Comprehensive Physiology Wiley Online Library

Sickle Cell Disease

Full Article on Wiley Online Library



Abstract

The sections in this article are:

1 Introduction
2 Erythrocytic Factors
2.1 HbS polymerization and microvascular transit times
2.2 Generation of dense sickle red cells
3 Microvascular Factors
4 Hemodynamic Behavior of Sickle red Cells
4.1 Ex vivo studies
4.2 In vivo studies in transgenic sickle mice
4.3 Intravital studies in sickle patients
5 Sickle Red Cell Interaction with Vascular Endothelium
5.1 Sites and characteristics of sickle red cell adhesion in the microcirculation
5.2 Adhesion molecules
6 Inflammation
7 The Role of Red Cell and Leukocyte Adhesion in Vaso‐Occlusion
8 Vascular Tone
8.1 Red cell rheological factors in vascular reactivity
8.2 NO bioavailability
8.3 Peroxynitrite, tetrahydrobiopterin, and eNOS
8.4 Nitrite pool
8.5 Non‐NO vasodilators
9 Anti‐Sickling Therapeutic Strategies
9.1 Fetal hemoglobin
9.2 Experimental modulation of red cell density
9.3 Gene therapy
9.4 Conclusions and future directions
Figure 1. Figure 1.

In hemoglobin S, single nucleotide substitution (GTG for GAG) in the 6th codon of the β‐globin gene results in the replacement of a glutamic acid residue by a valine residue. Deoxygenation results in polymerization of hemoglobin S (HbS) and shape changes (sickling) of HbS‐polymer‐containing red cells. Figure modified from reference 15, by permission of M.H. Steinberg. (See page 20 in colour section at the back of the book)

Figure 2. Figure 2.

Among red cell subpopulations, discocytes form the largest component of sickle blood and transform to typical sickled forms upon deoxygenation. On the other hand, dense sickle red cells (e.g. ISCs) show minimal transformation with deoxygenation. Figure modified from reference 20.

Figure 3. Figure 3.

(A–C) Arteriovenous Vrbc, wall shear rates and volumetric flow rate (Q) profiles in the arteriolar and venular branching orders (A2–A4 and V4–V2, respectively) in the resting cremaster muscle microcirculation of C57BL, BERK, BERK + γ mice 54. Microvascular blood flow in the BERK mice is characterized by a pronounced decline in arteriolar wall shear rates, and by a greater Q in A2 and V2 vessels. Note the normalization of wall shear rates and Q in BERK + γ (mice to control values. *P < 0.05 vs. C57BL and BERK mice (Kruskal‐Wallis test for ANOVA).

Reproduced with permission from Ref. 54
Figure 4. Figure 4.

Adhesion of sickle red cells in venules of the ex vivo mesocecum vasculature infused with a bolus of human sickle red cells during perfusion with Ringer‐albumin. (A) Adherent sickle red cells of discocyte morphology are seen deformed in the direction of the flow (arrow). (B) Increased adhesion of sickle red cells at venular bending and at junctions of small‐diameter immediate postcapillary venules. In this instance, the immediate postcapillary venules are completely blocked (arrows). (C) The inverse relationship between vessel diameter and sickle redcells adhesion in venules of the ex vivo mesocecum vasculature. The regression fits the equation y = aX−b, r = −0.81, p = < 0.001). Figure modified from reference 35.

Figure 5. Figure 5.

Ex vivo mesoceum microvasculature. (A) A clear vessel lumen (a, arteriole; v, venule is seen during perfusion with Ringer‐albumin solution. (B) The same area under epifluorescence illumination after a bolus infusion of SS1 reticulocytes (fluorescein isothiocyanate (FITC)‐labeled) and SS2 discocytes mixed in 1:1 ratio depicts preferential adhesion of SS1 red cells (small arrows) in the venule during flow (Large arrows indicates the flow direction). (C) Adherent SS1 and SS2 red cells in a venule. (D) The same area under epifluorescence illumination shows that the majority of adherent red cells are from SS1 fraction. Modified from Ref. 35.

Figure 6. Figure 6.

Selective trapping of dense SS4 red cells in postcapillary venules after a bolus infusion of a mixture of SS2 (discocytes) and FITC‐labeled SS4 cells (ISCs and dense discocytes) (3:1 ratio). (A) Areas of venular obstruction. Arrows indicate unobstructed areas showing only adherent sickle redcells. (B) The same area under epifluorescence illumination showing localization of FITC‐labeled dense SS4 red cells trapped in the obstructed venules and their absence in the areas with adhesion (arrows). Modified from Ref. 35.

Figure 7. Figure 7.

Videomicrographs showing in vivo adhesion of red cells in cremasteric venules of transgenic (S + S‐Antilles) sickle mice. (A) Adherent red cells (small arrows) in the venular flow (large arrow). (B) A rolling leukocyte (L) is distinguished from adherent red cells (small arrows) by its larger diameter. (C) A sickled cell with distinct spicules (small arrow) in the venular flow (large arrow). Bar in each case = 10 μm.

Reproduced with permission from Ref. 53
Figure 8. Figure 8.

Schematic representation of adhesion molecules involved in SS cell (RBC)‐endothelium interactions. EC matrix = extracellular matrix; IAP = integrin‐associated protein; ICAM‐4 = intercellular adhesion molecule‐4; Sulf. glycolipids = sulfated glycolipids. (See page 21 in colour section at the back of the book)

Figure 9. Figure 9.

A model for vaso‐occlusion in sickle cell disease. (A) Adhesion of deformable sickle red cells (arrows) and leukocytes (light colored) in postcapillary venules. (B) Adhesion of these cells is followed by reduction in local wall shear rates and selective trapping of dense sickle red cells, which could result in HbS polymerization in the trapped (dark red) and adhered sickle cells, and obstruction of the affected vessels. (See page 21 in colour section at the back of the book)

Figure 10. Figure 10.

Immunoperoxidase staining for eNOS in the cremaster muscle microvasculature of C57BL, BERK and BERK + γ mice. (A) Negative control. (B) The same vessels in an adjacent section from control C57BL cremaster muscle showing positive reaction for eNOS in the vessel wall (arrow heads). (C) BERK mice show a strongly positive reaction in the vessel wall (arrow heads), (d) In contrast to BERK mice, BERK + γ (mice show a distinct decrease in the intensity of staining for eNOS in vessels (arrow heads).

Reproduced with permission from 54. (See page 21 in colour section at the back of the book)
Figure 11. Figure 11.

Arteriolar diameter (% increase) responses to topical application of Ach (10−6 M) and SNP (10−6 M) in C57BL. BERK‐trait. BERK and BERK + γ mice. Note the attenuated response of arterioles in BERK mice to Ach (A) and SNP (B). Ach and SNP caused significant increases in arteriolar diameters of BERK + γ (mice as compared with that in BERK mice (∼33 and ∼50% increases, respectively). *p < 0.005–0.000001 compared with C57BL and BERK‐trait mice. + p < 0.00–0.002 compared with the diameter increase in BERK mice.

