Skip to main content
Log in

New Strategies for the Treatment of Pulmonary Hypertension in Sickle Cell Disease

The Rationale for Arginine Therapy

  • Review Article
  • Published:
Treatments in Respiratory Medicine

Abstract

Nitric oxide (NO) is inactivated in sickle cell disease (SCD), while bioavailability of arginine, the substrate for NO synthesis, is diminished. Impaired NO bioavailability represents the central feature of endothelial dysfunction, and is a key factor in the pathophysiology of SCD. Inactivation of NO correlates with the hemolytic rate and is associated with erythrocyte release of cell-free hemoglobin and arginase during hemolysis. Accelerated consumption of NO is enhanced further by the inflammatory environment of oxidative stress that exists in SCD. Based upon its critical role in mediating vasodilation and cell growth, decreased NO bioavailability has also been implicated in the pathogenesis of pulmonary arterial hypertension (PHT). Secondary PHT is a common life-threatening complication of SCD that also occurs in most hereditary and chronic hemolytic disorders. Aberrant arginine metabolism contributes to endothelial dysfunction and PHT in SCD, and is strongly associated with prospective patient mortality. The central mechanism responsible for this metabolic disorder is enhanced arginine turnover, occurring secondary to enhanced plasma arginase activity. This is consistent with a growing appreciation of the role of excessive arginase activity in human diseases, including asthma and PHT. Decompart-mentalization of hemoglobin into plasma consumes endothelial NO and thus drives a metabolic requirement for arginine, whose bioavailability is further limited by arginase activity. New treatments aimed at maximizing both arginine and NO bioavailability through arginase inhibition, suppression of hemolytic rate, or oral arginine supplementation may represent novel therapeutic strategies.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Table I
Fig. 2
Fig. 3

Similar content being viewed by others

References

  1. Bunn HF. Pathogenesis and treatment of sickle cell disease. N Engl J Med 1997; 337(11): 762–9

    PubMed  CAS  Google Scholar 

  2. Stuart MJ, Nagel RL. Sickle-cell disease. Lancet 2004; 364(9442): 1343–60

    PubMed  Google Scholar 

  3. Castro O, Hoque M, Brown M. Pulmonary hypertension in sickle cell disease: cardiac catheterization results and survival. Blood 2003; 101: 1257–61

    PubMed  CAS  Google Scholar 

  4. Gladwin M, Sachdev V, Jison M, et al. Pulmonary hypertension as a risk factor for death in patients with sickle cell disease. N Engl J Med 2004; 350: 22–31

    Google Scholar 

  5. Sutton LL, Castro O, Cross DJ, et al. Pulmonary hypertension in sickle cell disease. Am J Cardiol 1994; 74: 626–8

    PubMed  CAS  Google Scholar 

  6. Ataga KI, Sood N, De Gent G, et al. Pulmonary hypertension in sickle cell disease. Am J Med 2004; 117: 665–9

    PubMed  Google Scholar 

  7. Morris CR, Kato GJ, Poljakovic M, et al. Dysregulated arginine metabolism, hemolysis-associated pulmonary hypertension and mortality in sickle cell disease. JAMA 2005 Jul 6; 294(1): 81–90

    PubMed  CAS  Google Scholar 

  8. Vichinsky EP. Pulmonary hypertension in sickle cell disease: a time for intervention. N Engl J Med 2004; 350: 857–9

    PubMed  CAS  Google Scholar 

  9. Morris CR, Gardner J, Hagar W, et al. Pulmonary hypertension in sickle cell disease: a common complication for both adults and children [abstract no. 1666]. Blood 2004; 104: 463a

    Google Scholar 

  10. Hebbel RP, Osarogiagbon KD. The endothelial biology of sickle cell disease: inflammation and a chronic vasculopathy. Microcirculation 2004; 11: 129–51

    PubMed  CAS  Google Scholar 

  11. Frenette PS. Sickle cell vaso-occlusion: multistep and multicellular paradigm. Curr Opin Hematol 2002; 9(2): 101–6

    PubMed  Google Scholar 

  12. Parise LV, Telen MJ. Erythrocyte adhesion in sickle cell disease. Curr Hematol Rep 2003; 2(2): 102–8

    PubMed  Google Scholar 

  13. Voetsch B, Jin RC, Loscalzo J. Nitric oxide insufficiency and atherothrombosis. Histochem Cell Biol 2004; 122(4): 353–67

    PubMed  CAS  Google Scholar 

  14. Jison ML, Gladwin MT. Hemolytic anemia-associated pulmonary hypertension of sickle cell disease and the nitric oxide/arginine pathway. Am J Respir Crit Care Med 2003; 168: 3–4

    PubMed  Google Scholar 

  15. Rother RP, Bell L, Hillmen P, et al. The clinical sequelae of intravascular hemolysis and extracellular plasma hemoglobin: a novel mechanism of human disease. JAMA 2005; 293: 1653–62

    PubMed  CAS  Google Scholar 

  16. Morris C, Kuypers F, Kato G, et al. Hemolysis-associated pulmonary hypertension in thalassemia. Ann N Y Acad Sci 2005; 1054: 481–5

    PubMed  Google Scholar 

  17. Lin EE, Rodgers GP, Gladwin MT. Hemolytic anemia-associated pulmonary hypertension in sickle cell disease. Curr Hematol Rep 2005; 4(2): 117–25

    PubMed  CAS  Google Scholar 

  18. Reiter C, Wang X, Tanus-Santos J, et al. Cell-free hemoglobin limits nitric oxide bioavailability in sickle cell disease. Nat Med 2002; 8: 1383–9

    PubMed  CAS  Google Scholar 

  19. Rubin LJ. Primary pulmonary hypertension. N Engl J Med 1997; 336: 111–7

    PubMed  CAS  Google Scholar 

  20. Loscalzo J. Genetic clues to the cause of primary pulmonary hypertension. N Engl J Med 2001; 345: 367–71

    PubMed  CAS  Google Scholar 

  21. Moraes D, Loscalzo J. Pulmonary hypertension: newer concepts in diagnosis and management. Clin Cardiol 1997; 20: 676–82

    PubMed  CAS  Google Scholar 

  22. Farber HW, Loscalzo J. Pulmonary arterial hypertension. N Engl J Med 2004; 351: 1655–65

    PubMed  CAS  Google Scholar 

  23. Tuder RM, Cool CD, Yeager M, et al. The pathobiology of pulmonary hypertension: endothelium. Clin Chest Med 2001; 22: 405–18

    PubMed  CAS  Google Scholar 

  24. Budhiraja R, Tuder RM, Hassoun PM. Endothelial dysfunction in pulmonary hypertension. Circulation 2004; 109: 159–65

    PubMed  Google Scholar 

  25. Morris CR, Teehankee C, Kato G, et al. Decreased arginine bioavailability contributes to the pathogenesis of pulmonary artery hypertension. American College of Cardiology Annual Meeting; 2005 Mar 6–9; Orlando (FL)

  26. Xu W, Kaneko TF, Zheng S, et al. Increased arginase II and decreased NO synthesis in endothelial cells of patients with pulmonary arterial hypertension. FASEB J 2004; 18: 1746–8

    PubMed  CAS  Google Scholar 

  27. Collins FS, Orringer EP. Pulmonary hypertension and cor pulmonale in the sickle hemoglobinopathies. Am J Med 1982; 73: 814–21

