Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-r5zm4 Total loading time: 0 Render date: 2024-06-16T19:17:54.407Z Has data issue: false hasContentIssue false

12 - Animal Models of Hemoglobinopathies and Thalassemia

from SECTION TWO - PATHOPHYSIOLOGY OF HEMOGLOBIN AND ITS DISORDERS

Published online by Cambridge University Press:  03 May 2010

Martin H. Steinberg
Affiliation:
Boston University
Bernard G. Forget
Affiliation:
Yale University, Connecticut
Douglas R. Higgs
Affiliation:
MRC Institute of Molecular Medicine, University of Oxford
David J. Weatherall
Affiliation:
Albert Einstein College of Medicine, New York
Get access

Summary

INTRODUCTION

Transgenic mice are important adjuncts for the study of the pathophysiology and treatment of human hemoglobin disorders. Many inbred strains with spontaneous and induced mutations and engineered knockout and knockin lines are available (Table 12.1). Transgenic mice have both advantages and disadvantages for studying human disease. Most available mice are of mixed or poorly characterized genetic backgrounds and must be bred onto another inbred strain to obtain sustainable lines and consistent physiology. The choice of genetic background or the presence of a mixed background can affect both hematology and physiology.

Different inbred mouse strains, and male and female mice, can have different physiological characteristics, complicating the interpretation of some experiments. Mice have a higher plasma osmolarity than man and the 2,3-BPG content of mouse red cells is twice that of human erythrocytes. Inbred strains have five common α-globin chains and three common β-globin chains. The common C57BL/6 strain has only a single α- and β-globin chain; other strains can have two α- or two β-globin chains. When these are combined with human globin transgenes, many homotetramers due to the presence of interspecies αβ dimers result, some of which form tetramers with low oxygen affinity that will affect polymer formation and pathophysiology. These are some examples of factors likely to alter sickle cell mouse pathophysiology when compared with human.

THE TECHNOLOGY OF TRANSGENIC AND KNOCKOUT MICE

In the early 1980s cloned DNA was introduced into fertilized mouse embryos.

Type
Chapter
Information
Disorders of Hemoglobin
Genetics, Pathophysiology, and Clinical Management
, pp. 225 - 238
Publisher: Cambridge University Press
Print publication year: 2009

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Skow, LC, Burkhart, BA, Johnson, FM, et al. A mouse model for beta-thalassemia. Cell. 1983;34:1043–1052.CrossRefGoogle ScholarPubMed
Fabry, ME, Costantini, F, Pachnis, A, et al. High expression of human bS- and a-globins in transgenic mice: erythrocyte abnormalities, organ damage, and the effect of hypoxia. Proc Natl Acad Sci USA. 1992;89:12155–12159.CrossRefGoogle Scholar
Trudel, M, Garel, MC, Saadane, N, et al. Towards a mouse model for sickle cell disease: HB SAD. Nouv Rev Fr Hematol. 1990;32:407–408.Google ScholarPubMed
Rubin, EM, Witkowska, HE, Spangler, E, et al. Hypoxia-induced in vivo sickling of transgenic mouse red cells. J Clin Invest. 1991;87:639–647.CrossRefGoogle ScholarPubMed
Fabry, ME, Sengupta, A, Suzuka, SM, et al. A second generation transgenic mouse model expressing both hemoglobin S (HbS) and HbS-Antilles results in increased phenotypic severity. Blood. 1995;86:2419–2428.Google ScholarPubMed
Arcasoy, MO, Romana, M, Fabry, ME, et al. High levels of human gamma-globin gene expression in adult mice carrying a transgene of deletion-type hereditary persistence of fetal hemoglobin. Mol Cell Biol. 1997;17:2076–2089.CrossRefGoogle ScholarPubMed
Paszty, C, Mohandas, N, Stevens, ME, et al. Lethal alpha-thalassaemia created by gene targeting in mice and its genetic rescue. Nat Genet. 1995;11:33–39.CrossRefGoogle ScholarPubMed
Yang, B, Kirby, S, Lewis, J, et al. A mouse model for beta 0-thalassemia. Proc Natl Acad Sci USA. 1995;92:11608–11612.CrossRefGoogle ScholarPubMed
Ciavatta, DJ, Ryan, TM, Farmer, SC, Townes, TM. Mouse model of human beta zero thalassemia: targeted deletion of the mouse beta maj- and beta min-globin genes in embryonic stem cells. Proc Natl Acad Sci USA. 1995;92:9259–9263.CrossRefGoogle ScholarPubMed
Gilman, JG. Developmental changes of human Gg and Ag and mouse embryonic ey1, ey2, and bh1 in transgenic mice with HS4-Gg-Ag. Blood. 1995;86:648a.Google Scholar
