, Sitrin NF, Nussler AK et al: Molecular cloning, structure, and chromosomal localization of the human inducible nitric oxide synthase gene, J Biol Chem 269:6765–72, 1994.

Diefenbach A, Schindler H, Rollinghoff M, Yokoyama WM, Bogdan C: Requirement for type 2 NO synthase for IL-12 signaling in innate immunity, Science 284:951–5, 1999.

Koh KP, Wang Y, Yi T, Shiao SL, Lorber MI, Sessa WC et al: T cell-mediated vascular dysfunction of human allografts results from IFN-gamma dysregulation of NO synthase, J Clin Invest 114:846–56, 2004.

Kolodziejski PJ, Musial A, Koo JS, Eissa NT: Ubiquitination of inducible nitric oxide synthase is required for its degradation, Proc Natl Acad Sci USA 99:12315–20, 2002.

Nitric Oxide Synthase 3

Aicher A, Heeschen C, Mildner-Rihm C, Urbich C, Ihling C et al: Essential role of endothelial nitric oxide synthase for mobilization of stem and progenitor cells, Nature Med 9:1370–6, 2003.

Barouch LA, Harrison RW, Skaf MW, Rosas GO, Cappola TP et al: Nitric oxide regulates the heart by spatial confinement of nitric oxide synthase isoforms, Nature 416:337–40, 2002.

Casas JP, Bautista LE, Humphries SE, Hingorani AD: Endothelial nitric oxide synthase genotype and ischemic heart disease: Meta-analysis of 26 studies involving 23028 subjects, Circulation 109:1359–65, 2004.

Connelly L, Madhani M, Hobbs AJ: Resistance to endotoxic shock in endothelial nitric-oxide synthase (eNOS) knock-out mice: A pro-inflammatory role for eNOS-derived NO in vivo, J Biol Chem 280:10040–6, 2005.

Dimmeler S, Fleming I, Fisslthaler B, Hermann C, Busse R et al: Activation of nitric oxide synthase in endothelial cells by Akt-dependent phosphorylation, Nature 399:601–5, 1999.

Fulton D, Gratton JP, McCabe TJ, Fontana J, Fujio Y et al: Regulation of endothelium-derived nitric oxide production by the protein kinase Akt, Nature 399:597–601, 1999.

Huang PL, Huang Z, Mashimo H, Bloch KD, Moskowitz MA et al: Hypertension in mice lacking the gene for endothelial nitric oxide synthase, Nature 377:239–42, 1995.

Janssens SP, Shimouchi A, Quertermous T, Bloch DB, Bloch KD: Cloning and expression of a cDNA encoding human endothelium-derived relaxing factor/nitric oxide synthase, J Biol Chem 267:14519–22, 1992.

Liu S, Premont RT, Kontos CD, Zhu S, Rockey DC: A crucial role for GRK2 in regulation of endothelial cell nitric oxide synthase function in portal hypertension, Nature Med 11:952–8, 2005.

Marsden PA, Heng HHQ, Scherer SW, Stewart RJ, Hall AV et al: Structure and chromosomal localization of the human constitutive endothelial nitric oxide synthase gene, J Biol Chem 268:17478–88, 1993.

Nisoli E, Tonello C, Cardile A, Cozzi V, Bracale R et al: Calorie restriction promotes mitochondrial biogenesis by inducing the expression of eNOS, Science 310:314–7, 2005.

Ohashi Y, Kawashima S, Hirata K, Yamashita T, Ishida T et al: Hypotension and reduced nitric oxide-elicited vasorelaxation in transgenic mice overexpressing endothelial nitric oxide synthase, J Clin Invest 102:2061–71, 1998.

Raman CS, Li H, Martasak P, Kral V, Masters BSS et al: Crystal structure of constitutive endothelial nitric oxide synthase: A paradigm for pterin function involving a novel metal center, Cell 95:939–50, 1998.

Human protein reference data base, Johns Hopkins University and the Institute of Bioinformatics, at http://www.hprd.org/protein.

