1932

Abstract

Since the discovery of sickle cell disease (SCD) in 1910, enormous strides have been made in the elucidation of the pathogenesis of its protean complications, which has inspired recent advances in targeted molecular therapies. In SCD, a single amino acid substitution in the β-globin chain leads to polymerization of mutant hemoglobin S, impairing erythrocyte rheology and survival. Clinically, erythrocyte abnormalities in SCD manifest in hemolytic anemia and cycles of microvascular vaso-occlusion leading to end-organ ischemia-reperfusion injury and infarction. Vaso-occlusive events and intravascular hemolysis promote inflammation and redox instability that lead to progressive small- and large-vessel vasculopathy. Based on current evidence, the pathobiology of SCD is considered to be a vicious cycle of four major processes, all the subject of active study and novel therapeutic targeting: () hemoglobin S polymerization, () impaired biorheology and increased adhesion-mediated vaso-occlusion, () hemolysis-mediated endothelial dysfunction, and () concerted activation of sterile inflammation (Toll-like receptor 4– and inflammasome-dependent innate immune pathways). These molecular, cellular, and biophysical processes synergize to promote acute and chronic pain and end-organ injury and failure in SCD. This review provides an exhaustive overview of the current understanding of the molecular pathophysiology of SCD, how this pathophysiology contributes to complications of the central nervous and cardiopulmonary systems, and how this knowledge is being harnessed to develop current and potential therapies.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-pathmechdis-012418-012838
2019-01-24
2024-03-28
Loading full text...

Full text loading...

/deliver/fulltext/pathmechdis/14/1/annurev-pathmechdis-012418-012838.html?itemId=/content/journals/10.1146/annurev-pathmechdis-012418-012838&mimeType=html&fmt=ahah

Literature Cited

  1. 1.  Rees DC, Williams TN, Gladwin MT 2010. Sickle-cell disease. Lancet 376:2018–31
    [Google Scholar]
  2. 2.  Piel FB, Patil AP, Howes RE, Nyangiri OA, Gething PW et al. 2013. Global epidemiology of sickle haemoglobin in neonates: a contemporary geostatistical model-based map and population estimates. Lancet 381:142–51
    [Google Scholar]
  3. 3.  Piel FB, Patil AP, Howes RE, Nyangiri OA, Gething PW et al. 2010. Global distribution of the sickle cell gene and geographical confirmation of the malaria hypothesis. Nat. Commun. 1:104
    [Google Scholar]
  4. 4.  GBD Mortal, Causes Death Collab 2015. Global, regional, and national age-sex specific all-cause and cause-specific mortality for 240 causes of death, 1990–2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet 385:117–71
    [Google Scholar]
  5. 5.  Ingram VM 1956. A specific chemical difference between the globins of normal human and sickle-cell anaemia haemoglobin. Nature 178:792–94
    [Google Scholar]
  6. 6.  Ingram VM 1957. Gene mutations in human haemoglobin: the chemical difference between normal and sickle cell haemoglobin. Nature 180:326–28
    [Google Scholar]
  7. 7.  Bunn HF 1997. Pathogenesis and treatment of sickle cell disease. N. Engl. J. Med. 337:762–69
    [Google Scholar]
  8. 8.  Pauling L, Itano HA, Singer SJ, Wells IC 1949. Sickle cell anemia, a molecular disease. Science 110:543–48
    [Google Scholar]
  9. 9.  Barabino GA, Platt MO, Kaul DK 2010. Sickle cell biomechanics. Annu. Rev. Biomed. Eng. 12:345–67
    [Google Scholar]
  10. 10.  Noguchi CT, Rodgers GP, Serjeant G, Schechter AN 1988. Levels of fetal hemoglobin necessary for treatment of sickle cell disease. N. Engl. J. Med. 318:96–99
    [Google Scholar]
  11. 11.  Brittenham GM, Schechter AN, Noguchi CT 1985. Hemoglobin S polymerization: primary determinant of the hemolytic and clinical severity of the sickling syndromes. Blood 65:183–89
    [Google Scholar]
  12. 12.  Ware RE, de Montalembert M, Tshilolo L, Abboud MR 2017. Sickle cell disease. Lancet 390:311–23
    [Google Scholar]
  13. 13.  Manwani D, Frenette PS 2013. Vaso-occlusion in sickle cell disease: pathophysiology and novel targeted therapies. Blood 122:3892–98
    [Google Scholar]
  14. 14.  Zhang D, Xu C, Manwani D, Frenette PS 2016. Neutrophils, platelets, and inflammatory pathways at the nexus of sickle cell disease pathophysiology. Blood 127:801–9
    [Google Scholar]
  15. 15.  Kaul DK, Finnegan E, Barabino GA 2009. Sickle red cell-endothelium interactions. Microcirculation 16:97–111
    [Google Scholar]
  16. 16.  Bennewitz MF, Jimenez MA, Vats R, Tutuncuoglu E, Jonassaint J et al. 2017. Lung vaso-occlusion in sickle cell disease mediated by arteriolar neutrophil-platelet microemboli. JCI Insight 2:e89761
    [Google Scholar]
  17. 17.  Kato GJ, Steinberg MH, Gladwin MT 2017. Intravascular hemolysis and the pathophysiology of sickle cell disease. J. Clin. Invest. 127:750–60
    [Google Scholar]
  18. 18.  Gladwin MT, Ofori-Acquah SF 2014. Erythroid DAMPs drive inflammation in SCD. Blood 123:3689–90
    [Google Scholar]
  19. 19.  Kenny MW, George AJ, Stuart J 1980. Platelet hyperactivity in sickle-cell disease: a consequence of hyposplenism. J. Clin. Pathol. 33:622–25
    [Google Scholar]
  20. 20.  Mohan JS, Lip GY, Bareford D, Blann AD 2006. Platelet P-selectin and platelet mass, volume and component in sickle cell disease: relationship to genotype. Thromb. Res. 117:623–29
    [Google Scholar]
  21. 21.  Westwick J, Watson-Williams EJ, Krishnamurthi S, Marks G, Ellis V et al. 1983. Platelet activation during steady state sickle cell disease. J. Med. 14:17–36
    [Google Scholar]
  22. 22.  Curtis SA, Danda N, Etzion Z, Cohen HW, Billett HH 2015. Elevated steady state WBC and platelet counts are associated with frequent emergency room use in adults with sickle cell anemia. PLOS ONE 10:e0133116
    [Google Scholar]
  23. 23.  Frelinger AL 3rd, Jakubowski JA, Brooks JK, Carmichael SL, Berny-Lang MA et al. 2014. Platelet activation and inhibition in sickle cell disease (pains) study. Platelets 25:27–35
    [Google Scholar]
  24. 24.  Dominical VM, Samsel L, Nichols JS, Costa FF, McCoy JP Jr et al. 2014. Prominent role of platelets in the formation of circulating neutrophil-red cell heterocellular aggregates in sickle cell anemia. Haematologica 99:e214–17