Shear, HL, Grinberg, L, Gilman, J, et al. Transgenic mice expressing human fetal globin are protected from malaria by a novel mechanism. Blood. 1998;92:2520–2526.Google ScholarPubMed
Fabry, ME, Suzuka, SM, Weinberg, RS, et al. Second generation knockout sickle mice: the effect of HbF. Blood. 2001;97:410–418.CrossRefGoogle ScholarPubMed
Ryan, TM, Ciavatta, DJ, Townes, TM. Knockout-transgenic mouse model of sickle cell disease. Science. 1997;278:873–876.CrossRefGoogle ScholarPubMed
Paszty, C, Brion, CM, Manci, E, et al. Transgenic knockout mice with exclusively human sickle hemoglobin and sickle cell disease. Science. 1997;278:876–878.CrossRefGoogle ScholarPubMed
Chang, JC, Lu, R, Lin, C, et al. Transgenic knockout mice exclusively expressing human hemoglobin S after transfer of a 240-kb betas-globin yeast artificial chromosome: A mouse model of sickle cell anemia. Proc Natl Acad Sci USA. 1998;95:14886–14890.CrossRefGoogle ScholarPubMed
Wu, LC, Sun, CW, Ryan, TM, et al. Correction of sickle cell disease by homologous recombination in embryonic stem cells. Blood. 2006;108:1183–1188.CrossRefGoogle ScholarPubMed
Crispins, CG. Handbook on the Laboratory Mouse. Springfield, IL: Charles C Thomas; 1975.Google Scholar
Whitney, JB III. Mouse hemoglobinopathies: detection and characterization of thalassemias and globin-structure mutations. Prog Clin Biol Res. 1982;94:133–142.Google ScholarPubMed
Roy, RP, Nacharaju, P, Nagel, RL, Acharya AS. Symmetric interspecies hybrids of mouse and human hemoglobin: molecular basis of their abnormal oxygen affinity. J Protein Chem. 1995;14:81–88.CrossRefGoogle ScholarPubMed
Gordon, JW, Scangos, GA, Plotkin, DJ, Barbosa, JA, Ruddle, FH. Genetic transformation of mouse embryos by microinjection of purified DNA. Proc Natl Acad Sci USA. 1980;77:7380–7384.CrossRefGoogle ScholarPubMed
Costantini, F, Lacy, E. Introduction of a rabbit beta-globin gene into the mouse germ line. Nature. 1981;294:92–94.CrossRefGoogle ScholarPubMed
Tuan, D, Solomon, W, Li, Q, London, IM. The “beta-like-globin” gene domain in human erythroid cells. Proc Natl Acad Sci USA. 1985;82:6384–6388.CrossRefGoogle ScholarPubMed
Chada, K, Magram, J, Raphael, K, et al. Specific expression of a foreign beta-globin gene in erythroid cells of transgenic mice. Nature. 1985;314:377–380.CrossRefGoogle ScholarPubMed
Kollias, G, Wrighton, N, Hurst, J, Grosveld, F. Regulated expression of human A gamma-, beta-, and hybrid gamma beta-globin genes in transgenic mice: manipulation of the developmental expression patterns. Cell. 1986;46:89–94.CrossRefGoogle Scholar
Townes, TM, Lingrel, JB, Chen, HY, Brinster, RL, Palmiter, RD. Erythroid-specific expression of human beta-globin genes in transgenic mice. EMBO J. 1985;4:1715–1723.Google ScholarPubMed
Hogan, BLM, Costantini, F, Lacy, E. Manipulating the Mouse Embryo: A Laboratory Manual. Cold Spring Harbor: Cold Spring Harbor Laboratory; 1986.Google Scholar
Skow, LC, Burkhart, BA, Johnson, FM, et al. A mouse model for beta-thalassemia. Cell. 1983;34:1043–1052.CrossRefGoogle ScholarPubMed
Whitney, JB, Martinell, J, Popp, RA, Russell, LB, Anderson, WF. Deletions in the alpha-globin gene complex in alpha-thalassemic mice. Proc Natl Acad Sci USA. 1981;78:7644–7647.CrossRefGoogle ScholarPubMed
Martinell, J, Whitney, JB, Popp, RA, Russell, LB, Anderson, WF. Three mouse models of human thalassemia. Proc Natl Acad Sci USA. 1981;78:5056–5060.CrossRefGoogle ScholarPubMed
Rouyer Fessard, P, Scott, MD, Leroy Viard, K, et al. Fate of alpha-hemoglobin chains and erythrocyte defects in beta- thalassemia. Ann NY Acad Sci. 1990;612:106–117.CrossRefGoogle ScholarPubMed
Rivella, S, May, C, Chadburn, A, Riviere, I, Sadelain, M. A novel murine model of Cooley anemia and its rescue by lentiviral-mediated human beta-globin gene transfer. Blood. 2003;101:2932–2939.CrossRefGoogle ScholarPubMed
Rouyer Fessard, P, Leroy Viard, K, Domenget, C, Mrad, A, Beuzard, Y. Mouse beta thalassemia, a model for the membrane defects of erythrocytes in the human disease. J Biol Chem. 1990;265:20247–20251.Google ScholarPubMed
Whitney, JB, Popp, RA. Animal model of human disease: thalassemia: alpha-thalassemia in laboratory mice. Am J Pathol. 1984;116:523–525.Google ScholarPubMed
Franceschi, L, Rouyer-Fessard, P, Alper, SL, et al. Combination therapy of erythropoietin, hydroxyurea, and clotrimazole in a beta thalassemic mouse: a model for human therapy. Blood. 1996;87:1188–1195.Google Scholar
Shehee, WR, Oliver, P, Smithies, O. Lethal thalassemia after insertional disruption of the mouse major adult beta-globin gene. Proc Natl Acad Sci USA. 1993;90:3177–3181.CrossRefGoogle ScholarPubMed