Reproduced with permission from 54
Figure 12. Figure 12.

Plasma heme and its effect on NO consumption and microvascular response to SNP. (A) Arterial plasma in human patients with sickle cell disease contains an average of 4.2 + 1.1 μmol heme compared with 0.2 + 0.1 μmol heme in the plasma of normal human volunteers. (B) Heme concentration within plasma of sickle cell patients shows a significant correlation with NO consumption (orange circles; line represents the best fit, r = 0.9, P < 0.0001). (C) Relationship between plasma heme levels and the arteriolar diameter response to SNP in the cremaster microcirculation of C57BL (control), BERK, BERK‐trait and BERK + y mice. A strong correlation is observed between the percent arteriolar diameter increase in response to SNP and the extent of hemolysis (plasma heme). With a greater hemolysis in sickle (BERK) mice the diameter response was blunted. Low plasma heme levels in controls were associated with maximal arteriolar dilation, while BERK mice expressing 20% fetal hemoglobin showed a lower plasma heme and an improved diameter response compared with BERK mice.

Figures A and B are reproduced from reference 120 by permission. Figure C is based on the published data in reference 54. (See page 22 in colour section at the back of the book)
Figure 13. Figure 13.

Elevated nitrotyrosine levels and eNOS monomerization in sickle (BERK) mice. (A) Western blot analysis of cremaster muscle lysates for the expression of nitrotyrosine. Two prominent bands of nitrated proteins (66 and 26 kDa) were detected by the antibody to nitrotyrosine. BERK mice showed increased tyrosine nitration of both 66 and 26 kDa proteins (i.e. average increase: 5‐fold and ∼2‐fold respectively), while the BERK + γ mouse showed a smaller increase as compared with C57BL controls. The nitrotyrosine levels in BERK‐trait and β – thal mice showed no increase as compared with C57BL controls. Control lane depicts positive nitrotyrosine controls provided by the antibody manufacturer. Equal loading of the samples was ascertained using anti‐actin antibody. (B) Western blots of lung homogenates under nondenaturing conditions demonstrate 280 kDa dimer (active form) and 140 kDa eNOS monomer. Wild‐type (WT) and hemizygous (Hemi) sickle mice had more eNOS dimer than monomer, but sickle mice showed almost a complete lack of dimerized eNOS. Positive monomer controls show eNOS dissociated completely to monomeric form by boiling. (C) Lung nitrotyrosine, evidence of NO scavenging by superoxide, was elevated in sickle mice.

Figure A is reproduced with permission from reference 54. Figures B and C are reproduced from reference 117, by permission
Figure 14. Figure 14.

Immunoperoxidase staining for COX‐2 in the cremaster muscle microvasculature of C57BL, BERK and BERK + γ mice. (A) Negative control. (B) The same vessels in an adjacent section from the control C57BL cremaster muscle show negative to weakly positive reaction (arrow heads). (C) and (D) Strongly positive reaction for COX‐2 in vascular endothelium of blood vessels in BERK mice (arrow heads). (e) BERK (γ mice show negative or weakly positive reaction for COX‐2 in vessel walls (arrow heads).

Reproduced with permission from 54. (See page 22 in colour section at the back of the book)


Figure 1.

In hemoglobin S, single nucleotide substitution (GTG for GAG) in the 6th codon of the β‐globin gene results in the replacement of a glutamic acid residue by a valine residue. Deoxygenation results in polymerization of hemoglobin S (HbS) and shape changes (sickling) of HbS‐polymer‐containing red cells. Figure modified from reference 15, by permission of M.H. Steinberg. (See page 20 in colour section at the back of the book)



Figure 2.

Among red cell subpopulations, discocytes form the largest component of sickle blood and transform to typical sickled forms upon deoxygenation. On the other hand, dense sickle red cells (e.g. ISCs) show minimal transformation with deoxygenation. Figure modified from reference 20.



Figure 3.

(A–C) Arteriovenous Vrbc, wall shear rates and volumetric flow rate (Q) profiles in the arteriolar and venular branching orders (A2–A4 and V4–V2, respectively) in the resting cremaster muscle microcirculation of C57BL, BERK, BERK + γ mice 54. Microvascular blood flow in the BERK mice is characterized by a pronounced decline in arteriolar wall shear rates, and by a greater Q in A2 and V2 vessels. Note the normalization of wall shear rates and Q in BERK + γ (mice to control values. *P < 0.05 vs. C57BL and BERK mice (Kruskal‐Wallis test for ANOVA).

Reproduced with permission from Ref. 54


Figure 4.

Adhesion of sickle red cells in venules of the ex vivo mesocecum vasculature infused with a bolus of human sickle red cells during perfusion with Ringer‐albumin. (A) Adherent sickle red cells of discocyte morphology are seen deformed in the direction of the flow (arrow). (B) Increased adhesion of sickle red cells at venular bending and at junctions of small‐diameter immediate postcapillary venules. In this instance, the immediate postcapillary venules are completely blocked (arrows). (C) The inverse relationship between vessel diameter and sickle redcells adhesion in venules of the ex vivo mesocecum vasculature. The regression fits the equation y = aX−b, r = −0.81, p = < 0.001). Figure modified from reference 35.



Figure 5.

Ex vivo mesoceum microvasculature. (A) A clear vessel lumen (a, arteriole; v, venule is seen during perfusion with Ringer‐albumin solution. (B) The same area under epifluorescence illumination after a bolus infusion of SS1 reticulocytes (fluorescein isothiocyanate (FITC)‐labeled) and SS2 discocytes mixed in 1:1 ratio depicts preferential adhesion of SS1 red cells (small arrows) in the venule during flow (Large arrows indicates the flow direction). (C) Adherent SS1 and SS2 red cells in a venule. (D) The same area under epifluorescence illumination shows that the majority of adherent red cells are from SS1 fraction. Modified from Ref. 35.



Figure 6.

Selective trapping of dense SS4 red cells in postcapillary venules after a bolus infusion of a mixture of SS2 (discocytes) and FITC‐labeled SS4 cells (ISCs and dense discocytes) (3:1 ratio). (A) Areas of venular obstruction. Arrows indicate unobstructed areas showing only adherent sickle redcells. (B) The same area under epifluorescence illumination showing localization of FITC‐labeled dense SS4 red cells trapped in the obstructed venules and their absence in the areas with adhesion (arrows). Modified from Ref. 35.



Figure 7.

Videomicrographs showing in vivo adhesion of red cells in cremasteric venules of transgenic (S + S‐Antilles) sickle mice. (A) Adherent red cells (small arrows) in the venular flow (large arrow). (B) A rolling leukocyte (L) is distinguished from adherent red cells (small arrows) by its larger diameter. (C) A sickled cell with distinct spicules (small arrow) in the venular flow (large arrow). Bar in each case = 10 μm.

Reproduced with permission from Ref. 53


Figure 8.

Schematic representation of adhesion molecules involved in SS cell (RBC)‐endothelium interactions. EC matrix = extracellular matrix; IAP = integrin‐associated protein; ICAM‐4 = intercellular adhesion molecule‐4; Sulf. glycolipids = sulfated glycolipids. (See page 21 in colour section at the back of the book)



Figure 9.