    [Google Scholar]
  25. 25.  Polanowska-Grabowska R, Wallace K, Field JJ, Chen L, Marshall MA et al. 2010. P-selectin-mediated platelet-neutrophil aggregate formation activates neutrophils in mouse and human sickle cell disease. Arterioscler. Thromb. Vasc. Biol. 30:2392–99
    [Google Scholar]
  26. 26.  Wun T, Cordoba M, Rangaswami A, Cheung AW, Paglieroni T 2002. Activated monocytes and platelet-monocyte aggregates in patients with sickle cell disease. Clin. Lab. Haematol. 24:81–88
    [Google Scholar]
  27. 27.  Miller ST, Sleeper LA, Pegelow CH, Enos LE, Wang WC et al. 2000. Prediction of adverse outcomes in children with sickle cell disease. N. Engl. J. Med. 342:83–89
    [Google Scholar]
  28. 28.  Wongtong N, Jones S, Deng Y, Cai J, Ataga KI 2015. Monocytosis is associated with hemolysis in sickle cell disease. Hematology 20:593–97
    [Google Scholar]
  29. 29.  Chaturvedi S, Ghafuri DL, Glassberg J, Kassim AA, Rodeghier M, DeBaun MR 2016. Rapidly progressive acute chest syndrome in individuals with sickle cell anemia: a distinct acute chest syndrome phenotype. Am. J. Hematol. 91:1185–90
    [Google Scholar]
  30. 30.  Gardner K, Thein SL 2015. Super-elevated LDH and thrombocytopenia are markers of a severe subtype of vaso-occlusive crisis in sickle cell disease. Am. J. Hematol. 90:E206–7
    [Google Scholar]
  31. 31.  Alhandalous CH, Han J, Hsu L, Gowhari M, Hassan J et al. 2015. Platelets decline during vaso-occlusive crisis as a predictor of acute chest syndrome in sickle cell disease. Am. J. Hematol. 90:E228–29
    [Google Scholar]
  32. 32.  Novelli EM, Gladwin MT 2016. Crises in sickle cell disease. Chest 149:1082–93
    [Google Scholar]
  33. 33.  Hidalgo A, Chang J, Jang JE, Peired AJ, Chiang EY, Frenette PS 2009. Heterotypic interactions enabled by polarized neutrophil microdomains mediate thromboinflammatory injury. Nat. Med. 15:384–91
    [Google Scholar]
  34. 34.  Belcher JD, Chen C, Nguyen J, Milbauer L, Abdulla F et al. 2014. Heme triggers TLR4 signaling leading to endothelial cell activation and vaso-occlusion in murine sickle cell disease. Blood 123:377–90
    [Google Scholar]
  35. 35.  Wallace KL, Linden J 2010. Adenosine A2A receptors induced on iNKT and NK cells reduce pulmonary inflammation and injury in mice with sickle cell disease. Blood 116:5010–20
    [Google Scholar]
  36. 36.  Gutsaeva DR, Montero-Huerta P, Parkerson JB, Yerigenahally SD, Ikuta T, Head CA 2014. Molecular mechanisms underlying synergistic adhesion of sickle red blood cells by hypoxia and low nitric oxide bioavailability. Blood 123:1917–26
    [Google Scholar]
  37. 37.  Zennadi R, Moeller BJ, Whalen EJ, Batchvarova M, Xu K et al. 2007. Epinephrine-induced activation of LW-mediated sickle cell adhesion and vaso-occlusion in vivo. Blood 110:2708–17
    [Google Scholar]
  38. 38.  Turhan A, Weiss LA, Mohandas N, Coller BS, Frenette PS 2002. Primary role for adherent leukocytes in sickle cell vascular occlusion: a new paradigm. PNAS 99:3047–51
    [Google Scholar]
  39. 39.  Chang J, Patton JT, Sarkar A, Ernst B, Magnani JL, Frenette PS 2010. GMI-1070, a novel pan-selectin antagonist, reverses acute vascular occlusions in sickle cell mice. Blood 116:1779–86
    [Google Scholar]
  40. 40.  Chang J, Shi PA, Chiang EY, Frenette PS 2008. Intravenous immunoglobulins reverse acute vaso-occlusive crises in sickle cell mice through rapid inhibition of neutrophil adhesion. Blood 111:915–23
    [Google Scholar]
  41. 41.  Jang JE, Hod EA, Spitalnik SL, Frenette PS 2011. CXCL1 and its receptor, CXCR2, mediate murine sickle cell vaso-occlusion during hemolytic transfusion reactions. J. Clin. Invest. 121:1397–401
    [Google Scholar]
  42. 42.  Almeida CB, Scheiermann C, Jang JE, Prophete C, Costa FF et al. 2012. Hydroxyurea and a cGMP-amplifying agent have immediate benefits on acute vaso-occlusive events in sickle cell disease mice. Blood 120:2879–88
    [Google Scholar]
  43. 43.  Li J, Kim K, Hahm E, Molokie R, Hay N et al. 2014. Neutrophil AKT2 regulates heterotypic cell-cell interactions during vascular inflammation. J. Clin. Invest. 124:1483–96
    [Google Scholar]
  44. 44.  Kim K, Li J, Tseng A, Andrews RK, Cho J 2015. NOX2 is critical for heterotypic neutrophil-platelet interactions during vascular inflammation. Blood 126:1952–64
    [Google Scholar]
  45. 45.  Embury SH, Matsui NM, Ramanujam S, Mayadas TN, Noguchi CT et al. 2004. The contribution of endothelial cell P-selectin to the microvascular flow of mouse sickle erythrocytes in vivo. Blood 104:3378–85
    [Google Scholar]
  46. 46.  Gutsaeva DR, Parkerson JB, Yerigenahally SD, Kurz JC, Schaub RG et al. 2011. Inhibition of cell adhesion by anti-P-selectin aptamer: a new potential therapeutic agent for sickle cell disease. Blood 117:727–35
    [Google Scholar]
  47. 47.  Zennadi R 2014. MEK inhibitors, novel anti-adhesive molecules, reduce sickle red blood cell adhesion in vitro and in vivo, and vasoocclusion in vivo. PLOS ONE 9:e110306
    [Google Scholar]
  48. 48.  Zennadi R, Hines PC, De Castro LM, Cartron JP, Parise LV, Telen MJ 2004. Epinephrine acts through erythroid signaling pathways to activate sickle cell adhesion to endothelium via LW-alphavbeta3 interactions. Blood 104:3774–81
    [Google Scholar]
  49. 49.  Zennadi R, Whalen EJ, Soderblom EJ, Alexander SC, Thompson JW et al. 2012. Erythrocyte plasma membrane-bound ERK1/2 activation promotes ICAM-4-mediated sickle red cell adhesion to endothelium. Blood 119:1217–27
    [Google Scholar]
  50. 50.  Zhao Y, Schwartz EA, Palmer GM, Zennadi R 2016. MEK1/2 inhibitors reverse acute vascular occlusion in mouse models of sickle cell disease. FASEB J 30:1171–86
    [Google Scholar]
  51. 51.  Zhang D, Chen G, Manwani D, Mortha A, Xu C et al. 2015. Neutrophil ageing is regulated by the microbiome. Nature 525:528–32
    [Google Scholar]
  52. 52.  Lebensburger JD, Howard T, Hu Y, Pestina TI, Gao G et al. 2012. Hydroxyurea therapy of a murine model of sickle cell anemia inhibits the progression of pneumococcal disease by down-modulating E-selectin. Blood 119:1915–21