    PubMed  CAS  Google Scholar 

  28. Van Enk A, Visschers G, Jansen W, et al. Maternal death due to sickle cell chronic lung disease. Br J Obstet Gynaecol 1992; 99: 162–3

    PubMed  Google Scholar 

  29. Simmons BE, Santhanam V, Castaner A, et al. Two-dimensional echo and Doppler ultrasonographic findings in the hearts of adult patients with sickle cell anemia. Arch Intern Med 1988; 148: 1526–8

    PubMed  CAS  Google Scholar 

  30. Norris SL, Johnson C, Haywood LJ. Left ventricular filling pressure in sickle cell anemia. J Assoc Acad Minor Phys 1992; 3: 20–3

    PubMed  CAS  Google Scholar 

  31. Adedeji MO, Cespedes J, Allen K, et al. Pulmonary thrombotic arteriopathy in patients with sickle cell disease. Arch Pathol Lab Med 2001; 125: 1436–41

    PubMed  CAS  Google Scholar 

  32. Haque AK, Grinberg AR, Lencioni M, et al. Pulmonary hypertension in sickle cell hemoglobinopathy: a clinicopathologic study of 20 cases. Hum Pathol 2002; 33(33): 1037–43

    PubMed  Google Scholar 

  33. Morris CR, Morris Jr SM, Hagar W, et al. Arginine therapy: a new treatment for pulmonary hypertension in sickle cell disease? Am J Respir Crit Care Med 2003; 168: 63–9

    PubMed  Google Scholar 

  34. Castro O. Systemic fat embolism and pulmonary hypertension in sickle cell disease. Hematol Oncol Clin North Am 1996; 10: 1289–303

    PubMed  CAS  Google Scholar 

  35. Steward GW, Amess JA, Eber SW, et al. Thrombo-embolic disease after splenectomy for hereditary stomatocytosis. Br J Haematol 1996; 93: 303–10

    Google Scholar 

  36. Hoeper MM, Niedermeyer J, Hoffmeyer F, et al. Pulmonary hypertension after splenectomy? Ann Intern Med 1999; 130: 506–9

    PubMed  CAS  Google Scholar 

  37. Aessopos A, Stamatelow G, Skoumas V, et al. Pulmonary hypertension and right heart failure in patients with B-thalassemia intermedia. Chest 1995; 107: 50–3

    PubMed  CAS  Google Scholar 

  38. Powars DR, Pegelow CH. The spleen in sickle cell disease and thalassemia. Am J Pediatr Hematol Oncol 1979; 1(4): 343–53

    PubMed  CAS  Google Scholar 

  39. Aldrich TK, Dhuper SK, Patwa NS, et al. Pulmonary entrapment of sickle cells: the role of regional alveolar hypoxia. J Appl Physiol 1996; 80: 531–9

    PubMed  CAS  Google Scholar 

  40. Setty BN, Stuart MJ, Dampier C, et al. Hypoxaemia in sickle cell disease: biomarker modulation and relevance to pathophysiology. Lancet 2003; 362: 1450–5

    PubMed  CAS  Google Scholar 

  41. Setty BN, Stuart MJ. Vascular cell adhesion molecule-1 is involved in mediating hypoxia- induced sickle red blood cell adherence to endothelium: potential role in sickle cell disease. Blood 1996; 88: 2311–20

    PubMed  CAS  Google Scholar 

  42. Solovey A, Lin Y, Browne P, et al. Circulating activated endothelial cells in sickle cell anemia. N Engl J Med 1997; 337(22): 1584–90

    PubMed  CAS  Google Scholar 

  43. Vichinsky EP, Styles LA, Colangelo LH, et al. Acute chest syndrome in sickle cell disease: clinical presentation and course. Blood 1997; 89(5): 1787–92

    PubMed  CAS  Google Scholar 

  44. Phelan M, Perrine SP, Brauer M, et al. Sickle erythrocytes, after sickling, regulate the expression of the endothelin-1 gene and protein in human endothelial cells in culture. J Clin Invest 1995; 96: 1145–51

    PubMed  CAS  Google Scholar 

  45. Kaul DK, Hebbel RP. Hypoxia/reoxygenation causes inflammatory response in transgenic sickle mice but not in normal mice. J Clin Invest 2000; 106: 411–20

    PubMed  CAS  Google Scholar 

  46. Osarogiagbon UR, Choong S, Belcher JD, et al. Reperfusion injury pathophysiology in sickle cell transgenic mice. Blood 2000; 96: 314–20

    PubMed  CAS  Google Scholar 

  47. Key NS, Slungaard A, Dandelet L, et al. Whole blood tissue factor procoagulant activity is elevated in patients with sickle cell disease. Blood 1998 Jun 1; 91(11): 4216–23

    PubMed  CAS  Google Scholar 

  48. Powars D, Weidmen JA, Odom-Maryon T, et al. Sickle cell chronic lung disease: prior morbidity and the risk of pulmonary failure. Medicine (Baltimore) 1988; 67: 66–76

    CAS  Google Scholar 

  49. Liesner RJ, Vandenberghe FA. Sudden death in sickle cell disease. J R Soc Med 1993; 86: 484–5

    PubMed  CAS  Google Scholar 

  50. Romero Mestre JC, Hernandez A, Agramonte O, et al. Cardiovascular autonomic dysfunction in sickle cell anemia: a possible risk factor for sudden death? Clin Auton Res 1997; 7: 121–5

    PubMed  CAS  Google Scholar 

  51. Escoffery C, Shirley S. Causes of sudden natural death in Jamaica: a medicolegal (coroner’s) autopsy study from the University Hospital of the West Indies. Forensic Sci Int 2002; 129: 116–21

    PubMed  CAS  Google Scholar 

  52. McGoon M, Gutterman D, Steen V, et al. Screening, early detection and diagnosis of pulmonary artery hypertension. Chest 2004; 126: 14S–34S

    PubMed  Google Scholar 

  53. Minter K, Gladwin M. Pulmonary complications of sickle cell anemia: a need for increased recognition, treatment, and research. Am J Respir Crit Care Med 2001; 164: 2016–9

    PubMed  CAS  Google Scholar 

  54. Hammond TG, Mosesson MW. Fatal small-bowel necrosis and pulmonary hypertension in sickle cell disease. Arch Intern Med 1989; 149: 447–8

    PubMed  CAS  Google Scholar 

  55. Moncada S, Higgs A. The L-arginine-nitric oxide pathway. N Engl J Med 1993; 329: 2002–12

    PubMed  CAS  Google Scholar 

  56. Palmer RM, Ferrige AG, Moncada S. Nitric oxide release accounts for the biological activity of endothelium-derived relaxing factor. Nat Med 1987; 327: 524–6

    CAS  Google Scholar 

  57. Ignarro LJ. Heme-dependent activation of soluble guanylate cyclase by nitric oxide: regulation of enzyme activity by porphyrins and metalloporphyrins. Semin Hematol 1989; 26: 63–76

    PubMed  CAS  Google Scholar 

  58. Peng H-B, Spiecker M, Liao J. Inducible nitric oxide: an autoregulatory feedback inhibitor of vascular inflammation. J Immunol 1998; 161: 1970–6

    PubMed  CAS  Google Scholar 

  59. Kunes J, Hojna S, Kadlecova M, et al. Altered balance of vasoactive systems in experimental hypertension: the role of relative NO deficiency. Physiol Res 2004; 53Suppl. 1: S23–34