A model for vaso‐occlusion in sickle cell disease. (A) Adhesion of deformable sickle red cells (arrows) and leukocytes (light colored) in postcapillary venules. (B) Adhesion of these cells is followed by reduction in local wall shear rates and selective trapping of dense sickle red cells, which could result in HbS polymerization in the trapped (dark red) and adhered sickle cells, and obstruction of the affected vessels. (See page 21 in colour section at the back of the book)



Figure 10.

Immunoperoxidase staining for eNOS in the cremaster muscle microvasculature of C57BL, BERK and BERK + γ mice. (A) Negative control. (B) The same vessels in an adjacent section from control C57BL cremaster muscle showing positive reaction for eNOS in the vessel wall (arrow heads). (C) BERK mice show a strongly positive reaction in the vessel wall (arrow heads), (d) In contrast to BERK mice, BERK + γ (mice show a distinct decrease in the intensity of staining for eNOS in vessels (arrow heads).

Reproduced with permission from 54. (See page 21 in colour section at the back of the book)


Figure 11.

Arteriolar diameter (% increase) responses to topical application of Ach (10−6 M) and SNP (10−6 M) in C57BL. BERK‐trait. BERK and BERK + γ mice. Note the attenuated response of arterioles in BERK mice to Ach (A) and SNP (B). Ach and SNP caused significant increases in arteriolar diameters of BERK + γ (mice as compared with that in BERK mice (∼33 and ∼50% increases, respectively). *p < 0.005–0.000001 compared with C57BL and BERK‐trait mice. + p < 0.00–0.002 compared with the diameter increase in BERK mice.

Reproduced with permission from 54


Figure 12.

Plasma heme and its effect on NO consumption and microvascular response to SNP. (A) Arterial plasma in human patients with sickle cell disease contains an average of 4.2 + 1.1 μmol heme compared with 0.2 + 0.1 μmol heme in the plasma of normal human volunteers. (B) Heme concentration within plasma of sickle cell patients shows a significant correlation with NO consumption (orange circles; line represents the best fit, r = 0.9, P < 0.0001). (C) Relationship between plasma heme levels and the arteriolar diameter response to SNP in the cremaster microcirculation of C57BL (control), BERK, BERK‐trait and BERK + y mice. A strong correlation is observed between the percent arteriolar diameter increase in response to SNP and the extent of hemolysis (plasma heme). With a greater hemolysis in sickle (BERK) mice the diameter response was blunted. Low plasma heme levels in controls were associated with maximal arteriolar dilation, while BERK mice expressing 20% fetal hemoglobin showed a lower plasma heme and an improved diameter response compared with BERK mice.

Figures A and B are reproduced from reference 120 by permission. Figure C is based on the published data in reference 54. (See page 22 in colour section at the back of the book)


Figure 13.

Elevated nitrotyrosine levels and eNOS monomerization in sickle (BERK) mice. (A) Western blot analysis of cremaster muscle lysates for the expression of nitrotyrosine. Two prominent bands of nitrated proteins (66 and 26 kDa) were detected by the antibody to nitrotyrosine. BERK mice showed increased tyrosine nitration of both 66 and 26 kDa proteins (i.e. average increase: 5‐fold and ∼2‐fold respectively), while the BERK + γ mouse showed a smaller increase as compared with C57BL controls. The nitrotyrosine levels in BERK‐trait and β – thal mice showed no increase as compared with C57BL controls. Control lane depicts positive nitrotyrosine controls provided by the antibody manufacturer. Equal loading of the samples was ascertained using anti‐actin antibody. (B) Western blots of lung homogenates under nondenaturing conditions demonstrate 280 kDa dimer (active form) and 140 kDa eNOS monomer. Wild‐type (WT) and hemizygous (Hemi) sickle mice had more eNOS dimer than monomer, but sickle mice showed almost a complete lack of dimerized eNOS. Positive monomer controls show eNOS dissociated completely to monomeric form by boiling. (C) Lung nitrotyrosine, evidence of NO scavenging by superoxide, was elevated in sickle mice.

Figure A is reproduced with permission from reference 54. Figures B and C are reproduced from reference 117, by permission


Figure 14.

Immunoperoxidase staining for COX‐2 in the cremaster muscle microvasculature of C57BL, BERK and BERK + γ mice. (A) Negative control. (B) The same vessels in an adjacent section from the control C57BL cremaster muscle show negative to weakly positive reaction (arrow heads). (C) and (D) Strongly positive reaction for COX‐2 in vascular endothelium of blood vessels in BERK mice (arrow heads). (e) BERK (γ mice show negative or weakly positive reaction for COX‐2 in vessel walls (arrow heads).