    [Google Scholar]
  53. 53.  Jimenez MA, Novelli E, Shaw GD, Sundd P 2017. Glycoprotein Ibα inhibitor (CCP-224) prevents neutrophil-platelet aggregation in sickle cell disease. Blood Adv 1:1712–16
    [Google Scholar]
  54. 54.  Sparkenbaugh E, Pawlinski R 2013. Interplay between coagulation and vascular inflammation in sickle cell disease. Br. J. Haematol. 162:3–14
    [Google Scholar]
  55. 55.  Pakbaz Z, Wun T 2014. Role of the hemostatic system on sickle cell disease pathophysiology and potential therapeutics. Hematol. Oncol. Clin. North Am. 28:355–74
    [Google Scholar]
  56. 56.  Noubouossie D, Key NS, Ataga KI 2016. Coagulation abnormalities of sickle cell disease: relationship with clinical outcomes and the effect of disease modifying therapies. Blood Rev 30:245–56
    [Google Scholar]
  57. 57.  Liu Y, Jing F, Yi W, Mendelson A, Shi P et al. 2018. HO-1hi patrolling monocytes protect against vaso-occlusion in sickle cell disease. Blood 131:1600–10
    [Google Scholar]
  58. 58.  Ataga KI, Moore CG, Jones S, Olajide O, Strayhorn D et al. 2006. Pulmonary hypertension in patients with sickle cell disease: a longitudinal study. Br. J. Haematol. 134:109–15
    [Google Scholar]
  59. 59.  Bartolucci P, Brugnara C, Teixeira-Pinto A, Pissard S, Moradkhani K et al. 2012. Erythrocyte density in sickle cell syndromes is associated with specific clinical manifestations and hemolysis. Blood 120:3136–41
    [Google Scholar]
  60. 60.  Day TG, Drasar ER, Fulford T, Sharpe CC, Thein SL 2012. Association between hemolysis and albuminuria in adults with sickle cell anemia. Haematologica 97:201–5
    [Google Scholar]
  61. 61.  Fitzhugh CD, Lauder N, Jonassaint JC, Telen MJ, Zhao X et al. 2010. Cardiopulmonary complications leading to premature deaths in adult patients with sickle cell disease. Am. J. Hematol. 85:36–40
    [Google Scholar]
  62. 62.  Gladwin MT, Barst RJ, Gibbs JS, Hildesheim M, Sachdev V et al. 2014. Risk factors for death in 632 patients with sickle cell disease in the United States and United Kingdom. PLOS ONE 9:e99489
    [Google Scholar]
  63. 63.  Gladwin MT, Sachdev V 2012. Cardiovascular abnormalities in sickle cell disease. J. Am. Coll. Cardiol. 59:1123–33
    [Google Scholar]
  64. 64.  Gladwin MT, Sachdev V, Jison ML, Shizukuda Y, Plehn JF et al. 2004. Pulmonary hypertension as a risk factor for death in patients with sickle cell disease. N. Engl. J. Med. 350:886–95
    [Google Scholar]
  65. 65.  Kato GJ, McGowan V, Machado RF, Little JA, Taylor J et al. 2006. Lactate dehydrogenase as a biomarker of hemolysis-associated nitric oxide resistance, priapism, leg ulceration, pulmonary hypertension, and death in patients with sickle cell disease. Blood 107:2279–85
    [Google Scholar]
  66. 66.  Mehari A, Alam S, Tian X, Cuttica MJ, Barnett CF et al. 2013. Hemodynamic predictors of mortality in adults with sickle cell disease. Am. J. Respir. Crit. Care Med. 187:840–47
    [Google Scholar]
  67. 67.  Novelli EM, Hildesheim M, Rosano C, Vanderpool R, Simon M et al. 2014. Elevated pulse pressure is associated with hemolysis, proteinuria and chronic kidney disease in sickle cell disease. PLOS ONE 9:e114309
    [Google Scholar]
  68. 68.  Platt OS, Brambilla DJ, Rosse WF, Milner PF, Castro O et al. 1994. Mortality in sickle cell disease: life expectancy and risk factors for early death. N. Engl. J. Med. 330:1639–44
    [Google Scholar]
  69. 69.  Powars DR, Elliott-Mills DD, Chan L 1991. Chronic renal failure in sickle cell disease: risk factors, clinical course, and mortality. Ann. Intern. Med. 115:614–20
    [Google Scholar]
  70. 70.  Saraf SL, Zhang X, Kanias T, Lash JP, Molokie RE et al. 2014. Haemoglobinuria is associated with chronic kidney disease and its progression in patients with sickle cell anaemia. Br. J. Haematol. 164:729–39
    [Google Scholar]
  71. 71.  Belcher JD, Bryant CJ, Nguyen J, Bowlin PR, Kielbik MC et al. 2003. Transgenic sickle mice have vascular inflammation. Blood 101:3953–59
    [Google Scholar]
  72. 72.  Osarogiagbon UR, Choong S, Belcher JD, Vercellotti GM, Paller MS, Hebbel RP 2000. Reperfusion injury pathophysiology in sickle transgenic mice. Blood 96:314–20
    [Google Scholar]
  73. 73.  Connes P, Lamarre Y, Waltz X, Ballas SK, Lemonne N et al. 2014. Haemolysis and abnormal haemorheology in sickle cell anaemia. Br. J. Haematol. 165:564–72
    [Google Scholar]
  74. 74.  Nouraie M, Lee JS, Zhang Y, Kanias T, Zhao X et al. 2013. The relationship between the severity of hemolysis, clinical manifestations and risk of death in 415 patients with sickle cell anemia in the US and Europe. Haematologica 98:464–72
    [Google Scholar]
  75. 75.  Gladwin MT 2016. Cardiovascular complications and risk of death in sickle-cell disease. Lancet 387:2565–74
    [Google Scholar]
  76. 76.  Poludasu S, Ramkissoon K, Salciccioli L, Kamran H, Lazar JM 2013. Left ventricular systolic function in sickle cell anemia: a meta-analysis. J. Card. Fail. 19:333–41
    [Google Scholar]
  77. 77.  Sachdev V, Kato GJ, Gibbs JS, Barst RJ, Machado RF et al. 2011. Echocardiographic markers of elevated pulmonary pressure and left ventricular diastolic dysfunction are associated with exercise intolerance in adults and adolescents with homozygous sickle cell anemia in the United States and United Kingdom. Circulation 124:1452–60
    [Google Scholar]
  78. 78.  Milton JN, Rooks H, Drasar E, McCabe EL, Baldwin CT et al. 2013. Genetic determinants of haemolysis in sickle cell anaemia. Br. J. Haematol. 161:270–78
    [Google Scholar]
  79. 79.  Kato GJ, Gladwin MT, Steinberg MH 2007. Deconstructing sickle cell disease: reappraisal of the role of hemolysis in the development of clinical subphenotypes. Blood Rev 21:37–47
    [Google Scholar]
  80. 80.  DeBaun MR, Armstrong FD, McKinstry RC, Ware RE, Vichinsky E, Kirkham FJ 2012. Silent cerebral infarcts: a review on a prevalent and progressive cause of neurologic injury in sickle cell anemia. Blood 119:4587–96
    [Google Scholar]
  81. 81.  Ohene-Frempong K, Weiner SJ, Sleeper LA, Miller ST, Embury S et al. 1998. Cerebrovascular accidents in sickle cell disease: rates and risk factors. Blood 91:288–94
    [Google Scholar]