Fabry, ME, Suzuka, SM, Rubin, EM, Costantini, F, Gilman, J, Nagel, RL. Strategies for amelioration of sickle cell disease: Use of transgenic mice for validation of anti-sickling strategies. In: Beuzard, Y, Lubin, B, Rosa, J, eds. Sickle Cell Disease and Thalassemias: New Trends in Therapy. London: John Libbey Eurotext Ltd; 1995:253–262.Google Scholar
Ciavatta, DJ, Ryan, TM, Farmer, SC, Townes, TM. Mouse model of human beta zero thalassemia: targeted deletion of the mouse beta maj- and beta min-globin genes in embryonic stem cells. Proc Natl Acad Sci USA. 1995;92:9259–9263.CrossRefGoogle ScholarPubMed
Yang, B, Kirby, S, Lewis, J, et al. A mouse model for beta 0-thalassemia. Proc Natl Acad Sci USA. 1995;92:11608–11612.CrossRefGoogle ScholarPubMed
Paszty, C, Mohandas, N, Stevens, ME, et al. Lethal alpha-thalassaemia created by gene targeting in mice and its genetic rescue. Nat Genet. 1995;11:33–39.CrossRefGoogle ScholarPubMed
Costantini, F, Chada, K, Magram, J. Correction of murine beta-thalassemia by gene transfer into the germ line. Science. 1986;233:1192–1194.CrossRefGoogle ScholarPubMed
Rubin, EM, Lu, RH, Cooper, S, Mohandas, N, Kan, YW. Introduction and expression of the human βs-globin gene in transgenic mice. Am J Hum Genet. 1988;42:585–591.Google Scholar
Rhoda, MD, Domenget, C, Vidaud, M, et al. Mouse α-chains inhibit polymerization of hemoglobin induced by human βS or βS-Antilles chains. Biochim Biophys Acta. 1988;952:208–212.CrossRefGoogle Scholar
Greaves, DR, Fraser, P, Vidal, MA, et al. A transgenic mouse model of sickle cell disorder. Nature. 1990;343:183–185.CrossRefGoogle ScholarPubMed
Ryan, TM, Townes, TM, Reilly, MP, et al. Human sickle hemoglobin in transgenic mice. Science. 1990;247:566–568.CrossRefGoogle ScholarPubMed
Rubin, EM, Witkowska, HE, Spangler, E, et al. Hypoxia-induced in vivo sickling of transgenic mouse red cells. J Clin Invest. 1991;87:639–647.CrossRefGoogle ScholarPubMed
Trudel, M, Saadane, N, Garel, M-C, et al. Towards a transgenic mouse model of sickle cell disease: hemoglobin SAD. EMBO J. 1991;10:3157–3168.Google ScholarPubMed
Fabry, ME, Costantini, F, Pachnis, A, et al. High expression of human βS- and α-globins in transgenic mice: erythrocyte abnormalities, organ damage, and the effect of hypoxia. Proc Natl Acad Sci USA. 1992;89:12155–12159.CrossRefGoogle Scholar
Fabry, ME, Nagel, RL, Pachnis, A, Suzuka, SM, Costantini, F. High expression of human βS- and α-globins in transgenic mice: hemoglobin composition and hematological consequences. Proc Natl Acad Sci USA. 1992;89:12150–12154.CrossRefGoogle Scholar
Reilly, MP, Chomo, MJ, Obata, K, Asakura, T. Red blood cell membrane and density changes under ambient and hypoxic conditions in transgenic mice producing human sickle hemoglobin. Exp Hematol. 1994;22:501–509.Google ScholarPubMed
Fabry, ME, Sengupta, A, Suzuka, SM, et al. A second generation transgenic mouse model expressing both hemoglobin S (HbS) and HbS-Antilles results in increased phenotypic severity. Blood. 1995;86:2419–2428.Google ScholarPubMed
Popp, RA, Popp, DM, Shinpock, SG, et al. A transgenic mouse model of hemoglobin S Antilles disease. Blood. 1997;89:4204–4212.Google ScholarPubMed
Monplaisir, N, Merault, G, Poyart, C, et al. Hemoglobin S Antilles: a variant with lower solubility than hemoglobin S and producing sickle cell disease in heterozygotes. Proc Natl Acad Sci USA. 1986;83:9363–9367.CrossRefGoogle ScholarPubMed
Monplaisir, N, Galacteros, F, Arous, N, et al. [Abnormal hemoglobins identified in Martinique]. Nouv Rev Fr Hematol. 1985;27:11–14.Google Scholar
Li, X, Mirza, UA, Chait, BT, Manning, JM. Systematic enhancement of polymerization of recombinant sickle hemoglobin mutants: implications for transgenic mouse model for sickle cell anemia. Blood. 1997;90:4620–4627.Google ScholarPubMed
Paszty, C, Brion, CM, Manci, E, et al. Transgenic knockout mice with exclusively human sickle hemoglobin and sickle cell disease. Science. 1997;278:876–878.CrossRefGoogle ScholarPubMed
Ryan, TM, Ciavatta, DJ, Townes, TM. Knockout-transgenic mouse model of sickle cell disease. Science. 1997;278:873–876.CrossRefGoogle ScholarPubMed
Romero, J, Fabry, ME, Suzuka, SM, Nagel, RL, Canessa, M. Red blood cells of a transgenic mouse expressing high levels of human hemoglobin S exhibit deoxy-stimulated cation flux. J Membr Biol. 1997;159:187–196.CrossRefGoogle ScholarPubMed