Reproduced with permission from 54. (See page 22 in colour section at the back of the book)
References
 1. Hebbel RP, Boogaerts MA, Eaton JW and Steinberg MH. Erythrocyte adherence to endothelium in sickle‐cell anemia. A possible determinant of disease severity. New Eng J Med 302: 992–995, 1980.
 2. Hebbel RP, Osarogiagbon R and Kaul D. The endothelial biology of sickle cell disease: inflammation and a chronic vasculopathy. Microcirculation 11: 129–151, 2004.
 3. Kaul DK and Fabry ME. In vivo studies of sickle red blood cells. Microcirculation 11: 153–165, 2004.
 4. Kaul DK and Hebbel RP. Hypoxia/reoxygenation causes inflammatory response in transgenic sickle mice but not in normal mice [see comments]. J Clin Invest 106: 411–420, 2000.
 5. Nath KA, Katusic ZS and Gladwin MT. The perfusion paradox and vascular instability in sickle cell disease. Microcirculation 11: 179–193, 2004.
 6. Nagel RL and Fabry ME. The many pathophysiologies of sickle cell anemia [Review]. Am J Hematol 20: 195–199, 1985.
 7. Powars DR. Natural history of sickle cell disease‐the first ten years. Semin Hematol 12: 267–285, 1975.
 8. Nagel RL. Pleiotropic and epistatic effects in sickle cell anemia. Curr Opin Hematol 8: 105–110, 2001.
 9. Nagel RL, Fabry ME, Kaul DK, Billett H, Croizat H. Labie D and Canessa S. Known and potential sources for epistatic effects in sickle cell anemia [62] Ann NY Acad Sci 565: 228–238, 1989.
 10. Nagel RL and Steinberg MH. Role of epistatic (modifier) genes in the modulation of the phenotypic diversity of sickle cell anemia. Pediatr Pathol Mol Med 20: 123–136, 2001.
 11. Lu ZH and Steinberg MH. Fetal hemoglobin in sickle cell anemia: relation to regulatory sequences cis to the beta‐globin gene. Multicenter Study of Hydroxyurea. Blood 87: 1604–1611, 1996.
 12. Stuart MJ and Nagel RL. Sickle‐cell disease. Lancet 364: 1343–1360, 2004.
 13. Bunn HF. Pathogenesis and treatment of sickle cell disease. N Engl J Med 337: 762–769, 1997.
 14. Pauling L and Itano HA. Sickle cell anemia a molecular disease. Science 110: 543–548, 1949.
 15. Steinberg MH. Management of sickle cell disease. N Engl J Med 340: 1021–1030, 1999.
 16. Eaton WA and Hofrichter J. Hemoglobin S gelation and sickle cell disease [Review]. Blood 70: 1245–1266, 1987.
 17. Ferrone FA. Polymerization and sickle cell disease: a molecular view. Microcirculation 11: 115–128, 2004.
 18. Kaul DK, Fabry ME and Nagel RL. The pathophysiology of vascular obstruction in the sickle syndromes [Review]. Blood Rev 10: 29–44, 1996.
 19. Fabry ME and Nagel RL. Heterogeneity of red cells in the sickler: a characteristic with practical clinical and pathophysiological implications. Blood Cells 8: 9–15, 1982.
 20. Kaul DK, Fabry ME, Windisch P, Baez S and Nagel RL. Erythrocytes in sickle cell anemia are heterogeneous in their rheological and hemodynamic characteristics. J Clin Invest 72: 22–31, 1983.
 21. Bridges KR, Barabino GD, Brugnara C, Cho MR, Christoph GW, Dover G, Ewenstein BM, Golan DE, Guttmann CR, Hofrichter J, Mulkern RV, Eaton S and Zhang WA. A multiparameter analysis of sickle erythrocytes in patients undergoing hydroxyurea therapy. Blood 88: 4701–4710, 1996.
 22. Brugnara C, Bunn HF and Tosteson DC. Regulation of erythrocyte cation and water content in sickle cell anemia. Science 232: 388–390, 1986.
 23. Joiner CH. Cation transport and volume regulation in sickle red blood cells. Am J Physiol 264: C251–C270, 1993.
 24. Lux SE, John KM and Karnovsky MJ. Irreversible deformation of the spectrin‐actin lattice in irreversibly sickled cells. J Clin Invest 58: 955–963, 1976.
 25. Rodgers GP, Schechter AN, Noguchi CT, Klein HG, Nienhuis AW and Bonner RF. Periodic microcirculatory flow in patients with sickle‐cell disease. New Engl J Med 311: 1534–1538, 1984.
 26. Ahluwalia A, Foster P, Scotland RS, McLean PG, Mathur A, Perretti M, Moncada S and Hobbs AJ. Antiinflammatory activity of soluble guanylate cyclase: cGMP‐dependent down‐regulation of P‐selectin expression and leukocyte recruitment. Proc Natl Acad Sci USA 101: 1386–1391, 2004.
 27. Alp NJ and Channon KM. Regulation of endothelial nitric oxide synthase by tetrahydrobiopterin in vascular disease. Arterioscler Thromb Vasc Biol 24: 413–420, 2004.
 28. Aslan M, Ryan TM, Adler B, Townes TM, Parks DA, Thompson JA, Tousson A, Gladwin MT, Patel RP, Tarpey MM. Batinic‐Haberle I. White CR and Freeman BA. Oxygen radical inhibition of nitric oxide‐dependent vascular function in sickle cell disease. Proc Natl Acad Sci USA 98: 15215–15220, 2001.
 29. Mohandas N, Phillips WM and Bessis M. Red blood cell deformability and hemolytic anemias. Semin Hematol 16: 95–114, 1979.
 30. Chien S. Filterability and other methods of approaching red cell deformability. Determinants of blood viscosity and red cell deformability. Scand J Clin Lab Invest Suppl 156: 7–12, 1981.
 31. Chien S, Kaperonis AA, King RG, Lipowsky HH, Schmalzer EA, Sung LA, Sung KL and Usami S. Rheology of sickle cells and its role in microcirculatory dynamics. Prog Clin Biol Res 240: 151–165, 1987.
 32. Lipowsky HH, Sheikh NU and Katz DM. Intravital microscopy of capillary hemodynamics in sickle cell disease. J Clin Invest 80: 117–127, 1987.
 33. Lipowsky HH and Williams ME. Shear rate dependency of red cell sequestration in skin capillaries in sickle cell disease and its variation with vasoocclusive crisis. Microcirculation 4: 289–301, 1997.
 34. Baez S, Lamport H and Baez A. Pressure effects in living microscopic vessels. In: Flow Properties of Blood and Other Bilogical Systems, eds Copley AL and Stainsby G. Pergamon Press: London, 1960, pp. 122–136.
 35. Kaul DK, Fabry ME and Nagel RL. Microvascular sites and characteristics of sickle cell adhesion to vascular endothelium in shear flow conditions: pathophysiological implications. Proc Natl Acad Sci USA 86: 3356–3360, 1989.
 36. Green HD, Rapela C and Conard MD. Resistance (conductance) and capacitance phenomena in terminal vascular beds. In: Handbook of Physiology, eds Hamilton WF and Dow P. American Physiological Society: Washington, DC, 1963, pp. 122–136.