  82. 82.  Sachdev V, Machado RF, Shizukuda Y, Rao YN, Sidenko S et al. 2007. Diastolic dysfunction is an independent risk factor for death in patients with sickle cell disease. J. Am. Coll. Cardiol. 49:472–79
    [Google Scholar]
  83. 83.  Lundberg JO, Gladwin MT, Weitzberg E 2015. Strategies to increase nitric oxide signalling in cardiovascular disease. Nat. Rev. Drug Discov. 14:623–41
    [Google Scholar]
  84. 84.  Reiter CD, Wang X, Tanus-Santos JE, Hogg N, Cannon RO III et al. 2002. Cell-free hemoglobin limits nitric oxide bioavailability in sickle-cell disease. Nat. Med. 8:1383–89
    [Google Scholar]
  85. 85.  Rother RP, Bell L, Hillmen P, Gladwin MT 2005. The clinical sequelae of intravascular hemolysis and extracellular plasma hemoglobin: a novel mechanism of human disease. JAMA 293:1653–62
    [Google Scholar]
  86. 86.  Morris CR, Kato GJ, Poljakovic M, Wang X, Blackwelder WC et al. 2005. Dysregulated arginine metabolism, hemolysis-associated pulmonary hypertension, and mortality in sickle cell disease. JAMA 294:81–90
    [Google Scholar]
  87. 87.  Hsu LL, Champion HC, Campbell-Lee SA, Bivalacqua TJ, Manci EA et al. 2007. Hemolysis in sickle cell mice causes pulmonary hypertension due to global impairment in nitric oxide bioavailability. Blood 109:3088–98
    [Google Scholar]
  88. 88.  Gladwin MT 2006. Deconstructing endothelial dysfunction: soluble guanylyl cyclase oxidation and the NO resistance syndrome. J. Clin. Invest. 116:2330–32
    [Google Scholar]
  89. 89.  Ghosh S, Adisa OA, Chappa P, Tan F, Jackson KA et al. 2013. Extracellular hemin crisis triggers acute chest syndrome in sickle mice. J. Clin. Invest. 123:4809–20
    [Google Scholar]
  90. 90.  Almeida CB, Souza LE, Leonardo FC, Costa FT, Werneck CC et al. 2015. Acute hemolytic vascular inflammatory processes are prevented by nitric oxide replacement or a single dose of hydroxyurea. Blood 126:711–20
    [Google Scholar]
  91. 91.  Villagra J, Shiva S, Hunter LA, Machado RF, Gladwin MT, Kato GJ 2007. Platelet activation in patients with sickle disease, hemolysis-associated pulmonary hypertension, and nitric oxide scavenging by cell-free hemoglobin. Blood 110:2166–72
    [Google Scholar]
  92. 92.  Cardenes N, Corey C, Geary L, Jain S, Zharikov S et al. 2014. Platelet bioenergetic screen in sickle cell patients reveals mitochondrial complex V inhibition, which contributes to platelet activation. Blood 123:2864–72
    [Google Scholar]
  93. 93.  Helms CC, Marvel M, Zhao W, Stahle M, Vest R et al. 2013. Mechanisms of hemolysis-associated platelet activation. J. Thromb. Haemost. 11:2148–54
    [Google Scholar]
  94. 94.  Ataga KI 2009. Hypercoagulability and thrombotic complications in hemolytic anemias. Haematologica 94:1481–84
    [Google Scholar]
  95. 95.  Chen GY, Nunez G 2010. Sterile inflammation: sensing and reacting to damage. Nat. Rev. Immunol. 10:826–37
    [Google Scholar]
  96. 96.  Dutra FF, Alves LS, Rodrigues D, Fernandez PL, de Oliveira RB et al. 2014. Hemolysis-induced lethality involves inflammasome activation by heme. PNAS 111:E4110–18
    [Google Scholar]
  97. 97.  Chen G, Zhang D, Fuchs TA, Wagner DD, Frenette PS 2014. Heme-induced neutrophil extracellular traps contribute to the pathogenesis of sickle cell disease. Blood 123:3818–27
    [Google Scholar]
  98. 98.  Setty BN, Betal SG, Zhang J, Stuart MJ 2008. Heme induces endothelial tissue factor expression: potential role in hemostatic activation in patients with hemolytic anemia. J. Thromb. Haemost. 6:2202–9
    [Google Scholar]
  99. 99.  Dutra FF, Bozza MT 2014. Heme on innate immunity and inflammation. Front. Pharmacol. 5:115
    [Google Scholar]
  100. 100.  Camus SM, De Moraes JA, Bonnin P, Abbyad P, Le Jeune S et al. 2015. Circulating cell membrane microparticles transfer heme to endothelial cells and trigger vasoocclusions in sickle cell disease. Blood 125:3805–14
    [Google Scholar]
  101. 101.  Monteiro AP, Pinheiro CS, Luna-Gomes T, Alves LR, Maya-Monteiro CM et al. 2011. Leukotriene B4 mediates neutrophil migration induced by heme. J. Immunol. 186:6562–67
    [Google Scholar]
  102. 102.  Figueiredo RT, Fernandez PL, Mourao-Sa DS, Porto BN, Dutra FF et al. 2007. Characterization of heme as activator of Toll-like receptor 4. J. Biol. Chem. 282:20221–29
    [Google Scholar]
  103. 103.  Arruda MA, Rossi AG, de Freitas MS, Barja-Fidalgo C, Graca-Souza AV 2004. Heme inhibits human neutrophil apoptosis: involvement of phosphoinositide 3-kinase, MAPK, and NF-κB. J. Immunol. 173:2023–30
    [Google Scholar]
  104. 104.  Kono M, Saigo K, Takagi Y, Takahashi T, Kawauchi S et al. 2014. Heme-related molecules induce rapid production of neutrophil extracellular traps. Transfusion 54:2811–19
    [Google Scholar]
  105. 105.  Graca-Souza AV, Arruda MA, de Freitas MS, Barja-Fidalgo C, Oliveira PL 2002. Neutrophil activation by heme: implications for inflammatory processes. Blood 99:4160–65
    [Google Scholar]
  106. 106.  Porto BN, Alves LS, Fernandez PL, Dutra TP, Figueiredo RT et al. 2007. Heme induces neutrophil migration and reactive oxygen species generation through signaling pathways characteristic of chemotactic receptors. J. Biol. Chem. 282:24430–36
    [Google Scholar]
  107. 107.  Jorch SK, Kubes P 2017. An emerging role for neutrophil extracellular traps in noninfectious disease. Nat. Med. 23:279–87
    [Google Scholar]
  108. 108.  Schimmel M, Nur E, Biemond BJ, van Mierlo GJ, Solati S et al. 2013. Nucleosomes and neutrophil activation in sickle cell disease painful crisis. Haematologica 98:1797–803
    [Google Scholar]
  109. 109.  Malik Z, Creter D, Cohen A, Djaldetti M 1983. Haemin affects platelet aggregation and lymphocyte mitogenicity in whole blood incubations. Cytobios 38:33–38
    [Google Scholar]
  110. 110.  Gladwin MT, Vichinsky E 2008. Pulmonary complications of sickle cell disease. N. Engl. J. Med. 359:2254–65
    [Google Scholar]
  111. 111.  Vichinsky EP, Neumayr LD, Earles AN, Williams R, Lennette ET et al. 2000. Causes and outcomes of the acute chest syndrome in sickle cell disease. National Acute Chest Syndrome Study Group. N. Engl. J. Med. 342:1855–65
    [Google Scholar]