    PubMed  CAS  Google Scholar 

  60. Pieper GM. Review of alterations in endothelial nitric oxide production in diabetes: protective role of arginine on endothelial dysfunction. Hypertension 1998; 31: 1047–60

    PubMed  CAS  Google Scholar 

  61. de Boer J, Duyvendak M, Schuurman FE, et al. Role of L-arginine in the deficiency of nitric oxide and airway hyperreactivity after the allergen-induced early asthmatic reaction in guinea-pigs. Br J Pharmacol 1999; 128: 1114–20

    PubMed  CAS  Google Scholar 

  62. de Boer J, Meurs H, Coers W, et al. Deficiency of nitric oxide in allergen-induced airway hyperreactivity to contractile agonists after the early asthmatic reaction: an ex vivo study. Br J Pharmacol 1996; 119: 1109–16

    PubMed  Google Scholar 

  63. de Gouw HW, Hendriks J, Woltman AM, et al. Exhaled nitric oxide (NO) is reduced shortly after bronchoconstriction to direct and indirect stimuli in asthma. Am J Respir Crit Care Med 1998; 158: 315–9

    PubMed  Google Scholar 

  64. Creager MA, Gallagher SJ, Girerd XJ, et al. L-arginine improves endothelium-dependent vasodilation in hypercholesterolemic humans. J Clin Invest 1992; 90: 1248–53

    PubMed  CAS  Google Scholar 

  65. Tsao PS, McEvoy LM, Drexler H, et al. Enhanced endothelial adhesiveness in hypercholesterolemia is attenuated by L-arginine. Circulation 1994; 89(5): 2176–82

    PubMed  CAS  Google Scholar 

  66. Napoli C, Loscalzo J. Nitric oxide and other novel therapies for pulmonary hypertension. J Cardiovasc Pharmacol Ther 2004; 9: 1–8

    PubMed  CAS  Google Scholar 

  67. Hampl V, Herget J. Role of nitric oxide in the pathogenesis of chronic pulmonary hypertension. Physiol Rev 2000; 80: 1337–72

    PubMed  CAS  Google Scholar 

  68. Reiter CD, Gladwin MT. An emerging role for nitric oxide in sickle cell disease vascular homeostasis and therapy. Curr Opin Hematol 2003; 10: 99–107

    PubMed  CAS  Google Scholar 

  69. Morris CR, Kuypers FA, Larkin S, et al. Patterns of arginine and nitric oxide in sickle cell disease patients with vaso-occlusive crisis and acute chest syndrome. J Pediatr Hematol Oncol 2000; 22: 515–20

    PubMed  CAS  Google Scholar 

  70. Gladwin M, Schechter A, Ognibene F, et al. Divergent nitric oxide bioavailability in men and women with sickle cell disease. Circulation 2003; 107: 271–8

    PubMed  CAS  Google Scholar 

  71. Kaul DK, Liu X, Chang H, et al. Effect of fetal hemoglobin on microvascular regulation in sickle transgenic-knockout mice. J Clin Invest 2004; 114: 1136–45

    PubMed  CAS  Google Scholar 

  72. Reiter C, Wang X, Tanus-Santos J, et al. Cell-free hemoglobin limits nitric oxide bioavailability in sickle-cell disease. Nat Med 2002; 8: 1383–9

    PubMed  CAS  Google Scholar 

  73. Nath KA, Katusic ZS, Gladwin MT. The perfusion paradox and vascular instability in sickle cell disease. Microcirculation 2004; 11(2): 179–93

    PubMed  CAS  Google Scholar 

  74. Asian M, Ryan T, Adler B, et al. Oxygen radical inhibition of nitric oxide-dependent vascular function in sickle cell disease. Proc Natl Acad Sci U S A 2001; 98: 15215–20

    Google Scholar 

  75. Kaul DK, Liu XD, Fabry ME, et al. Impaired nitric oxide-mediated vasodilation in transgenic sickle mouse. Am J Physiol Heart Circ Physiol 2000; 278: H1799–806

    PubMed  CAS  Google Scholar 

  76. Nath KA, Shah V, Haggard JJ, et al. Mechanisms of vascular instability in a transgenic mouse model of sickle cell disease. Am J Physiol Regul Integr Comp Physiol 2000; 279(6): R1949–55

    PubMed  CAS  Google Scholar 

  77. Eberhardt RT, McMahon L, Duffy SJ, et al. Sickle cell anemia is associated with reduced nitric oxide bioactivity in peripheral conduit and resistance vessels. Am J Hematol 2003; 74: 104–11

    PubMed  CAS  Google Scholar 

  78. Belhassen L, Pelle G, Sediame S, et al. Endothelial dysfunction in patients with sickle cell disease is related to selective impairment of shear stress-mediated vasodilation. Blood 2001; 97: 1584–9

    PubMed  CAS  Google Scholar 

  79. Rees DC, Cervi P, Grimwade D, et al. The metabolites of nitric oxide in sickle-cell disease. Br J Haematol 1995; 91: 834–7

    PubMed  CAS  Google Scholar 

  80. Lonsdorfer J, Bogui P, Otayeck A, et al. Cardiorespiratory adjustments in chronic sickle cell anemia. Bull Eur Physiopathol Respir 1983; 19: 339–44

    PubMed  CAS  Google Scholar 

  81. Johnson CS, Giorgio AJ. Arterial blood pressure in adults with sickle cell disease. Arch Intern Med 1981; 141: 891–3

    PubMed  CAS  Google Scholar 

  82. Karayaylali I, Onal M, Yildizer K, et al. Low blood pressure, decreased incidence of hypertension, and renal cardiac, and autonomic nervous system functions in patients with sickle cell syndromes. Nephron 202; 91: 535–7

  83. 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(3): 150–6

    PubMed  CAS  Google Scholar 

  84. 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(2): 171–7

    PubMed  CAS  Google Scholar 

  85. Lopez BL, Barnett J, Ballas SK, et al. Nitric oxide metabolite levels in acute vaso-occlusive sickle-cell crisis. Acad Emerg Med 1996; 3: 1098–103

    PubMed  CAS  Google Scholar 

  86. Lopez B, Davis-Moon L, Ballas S. Sequential nitric oxide measurements during the emergency department treatment of acute vasoocclusive sickle cell crisis. Am J Hematol 2000; 64: 15–9

    PubMed  CAS  Google Scholar 

  87. Dias-Da-Motta P, Arruda V, Muscara M, et al. The release of nitric oxide and Superoxide anion by neutrophils and mononuclear cells from patients with sickle cell anaemia. Br J Haematol 1996; 93: 333–40

    PubMed  CAS  Google Scholar 

  88. Demiryurek A, Dakici I, Danzik I. Peroxynitrite: a putative cytotoxin. Pharm Toxicol 1998; 82: 113–7

    CAS  Google Scholar 

  89. Xia Y, Dawson V, Dawson T, et al. Nitric oxide synthase generates Superoxide and nitric oxide in arginine-depleted cells leading to peroxynitrite-mediated cellular injury. Proc Natl Acad Sci U S A 1996; 93: 6770–4

    PubMed  CAS  Google Scholar 

  90. Osei SY, Ahima RS, Fabry ME, et al. Immunohistochemical localization of hepatic nitric oxide synthase in normal and transgenic sickle cell mice: the effect of hypoxia. Blood 1996; 88: 3583–8