 37. Whittaker SRF and Winton FR. The apparent viscosity of blood flowing in the isolated hind limb of the dog and its variation with corpuscular concentration. J Physiol (Lond) 78: 339–451, 1933.
 38. Kaul DK, Nagel RL and Baez S. Pressure effects on the flow behavior of sickle (HbSS) red cells in isolated (ex‐vivo) microvascular system. Microvasc Res 26: 170–181, 1983.
 39. Baez S, Kaul DK and Nagel RL. Microvascular determinants of blood flow behavior and HbSS erythrocyte plugging in microcirculation. Blood Cell 8: 127–137, 1982.
 40. Nash GB, Johnson CS and Meiselman HJ. Influence of oxygen tension on the viscoelastic behavior of red blood cells in sickle cell disease. Blood 67: 110–118, 1986.
 41. Fabry ME, Costantini F, Pachnis A, Suzuka SM, Bank N, Aynedjian HS, Factor SM and Nagel RL. High expression of human beta S‐ and alpha‐globins in transgenic mice: erythrocyte abnormalities, organ damage, and the effect of hypoxia. Proc Natl Acad Sci USA 89: 12155–12159, 1992.
 42. Greaves DR, Fraser P, Vidal MA, Hedges MJ, Ropers D, Luzzatto L and Grosveld F. A transgenic mouse model of sickle cell disorder [see comments]. Nature 343: 183–185, 1990.
 43. Rubin EM, Witkowska HE, Spangler E, Curtin P, Lubin BH, Mohandas N and Clift SM. Hypoxia‐induced in vivo sickling of transgenic mouse red cells. J Clin Invest 87: 639–647, 1991.
 44. Ryan TM, Ciavatta DJ and Townes TM. Knockout‐transgenic mouse model of sickle cell disease. Science 278: 873–876, 1997.
 45. Trudel M, Saadane N, Garel MC, Bardakdjian‐Michau J, Blouquit Y, Guerquin‐Kern JL, Rouyer‐Fessard P, Vidaud D, Pachnis A, Romeo PH, et al. Towards a transgenic mouse model of sickle cell disease: hemoglobin SAD. EMBO J 10: 3157–3165, 1991.
 46. Fabry ME, Sengupta A, Suzuka SM, Costantini F, Rubin EM, Hofrichter S, Christoph G, Manci E, Culberson D and Factor SM. A second generation transgenic mouse model expressing both hemoglobin S (HbS) and HbS‐Antilles results in increased phenotypic severity. Blood 86: 2419–2428, 1995.
 47. Rhoda MD, Domenget C, Vidaud D, Bardakdjian‐Michau J, Rouyer‐Fessard P, Rosa J and Beuzard Y. Mouse alpha chains inhibit polymerization of hemoglobin induced by human beta S and beta S‐Antilles chains. Biochimica et Biophysica Acta 952: 208, 1992.
 48. Paszty C, Brion CM, Manci E, Witkowska HE, Stevens ME. Mohandas N and Rubin EM. Transgenic knockout mice with exclusively human sickle hemoglobin and sickle cell disease [see comments]. Science 278: 876–878, 1997.
 49. Fabry ME, Suzuka SM, Weinberg RS, Lawrence C, Factor SM, Gilman JG, Costantini F and Nagel RL. Second generation knockout sickle mice: the effect of HbF. Blood 97: 410–418, 2001.
 50. Fabry M. Transgenic animal models. In: Sickle Cell Disease: Basic Principles and Clinical Practice, eds Embury SH, Hebbel RP, Mohandas N and Steinberg MH. Raven Press, Ltd.: New York, 1994, pp. 105–120.
 51. Nagel RL. A knockout of a transgenic mouse‐animal models of sickle cell anemia. New Eng J Med 339: 194–195, 1998.
 52. Baez S. An open cremaster muscle preparation for the study of blood vessels by in vivo microscopy. Microvasc Res 5: 384–394, 1973.
 53. Kaul DK, Fabry ME, Costantini F, Rubin EM and 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 96: 2845–2853, 1995.
 54. Kaul DK, Liu XD, Chang HY, Nagel RL and Fabry ME. Effect of fetal hemoglobin on microvascular regulation in sickle transgenic‐knockout mice. J Clin Invest 114: 1136–1145, 2004.
 55. Kaul DK, Liu XD, Zhang X, Ma L, Hsia CJ and Nagel RL. Inhibition of sickle red cell adhesion and vasoocclusion in the microcirculation by antioxidants. Am J Physiol Heart Circ Physiol 291: H167–hH175, 2006.
 56. Finnegan EM, Barabino GA, Liu XD, Chang HY, Jonczyk A and Kaul DK. Small‐molecule cyclic alpha V beta 3 antagonists inhibit sickle red cell adhesion to vascular endothelium and vasoocclusion. Am J Physiol Heart Circ Physiol 293: H1038–H1045, 2007.
 57. Barabino GA, McIntire LV, Eskin SG, Sears DA and Udden M. Endothelial cell interactions with sickle cell, sickle trait, mechanically injured, and normal erythrocytes under controlled flow. Blood 70: 152–157, 1987.
 58. Mohandas N and Evans E. Sickle erythrocyte adherence to vascular endothelium. Morphologic correlates and the requirement for divalent cations and collagen‐binding plasma proteins. J Clin Invest 76: 1605–1612, 1985.
 59. Hebbel RP, Yamada O, Moldow CF, Jacob HS, White JG and Eaton JW. Abnormal adherence of sickle erythrocytes to cultured vascular endothelium: possible mechanism for microvascular occlusion in sickle cell disease. J Clin Invest 65: 154–160, 1980.
 60. Hoover R, Rubin R, Wise G and Warren R. Adhesion of normal and sickle erythrocytes to endothelial monolayer cultures. Blood 54: 872–876, 1979.
 61. Hebbel RP. Adhesive interactions of sickle erythrocytes with endothelium [Review]. J Clin Invest 100: S83–S86, 1997.
 62. Hebbel RP, Berger EM and Eaton JW. Effect of increased maternal hemoglobin oxygen affinity on fetal growth in the rat. Blood 55: 969–974, 1980.
 63. Kaul DK, Nagel RL, Chen D and Tsai HM. Sickle erythrocyte‐endothelial interactions in microcirculation: the role of von Willebrand factor and implications for vasoocclusion. Blood 81: 2429–2438, 1993.
 64. Kaul DK, Tsai HM, Liu XD, Nakada MT, Nagel RL and Coller BS. Monoclonal antibodies to alpha Vbeta3 (7E 609) inhibit sickle red blood cell‐endothelium interactions induced by platelet‐activating factor [see comments]. Blood 95: 368–374, 2000.
 65. Kaul DK, Tsai HM, Nagel RL and Chen D. Platelet‐activating factor enhances adhesion of sickle erythrocytes to vascular endothelium. In: Sickle Cell Disease and Thalassemias: New Trends in Therapy (INSERM Symposium), eds Beuzard Y, Lubin BH and Rosa J. INSERM/John Libbey Eurotext: Montrouge, France, 1995, pp. 497–500.
 66. Hatch FE, Crowe LR, Miles DE, Young JP and Portner ME. Altered vascular reactivity in sickle hemoglobinopathy. A possible protective factor from hypertension. Am J Hyperten 2: 2–8, 1989.
 67. Hebbel RP, Ney PA and Foker W. Autoxidation, dehydration, and adhesivity may be related abnormalities of sickle erythrocytes. Am J Physiol 256: C579–C583, 1989.