  112. 112.  Martins R, Maier J, Gorki AD, Huber KV, Sharif O et al. 2016. Heme drives hemolysis-induced susceptibility to infection via disruption of phagocyte functions. Nat. Immunol. 17:1361–72
    [Google Scholar]
  113. 113.  Kaul DK, Hebbel RP 2000. Hypoxia/reoxygenation causes inflammatory response in transgenic sickle mice but not in normal mice. J. Clin. Invest. 106:411–20
    [Google Scholar]
  114. 114.  Belcher JD, Mahaseth H, Welch TE, Vilback AE, Sonbol KM et al. 2005. Critical role of endothelial cell activation in hypoxia-induced vasoocclusion in transgenic sickle mice. Am. J. Physiol. Heart Circ. Physiol. 288:H2715–25
    [Google Scholar]
  115. 115.  Eltzschig HK, Eckle T 2011. Ischemia and reperfusion—from mechanism to translation. Nat. Med. 17:1391–401
    [Google Scholar]
  116. 116.  Hotchkiss RS, Strasser A, McDunn JE, Swanson PE 2009. Cell death. N. Engl. J. Med. 361:1570–83
    [Google Scholar]
  117. 117.  Krysko DV, Agostinis P, Krysko O, Garg AD, Bachert C et al. 2011. Emerging role of damage-associated molecular patterns derived from mitochondria in inflammation. Trends Immunol 32:157–64
    [Google Scholar]
  118. 118.  Xu H, Wandersee NJ, Guo Y, Jones DW, Holzhauer SL et al. 2014. Sickle cell disease increases high mobility group box 1: a novel mechanism of inflammation. Blood 124:3978–81
    [Google Scholar]
  119. 119.  Elliott EI, Sutterwala FS 2015. Initiation and perpetuation of NLRP3 inflammasome activation and assembly. Immunol. Rev. 265:35–52
    [Google Scholar]
  120. 120.  Man SM, Kanneganti TD 2015. Regulation of inflammasome activation. Immunol. Rev. 265:6–21
    [Google Scholar]
  121. 121.  Warnatsch A, Ioannou M, Wang Q, Papayannopoulos V 2015. Neutrophil extracellular traps license macrophages for cytokine production in atherosclerosis. Science 349:316–20
    [Google Scholar]
  122. 122.  Pitanga TN, Oliveira RR, Zanette DL, Guarda CC, Santiago RP et al. 2016. Sickle red cells as danger signals on proinflammatory gene expression, leukotriene B4 and interleukin-1 beta production in peripheral blood mononuclear cell. Cytokine 83:75–84
    [Google Scholar]
  123. 123.  Alagbe AE, Justo AS Jr., Ruas LP, Tonasse WV, Santana RM et al. 2017. Interleukin-27 and interleukin-37 are elevated in sickle cell anemia patients and inhibit in vitro secretion of interleukin-8 in neutrophils and monocytes. Cytokine 107:85–92
    [Google Scholar]
  124. 124.  Bakele M, Joos M, Burdi S, Allgaier N, Poschel S et al. 2014. Localization and functionality of the inflammasome in neutrophils. J. Biol. Chem. 289:5320–29
    [Google Scholar]
  125. 125.  Hottz ED, Lopes JF, Freitas C, Valls-de-Souza R, Oliveira MF et al. 2013. Platelets mediate increased endothelium permeability in dengue through NLRP3-inflammasome activation. Blood 122:3405–14
    [Google Scholar]
  126. 126.  Hottz ED, Monteiro AP, Bozza FA, Bozza PT 2015. Inflammasome in platelets: allying coagulation and inflammation in infectious and sterile diseases?. Mediat. Inflamm. 2015:435783
    [Google Scholar]
  127. 127.  Xia M, Boini KM, Abais JM, Xu M, Zhang Y, Li PL 2014. Endothelial NLRP3 inflammasome activation and enhanced neointima formation in mice by adipokine visfatin. Am. J. Pathol. 184:1617–28
    [Google Scholar]
  128. 128.  Telen MJ 2016. Beyond hydroxyurea: new and old drugs in the pipeline for sickle cell disease. Blood 127:810–19
    [Google Scholar]
  129. 129.  Belcher JD, Young M, Chen C, Nguyen J, Burhop K et al. 2013. MP4CO, a pegylated hemoglobin saturated with carbon monoxide, is a modulator of HO-1, inflammation, and vaso-occlusion in transgenic sickle mice. Blood 122:2757–64
    [Google Scholar]
  130. 130.  Fleischmann RM, Tesser J, Schiff MH, Schechtman J, Burmester GR et al. 2006. Safety of extended treatment with anakinra in patients with rheumatoid arthritis. Ann. Rheum. Dis. 65:1006–12
    [Google Scholar]
  131. 131.  Lachmann HJ, Kone-Paut I, Kuemmerle-Deschner JB, Leslie KS, Hachulla E et al. 2009. Use of canakinumab in the cryopyrin-associated periodic syndrome. N. Engl. J. Med. 360:2416–25
    [Google Scholar]
  132. 132.  Nath KA, Katusic ZS, Gladwin MT 2004. The perfusion paradox and vascular instability in sickle cell disease. Microcirculation 11:179–93
    [Google Scholar]
  133. 133.  Powars D, Wilson B, Imbus C, Pegelow C, Allen J 1978. The natural history of stroke in sickle cell disease. Am. J. Med. 65:461–71
    [Google Scholar]
  134. 134.  Rothman SM, Fulling KH, Nelson JS 1986. Sickle cell anemia and central nervous system infarction: a neuropathological study. Ann. Neurol. 20:684–90
    [Google Scholar]
  135. 135.  Merkel KH, Ginsberg PL, Parker JC Jr, Post MJ 1978. Cerebrovascular disease in sickle cell anemia: a clinical, pathological and radiological correlation. Stroke 9:45–52
    [Google Scholar]
  136. 136.  Prohovnik I, Pavlakis SG, Piomelli S, Bello J, Mohr JP et al. 1989. Cerebral hyperemia, stroke, and transfusion in sickle cell disease. Neurology 39:344–48
    [Google Scholar]
  137. 137.  Kim YS, Nur E, van Beers EJ, Truijen J, Davis SC et al. 2009. Dynamic cerebral autoregulation in homozygous sickle cell disease. Stroke 40:808–14
    [Google Scholar]
  138. 138.  Nur E, Kim YS, Truijen J, van Beers EJ, Davis SC et al. 2009. Cerebrovascular reserve capacity is impaired in patients with sickle cell disease. Blood 114:3473–78
    [Google Scholar]
  139. 139.  Fields ME, Guilliams KP, Ragan DK, Binkley MM, Eldeniz C et al. 2018. Regional oxygen extraction predicts border zone vulnerability to stroke in sickle cell disease. Neurology 90:e1134–42
    [Google Scholar]
  140. 140.  Jordan LC, Gindville MC, Scott AO, Juttukonda MR, Strother MK et al. 2016. Non-invasive imaging of oxygen extraction fraction in adults with sickle cell anaemia. Brain 139:738–50
    [Google Scholar]
  141. 141.  Adams RJ, Nichols FT, McKie V, McKie K, Milner P, Gammal TE 1988. Cerebral infarction in sickle cell anemia: mechanism based on CT and MRI. Neurology 38:1012–17
    [Google Scholar]
  142. 142.  Guilliams KP, Fields ME, Ragan DK, Chen Y, Eldeniz C et al. 2017. Large-vessel vasculopathy in children with sickle cell disease: a magnetic resonance imaging study of infarct topography and focal atrophy. Pediatr. Neurol. 69:49–57