    PubMed  CAS  Google Scholar 

  91. Bank N, Aynedjian H, Qiu J, et al. Renal nitric oxide synthases in transgenic sickle cell mice. Kidney Int 1996; 50: 184–9

    PubMed  CAS  Google Scholar 

  92. Bassenge E. Endothelial function in different organs. Prog Cardiovasc Dis 1996; 39: 209–28

    PubMed  CAS  Google Scholar 

  93. Graido-Gonzalez E, Doherty LC, Bergreen EW, et al. Plasma endothelin-1, cytokine, and prostaglandin E2 levels in sickle cell disease and acute vasoocclusive sickle. Blood 1998; 92: 2551–5

    PubMed  CAS  Google Scholar 

  94. Rubin L, Badesch D, Barst R, et al. Bosentan therapy for pulmonary arterial hypertension. N Engl J Med 2002; 346: 896–903

    PubMed  CAS  Google Scholar 

  95. Dupuis J. Endothelin-receptor antagonists in pulmonary hypertension. Lancet 2001; 358: 1113–4

    PubMed  CAS  Google Scholar 

  96. Schnog JB, Rojer RA, MacGillavry MR, et al. Steady-state sVCAM-1 serum levels in adults with sickle cell disease. Ann Hematol 2003; 82: 109–13

    PubMed  CAS  Google Scholar 

  97. Duits AJ, Pieters RC, Saleh AW, et al. Enhanced levels of soluble VCAM-1 in sickle cell patients and their specific increment during vasoocclusive crisis. Clin Immunol Immunopathol 1996; 81: 96–8

    PubMed  CAS  Google Scholar 

  98. Natarajan M, Udden MM, McIntire LV. Adhesion of sickle red blood cells and damage to interleukin-1 beta stimulated endothelial cells under flow in vitro. Blood 1996; 87(11): 4845–52

    PubMed  CAS  Google Scholar 

  99. Swerlick RA, Eckman JR, Kumar A, et al. Alpha 4 beta 1-integrin expression on sickle reticulocytes: vascular cell adhesion molecule-1-dependent binding to endothelium. Blood 1993; 82(6): 1891–9

    PubMed  CAS  Google Scholar 

  100. Gee BE, Platt OS. Sickle reticulocytes adhere to VCAM-1. Blood 1995; 85(1): 268–74

    PubMed  CAS  Google Scholar 

  101. Stuart M, Setty Y. Sickle cell acute chest syndrome: pathogenesis and rational for treatment. Blood 1999; 94: 1555–60

    PubMed  CAS  Google Scholar 

  102. Kato GJ, Martyr S, Blackwelder WC, et al. Endothelial dysfunction in patients with sickle cell disease is associated with haemolytic rate, multi-organ failure, and risk of death. Blood; Epub 2005 Nov 15

    Google Scholar 

  103. Hallemeesch MM, Lamers WH, Deutz NE. Reduced arginine availability and nitric oxide production. Clin Nutr 2002; 21(4): 273–9

    PubMed  CAS  Google Scholar 

  104. Enwonwu CO. Increased metabolic demand for arginine in sickle cell anaemia. Med Sci Res 1989; 17: 997–8

    Google Scholar 

  105. Enwonwu CO, Xu X, Turner E. Nitrogen metabolism in sickle cell anemia: free amino acids in plasma and urine. Am J Med Sci 1990; 300: 366–71

    PubMed  CAS  Google Scholar 

  106. Waugh W, Daeschner C, Files B, et al. Evidence that L-arginine is a key amino acid in sickle cell anemia: a preliminary report. Nutr Res 1999; 19: 501–18

    CAS  Google Scholar 

  107. Mendes Ribeiro AC, Brunini TM. L-arginine transport in disease. Curr Med Chem Cardiovasc Hematol Agents 2004; 2: 123–31

    PubMed  Google Scholar 

  108. Morris CR, Kuypers FA, Larkin S, et al. Arginine therapy: a novel strategy to increase nitric oxide production in sickle cell disease. Br J Haematol 2000; 111: 498–500

    PubMed  CAS  Google Scholar 

  109. Lopez B, Kreshak A, Morris CR, et al. L-arginine levels are diminished in adult acute vaso-occlusive sickle cell crisis in the emergency department. Br J Haematol 2003; 120: 532–4

    PubMed  CAS  Google Scholar 

  110. Girgis RE, Qureshi MA, Abrams J, et al. Decreased exhaled nitric oxide in sickle cell disease: relationship to chronic lung involvement. Am J Hematol 2003; 72: 177–84

    PubMed  CAS  Google Scholar 

  111. Sullivan K, Kissoon N, Duckworth L, et al. Low exhaled nitric oxide and a polymorphism in the NOS I gene is associated with acute chest syndrome. Am J Respir Crit Care Med 2001; 164: 2186–90

    PubMed  CAS  Google Scholar 

  112. Heinzel B, John M, Klatt P, et al. Ca2+/calmodulin-dependent formation of hydrogen peroxide by brain nitric oxide synthase. Biochem J 1992; 281: 627–30

    PubMed  CAS  Google Scholar 

  113. Lee J, Ryu H, Ferrante R, et al. Translational control of inducible nitric oxide synthase expression by arginine can explain the arginine paradox. Proc Natl Acad Sci U S A 2003; 100: 4843–8

    PubMed  CAS  Google Scholar 

  114. El-Gayar S, Thuring-Nahler H, Pfeilschifter J, et al. Translational control of inducible nitric oxide synthase by IL-13 and arginine availability in inflammatory macrophages. J Immunol 2003; 171: 4561–8

    PubMed  CAS  Google Scholar 

  115. Kamada Y, Nagaretani H, Tamura S, et al. Vascular endothelial dysfunction resulting from L-arginine deficiency in a patient with lysinuric protein intolerance. J Clin Invest 2001; 108: 717–24

    PubMed  CAS  Google Scholar 

  116. Morris Jr SM. Regulation of enzymes of the urea cycle and arginine metabolism. Annu Rev Nutr 2002; 22: 87–105

    PubMed  CAS  Google Scholar 

  117. Morris Jr SM, Bhamidipati D, Kepka-Lenhart D. Human type II arginase: sequence analysis and tissue-specific expression. Gene 1997; 193: 157–61

    PubMed  CAS  Google Scholar 

  118. Vockley JG, Jenkinson CP, Shukula H, et al. Cloning and characterization of the human type II arginase gene. Genomics 1996; 38: 118–23

    PubMed  CAS  Google Scholar 

  119. Gotoh T, Sonoki T, Nagasaki A, et al. Molecular cloning of cDNA for nonhepatic mitochondrial arginase (arginase II) and comparison of its induction with nitric oxide synthase in a murine macrophage-like cell line. FEBS Lett 1996; 395: 119–22

    PubMed  CAS  Google Scholar 

  120. Kim P, Iyer R, Lu K, et al. Expression of the liver form of arginase in erythrocytes. Mol Genet Metab 2002; 76: 100–10

    PubMed  CAS  Google Scholar 

  121. Azizi E, Dror Y, Wallis K. Arginase activity in erythrocytes of healthy and ill children. Clin Chim Acta 1970; 28: 391–6

    PubMed  CAS  Google Scholar 

  122. Morris Jr SM. Regulation of arginine availability and its impact on NO synthesis. In: Ignarro L, editor. Nitric oxide: biology and pathobiology. San Diego (CA): Academic Press, 2000: 187–97