Manci, EA, Hillery, CA, Bodian, CA, et al. Pathology of Berkeley sickle cell mice: similarities and differences with human sickle cell disease. Blood. 2006;107:1651–1658.CrossRefGoogle ScholarPubMed
Gilman, JG, Fabry, ME, Suzuka, SM, Nagel, RL. Human G-gamma:A-gamma globin ratios switch during prenatal development in a transgenic mouse with HS4-G-gamma(-158T)-A-gamma. Blood. 1995;86:648a.Google Scholar
Fabry, ME, Suzuka, SM, Weinberg, RS, et al. Second generation knockout sickle mice: the effect of HbF. Blood. 2001;97:410–418.CrossRefGoogle ScholarPubMed
Fabry, ME, Witt, WM, Suzuka, SM, Nagel, RL. Impact of HbF and gender on mean survival time of NY1KO sickle transgenic ice expressing exclusively human hemoglobins. Blood. 2001;98:750aa.Google Scholar
Gilman, JG. Developmental changes of human Gγ and Aγ and mouse embryonic εy1, εy2, and βh1 in transgenic mice with HS4-Gγ-Aγ. Blood. 1995;86:648a.Google Scholar
Fabry, ME, Romero, JR, Suzuka, SM, et al. Hemoglobin C in transgenic mice: effect of HbC expression from founders to full mouse globin knockouts. Blood Cell Mol Dis. 2000;26:331–347.CrossRefGoogle ScholarPubMed
Noguchi, CT, Gladwin, M, Diwan, B, et al. Pathophysiology of a sickle cell trait mouse model: human alpha(beta)(S) transgenes with one mouse beta-globin allele. Blood Cell Mol Dis. 2001;27:971–977.CrossRefGoogle ScholarPubMed
Kiefmann, R, Rifkind, JM, Nagababu, E, Bhattacharya, J. Red blood cells induce hypoxic lung inflammation. Blood. 2008;111:5205–5214.CrossRefGoogle ScholarPubMed
Rifkind, JM, Nagababu, E, Ramasamy, S, Ravi, LB. Hemoglobin redox reactions and oxidative stress. Redox Rep. 2003;8:234–237.CrossRefGoogle ScholarPubMed
Abugo, OO, Rifkind, JM. Oxidation of hemoglobin and the enhancement produced by nitroblue tetrazolium. J Biol Chem. 1994;269:24845–24853.Google ScholarPubMed
Rifkind, JM, Zhang, L, Heim, JM, Levy, A. The role of hemoglobin in generating oxyradicals. Basic Life Sci. 1988;49:157–162.Google ScholarPubMed
Ieremia, J, Blau, CA. Limitations of a mouse model of sickle cell anemia. Blood Cell Mol Dis. 2002;28:146–151.CrossRefGoogle ScholarPubMed
Chang, JC, Lu, R, Lin, C, et al. Transgenic knockout mice exclusively expressing human hemoglobin S after transfer of a 240-kb betas-globin yeast artificial chromosome: A mouse model of sickle cell anemia. Proc Natl Acad Sci USA. 1998;95:14886–14890.CrossRefGoogle ScholarPubMed
Ye, L, Chang, JC, Lu, R, Kan, YW. High oxygen environment during pregnancy rescues sickle cell anemia mice from prenatal death. Blood Cell Mol Dis. 2008;41:67–72.CrossRefGoogle ScholarPubMed
Wu, LC, Sun, CW, Ryan, TM, et al. Correction of sickle cell disease by homologous recombination in embryonic stem cells. Blood. 2006;108:1183–1188.CrossRefGoogle ScholarPubMed
Scott, AF, Bunn, HF, Brush, AH. Functional aspects of hemoglobin evolution in the mammals. J Mol Evol. 1976;8:311–316.CrossRefGoogle ScholarPubMed
Swerdlow, PH, Bryan, RA, Bertles, JF, et al. Effect of 2, 3-diphosphoglycerate on the solubility of deoxy-sickle hemoglobin. Hemoglobin. 1977;1:527–537.CrossRefGoogle ScholarPubMed
Tosteson, DC, Carlsen, E, Dunham, ET. The effect of sickling on ion transport. I. The effect of sickling on potassium transport. J Gen Physiol. 1955;39:31–53.CrossRefGoogle ScholarPubMed
Vandorpe, DH, Shmukler, BE, Jiang, L, et al. cDNA cloning and functional characterization of the mouse Ca2+-gated K+ channel, mIK1. Roles in regulatory volume decrease and erythroid differentiation. J Biol Chem. 1998;273;21542–21553.CrossRefGoogle ScholarPubMed
Franceschi, L, Brugnara, C, Rouyer-Fessard, P, Jouault, H, Beuzard, Y. Formation of dense erythrocytes in SAD mice exposed to chronic hypoxia: evaluation of different therapeutic regimens and of a combination of oral clotrimazole and magnesium therapies. Blood. 1999;94:4307–4313.Google ScholarPubMed
Franceschi, L, Saadane, N, Trudel, M, et al. Treatment with oral clotrimazole blocks Ca(2+)-activated K+ transport and reverses erythrocyte dehydration in transgenic SAD mice. A model for therapy of sickle cell disease. J Clin Invest. 1994;93:1670–1676.CrossRefGoogle ScholarPubMed
Stocker, JW, Franceschi, L, McNaughton-Smith, GA, et al. ICA-17043, a novel Gardos channel blocker, prevents sickled red blood cell dehydration in vitro and in vivo in SAD mice. Blood. 2003;101:2412–2418.CrossRefGoogle ScholarPubMed
Romero, JR, Suzuka, SM, Nagel, RL, Fabry, ME. Arginine supplementation of sickle transgenic mice reduces red cell density and Gardos channel activity. Blood. 2002;99:1103–1108.CrossRefGoogle ScholarPubMed