 68. Kaul DK, Chen D and Zhan J. Adhesion of sickle cells to vascular endothelium is critically dependent on changes in density and shape of the cells. Blood 83: 3006–3017, 1994.
 69. Zennadi R, Hines PC, De Castro LM, Cartron JP, Parise LV and Telen MJ. Epinephrine acts through erythroid signaling pathways to activate sickle cell adhesion to endothelium via LW‐alphavbeta3 interactions. Blood 104: 3774–3781, 2004.
 70. Hillery CA, Du MC, Montgomery RR and Scott JP. Increased adhesion of erythrocytes to components of the extracellular matrix: Isolation and characterization of a red blood cell lipid that binds thrombospondin and laminin. Blood 87: 4879–4886, 1996.
 71. Aslan M, Thornley‐Brown D and Freeman BA. Reactive species in sickle cell disease. Ann NY Acad Sci 899: 375–391, 2000.
 72. Brittain HA, Eckman JR, Swerlick RA, Howard RJ and Wick TM. Thrombospondin from activated platelets promotes sickle erythrocyte adherence to human microvascular endothelium under physiologic flow: a potential role for platelet activation in sickle cell vaso‐occlusion. Blood 81: 2137–2143, 1993.
 73. Sugihara K, Sugihara T, Mohandas N and Hebbel RP. Thrombospondin mediates adherence of CD36+ sickle reticulocytes to endothelial cells. Blood 80: 2634–2642, 1992.
 74. Swerlick RA, Eckman JR, Kumar A, Jeitler M and Wick TM. Alpha 4 beta 1‐integrin expression on sickle reticulocytes: vascular cell adhesion molecule‐1‐dependent binding to endothelium. Blood 82: 1891–1899, 1993.
 75. Kumar A, Eckmam JR, Swerlick RA and Wick TM. Phorbol ester stimulation increases sickle erythrocyte adherence to endothelium: a novel pathway involving alpha 4 beta 1 integrin receptors on sickle reticulocytes and fibronectin. Blood 88: 4348–4358, 1996.
 76. Setty BN and 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 88: 2311–2320, 1996.
 77. Stuart MJ and Setty BN. Acute chest syndrome of sickle cell disease: New light on an old problem. Curr Opin Hematol 8: 111–122, 2001.
 78. Vichinsky EP, Styles LA, Colangelo LH, Wright EC, Castro O and Nickerson B. Acute chest syndrome in sickle cell disease: clinical presentation and course. Cooperative Study of Sickle Cell Disease. Blood 89: 1787–1792, 1997.
 79. Barabino GA, Liu XD, Ewenstein BM and Kaul DK. Anionic polysaccharides inhibit adhesion of sickle erythrocytes to the vascular endothelium and result in improved hemodynamic behavior. Blood 93: 1422–1429, 1999.
 80. Brittain JE, Mlinar KJ, Anderson CS, Orringer EP and Parise LV. Activation of sickle red blood cell adhesion via integrin‐associated protein/CD47‐induced signal transduction. J Clin Invest 107: 1555–1562, 2001.
 81. Hines PC, Zen Q, Burney SN, Shea DA, Ataga KI, Orringer EP, Telen MJ and Parise LV. Novel epinephrine and cyclic AMP‐mediated activation of BCAM/Lu‐dependent sickle (SS) RBC adhesion. Blood 101: 3281–3287, 2003.
 82. Mankelow TJ, Spring FA, Parsons SF, Brady RL, Mohandas N, Chasis JA and Anstee DJ. Identification of critical amino‐acid residues on the erythroid intercellular adhesion molecule‐4 (ICAM‐4) mediating adhesion to alpha V integrins. Blood, 2003.
 83. Kaul DK, Liu XD, Zhang Z, Mankelow T, Parsons S, Spring F, An X, Narla M, Anstee D and Chassis JA. Inhibiting binding of sickle red cell ICAM‐4 to endothelial cell alpha V beta 3 integrin decreases red cell adhesion and vaso‐occlusion. Blood 104 (11 Suppl.): 106a, 2004.
 84. Wick TM, Moake JL, Udden MM and McIntire LV. Unusually large von Willebrand factor multimers preferentially promote young sickle and nonsickle erythrocyte adhesion to endothelial cells. Am J Hematol 42: 284–292, 1993.
 85. Felding‐Habermann B and Cheresh DA. Vitronectin and its receptors [Review] [51 refs]. Curr Opin Cell Biol 5: 864–868, 1993.
 86. Mackie I, Bull H and Brozovic M. Altered factor VIII complexes in sickle cell disease. Br J Haematol 46: 499–502, 1980.
 87. Richardson SG, Matthews KB, Stuart J, Geddes AM and Wilcox RM. Serial changes in coagulation and viscosity during sickle‐cell crisis. Brit J Haematol 41: 95–103, 1979.
 88. Cheresh DA. Human endothelial cells synthesize and express an Arg‐Gly‐Asp‐directed adhesion receptor involved in attachment to fibrinogen and von Willebrand factor. Proc Natl Acad Scie USA 84: 6471–6475, 1987.
 89. Kramer RH, Cheng YF and Clyman R. Human microvascular endothelial cells use beta 1 and beta 3 integrin receptor complexes to attach to laminin. J Cell Biol 111: 1233–1243, 1990.
 90. Hebbel RP. Blockade of adhesion of sickle cells to endothelium by monoclonal antibodies. N Engl J Med 342: 1910–1912, 2000.
 91. Solovey A, Lin Y, Browne P, Choong S, Wayner E and Hebbel RP. Circulating activated endothelial cells in sickle cell anemia [see comments]. New Engl J Med 337: 1584–1590, 1997.
 92. Sowemimo‐Coker SO, Meiselman HJ and Francis RB, Jr. Increased circulating endothelial cells in sickle cell crisis. Am J Hematol 31: 263–265, 1989.
 93. Boggs DR, Hyde F and Srodes C. An unusual pattern of neutrophil kinetics in sickle cell anemia. Blood 41: 59–65, 1973.
 94. Platt OS. Sickle cell anemia as an inflammatory disease. J Clin Invest 106: 337–338, 2000.
 95. Osarogiagbon UR, Choong S, Belcher JD, Vercellotti GM, Paller MS and Hebbel RP. Reperfusion injury pathophysiology in sickle transgenic mice. Blood 96: 314–320, 2000.
 96. Griendling KK, Sorescu D and Ushio‐Fukai M. NAD(P)H oxidase: role in cardiovascular biology and disease. Circ Res 86: 494–501, 2000.
 97. Wood KC, Hebbel RP and Granger DN. Endothelial cell NADPH oxidase mediates the cerebral microvascular dysfunction in sickle cell transgenic mice. FASEB J 19: 989–991, 2005.
 98. Vasquez‐Vivar J, Kalyanaraman B, Martasek P, Hogg N, Masters BS, Karoui H, Tordo P and Pritchard KA, Jr. Superoxide generation by endothelial nitric oxide synthase: the influence of cofactors. Proc Natl Acad Sci USA 95: 9220–9225, 1998.
 99. Kalambur VS, Mahaseth H, Bischof JC, Kielbik MC, Welch TE, Vilback A, Swanlund DJ, Hebbel RP, Belcher JD and Vercellotti GM. Microvascular blood flow and stasis in transgenic sickle mice: utility of a dorsal skin fold chamber for intravital microscopy. Am J Hematol 77: 117–125, 2004.