BIBLIOGRAPHY 725

15.13. Herpes Virus Thymidine Kinase Gene

Ohno T, Gordon D, San H, Pompili VJ, Imperiale MJ et al: Gene therapy for vascular smooth muscle cell proliferation after arterial injury, Science 265(5173):781–4, 1994.

Chang MW, Ohno T, Gordon D, Lu MM, Nabel GJ et al: Adenovirus-mediated transfer of the herpes simplex virus thymidine kinase gene inhibits vascular smooth muscle cell proliferation and neointima formation following balloon angioplasty of the rat carotid artery, Mol Med 1(2):172–81, 1995.

15.14. Dominant-Negative Mutant Mitogenic Genes

Reusch HP, Schaefer M, Plum C, Schultz G, Paul M: Gbeta gamma mediate differentiation of vascular smooth muscle cells, J Biol Chem 276(22):19540–7, 2001.

Ginnan R, Pfleiderer PJ, Pumiglia K, Singer HA: PKC-delta and CaMKII-delta 2 mediate ATPdependent activation of ERK1/2 in vascular smooth muscle, Am J Physiol Cell Physiol 286(6): C128–9, 2004.

Miura S, Matsuo Y, Kawamura A, Saku K: JTT-705 blocks cell proliferation and angiogenesis through p38 kinase/p27(kip1) and Ras/p21(waf1) pathways, Atherosclerosis 182(2):267–75, 2005.

15.15. Construction of Polymeric Arterial Substitutes with Endothelial Cell Seeding

Brewster DC, Rutherford RB: Prosthetic grafts, in: Vascular Surgery, Rutherford RB, ed, Saunders, Philadelphia, 2000, pp 492–521.

O’Donnell TF: Correlation of operative findings with angiographic and noninvasive hemodynamic factors associated with failure of PTFE grafts, J Vasc Surg 1:136–48, 1984.

Bowlin GL, Meyer A, Fields C, Cassano A, Makhoul RG et al: The persistence of electrostatically seeded endothelial cells lining a small diameter expanded polytetrafluoroethylene vascular graft, J Biomater Appl 16:157–73, 2001.

Braddon LG, Karoyli D, Harrison DG, Nerem RM: Maintenance of a functional endothelial cell monolayer on a fibroblast/polymer substrate under physiologically relevant shear stress conditions, Tissue Eng 8:695–708, 2002.

Deutsch M, Meinhart J, Fischlein T, Preiss P, Zilla P: Clinical autologous in vitro endothelialization of infrainguinal ePTFE grafts in 100 patients: A 9-year experience, Surgery 126:847–55, 1999.

Herring MB, Dilley R, Jersild RA Jr, Boxer L, Gardner A et al: Seeding arterial prostheses with vascular endothelium. The nature of the lining, Ann Surg 190:84–90, 1979.

Herring MB, Compton RS, LeGrand DR, Gardner AL, Madison DL et al: Endothelial seeding of polytetrafluoroethylene popliteal bypasses. A preliminary report, J Vasc Surg 6:114–8, 1987.

Herring MB, LeGrand DR: The histology of seeded PTFE grafts in humans, Ann Vasc Surg 3:96–103, 1989.

Herring MB, Compton R, Legrand DR, Gardner AL: Endothelial cell seeding in the management of vascular thrombosis, Semin Thromb Hemost 15:200–5, 1989.

Herring MB: The use of endothelial seeding of prosthetic arterial bypass grafts, Surg Ann 23(Pt 2):157–71, 1991.

Herring MB: Endothelial cell seeding, J Vasc Surg 13:731–2, 1991.

Kesler KA, Herring MB, Arnold MP, Park HM, Baughman S et al: Short-term in vivo stability of endothelial-lined polyester elastomer and polytetrafluoroethylene grafts, Ann Vasc Surg 1:60–5, 1986.

Ratcliffe A: Tissue engineering of vascular grafts, Matrix Biol 19:353–7, 2000.