Rivera, A, Rotter, MA, Brugnara, C. Endothelins activate Ca(2+)-gated K(+) channels via endothelin B receptors in CD-1 mouse erythrocytes. Am J Physiol. 1999;277:C746–C754.CrossRefGoogle ScholarPubMed
Rivera, A, Jarolim, P, Brugnara, C. Modulation of Gardos channel activity by cytokines in sickle erythrocytes. Blood. 2002;99:357–603.CrossRefGoogle ScholarPubMed
Romero, JR, Rivera, A, Muniz, A, Suzuka, SM, Nagel, RL, Fabry, ME. Effect of arginine diet for sickle transgenic mice on plasma cytokine levels and the mechanism of reduction of Gardos channel activity. Blood. 2003;102:260a.Google Scholar
Rivera, A. Reduced sickle erythrocyte dehydration in vivo by endothelin-1 receptor antagonists. Am J Physiol Cell Physiol. 2007;293:C960–966.CrossRefGoogle ScholarPubMed
Romero, JR, Fabry, ME, Suzuka, SM, et al. K:Cl cotransport in red cells of transgenic mice expressing high levels of human hemoglobin S. Am J Hematol. 1997;55:112–114.3.0.CO;2-G>CrossRefGoogle ScholarPubMed
Armsby, CC, Brugnara, C, Alper, SL. Cation transport in mouse erythrocytes: role of K(+)-Cl- cotransport in regulatory volume decrease. Am J Physiol. 1995;268(Pt 1):C894–902.CrossRefGoogle Scholar
Romero, JR, Suzuka, SM, Nagel, RL, Fabry, ME. Expression of HbC and HbS, but not HbA, results in activation of K-Cl cotransport activity in transgenic mouse red cells. Blood. 2004;103:2384–2390.CrossRefGoogle Scholar
Franceschi, L, Beuzard, Y, Jouault, H, Brugnara, C. Modulation of erythrocyte potassium chloride cotransport, potassium content, and density by dietary magnesium intake in transgenic SAD mouse. Blood. 1996;88:2738–2744.Google ScholarPubMed
Su, W, Shmukler, BE, Chernova, MN, et al. Mouse K-Cl cotransporter KCC1: cloning, mapping, pathological expression, and functional regulation. Am J Physiol. 1999;277:C899–C912.CrossRefGoogle ScholarPubMed
Pellegrino, CM, Rybicki, AC, Musto, S, Nagel, RL, Schwartz, RS. Molecular identification and expression of erythroid K:Cl cotransporter in human and mouse erythroleukemic cells. Blood Cell Mol Dis. 1998;24:31–40.CrossRefGoogle ScholarPubMed
Gillen, CM, Brill, S, Payne, JA, Forbush, B. Molecular cloning and functional expression of the K-Cl cotransporter from rabbit, rat, and human. A new member of the cation-chloride cotransporter family. J Biol Chem. 1996;271:16237–16244.CrossRefGoogle Scholar
Crable, SC, Hammond, SM, Papes, R, et al. Multiple isoforms of the KC1 cotransporter are expressed in sickle and normal erythroid cells. Exp Hematol. 2005;33:624–631.CrossRefGoogle ScholarPubMed
Rust, MB, Alper, SL, Rudhard, Y, et al. Disruption of erythroid K-Cl cotransporters alters erythrocyte volume and partially rescues erythrocyte dehydration in SAD mice. J Clin Invest. 2007;117:1708–1717.CrossRefGoogle ScholarPubMed
Reilly, MP, Obata, K, Chomo, M, Asakura, T. Red blood cell membrane alterations and dense cell formation in transgenic mice expressing different levels of human sickle hemoglobin. National Sickle Cell Disease Program 18th, 1a. 1993.
Franceschi, L, Beuzard, Y, Brugnara, C. Sulfhydryl oxidation and activation of red cell K(+)-Cl- cotransport in the transgenic SAD mouse. Am J Physiol. 1995;269(Pt 1):C899–906.CrossRefGoogle ScholarPubMed
Jong, K, Emerson, RK, Butler, J, et al. Short survival of phosphatidylserine-exposing red blood cells in murine sickle cell anemia. Blood. 2001;98:1577–1584.CrossRefGoogle ScholarPubMed
Brugnara, C. Therapeutic strategies for prevention of sickle cell dehydration. Blood Cell Mol Dis. 2001;27:71–80.CrossRefGoogle ScholarPubMed
Brugnara, C. Sickle cell disease: from membrane pathophysiology to novel therapies for prevention of erythrocyte dehydration. J Pediatr Hematol Oncol. 2003;25:927–933.CrossRefGoogle ScholarPubMed
Lew, VL, Bookchin, RM. Ion transport pathology in the mechanism of sickle cell dehydration. Physiol Rev. 2005;85:179–200.CrossRefGoogle ScholarPubMed
Wulff, H, Kolski-Andreaco, A, Sankaranarayanan, A, Sabatier, JM, Shakkottai, V. Modulators of small- and intermediate-conductance calcium-activated potassium channels and their therapeutic indications. Curr Med Chem. 2007;14:1437–1457.CrossRefGoogle ScholarPubMed
Trudel, M, Paepe, ME, Chretien, N, et al. Sickle cell disease of transgenic SAD mice. Blood. 1994;84:3189–3197.Google ScholarPubMed
Bank, N, Aynedjian, HS, Qiu, JH, et al. Renal nitric oxide synthases in transgenic sickle cell mice. Kidney Int. 1996;50:184–189.CrossRefGoogle ScholarPubMed