    Google Scholar 

  123. Mori M, Gotoh T. Relationship between arginase activity and nitric oxide production. In: Ignarro L, editor. Nitric oxide: biology and pathology. San Diego (CA): Academic Press, 2000: 199–208

    Google Scholar 

  124. Belfiore F. Enzymatic activities of the blood serum in thalassemia and in thalassodrepanocytosis. Riforma Med 1964; 78: 1052–5

    PubMed  CAS  Google Scholar 

  125. Reynolds J, Follette JH, Valentine WH. The arginase activity of erythrocytes and leukocytes with particular reference to pernicious anemia and thalassemia major. J Lab Clin Med 1957; 50: 78–92

    PubMed  CAS  Google Scholar 

  126. Verresen D, De Backer W, Van Meerbeeck J, et al. Spherocytosis and pulmonary hypertension: coincidental occurrence or causal relationship? Eur Respir J 1991; 4: 629–31

    PubMed  CAS  Google Scholar 

  127. Heller PG, Grinberg AR, Lencioni M, et al. Pulmonary hypertension in paroxysmal nocturnal hemoglobinuria. Chest 1992; 102: 642–3

    PubMed  CAS  Google Scholar 

  128. Labrune P, Sittoun J, Duvaltier I, et al. Haemolytic uraemic syndrome and pulmonary hypertension in a patient with methionine synthase deficiency. Eur J Pediatr 1999; 158: 734–9

    PubMed  CAS  Google Scholar 

  129. Stuard ID, Heusinkveld RS, Moss AJ. Microangiopathic haemolytic anemia and thrombocytopenia in primary pulmonary hypertension. N Engl J Med 1972; 287: 869–70

    PubMed  CAS  Google Scholar 

  130. Jubelirer SJ. Primary pulmonary hypertension: its association with microangiopathic haemolytic anemia and thrombocytopenia. Arch Intern Med 1991; 151: 1221–3

    PubMed  CAS  Google Scholar 

  131. Chou R, DeLoughery TG. Recurrent thromboembolic disease following splenectomy for pyruvate kinase deficiency. Am J Hematol 2001; 67: 197–9

    PubMed  CAS  Google Scholar 

  132. Mehta S, Stewart D, Langleben D, et al. Short-term pulmonary vasodilation with L-arginine in pulmonary hypertension. Circulation 1995; 92: 1539–45

    PubMed  CAS  Google Scholar 

  133. Archer S, Djaballah K, Humbert M, et al. Nitric oxide deficiency in fenfluramine and dexfenfluramine-induced pulmonary hypertension. Am J Respir Crit Care Med 1998; 158: 1061–7

    PubMed  CAS  Google Scholar 

  134. Cooper C, Landzberg M, Anderson T, et al. Role of nitric oxide in the local regulation of pulmonary vascular resistance in humans. Circulation 1996; 93: 266–71

    PubMed  CAS  Google Scholar 

  135. Kaneko F, Arroliga A, Dweik R, et al. Biochemical reaction products of nitric oxide as quantitative markers of primary pulmonary hypertension. Am J Respir Crit Care Med 1998; 158: 917–23

    PubMed  CAS  Google Scholar 

  136. Jison ML, Munson PJ, Barb JJ, et al. Blood mononuclear cell gene expression profiles characterize the oxidant, hemolytic, and inflammatory stress of sickle cell disease. Blood 2004; 104: 270–80

    PubMed  CAS  Google Scholar 

  137. Morris CR, Poljakovic M, Lavrisha L, et al. Decreased arginine bioavailability and increased serum arginase activity in asthma. Am J Respir Crit Care Med 2004; 170: 148–53

    PubMed  Google Scholar 

  138. Meurs H, Schuurman FE, Duyvendak M, et al. Deficiency of nitric oxide in polycation-induced airway hyperreactivity. Br J Pharmacol 1999; 126: 559–62

    PubMed  CAS  Google Scholar 

  139. Ricciardolo FL, Geppetti P, Mistretta A, et al. Randomized double-blind placebo-controlled study of the effect of inhibition of nitric oxide synthesis in bradykinin-induced asthma. Lancet 1996; 348: 374–7

    PubMed  CAS  Google Scholar 

  140. Ricciardolo FL, Di Maria GU, Mistretta A, et al. Impairment of bronchoprotection by nitric oxide in severe asthma. Lancet 1997; 350: 1297–8

    PubMed  CAS  Google Scholar 

  141. Ricciardolo FL, Timmers MC, Gepetti P, et al. Allergen-induced impairment of bronchoprotective nitric oxide synthesis in asthma. J Allergy Clin Immunol 2001; 108: 198–204

    PubMed  CAS  Google Scholar 

  142. Zimmerman N, King NE, Laporte J, et al. Dissection of expeirmental asthma with DNA microarray analysis identifies arginase in asthma pathogensisi. J Clin Invest 2003; 111: 1863–74

    Google Scholar 

  143. Meurs H, McKay S, Maarsingh H, et al. Increased arginase activity underlies allergen-induced deficiency of cNOS-derived nitric oxide and airway hyper-responsiveness. Br J Pharmacol 2002; 136: 391–8

    PubMed  CAS  Google Scholar 

  144. Meurs H, Hamer MA, Pethe S, et al. Modulation of cholinergic airway reactivity and nitric oxide production by endogenous arginase activity. Br J Pharmacol 2000; 130: 1793–8

    PubMed  CAS  Google Scholar 

  145. Meurs H, Maarsingh H, Zaagsma J. Arginase and asthma: novel insights into nitric oxide homeostasis and airway hyperresponsiveness. Trends Pharmacol Sci 2003; 24: 450–5

    PubMed  CAS  Google Scholar 

  146. Raghupathy R, Haider MZ, Azizieh F, et al. Th1 and Th2 cytokine profiles in sickle cell disease. Acta Haematol 2000; 103: 197–202

    PubMed  CAS  Google Scholar 

  147. Komai M, Tanaka H, Masuda T, et al. Role of Th2 responses in the development of allergen-induced airway remodeling in a murine model of allergic asthma. Br J Pharmacol 2003; 138: 912–20

    PubMed  CAS  Google Scholar 

  148. Chung KF, Barnes PJ. Cytokines in asthma. Thorax 1999; 54: 825–57

    PubMed  CAS  Google Scholar 

  149. Ahmed S, Siddiqui AK, Sadiq A, et al. Echocardiographic abnormalities in sickle cell disease. Am J Hematol 2004; 76(3): 195–8

    PubMed  Google Scholar 

  150. Atz AM, Wessel DL. Inhaled nitric oxide in sickle cell disease with acute chest syndrome. Anesthesiology 1997; 87: 988–90

    PubMed  CAS  Google Scholar 

  151. Sullivan KJ, Goodwin SR, Evangelist J, et al. Nitric oxide successfully used to treat acute chest syndrome of sickle cell disease in a young adolescent. Crit Care Med 1999; 27: 2563–8

    PubMed  CAS  Google Scholar 

  152. Vichinsky E, Neumayr L, Earles A, et al. Causes and outcomes of the acute chest syndrome in sickle cell disease. National Acute Chest Syndrome Study Group. N Engl J Med 2000; 342: 1855–65

    PubMed  CAS  Google Scholar 

  153. Koumbourlis A, Zar H, Hurlet-Jensen A, et al. Prevalence and reversibility of lower airway obstruction in children with sickle cell disease. J Pediatr 2001; 138: 188–92