    [Google Scholar]
  143. 143.  Kirk GR, Haynes MR, Palasis S, Brown C, Burns TG et al. 2009. Regionally specific cortical thinning in children with sickle cell disease. Cereb. Cortex 19:1549–56
    [Google Scholar]
  144. 144.  Kawadler JM, Clayden JD, Kirkham FJ, Cox TC, Saunders DE, Clark CA 2013. Subcortical and cerebellar volumetric deficits in paediatric sickle cell anaemia. Br. J. Haematol. 163:373–76
    [Google Scholar]
  145. 145.  Kawadler JM, Clark CA, McKinstry RC, Kirkham FJ 2017. Brain atrophy in paediatric sickle cell anaemia: findings from the silent infarct transfusion (SIT) trial. Br. J. Haematol. 177:151–53
    [Google Scholar]
  146. 146.  Earley CJ, Kittner SJ, Feeser BR, Gardner J, Epstein A et al. 1998. Stroke in children and sickle-cell disease: Baltimore-Washington Cooperative Young Stroke Study. Neurology 51:169–76
    [Google Scholar]
  147. 147.  Dhabangi A, Ainomugisha B, Cserti-Gazdewich C, Ddungu H, Kyeyune D et al. 2016. Cerebral oximetry in Ugandan children with severe anemia: clinical categories and response to transfusion. JAMA Pediatr 170:995–1002
    [Google Scholar]
  148. 148.  Adams RJ, McKie VC, Hsu L, Files B, Vichinsky E et al. 1998. Prevention of a first stroke by transfusions in children with sickle cell anemia and abnormal results on transcranial Doppler ultrasonography. N. Engl. J. Med. 339:5–11
    [Google Scholar]
  149. 149.  Meier ER, Wright EC, Miller JL 2014. Reticulocytosis and anemia are associated with an increased risk of death and stroke in the newborn cohort of the Cooperative Study of Sickle Cell Disease. Am. J. Hematol. 89:904–6
    [Google Scholar]
  150. 150.  Balkaran B, Char G, Morris JS, Thomas PW, Serjeant BE, Serjeant GR 1992. Stroke in a cohort of patients with homozygous sickle cell disease. J. Pediatr. 120:360–66
    [Google Scholar]
  151. 151.  Belisario AR, Sales RR, Toledo NE, Muniz MB, Velloso-Rodrigues C et al. 2016. Reticulocyte count is the most important predictor of acute cerebral ischemia and high-risk transcranial Doppler in a newborn cohort of 395 children with sickle cell anemia. Ann. Hematol. 95:1869–80
    [Google Scholar]
  152. 152.  Ware RE, Davis BR, Schultz WH, Brown RC, Aygun B et al. 2016. Hydroxycarbamide versus chronic transfusion for maintenance of transcranial Doppler flow velocities in children with sickle cell anaemia—TCD With Transfusions Changing to Hydroxyurea (TWiTCH): a multicentre, open-label, phase 3, non-inferiority trial. Lancet 387:661–70
    [Google Scholar]
  153. 153.  Manci EA, Hillery CA, Bodian CA, Zhang ZG, Lutty GA, Coller BS 2006. Pathology of Berkeley sickle cell mice: similarities and differences with human sickle cell disease. Blood 107:1651–58
    [Google Scholar]
  154. 154.  Paszty C, Brion CM, Manci E, Witkowska HE, Stevens ME et al. 1997. Transgenic knockout mice with exclusively human sickle hemoglobin and sickle cell disease. Science 278:876–78
    [Google Scholar]
  155. 155.  Cui MH, Suzuka SM, Branch NA, Ambadipudi K, Thangaswamy S et al. 2017. Brain neurochemical and hemodynamic findings in the NY1DD mouse model of mild sickle cell disease. NMR Biomed 30:e3692
    [Google Scholar]
  156. 156.  Hyacinth HI, Sugihara CL, Spencer TL, Archer DR, Shih AY 2017. Higher prevalence of spontaneous cerebral vasculopathy and cerebral infarcts in a mouse model of sickle cell disease. J. Cereb. Blood Flow Metab. https://doi.org/10.1177/0271678X17732275
    [Crossref]
  157. 157.  Cahill LS, Gazdzinski LM, Tsui AK, Zhou YQ, Portnoy S et al. 2017. Functional and anatomical evidence of cerebral tissue hypoxia in young sickle cell anemia mice. J. Cereb. Blood Flow Metab. 37:994–1005
    [Google Scholar]
  158. 158.  Hillery CA, Panepinto JA 2004. Pathophysiology of stroke in sickle cell disease. Microcirculation 11:195–208
    [Google Scholar]
  159. 159.  Kato GJ, Hsieh M, Machado R, Taylor J VI, Little J et al. 2006. Cerebrovascular disease associated with sickle cell pulmonary hypertension. Am. J. Hematol. 81:503–10
    [Google Scholar]
  160. 160.  Bernaudin F, Verlhac S, Chevret S, Torres M, Coic L et al. 2008. G6PD deficiency, absence of alpha-thalassemia, and hemolytic rate at baseline are significant independent risk factors for abnormally high cerebral velocities in patients with sickle cell anemia. Blood 112:4314–17
    [Google Scholar]
  161. 161.  Kwiatkowski JL, Yim E, Miller S, Adams RJ, STOP 2 Study Investig 2011. Effect of transfusion therapy on transcranial Doppler ultrasonography velocities in children with sickle cell disease. Pediatr. Blood Cancer 56:777–82
    [Google Scholar]
  162. 162.  Adams RJ, Brambilla D, Optim. Prim. Stroke Prev. Sickle Cell Anemia STOP 2 Trial Investig. 2005. Discontinuing prophylactic transfusions used to prevent stroke in sickle cell disease. N. Engl. J. Med. 353:2769–78
    [Google Scholar]
  163. 163.  Wang WC, Pavlakis SG, Helton KJ, McKinstry RC, Casella JF et al. 2008. MRI abnormalities of the brain in one-year-old children with sickle cell anemia. Pediatr. Blood Cancer 51:643–46
    [Google Scholar]
  164. 164.  Bernaudin F, Verlhac S, Arnaud C, Kamdem A, Chevret S et al. 2011. Impact of early transcranial Doppler screening and intensive therapy on cerebral vasculopathy outcome in a newborn sickle cell anemia cohort. Blood 117:1130–40; quiz 1436
    [Google Scholar]
  165. 165.  Calvet D, Tuilier T, Mele N, Turc G, Habibi A et al. 2017. Low fetal hemoglobin percentage is associated with silent brain lesions in adults with homozygous sickle cell disease. Blood Adv 1:2503–9
    [Google Scholar]
  166. 166.  Kassim AA, Pruthi S, Day M, Rodeghier M, Gindville MC et al. 2016. Silent cerebral infarcts and cerebral aneurysms are prevalent in adults with sickle cell anemia. Blood 127:2038–40
    [Google Scholar]
  167. 167.  Casella JF, King AA, Barton B, White DA, Noetzel MJ et al. 2010. Design of the silent cerebral infarct transfusion (SIT) trial. Pediatr. Hematol. Oncol. 27:69–89
    [Google Scholar]
  168. 168.  DeBaun MR, Schatz J, Siegel MJ, Koby M, Craft S et al. 1998. Cognitive screening examinations for silent cerebral infarcts in sickle cell disease. Neurology 50:1678–82