Xia, Y, Dawson, VL, Dawson, TM, Snyder, SH, Zweier, JL. Nitric oxide synthase generates superoxide and nitric oxide in arginine-depleted cells leading to peroxynitrite-mediated cellular injury. Proc Natl Acad Sci USA. 1996;93:6770–6774.CrossRefGoogle ScholarPubMed
Bank, N, Kiroycheva, M, Ahmed, F, et al. Peroxynitrite formation and apoptosis in transgenic sickle cell mouse kidneys. Kidney Int. 1998;54:1520–1528.CrossRefGoogle ScholarPubMed
Enwonwu, CO. Increased metabolic demand for arginine in sickle cell anemia. Med Sci Res. 1989;17:997–998.Google Scholar
Lipowsky, HL, Sheikh, NU, Katz, DM. Intravital microscopy of capillary hemodynamics in sickle cell disease. J Clin Invest. 1987;80:117–127.CrossRefGoogle ScholarPubMed
Rodgers, GP, Walker, EC, Podgor, MJ. Is “relative” hypertension a risk factor for vaso-occlusive complications in sickle cell disease?Am J Med Sci. 1993;305:150–156.CrossRefGoogle Scholar
Pegelow, CH, Colangelo, L, Steinberg, M, et al. Natural history of blood pressure in sickle cell disease: risks for stroke and death associated with relative hypertension in sickle cell anemia. Am J Med. 1997;102:171–177.CrossRefGoogle ScholarPubMed
Kaul, DK, Liu, XD, Fabry, ME, Nagel, RL. Increased nitric oxide synthase expression in transgenic sickle mice is associated with relative hypotension and altered microvascular response. Blood. 1998;92:330a.Google Scholar
Fabry, ME, Kennan, RP, Paszty, C, et al. Magnetic resonance evidence of hypoxia in a homozygous a-knockout of a transgenic mouse model for sickle cell disease. J Clin Invest. 1996;98:2450–2455.CrossRefGoogle Scholar
Brezis, M, Rosen, S. Hypoxia of the renal medulla – its implications for disease. N Engl J Med. 1995;332:647–655.CrossRefGoogle ScholarPubMed
Kobayashi, H, Kawamoto, S, Brechbiel, MW, et al. Micro-MRI methods to detect renal cysts in mice. Kidney Int. 2004;65:1511–1516.CrossRefGoogle ScholarPubMed
Bank, N, Kiroycheva, M, Singhal, PC, et al. Inhibition of nitric oxide synthase ameliorates cellular injury in sickle cell mouse kidneys. Kidney Int. 2000;58:82–89.CrossRefGoogle ScholarPubMed
Nath, KA, Grande, JP, Haggard, JJ, et al. Oxidative stress and induction of heme oxygenase-1 in the kidney in sickle cell disease. Am J Pathol. 2001;158:893–903.CrossRefGoogle ScholarPubMed
Rybicki, AC, Fabry, ME, Does, MD, Kaul, DK, Nagel, RL. Differential gene expression in the kidney of sickle cell transgenic mice: upregulated genes. Blood Cell Mol Dis. 2003;31:370–380.CrossRefGoogle ScholarPubMed
Belcher, JD, Bryant, CJ, Nguyen, J, et al. Transgenic sickle mice have vascular inflammation. Blood. 2003;101:3953–3959.CrossRefGoogle ScholarPubMed
Nath, KA, Grande, JP, Croatt, AJ, et al. Transgenic sickle mice are markedly sensitive to renal ischemia-reperfusion injury. Am J Pathol. 2005;166:963–972.CrossRefGoogle ScholarPubMed
Diwan, BA, Gladwin, MT, Noguchi, CT, et al. Renal pathology in hemizygous sickle cell mice. Toxicol Pathol. 2002;30:254–262.CrossRefGoogle ScholarPubMed
Bonnin, P, Sabaa, N, Flamant, M, Debbabi, H, Tharaux, PL. Ultrasound imaging of renal vaso–occlusive events in transgenic sickle mice exposed to hypoxic stress. Ultrasound Med Biol. 2008;34:1076–1084.CrossRefGoogle ScholarPubMed
Lutty, GA, McLeod, DS, Pachnis, A, et al. Retinal and choroidal neovascularization in a transgenic mouse model of sickle cell disease. Am J Pathol. 1994;145:490–497.Google Scholar
Lutty, GA, Merges, C, McLeod, DS, et al. Nonperfusion of retina and choroid in transgenic mouse models of sickle cell disease. Curr Eye Res. 1998;17:438–444.CrossRefGoogle ScholarPubMed
McLeod, DS, Merges, C, Fukushima, A, Goldberg, MF, Lutty, GA. Histopathologic features of neovascularization in sickle cell retinopathy. Am J Ophthalmol. 1997;124:455–472.CrossRefGoogle ScholarPubMed
Hebbel, RP, Yamada, O, Moldow, CF, et al. Abnormal adherence of sickle erythrocytes to cultured vascular endothelium: possible mechanism for microvascular occlusion in sickle cell disease. J Clin Invest. 1980;65:154–160.CrossRefGoogle ScholarPubMed
Hebbel, RP, Boogaerts, MAB, Eaton, JW, Steinberg, MH. Erythrocyte adherence in sickle cell disorders. N Engl J Med. 1980;302:992.CrossRefGoogle Scholar
Kaul, DK, Fabry, ME, Costantini, F, Rubin, EM, Nagel, RL. In vivo demonstration of red cell-endothelial interaction, sickling and altered microvascular response to oxygen in the sickle transgenic mouse. J Clin Invest. 1995;96:2845–2853.CrossRefGoogle ScholarPubMed