    PubMed  CAS  Google Scholar 

  154. Leong M, Dampier C, Varlotta L, et al. Airway hyperreactivity in children with sickle cell disease. J Pediatr 1997; 131: 278–83

    PubMed  CAS  Google Scholar 

  155. Rastogi D, Ngai P, Barst RJ, et al. Lower airway obstruction, bronchial hyperresponsiveness, and primary pulmonary hypertension in children. Pediatr Pulmonol 2004; 37: 50–5

    PubMed  Google Scholar 

  156. Arnal J-F, Munzel T, Venema R, et al. Interactions between L-arginine and L-glutamine change endothelial NO production; an effect independent of NO synthase substrate availability. J Clin Invest 1995; 95: 2565–72

    PubMed  CAS  Google Scholar 

  157. Kurz S, Harrison D. Insulin and the arginine paradox. J Clin Invest 1997; 99: 369–70

    PubMed  CAS  Google Scholar 

  158. Wu G, Morris Jr SM. Arginine metabolism: nitric oxide and beyond. Biochem J 1998; 336: 1–17

    PubMed  CAS  Google Scholar 

  159. Graf P, Forstermann U, Closs E. The transport activity of the human cationic amino acid transporter hCAT-1 is downregulated by activation of protein kinase C. Br J Pharmacol 2001; 132: 1193–200

    PubMed  CAS  Google Scholar 

  160. Zharikov S, Block E. Association of L-arginine transporters with fodrin: implications for hypoxic inhibition of arginine uptake. Am J Physiol Lung Cell Mol Physiol 2000; 278: L111–7

    PubMed  CAS  Google Scholar 

  161. Closs EI, Mann GE. Membrane transport of L-arginine and cationic amino acid analogs. San Diego (CA): Academic Press, 2000

    Google Scholar 

  162. Closs EI. Expression, regulation and function of carrier proteins for cationic amino acids. Curr Opin Nephrol Hypertens 2002; 11: 99–107

    PubMed  Google Scholar 

  163. Wu G, Morris Jr SM. Metabolic and therapeutic aspects of amino acids in clinical nutrition. CRC Press: Boca Raton (FL), 2004

    Google Scholar 

  164. Schnog JB, Jager EH, van der Dijs FP, et al. Evidence for a metabolic shift of arginine metabolism in sickle cell disease. Ann Hematol 2004; 83: 371–5

    PubMed  CAS  Google Scholar 

  165. Natta CL, Motyczka AA, Kremzner LT. Polyamines in sickle cell disease. Biochem Med 1980; 23(2): 144–9

    PubMed  CAS  Google Scholar 

  166. Natta CL, Motyczka AA, Kremzner LT. Polyamines and globin binding in sickle cell disease. Am J Pediatr Hematol Oncol 1982; 4(1): 73–6

    PubMed  CAS  Google Scholar 

  167. VanderJagt DJ, Kanellis GJ, Isichei C, et al. Serum and urinary amino acid levels in sickle cell disease. J Trop Pediatr 1997; 43: 220–5

    PubMed  CAS  Google Scholar 

  168. Zimmermann N, King NE, Laporte J, et al. Dissection of experimental asthma with DNA microarray analysis identifies arginase in asthma pathogenesis. J Clin Invest 2003; 111: 1863–74

    PubMed  CAS  Google Scholar 

  169. Wei LH, Wu Jr SM, Ignarro LJ. Elevated arginase I expression in rat aortic smooth muscle cells increases cell proliferation. Proc Natl Acad Sci U S A 2001; 98: 9260–4

    PubMed  CAS  Google Scholar 

  170. Li H, Meininger CJ, Kelly KA, et al. Activities of arginase I and II are limiting for endothelial cell proliferation. Am J Physiol Regul Integr Comp Physiol 2002; 282: R64–9

    PubMed  CAS  Google Scholar 

  171. Vallance P. The asymmetrical dimethylarginine/dimethylarginine dimethylaminohydrolase pathway in the regulation of nitric oxide generation. Clin Sci 2001; 100: 159–60

    PubMed  CAS  Google Scholar 

  172. Cooke JP, Mont-Reynaud R, Tsao PS, et al. Nitric oxide and vascular disease. In: Ignarro LJ, editor. Nitric oxide: biology and pathology. New York: Academic Press, 2000: 759–83

    Google Scholar 

  173. Stuhlinger MC, Oka RK, Graf EE, et al. Endothelial dysfunction induced by hyperhomocyst (e)inemia: role of asymmetric dimethylarginine. Circulation 2003; 108: 933–8

    PubMed  Google Scholar 

  174. Boger RH, Bode-Boger SM. Asymmetric dimethylarginine, derangements of the endothelial nitric oxide synthase pathway, and cardiovascular disease. Semin Thromb Hemost 2000; 26: 539–45

    PubMed  CAS  Google Scholar 

  175. Millatt LJ, Whitley GS, Li D, et al. Evidence for dysregulation of dimethylarginine dimethylaminohydrolase I in chronic hypoxia-induced pulmonary hypertension. Circulation 2003; 108(12): 1493–8

    PubMed  CAS  Google Scholar 

  176. Gorenflo M, Zheng C, Werle E, et al. Plasma levels of asymmetrical dimethyl-L-arginine in patients with congenital heart disease and pulmonary hypertension. J Cardiovasc Pharmacol 2001; 37(4): 489–92

    PubMed  CAS  Google Scholar 

  177. Wu G, Meininger CJ, Knabe DA, et al. Arginine nutrition in development, health and disease. Curr Opin Clin Nutr Metab Care 2000; 3(1): 59–66

    PubMed  CAS  Google Scholar 

  178. Wu G. Intestinal mucosal amino acid catabolism. J Nutr 1998; 128: 1249–52

    PubMed  CAS  Google Scholar 

  179. Waugh WH, Daeschner CW, Files BA, et al. Oral citrulline as arginine precursor may be beneficial in sickle cell disease: early phase two results. J Natl Med Assoc 2001; 93: 363–71

    PubMed  CAS  Google Scholar 

  180. Merimee TJ, Rabinowitz D, Riggs L, et al. Plasma growth hormone after arginine infusion. N Engl J Med 1967; 276: 434–9

    PubMed  CAS  Google Scholar 

  181. Merimee TJ, Rabinowitz D, Fineberg S. Arginine-initiated release of human growth hormone. N Engl J Med 1969; 28: 1434–8

    Google Scholar 

  182. Barbul A, Rettura G, Levenson SM, et al. Wound healing and thymotropic effects of arginine: a pituitary mechanism of action. Am J Clin Nutr 1983; 37: 786–94

    PubMed  CAS  Google Scholar 

  183. Solomons C, Cotton EK, Dubois R. The use of buffered L-arginine in the treatment of cystic fibrosis. Pediatrics 1971; 47: 384–90

    PubMed  CAS  Google Scholar 

  184. Lerman A, Burnett JC, Higano ST, et al. Long-term L-arginine supplementation improves small-vessel coronary endothelial function in human. Circulation 1998; 97: 2123–8

    PubMed  CAS  Google Scholar 

  185. Perrine SP, Olivieri NF, Faller DV, et al. Butyrate derivatives: new agents for stimulating fetal globin production in the beta-globin disorders. Am J Pediatr Hematol Oncol 1994; 16: 67–71