 100. Kaul DK, Liu XD, Choong S, Belcher JD, Vercellotti GM and Hebbel RP. Anti‐inflammatory therapy ameliorates leukocyte adhesion and microvascular flow abnormalities in transgenic sickle mice. Am J Physiol Heart Circ Physiol 287: H293–hH301, 2004.
 101. Wood KC, Hebbel RP and 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 286: H1608–hH1614, 2004.
 102. Turhan A, Weiss LA, Mohandas N, Coller BS and Frenette PS. Primary role for adherent leukocytes in sickle cell vascular occlusion: a new paradigm. Proc Natl Acad Sci USA 99: 3047–3051, 2002.
 103. Embury SH, Matsui NM, Ramanujam S, Mayadas TN, Noguchi CT, Diwan BA, Mohandas N and Cheung AT. The contribution of endothelial cell P‐selectin to the microvascular flow of mouse sickle erythrocytes in vivo. Blood 104: 3374–3385, 2004.
 104. Graido‐Gonzalez E, Doherty JC, Bergreen EW, Organ G, Telfer M and McMillen MA. Plasma endothelin‐1. cytokine, and prostaglandin E2 levels in sickle cell disease and acute vaso‐occlusive sickle crisis. Blood 92: 2551–2555, 1998.
 105. Kaul DK, Fabry ME and Nagel RL. Erythrocytic and vascular factors influencing the microcirculatory behavior of blood in sickle cell anemia. [35] Ann NY Acad Sci 565: 316–326, 1989.
 106. Kaul DK. Flow properties and endothelial adhesion of sickle erythrocytes in an ex vivo microvascular preparation. In: Membrane Abnormalities in Sickle Cell Disease and in Other Red Blood Cell Disorders, eds Ohnishi ST and Ohnishi T. CRC Press: Boca Raton, FL, 1994, pp. 217–241.
 107. Embury SH. The not‐so‐simple process of sickle cell vasoocclusion. Microcirculation 11: 101–113, 2004.
 108. Belhassen L, Pelle G, Sediame S, Bachir D, Carville C, Bucherer C, Lacombe C, Galacteros F and Adnot S. Endothelial dysfunction in patients with sickle cell disease is related to selective impairment of shear stress‐mediated vasodilation. Blood 97: 1584–1589, 2001.
 109. Lonsdorfer J, Bogui P, Otayeck A, Bursaux E, Poyart C and Cabannes R. Cardiorespiratory adjustments in chronic sickle cell anemia. Bulletin Europeen de Physiopathologie Respiratoire 19: 339–344, 1983.
 110. Johnson CS and Giorgio AJ. Arterial blood pressure in adults with sickle cell disease. Arch Int Med 141: 891–893, 1981.
 111. Kubes P, Suzuki M and Granger DN. Nitric oxide: an endogenous modulator of leukocyte adhesion. Proc Natl Acad Sci USA 88: 4651–4655, 1991.
 112. Moncada S, Palmer RM and Higgs EA. Nitric oxide: physiology, pathophysiology, and pharmacology [404]. Pharmacol Rev 43: 109–142, 1991.
 113. Ohashi Y, Kawashima S, Hirata K, Yamashita T, Ishida T, Inoue N, Sakoda T, Kurihara H, Yazaki Y and Yokoyama M. Hypotension and reduced nitric oxide‐elicited vasorelaxation in transgenic mice overexpressing endothelial nitric oxide synthase [see comments]. J Clin Invest 102: 2061–2071, 1998.
 114. Harrison DG. Cellular and molecular mechanisms of endothelial cell dysfunction [Review]. J Clin Invest 100: 2153–2157, 1997.
 115. Morris CR, Kuypers FA, Larkin S, Vichinsky EP and Styles LA. Patterns of arginine and nitric oxide in patients with sickle cell disease with vaso‐occlusive crisis and acute chest syndrome. J Pediatr Hematol Oncol 22: 515–520, 2000.
 116. Rees DC, Cervi P, Grimwade D, O'Driscoll A, Hamilton M, Parker NE and Porter JB. The metabolites of nitric oxide in sickle‐cell disease. Br J Haematol 91: 834–837, 1995.
 117. Hsu LL, Champion HC, Campbell‐Lee SA, Bivalacqua TJ, Manci EA, Diwan BA, Schimel DM, Cochard AE, Wang X, Schechter AN, Noguchi CT and Gladwin MT. Hemolysis in sickle cell mice causes pulmonary hypertension due to global impairment in nitric oxide bioavailability. Blood 109: 3088–3098, 2007.
 118. Morris CR, Kato GJ, Poljakovic M, Wang X, Blackwelder WC, Sachdev V, Hazen SL, Vichinsky EP, Morris SM, Jr and Gladwin MT. Dysregulated arginine metabolism, hemolysis‐associated pulmonary hypertension, and mortality in sickle cell disease. JAMA 294: 81–90, 2005.
 119. Gladwin MT, Lancaster JR, Jr., Freeman BA and Schechter AN. Nitric oxide's reactions with hemoglobin: a view through the SNO‐storm. Nat Med 9: 496–500, 2003.
 120. Reiter CD, Wang X, Tanus‐Santos JE, Hogg N, Cannon RO, III, Schechter AN and Gladwin MT. Cell‐free hemoglobin limits nitric oxide bioavailability in sickle‐cell disease. Nat Med 8: 1383–1389, 2002.
 121. Gladwin MT, Schechter AN, Ognibene FP, Coles WA, Reiter CD, Schenke WH, Csako G, Waclawiw MA, Panza JA and Cannon RO, III. Divergent nitric oxide bioavailability in men and women with sickle cell disease. Circulation 107: 271–278, 2003.
 122. Kaul DK, Liu XD, Fabry ME and Nagel RL. Impaired nitric oxide‐mediated vasodilation in transgenic sickle mouse. Am J Physiol Heart Circ Physiol 278: H1799–H1806, 2000.
 123. Nath KA, Shah V, Haggard JJ, Croatt AJ, Smith LA, Hebbel RP and Katusic ZS. Mechanisms of vascular instability in a transgenic mouse model of sickle cell disease. Am J Physiol Regul Integr Comp Physiol 279: R1949–R1955, 2000.
 124. Gladwin MT and Kato GJ. Cardiopulmonary complications of sickle cell disease: role of nitric oxide and hemolytic anemia. Hematol Am Soc Hematol Educ Program: 51‐57, 2005.
 125. Eberhardt RT, McMahon L, Duffy SJ, Steinberg MH, Perrine SP, Loscalzo J, Coffman JD and Vita JA. Sickle cell anemia is associated with reduced nitric oxide bioactivity in peripheral conduit and resistance vessels. Am J Hematol 74: 104–111, 2003.
 126. Conger JD and Weil JV. Abnormal vascular function following ischemia‐reperfusion injury [Review]. J Invest Med 43: 431–442, 1995.
 127. Katusic ZS and d'Uscio LV. Tetrahydrobiopterin: mediator of endothelial protection. Arterioscler Thromb Vasc Biol 24: 397–398, 2004.
 128. Landmesser U, Dikalov S, Price SR, McCann L, Fukai T, Holland SM, Mitch WE and Harrison DG. Oxidation of tetrahydrobiopterin leads to uncoupling of endothelial cell nitric oxide synthase in hypertension. J Clin Invest 111: 1201–1209, 2003.
 129. Wood KC, Hebbel RP, Lefer DJ and Granger DN. Critical role of endothelial cell‐derived nitric oxide synthase in sickle cell disease‐induced microvascular dysfunction. Free Radic Biol Med 40: 1443–1453, 2006.
 130. Dejam A, Hunter CJ, Pelletier MM, Hsu LL, Machado RF, Shiva S, Power GG, Kelm M, Gladwin MT and Schechter AN. Erythrocytes are the major intravascular storage sites of nitrite in human blood. Blood 106: 734–739, 2005.