Kaul, DK, Liu, XD, Fabry, ME, Nagel, RL. Impaired nitric oxide-mediated vasodilation in transgenic sickle mouse. Am J Physiol Heart Circ Physiol. 2000;278:H1799–H1806.CrossRefGoogle ScholarPubMed
Embury, SH, Mohandas, N, Paszty, C, Cooper, P, Cheung, AT. In vivo blood flow abnormalities in the transgenic knockout sickle cell mouse. J Clin Invest. 1999;103:915–920.CrossRefGoogle ScholarPubMed
Kaul, DK, Hebbel, RP. Hypoxia/reoxygenation causes inflammatory response in transgenic sickle mice but not in normal mice. J Clin Invest. 2000;106:411–420.CrossRefGoogle ScholarPubMed
Aslan, M, Ryan, TM, Adler, B, et al. Oxygen radical inhibition of nitric oxide-dependent vascular function in sickle cell disease. Proc Natl Acad Sci USA. 2001;98:15215–15220.CrossRefGoogle ScholarPubMed
Turhan, A, Weiss, , Mohandas, N, Coller, BS, Frenette, PS. Primary role for adherent leukocytes in sickle cell vascular occlusion: a new paradigm. Proc Natl Acad Sci USA. 2002;99:3047–3051.CrossRefGoogle ScholarPubMed
Chang, J, Shi, PA, Chiang, EY, Frenette, PS. Intravenous immunoglobulins reverse acute vaso-occlusive crises in sickle cell mice through rapid inhibition of neutrophil adhesion. Blood. 2008;111:915–923.CrossRefGoogle ScholarPubMed
Wood, KC, Hebbel, RP, Lefer, DJ, Granger, DN. Critical role of endothelial cell-derived nitric oxide synthase in sickle cell disease-induced microvascular dysfunction. Free Radic Biol Med. 2006;40:1443–1453.CrossRefGoogle ScholarPubMed
Kaul, DK, Zhang, X, Dasgupta, T, Fabry, ME. Arginine therapy of transgenic-knockout sickle mice improves microvascular function by reducing non-nitric oxide vasodilators, hemolysis and oxidative stress. Am J Physiol Heart Circ Physiol. 2008;295:H39–47.CrossRefGoogle ScholarPubMed
Osei, SY, Ahima, RS, Fabry, ME, Nagel, RL, Bank, N. Immunohistochemical localization of hepatic nitric oxide synthase in normal and transgenic sickle cell mice: the effect of hypoxia. Blood. 1996;88:3583–3588.Google ScholarPubMed
Fabry, ME, Romero, JR, Suzuka, SM, et al. Hemoglobin C in transgenic mice: effect of HbC expression from founders to full mouse globin knockouts. Blood Cell Mol Dis. 2000;26:331–347.CrossRefGoogle ScholarPubMed
Nagar, S, Remmel, RP, Hebbel, RP, Zimmerman, CL. Metabolism of opioids is altered in liver microsomes of sickle cell transgenic mice. Drug Metab Dispos. 2004;32:98–104.CrossRefGoogle ScholarPubMed
May, C, Rivella, S, Callegari, J, et al. Therapeutic haemoglobin synthesis in beta-thalassaemic mice expressing lentivirus-encoded human beta-globin. Nature. 2000;406:82–86.CrossRefGoogle ScholarPubMed
Pawliuk, R, Westerman, KA, Fabry, ME, et al. Correction of sickle cell disease in transgenic mouse models by gene therapy. Science. 2001;294:2368–2371.CrossRefGoogle ScholarPubMed
Levasseur, DN, Ryan, TM, Pawlik, KM, Townes, TM. Correction of a mouse model of sickle cell disease: lentiviral/antisickling beta-globin gene transduction of unmobilized, purified hematopoietic stem cells. Blood. 2003;102:4312–4319.CrossRefGoogle ScholarPubMed
Imren, S, Payen, E, Westerman, KA, et al. Permanent and panerythroid correction of murine beta thalassemia by multiple lentiviral integration in hematopoietic stem cells. Proc Natl Acad Sci USA. 2002;99:14380–14385.CrossRefGoogle ScholarPubMed
Franceschi, L, Baron, A, Scarpa, A et al. Inhaled nitric oxide protects transgenic SAD mice from sickle cell disease-specific lung injury induced by hypoxia/reoxygenation. Blood. 2003; 102:1087–1096.CrossRefGoogle ScholarPubMed
Minneci, PC, Deans, KJ, Zhi, H, et al. Hemolysis-associated endothelial dysfunction mediated by accelerated NO inactivation by decompartmentalized oxyhemoglobin. J Clin Invest. 2005;115:3409–3417.CrossRefGoogle ScholarPubMed
Pritchard, KA, Ou, J, Ou, Z, et al. Hypoxia-induced acute lung injury in murine models of sickle cell disease. Am J Physiol Lung Cell Mol Physiol. 2004;286:L705–L714.CrossRefGoogle ScholarPubMed
Holtzclaw, JD, Jack, D, Aguayo, SM, et al. Enhanced pulmonary and systemic response to endotoxin in transgenic sickle mice. Am J Respir Crit Care Med. 2004;169:687–695.CrossRefGoogle ScholarPubMed
Zhang, S, Li, H, Ma, L, et al. Polynitroxyl-albumin (PNA) plus tempol attenuate lung capillary leak elicited by prolonged intestinal ischemia and reperfusion. Free Radic Biol Med. 2000;29:42–50.CrossRefGoogle ScholarPubMed
Franceschi, L, Malpeli, G, Scarpa, A, et al. Protective effects of S-nitrosoalbumin on lung injury induced by hypoxia-reoxygenation in mouse model of sickle cell disease. Am J Physiol Lung Cell Mol Physiol. 2006;291:L457–L465.CrossRefGoogle ScholarPubMed