    PubMed  CAS  Google Scholar 

  186. Perrine SP, Ginder GD, Faller DV, et al. A short-term trial of butyrate to stimulate fetal-globin-gene expression in the beta-globin disorders. N Engl J Med 1993; 328(2): 81–6

    PubMed  CAS  Google Scholar 

  187. Evans RW, Fernstrom JD, Thompson J, et al. Biochemical responses of healthy subjects during dietary supplementation with L-arginine. J Nutr Biochem 2004; 15: 534–9

    PubMed  CAS  Google Scholar 

  188. Boger RH, Bode-Boger SM. The clinical pharmacology of L-arginine. Annu Rev Pharmacol Toxicol 2001; 41: 79–99

    PubMed  CAS  Google Scholar 

  189. Martin WJ, Matzke GR. Treating severe metabolic alkalosis. Clin Pharm 1982; 1(1): 42–8

    PubMed  CAS  Google Scholar 

  190. Barbul A. Arginine: biochemistry, physiology and therapeutic implications. JPEN J Parenter Enterai Nutr 1986; 10: 227–38

    CAS  Google Scholar 

  191. Adams MR, Forsyth CJ, Jessup W, et al. Oral L-arginine inhibits platelet aggregation but does not enhance endothelium-dependent dilation in healthy young men. J Am Coll Cardiol 1995; 26: 1054–61

    PubMed  CAS  Google Scholar 

  192. Tangphao O, Chalon S, Moreno H, et al. Pharmacokinetics of L-arginine during chronic administration to patients with hypercholesterolaemia. Clin Sci 1999; 96: 199–207

    PubMed  CAS  Google Scholar 

  193. Morris Jr SM. Enzymes of arginine metabolism. J Nutr 2004; 134: 2743S–7S

    PubMed  CAS  Google Scholar 

  194. Morris Jr SM. Recent advances in arginine metabolism. Curr Opin Clin Nutr Metab Care 2004; 7: 45–51

    PubMed  CAS  Google Scholar 

  195. Maxwell AJ, Cooke JP. Cardiovascular effects of L-arginine. Curr Opin Nephrol Hypertens 1998; 7: 63–70

    PubMed  CAS  Google Scholar 

  196. Morris CR, Vichinsky EP, van Warmerdam J, et al. Hydroxyurea and arginine therapy: impact on nitric oxide production in sickle cell disease. J Pediatr Hematol Oncol 2003; 25: 629–34

    PubMed  Google Scholar 

  197. American Society of Health Systems Pharmacists. In: McEvoy G, editor. Bethesda (MD): American Hospital Formulary Service: Drug Information, 1999: 2185–6

    Google Scholar 

  198. Siani A, Pagano E, Iacone R, et al. Blood pressure and metabolic changes during dietary L-arginine supplementation in humans. Am J Hypertens 2000; 13 (5 Pt 1): 547–51

    PubMed  CAS  Google Scholar 

  199. Reyes AA, Karl IE, Klahr S. Role of arginine in health and in renal disease. Am J Physiol 1994; 267 (3 Pt 2): F331–46

    PubMed  CAS  Google Scholar 

  200. Tews JK, Bradford AM, Harper AE. Induction of lysine imbalance in rats: relationships between tissue amino acids and diet. J Nutr 1981; 111: 968–78

    PubMed  CAS  Google Scholar 

  201. Krauss JS, Freant LJ, Lee JR. Gastrointestinal pathology in sickle cell disease. Ann Clin Lab Sci 1998; 28(1): 19–23

    PubMed  CAS  Google Scholar 

  202. Bauer TW, Moore GW, Hutchins GM. The liver in sickle cell disease: a clinicopathologic study of 70 patients. Am J Med 1980; 69(6): 833–7

    PubMed  CAS  Google Scholar 

  203. Hertz P, Richardson J. Arginine-induced hyperkalemia in renal failure patients. Arch Intern Med 1972; 130: 778–80

    PubMed  CAS  Google Scholar 

  204. Gerard JM, Luisiri A. A fatal overdose of arginine hydrochloride. J Toxicol Clin Toxicol 1997; 35(6): 621–5

    PubMed  CAS  Google Scholar 

  205. Kharitonov SA, Lubec G, Lubec B, et al. L-arginine increases exhaled nitric oxide in normal human subjects. Clin Sci (Colch) 1995; 88(2): 135–9

    CAS  Google Scholar 

  206. Solomons C, Hathaway W, Cotton E. L-arginine, the sickling phenomenon, and cystic fibrosis. Pediatrics 1972; 49: 933

    PubMed  CAS  Google Scholar 

  207. Grasemann H, Grasemann C, Kurtz F, et al. Oral L-arginine supplementation in cystic fibrosis patients: a placebo-controlled study. Eur Respir J 2005; 25(1): 62–8

    PubMed  CAS  Google Scholar 

  208. Everard ML, Donnelly D. A pilot study of oral L-arginine in cystic fibrosis. J Cyst Fibros 2005; 4(1): 67–9

    PubMed  CAS  Google Scholar 

  209. Cooke JP, Singer AH, Tsao P, et al. Antiatherogenic effects of L-arginine in the hypercholesterolemic rabbit. J Clin Invest 1992; 90: 1168–72

    PubMed  CAS  Google Scholar 

  210. Nijkamp FP, Folkerts G. Nitric oxide and bronchial hyperresponsiveness. Arch Int Pharmacodyn Ther 1995; 329: 81–96

    PubMed  CAS  Google Scholar 

  211. Hamid Q, Springall DR, Riveros-Morena V, et al. Induction of nitric oxide synthase in asthma. Lancet 1993; 342: 1510–3

    PubMed  CAS  Google Scholar 

  212. Gornik HL, Creager MA. Arginine and endothelial and vascular health. J Nutr 2004; 134 (10 Suppl.): 2880S–7S

    PubMed  CAS  Google Scholar 

  213. Appleton J. Arginine: clinical potential of a semi-essential amino. Altern Med Rev 2002; 7(6): 512–22

    PubMed  Google Scholar 

  214. Kawano T, Nomura M, Nisikado A, et al. Supplementation of L-arginine improves hypertension and lipid metabolism but not insulin resistance in diabetic rats. Life Sci 2003; 73(23): 3017–26

    PubMed  CAS  Google Scholar 

  215. Drexler H, Zeiher AM, Meinzer K, et al. Correction of endothelial dysfunction in coronary microcirculation of hypercholesterolaemic patients by L-arginine. Lancet 1991; 338: 1546–50

    PubMed  CAS  Google Scholar 

  216. Kernohan AF, McIntyre M, Hughes DM, et al. An oral yohimbine/L-arginine combination (NMI 861) for the treatment of male erectile dysfunction: a pharmacokinetic, pharmacodynamic and interaction study with intravenous nitroglycerine in healthy male subjects. Br J Clin Pharmacol 2005; 59(1): 85–93

    PubMed  CAS  Google Scholar 

  217. Klotz T, Mathers MJ, Braun M, et al. Effectiveness of oral L-arginine in first-line treatment of erectile dysfunction in a controlled crossover study. Urol Int 1999; 63(4): 220–3

    PubMed  CAS  Google Scholar 

  218. Rytlewski K, Olszanecki R, Korbut R, et al. Effects of prolonged oral supplementation with l-arginine on blood pressure and nitric oxide synthesis in preeclampsia. Eur J Clin Invest 2005; 35(1): 32–7