 131. Gladwin MT, Crawford JH and Patel RP. The biochemistry of nitric oxide, nitrite, and hemoglobin: role in blood flow regulation. Free Radic Biol Med 36: 707–717, 2004.
 132. Cosby K, Partovi KS, Crawford JH, Patel RP, Reiter CD, Martyr S, Yang BK, Waclawiw MA, Zalos G, Xu X, Huang KT, Shields H, Kim‐Shapiro DB, Schechter AN, Cannon RO and Gladwin MT. Nitrite reduction to nitric oxide by deoxyhemoglobin vasodilates the human circulation. Nat Med 9: 1498–1505, 2003.
 133. Huang KT, Keszler A, Patel N, Patel RP, Gladwin MT, Kim‐Shapiro DB and Hogg N. The reaction between nitrite and deoxyhemoglobin. Reassessment of reaction kinetics and stoichiometry. J Biol Chem 280: 31126–31131, 2005.
 134. Tsai AG, Johnson PC and Intaglietta M. Oxygen gradients in the microcirculation. Physiol Rev 83: 933–963, 2003.
 135. Jison ML, Munson PJ, Barb JJ, Suffredini AF, Talwar S, Logun C, Raghavachari N, Beigel JH, Shelhamer JH, Danner RL and Gladwin MT. Blood mononuclear cell gene expression profiles characterize the oxidant, hemolytic, and inflammatory stress of sickle cell disease. Blood 104: 270–280, 2004.
 136. Belcher JD, Mahaseth H, Welch TE, Otterbein LE, Hebbel RP and Vercellotti GM. Heme oxygenase‐1 is a modulator of inflammation and vaso‐occlusion in transgenic sickle mice. J Clin Invest 116: 808–816, 2006.
 137. Ergul S, Brunson CY, Hutchinson J, Tawfik A, Kutlar A, Webb RC and Ergul A. Vasoactive factors in sickle cell disease: in vitro evidence for endothelin‐1‐mediated vasoconstriction. Am J Hematol 76: 245–251, 2004.
 138. Oltman CL, Kane NL, Miller FJ, Jr., Spector AA, Weintraub NL and Dellsperger KC. Reactive oxygen species mediate arachidonic acid‐induced dilation in porcine coronary microvessels. Am J Physiol Heart Circ Physiol 285: H2309–H2315, 2003.
 139. Wu G, Mannam AP, Wu J, Kirbis S, Shie JL, Chen C, Laham RJ, Sellke FW and Li J. Hypoxia induces myocyte‐dependent COX‐2 regulation in endothelial cells: Role of VEGF. Am J Physiol Heart Circ Physiol 285: H2420–H2429, 2003.
 140. Nath KA, Grande JP, Haggard JJ, Croatt AJ, Katusic ZS, Solovey A and Hebbel RP. Oxidative stress and induction of heme oxygenase‐1 in the kidney in sickle cell disease. Am J Pathol 158: 893–903, 2001.
 141. Thengchaisri N and Kuo L. Hydrogen peroxide induces endothelium‐dependent and ‐independent coronary arteriolar dilation: role of cyclooxygenase and potassium channels. Am J Physiol Heart Circ Physiol 285: H2255–H2263, 2003.
 142. Kooy NW and Lewis SJ. Nitrotyrosine attenuates the hemodynamic effects of adrenoceptor agonists in vivo: relevance to the pathophysiology of peroxynitrite. Eur J Pharmacol 310: 155–161, 1996.
 143. Kooy NW and Lewis SJ. The peroxynitrite product 3‐nitro‐L‐tyrosine attenuates the hemodynamic responses to angiotensin II in vivo. Eur J Pharmacol 315: 165–170, 1996.
 144. Charache S, Barton FB, Moore RD, Terrin ML, Steinberg MH, Dover GJ, Ballas SK, McMahon RP, Castro O and Orringer EP. Hydroxyurea and sickle cell anemia. Clinical utility of a myelosuppressive “switching” agent. The multicenter study of hydroxyurea in sickle cell anemia. Medicine 75: 300–326, 1996.
 145. Steinberg MH, Lu ZH, Barton FB, Terrin ML, Charache S and Dover GJ. Fetal hemoglobin in sickle cell anemia: determinants of response to hydroxyurea. Multicenter study of hydroxyurea. Blood 89: 1078–1088, 1997.
 146. Cokic VP, Smith RD, Beleslin‐Cokic BB, Njoroge JM, Miller JL, Gladwin MT and Schechter AN. Hydroxyurea induces fetal hemoglobin by the nitric oxide‐dependent activation of soluble guanylyl cyclase. J Clin Invest 111: 231–239, 2003.
 147. Dasgupta T, Hebbel RP and Kaul DK. Protective effect of arginine on oxidative stress in transgenic sickle mouse models. Free Radic Biol Med 41: 1771–1780, 2006.
 148. Ogawa T, Nussler AK, Tuzuner E, Neuhaus P, Kaminishi M, Mimura Y and Beger HG. Contribution of nitric oxide to the protective effects of ischemic preconditioning in ischemia‐reperfused rat kidneys. J Lab Clin Med 138: 50–58, 2001.
 149. Brugnara C, De Franceschi L and Alper SL. Inhibition of Ca(2+)‐dependent K+ transport and cell dehydration in sickle erythrocytes by clotrimazole and other imidazole derivatives [see comments]. J Clin Invest 92: 520–526, 1993.
 150. De Franceschi L, Saadane N, Trudel M, Alper SL, Brugnara C and Beuzard S. 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 93: 1670–1676, 1994.
 151. Romero JR, Fabry ME, Suzuka SM, Costantini F, Nagel RL and Canessa M. K:Cl cotransport in red cells of transgenic mice expressing high levels of human hemoglobin S. Am J Hematol 55: 112–114, 1997.
 152. Brugnara C. Sickle cell disease: from membrane pathophysiology to novel therapies for prevention of erythrocyte dehydration. J Pediatr Hematol Oncol 25: 927–933, 2003.
 153. Pawliuk R, Westerman KA, Fabry ME, Payen E, Tighe R, Bouhassira EE, Acharya SA, Ellis J, London IM, Eaves CJ, Humphries RK, Beuzard Y, Nagel RL and Leboulch P. Correction of sickle cell disease in transgenic mouse models by gene therapy. Science 294: 2368–2371, 2001.
 154. Levasseur DN, Ryan TM, Pawlik KM and Townes TM. Correction of a mouse model of sickle cell disease: Lentiviral/antisickling beta‐globin gene transduction of unmobilized, purified hematopoietic stem cells. Blood 102: 4312–4319, 2003.
 155. Fabry ME, Nagel RL, Pachnis A, Suzuka SM and Costantini F. High expression of human beta S‐ and alpha‐globins in transgenic mice: Hemoglobin composition and hematological consequences. Proc Natl Acad Sci USA 89: 12150–12154, 1992.
 156. Noguchi CT, Gladwin M, Diwan B, Merciris P, Smith R, Yu X, Buzard G, Fitzhugh A, Keefer LK, Schechter AN and Mohandas N. Pathophysiology of a sickle cell trait mouse model: human alpha(beta)(S) transgenes with one mouse beta‐globin allele. Blood Cells Mol Dis 27: 971–977, 2001.

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Dhananjay K Kaul. Sickle Cell Disease. Compr Physiol 2011, Supplement 9: Handbook of Physiology, The Cardiovascular System, Microcirculation: 769-793. First published in print 2008. doi: 10.1002/cphy.cp020417