Hsu, LL, Champion, HC, Campbell-Lee, SA, et al. Hemolysis in sickle cell mice causes pulmonary hypertension due to global impairment in nitric oxide bioavailability. Blood. 2007;109:3088–3098.Google ScholarPubMed
Bartolucci, P, Ngo, MT, Beuzard, Y, et al. Decrease in lung nitric oxide production after peritonitis in mice with sickle cell disease. Crit Care Med. 2007;35:502–509.CrossRefGoogle ScholarPubMed
Franceschi, L, Platt, OS, Malpeli, G, et al. Protective effects of phosphodiesterase-4 (PDE-4) inhibition in the early phase of pulmonary arterial hypertension in transgenic sickle cell mice. FASEB J. 2008;22:1849–1860.CrossRefGoogle ScholarPubMed
Kennan, RP, Suzuka, SM, Nagel, RL, Fabry, ME. Decreased cerebral perfusion correlates with increased BOLD hyperoxia response in transgenic mouse models of sickle cell disease. Magn Reson Med. 2004;51:525–532.CrossRefGoogle ScholarPubMed
Wood, KC, Hebbel, RP, Granger, DN. Endothelial cell P-selectin mediates a proinflammatory and prothrombogenic phenotype in cerebral venules of sickle cell transgenic mice. Am J Physiol Heart Circ Physiol. 2004;286:H1608–H1614.CrossRefGoogle ScholarPubMed
Wood, KC, Hebbel, RP, Granger, DN. Endothelial cell NADPH oxidase mediates the cerebral microvascular dysfunction in sickle cell transgenic mice. FASEB J. 2005;19:989–991.CrossRefGoogle ScholarPubMed
White, SP, Birch, P, Kumar, R. Interactions at the alpha 1 beta 1 interface in hemoglobin: a single amino acid change affects dimer ratio in transgenic mice expressing human hemoglobin. Hemoglobin. 1994;18:413–426.CrossRefGoogle Scholar
Arcasoy, MO, Romana, M, Fabry, ME, et al. High levels of human gamma-globin gene expression in adult mice carrying a transgene of deletion-type hereditary persistence of fetal hemoglobin. Mol Cell Biol. 1997;17:2076–2089.CrossRefGoogle ScholarPubMed
Nagel, RL, Sharma, A, Kumar, R, Fabry, ME. Severe red cell abnormalities in transgenic mice expressing high levels of normal human delta chains. Blood. 1995;86(10):990A.Google Scholar
Chen, Q, Bouhassira, EE, Besse, A, et al. Generation of transgenic mice expressing human hemoglobin E. Blood Cell Mol Dis. 2004;33:303–307.CrossRefGoogle ScholarPubMed
Alami, R, Gilman, JG, Feng, YQ, et al. Anti-bS-ribozyme reduces bS mRNA levels in transgenic mice: potential application to the gene therapy of sickle cell anemia. Blood Cell Mol Dis. 1999;25:110–119.CrossRefGoogle Scholar
Seibler, J, Schubeler, D, Fiering, S, Groudine, M, Bode, J. DNA cassette exchange in ES cells mediated by Flp recombinase: an efficient strategy for repeated modification of tagged loci by marker-free constructs. Biochemistry. 1998;37:6229–6234.CrossRefGoogle ScholarPubMed
Bouhassira, EE, Westerman, K, Leboulch, P. Transcriptional behavior of LCR enhancer elements integrated at the same chromosomal locus by recombinase-mediated cassette exchange. Blood. 1997;90:3332–3344.Google ScholarPubMed
Aird, WC. Phenotypic heterogeneity of the endothelium: I. Structure, function, and mechanisms. Circ Res. 2007;100:158–173.CrossRefGoogle ScholarPubMed
Aird, WC. Phenotypic heterogeneity of the endothelium: II. Representative vascular beds. Circ Res. 2007;100:174–190.CrossRefGoogle ScholarPubMed
Nagel, RL, Fabry, ME. The panoply of animal models for sickle cell anaemia. Br J Haematol. 2001;112:19–25.CrossRefGoogle ScholarPubMed
Beuzard, Y. Mouse models of sickle cell disease. Transfus Clin Biol. 2008;15:7–11.CrossRefGoogle ScholarPubMed
Sadelain, M. Recent advances in globin gene transfer for the treatment of beta-thalassemia and sickle cell anemia. Curr Opin Hematol. 2006;13:142–148.CrossRefGoogle ScholarPubMed
Pei, D. The magic continues for the iPS strategy. Cell Res. 2008;18:221–223.CrossRefGoogle ScholarPubMed
Brugnara, C, Franceschi, L. Clinical trials of new therapeutic pharmacology for sickle cell disease. Sante. 2006;16:263–268.Google ScholarPubMed
Wood, KC, Granger, DN. Sickle cell disease: role of reactive oxygen and nitrogen metabolites. Clin Exp Pharmacol Physiol. 2007;34:926–932.CrossRefGoogle ScholarPubMed
Wood, KC, Hsu, LL, Gladwin, MT. Sickle cell disease vasculopathy: A state of nitric oxide resistance. Free Radic Biol Med. 2008;44:1506–1528.CrossRefGoogle ScholarPubMed
Kaul, DK. Sickle red cell adhesion: many issues and some answers. Transfus Clin Biol. 2008;15:51–55.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×