    PubMed  CAS  Google Scholar 

  219. Preli RB, Klein KP, Herrington DM. Vascular effects of dietary L-arginine supplementation. Atherosclerosis 2002; 162(1): 1–15

    PubMed  CAS  Google Scholar 

  220. Rector TS, Bank AJ, Mullen KA, et al. Randomized, double-blind, placebo controlled study of supplemental oral L-arginine in patients with heart failure. Circulation 1996; 93: 2135–41

    PubMed  CAS  Google Scholar 

  221. Nagaya N, Uematsu M, Oya H, et al. Short-term oral administration of L-arginine improves hemodynamics and exercise capacity in patients with precapillary pulmonary hypertension. Am J Respir Crit Care Med 2001; 163: 887–91

    PubMed  CAS  Google Scholar 

  222. McCaffrey M, Bose C, Reiter P, et al. Effect of L-arginine infusion on infants with persistent pulmonary hypertension of the newborn. Biol Neonate 1995; 67: 240–3

    PubMed  CAS  Google Scholar 

  223. Baudouin SV, Bath P, Martin JF, et al. L-arginine infusion has no effect on systemic haemodynamics in normal volunteers, or systemic and pulmonary haemodynamics in patients with elevated pulmonary vascular resistance. Br J Clin Pharmacol 1993; 36: 45–9

    PubMed  CAS  Google Scholar 

  224. Mitani Y, Maruyama K, Sakurai M. Prolonged administration of L-arginine ameliorates chronic pulmonary hypertension and pulmonary vascular remodeling in rats. Circulation 1997; 96(2): 689–97

    PubMed  CAS  Google Scholar 

  225. Surdacki A, Zmudka K, Bieron K, et al. Lack of beneficial effects of L-arginine infusion in primary pulmonary hypertension. Wien Klin Wochenschr 1994; 106: 521–6

    PubMed  CAS  Google Scholar 

  226. Sher GD, Olivieri NG. Rapid healing of leg ulcers during arginine butyrate therapy in patients with sickle cell disease and thalassemia. Blood 1994; 84: 2378–80

    PubMed  CAS  Google Scholar 

  227. Sher GD, Ginder GD, Little J, et al. Extended therapy with intravenous arginine butyrate in patients with b-hemoglobinopathies. N Engl J Med 1995; 332: 1606–10

    PubMed  CAS  Google Scholar 

  228. Sutton M, Weinberg R, Padilla M, et al. Development of pulmonary hypertension in sickle cell disease in spite of a response to hydroxyurea. In: Ohene-Frempong K, editor. 24th Annual Meeting of the National Sickle Cell Disease Program; 2000 Apr; Philadelphia (PA), 2000: 87a.

  229. Gladwin M, Shelhamer J, Ognibene F, et al. Nitric oxide donor properties of hydroxyurea in patients with sickle cell disease. Br J Haematol 2002; 116: 436–44

    PubMed  CAS  Google Scholar 

  230. Nahavandi M, Wyche MO, Perlin E, et al. Nitric oxide metabolites in sickle cell anemia patients after oral administration of hydroxyurea. Hematology 2000; 5: 235–9

    Google Scholar 

  231. Glover R, Ivy E, Orringer E, et al. Detection of nitrosyl hemoglobin in venous blood in the treatment of sickle cell anemia with hydroxyurea. Mol Pharmacol 1999; 55: 1006–10

    PubMed  CAS  Google Scholar 

  232. Kim-Shapiro DB, King SB, Bonifant CL, et al. Time resolved absorption study of the reaction of hydroxyurea with sickle cell hemoglobin. Biochim Biophys Acta 1998; 1380(1): 64–74

    PubMed  CAS  Google Scholar 

  233. Romero J, Suzuka S, Nagel R, et al. Arginine supplementation of sickle transgenic mice reduces red cell density and Gardos channel activity. Blood 2002; 99: 1103–8

    PubMed  CAS  Google Scholar 

  234. Gladwin M, Schechter A. Nitric oxide therapy in sickle cell disease. Semin Hematol 2001; 38: 333–42

    PubMed  CAS  Google Scholar 

  235. Weiner DL, Hibberd PL, Betit P, et al. Preliminary assessment of inhaled nitric oxide for acute vaso-occlusive crisis in pediatric patients with sickle cell disease. JAMA 2003; 289: 1136–42

    PubMed  CAS  Google Scholar 

  236. Oppert M, Jorres A, Barckow D, et al. Inhaled nitric oxide for ARDS due to sickle cell disease. Swiss Med Wkly 2004; 134: 165–7

    PubMed  CAS  Google Scholar 

  237. de 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: 1087–96

    PubMed  Google Scholar 

  238. Roberts J, Fineman J, Morin F, et al. Inhaled nitric oxide and persistent pulmonary hypertension of the newborn. N Engl J Med 1997; 336: 605–10

    PubMed  CAS  Google Scholar 

  239. Pepke-Zaba J, Higenbottam T, Dinh-Xuan A, et al. Inhaled nitric oxide as a cause of selective pulmonary vasodilation in pulmonary hypertension. Lancet 1991; 338: 1173–4

    PubMed  CAS  Google Scholar 

  240. Sasaki S, Asano M, Ukai T, et al. Nitric oxide formation and plasma L-arginine levels in pulmonary hypertensive rats. Respir Med 2004; 98: 205–12

    PubMed  Google Scholar 

  241. Vosatka R, Kashyap S, Trifiletti R. Arginine deficiency accompanies persistent pulmonary hypertension of the newborn. Biol Neonate 1994; 66: 65–70

    PubMed  CAS  Google Scholar 

  242. Pearson D, Dawling S, Walsh W, et al. Neonatal pulmonary hypertension: ureacycle intermediates, nitric oxide production, and carbamoyl-phosphate synthetase function. N Engl J Med 2001; 344: 1832–8

    PubMed  CAS  Google Scholar 

  243. Schnader J. Top ten list in pulmonary vascular disease. Chest 2005; 127(2): 652–4

    PubMed  Google Scholar 

  244. Stuehr DJ, Kwon N, Nathan CF, et al. N-Hydroxyl-L-arginine is an intermediate in the biosynthesis of nitric oxide for L-arginine. J Biol Chem 1991; 266: 6259–63

    PubMed  CAS  Google Scholar 

  245. Boucher JL, Moali C, Tenu JP. Nitric oxide biosynthesis, nitric oxide synthase inhibitors, and arginase competition for L-arginine utilization. Cell Mol Life Sci 1999; 55: 1015–28

    PubMed  CAS  Google Scholar 

  246. Humbert M, Sitbon O, Simonneau G. Treatment of pulmonary arterial hypertension. N Engl J Med 2004; 351(14): 1425–36

    PubMed  CAS  Google Scholar 

Download references

Acknowledgments

Supported in part by National Institutes of Health grants HL-04386-05 and MO1-RR01271, Pediatric Clinical Research Center.

The author wishes to acknowledge the contributions of Drs Sidney Morris, Jr and Mark Gladwin for helpful discussions and Dr Sidney Morris, Jr for critical review of the manuscript.

The author has no conflicts of interest directly relevant to the content of this review.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Claudia R. Morris.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Morris, C.R. New Strategies for the Treatment of Pulmonary Hypertension in Sickle Cell Disease. Treat Respir Med 5, 31–45 (2006). https://doi.org/10.2165/00151829-200605010-00003

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00151829-200605010-00003

Keywords

Navigation