    [Google Scholar]
  169. 169.  King AA, Rodeghier MJ, Panepinto JA, Strouse JJ, Casella JF et al. 2014. Silent cerebral infarction, income, and grade retention among students with sickle cell anemia. Am. J. Hematol. 89:E188–92
    [Google Scholar]
  170. 170.  Schatz J, Brown RT, Pascual JM, Hsu L, DeBaun MR 2001. Poor school and cognitive functioning with silent cerebral infarcts and sickle cell disease. Neurology 56:1109–11
    [Google Scholar]
  171. 171.  van der Land V, Hijmans CT, de Ruiter M, Mutsaerts HJ, Cnossen MH et al. 2015. Volume of white matter hyperintensities is an independent predictor of intelligence quotient and processing speed in children with sickle cell disease. Br. J. Haematol. 168:553–56
    [Google Scholar]
  172. 172.  DeBaun MR, Gordon M, McKinstry RC, Noetzel MJ, White DA et al. 2014. Controlled trial of transfusions for silent cerebral infarcts in sickle cell anemia. N. Engl. J. Med. 371:699–710
    [Google Scholar]
  173. 173.  Miller ST, Macklin EA, Pegelow CH, Kinney TR, Sleeper LA et al. 2001. Silent infarction as a risk factor for overt stroke in children with sickle cell anemia: a report from the Cooperative Study of Sickle Cell Disease. J. Pediatr. 139:385–90
    [Google Scholar]
  174. 174.  Koshy M, Thomas C, Goodwin J 1990. Vascular lesions in the central nervous system in sickle cell disease (neuropathology). J. Assoc. Acad. Minor. Phys. 1:71–78
    [Google Scholar]
  175. 175.  Novelli EM, Sarles CE, Aizenstein HJ, Ibrahim TS, Butters MA et al. 2015. Brain venular pattern by 7T MRI correlates with memory and haemoglobin in sickle cell anaemia. Psychiatry Res 233:18–22
    [Google Scholar]
  176. 176.  Winchell AM, Taylor BA, Song R, Loeffler RB, Grundlehner P et al. 2014. Evaluation of SWI in children with sickle cell disease. AJNR Am. J. Neuroradiol. 35:1016–21
    [Google Scholar]
  177. 177.  Steen RG, Fineberg-Buchner C, Hankins G, Weiss L, Prifitera A, Mulhern RK 2005. Cognitive deficits in children with sickle cell disease. J. Child Neurol. 20:102–7
    [Google Scholar]
  178. 178.  Vichinsky EP, Neumayr LD, Gold JI, Weiner MW, Rule RR et al. 2010. Neuropsychological dysfunction and neuroimaging abnormalities in neurologically intact adults with sickle cell anemia. JAMA 303:1823–31
    [Google Scholar]
  179. 179.  van der Land V, Zwanenburg JJ, Fijnvandraat K, Biemond BJ, Hendrikse J et al. 2015. Cerebral lesions on 7 tesla MRI in patients with sickle cell anemia. Cerebrovasc. Dis. 39:181–89
    [Google Scholar]
  180. 180.  Dobson SR, Holden KR, Nietert PJ, Cure JK, Laver JH et al. 2002. Moyamoya syndrome in childhood sickle cell disease: a predictive factor for recurrent cerebrovascular events. Blood 99:3144–50
    [Google Scholar]
  181. 181.  Birkeland P, Gardner K, Kesse-Adu R, Davies J, Lauritsen J et al. 2016. Intracranial aneurysms in sickle-cell disease are associated with the hemoglobin SS genotype but not with Moyamoya syndrome. Stroke 47:1710–13
    [Google Scholar]
  182. 182.  Nabavizadeh SA, Vossough A, Ichord RN, Kwiatkowski J, Pukenas BA et al. 2016. Intracranial aneurysms in sickle cell anemia: clinical and imaging findings. J. Neurointerv. Surg. 8:434–40
    [Google Scholar]
  183. 183.  Kato GJ, Hebbel RP, Steinberg MH, Gladwin MT 2009. Vasculopathy in sickle cell disease: biology, pathophysiology, genetics, translational medicine, and new research directions. Am. J. Hematol. 84:618–25
    [Google Scholar]
  184. 184.  Lai YC, Potoka KC, Champion HC, Mora AL, Gladwin MT 2014. Pulmonary arterial hypertension: the clinical syndrome. Circ. Res. 115:115–30
    [Google Scholar]
  185. 185.  Hoeper MM, Bogaard HJ, Condliffe R, Frantz R, Khanna D et al. 2013. Definitions and diagnosis of pulmonary hypertension. J. Am. Coll. Cardiol. 62:D42–50
    [Google Scholar]
  186. 186.  Maron BA, Brittain EL, Choudhary G, Gladwin MT 2018. Redefining pulmonary hypertension. Lancet Respir. Med. 6:168–70
    [Google Scholar]
  187. 187.  Maron BA, Wertheim BM, Gladwin MT 2018. Under pressure to clarify pulmonary hypertension clinical risk. Am. J. Respir. Crit. Care Med. 197:423–26
    [Google Scholar]
  188. 188.  De Castro LM, Jonassaint JC, Graham FL, Ashley-Koch A, Telen MJ 2008. Pulmonary hypertension associated with sickle cell disease: clinical and laboratory endpoints and disease outcomes. Am. J. Hematol. 83:19–25
    [Google Scholar]
  189. 189.  Lorch D, Spevack D, Little J 2011. An elevated estimated pulmonary arterial systolic pressure, whenever measured, is associated with excess mortality in adults with sickle cell disease. Acta Haematol 125:225–29
    [Google Scholar]
  190. 190.  Klings ES, Machado RF, Barst RJ, Morris CR, Mubarak KK et al. 2014. An official American Thoracic Society clinical practice guideline: diagnosis, risk stratification, and management of pulmonary hypertension of sickle cell disease. Am. J. Respir. Crit. Care Med. 189:727–40
    [Google Scholar]
  191. 191.  Caughey MC, Poole C, Ataga KI, Hinderliter AL 2015. Estimated pulmonary artery systolic pressure and sickle cell disease: a meta-analysis and systematic review. Br. J. Haematol. 170:416–24
    [Google Scholar]
  192. 192.  Damy T, Bodez D, Habibi A, Guellich A, Rappeneau S et al. 2016. Haematological determinants of cardiac involvement in adults with sickle cell disease. Eur. Heart J. 37:1158–67
    [Google Scholar]
  193. 193.  Machado RF, Anthi A, Steinberg MH, Bonds D, Sachdev V et al. 2006. N-terminal pro-brain natriuretic peptide levels and risk of death in sickle cell disease. JAMA 296:310–18
    [Google Scholar]
  194. 194.  Mehari A, Gladwin MT, Tian X, Machado RF, Kato GJ 2012. Mortality in adults with sickle cell disease and pulmonary hypertension. JAMA 307:1254–56
    [Google Scholar]
  195. 195.  Parent F, Bachir D, Inamo J, Lionnet F, Driss F et al. 2011. A hemodynamic study of pulmonary hypertension in sickle cell disease. N. Engl. J. Med. 365:44–53
    [Google Scholar]
  196. 196.  Fonseca GH, Souza R, Salemi VM, Jardim CV, Gualandro SF 2012. Pulmonary hypertension diagnosed by right heart catheterisation in sickle cell disease. Eur. Respir. J. 39:112–18
    [Google Scholar]
  197. 197.  Bakeer N, James J, Roy S, Wansapura J, Shanmukhappa SK et al. 2016. Sickle cell anemia mice develop a unique cardiomyopathy with restrictive physiology. PNAS 113:E5182–91
    [Google Scholar]
  198. 198.  Niss O, Quinn CT, Lane A, Daily J, Khoury PR et al. 2016. Cardiomyopathy with restrictive physiology in sickle cell disease. JACC Cardiovasc. Imaging 9:243–52
    [Google Scholar]
  199. 199.  Niss O, Fleck R, Makue F, Alsaied T, Desai P et al. 2017. Association between diffuse myocardial fibrosis and diastolic dysfunction in sickle cell anemia. Blood 130:205–13
    [Google Scholar]
  200. 200.  Ingoglia G, Sag CM, Rex N, De Franceschi L, Vinchi F et al. 2017. Hemopexin counteracts systolic dysfunction induced by heme-driven oxidative stress. Free Radic. Biol. Med. 108:452–64
    [Google Scholar]
  201. 201.  Guasch A, Navarrete J, Nass K, Zayas CF 2006. Glomerular involvement in adults with sickle cell hemoglobinopathies: prevalence and clinical correlates of progressive renal failure. J. Am. Soc. Nephrol. 17:2228–35
    [Google Scholar]
  202. 202.  Powars DR, Chan LS, Hiti A, Ramicone E, Johnson C 2005. Outcome of sickle cell anemia: a 4-decade observational study of 1056 patients. Medicine 84:363–76
    [Google Scholar]
  203. 203.  da Silva GB Jr, Libório AB, Daher Ede F 2011. New insights on pathophysiology, clinical manifestations, diagnosis, and treatment of sickle cell nephropathy. Ann. Hematol 90:1371–79
    [Google Scholar]
  204. 204.  Charache S, Scott JC, Charache P 1979. “Acute chest syndrome” in adults with sickle cell anemia: microbiology, treatment, and prevention. Arch. Intern. Med. 139:67–69
    [Google Scholar]
  205. 205.  Charache S, Terrin ML, Moore RD, Dover GJ, Barton FB et al. 1995. Effect of hydroxyurea on the frequency of painful crises in sickle cell anemia. Investigators of the Multicenter Study of Hydroxyurea in Sickle Cell Anemia. N. Engl. J. Med. 332:1317–22
    [Google Scholar]
  206. 206.  Mekontso Dessap A, Deux JF, Habibi A, Abidi N, Godeau B et al. 2014. Lung imaging during acute chest syndrome in sickle cell disease: computed tomography patterns and diagnostic accuracy of bedside chest radiograph. Thorax 69:144–51
    [Google Scholar]
  207. 207.  Matthay MA, Ware LB, Zimmerman GA 2012. The acute respiratory distress syndrome. J. Clin. Invest. 122:2731–40
    [Google Scholar]
  208. 208.  Vichinsky EP, Styles LA, Colangelo LH, Wright EC, Castro O, Nickerson B 1997. Acute chest syndrome in sickle cell disease: clinical presentation and course. Cooperative Study of Sickle Cell Disease. Blood 89:1787–92
    [Google Scholar]
  209. 209.  Leong MA, Dampier C, Varlotta L, Allen JL 1997. Airway hyperreactivity in children with sickle cell disease. J. Pediatr. 131:278–83
    [Google Scholar]
  210. 210.  DeBaun MR, Strunk RC 2016. The intersection between asthma and acute chest syndrome in children with sickle-cell anaemia. Lancet 387:2545–53
    [Google Scholar]
  211. 211.  Willen SM, Rodeghier M, Strunk RC, Bacharier LB, Rosen CL et al. 2018. Aeroallergen sensitization predicts acute chest syndrome in children with sickle cell anaemia. Br. J. Haematol. 180:571–77
    [Google Scholar]
  212. 212.  Vichinsky E, Williams R, Das M, Earles AN, Lewis N et al. 1994. Pulmonary fat embolism: a distinct cause of severe acute chest syndrome in sickle cell anemia. Blood 83:3107–12
    [Google Scholar]
  213. 213.  Styles LA, Aarsman AJ, Vichinsky EP, Kuypers FA 2000. Secretory phospholipase A(2) predicts impending acute chest syndrome in sickle cell disease. Blood 96:3276–78
    [Google Scholar]
  214. 214.  Godeau B, Schaeffer A, Bachir D, Fleury-Feith J, Galacteros F et al. 1996. Bronchoalveolar lavage in adult sickle cell patients with acute chest syndrome: value for diagnostic assessment of fat embolism. Am. J. Respir. Crit. Care Med. 153:1691–96
    [Google Scholar]
  215. 215.  Castro O 1996. Systemic fat embolism and pulmonary hypertension in sickle cell disease. Hematol. Oncol. Clin. North Am. 10:1289–303
    [Google Scholar]
  216. 216.  Adisa OA, Hu Y, Ghosh S, Aryee D, Osunkwo I, Ofori-Acquah SF 2013. Association between plasma free haem and incidence of vaso-occlusive episodes and acute chest syndrome in children with sickle cell disease. Br. J. Haematol. 162:702–5
    [Google Scholar]
  217. 217.  Stankovic Stojanovic K, Steichen O, Lefevre G, Bachmeyer C, Avellino V et al. 2012. High lactate dehydrogenase levels at admission for painful vaso-occlusive crisis is associated with severe outcome in adult SCD patients. Clin. Biochem. 45:1578–82
    [Google Scholar]
  218. 218.  Anea CB, Lyon M, Lee IA, Gonzales JN, Adeyemi A et al. 2016. Pulmonary platelet thrombi and vascular pathology in acute chest syndrome in patients with sickle cell disease. Am. J. Hematol. 91:173–78
    [Google Scholar]
  219. 219.  Mekontso Dessap A, Deux JF, Abidi N, Lavenu-Bombled C, Melica G et al. 2011. Pulmonary artery thrombosis during acute chest syndrome in sickle cell disease. Am. J. Respir. Crit. Care Med. 184:1022–29
    [Google Scholar]
  220. 220.  Rucknagel DL 2001. The role of rib infarcts in the acute chest syndrome of sickle cell diseases. Pediatr. Pathol. Mol. Med. 20:137–54
    [Google Scholar]
  221. 221.  Bellet PS, Kalinyak KA, Shukla R, Gelfand MJ, Rucknagel DL 1995. Incentive spirometry to prevent acute pulmonary complications in sickle cell diseases. N. Engl. J. Med. 333:699–703
    [Google Scholar]
  222. 222.  Knight-Madden JM, Forrester TS, Lewis NA, Greenough A 2010. The impact of recurrent acute chest syndrome on the lung function of young adults with sickle cell disease. Lung 188:499–504
    [Google Scholar]
  223. 223.  Field JJ, Burdick MD, DeBaun MR, Strieter BA, Liu L et al. 2012. The role of fibrocytes in sickle cell lung disease. PLOS ONE 7:e33702
    [Google Scholar]
  224. 224.  Mehrad B, Burdick MD, Wandersee NJ, Shahir KS, Zhang L et al. 2017. Circulating fibrocytes as biomarkers of impaired lung function in adults with sickle cell disease. Blood Adv 1:2217–24
    [Google Scholar]
/content/journals/10.1146/annurev-pathmechdis-012418-012838
Loading
/content/journals/10.1146/annurev-pathmechdis-012418-012838
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error