Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-m9kch Total loading time: 0 Render date: 2024-05-31T06:58:31.728Z Has data issue: false hasContentIssue false

Section I - Developmental Hematology

Published online by Cambridge University Press:  30 January 2021

Pedro A. de Alarcón
Affiliation:
University of Illinois College of Medicine
Eric J. Werner
Affiliation:
Children's Hospital of the King's Daughters
Robert D. Christensen
Affiliation:
University of Utah
Martha C. Sola-Visner
Affiliation:
Harvard University, Massachusetts
Get access

Summary

Ancient concepts of the blood were described by Hippocrates and Galen 2000 years ago in their doctrine of “humors.” It was believed that the body was made up of four humors – blood, phlegm, black bile, and yellow bile – and that these four components had the qualities of heat (hot-blooded!), cold, moist, and dry. The Galenic concept of the blood prevailed through the Middle Ages. Health or disease were a result of an imbalance, between these humors. This was the basis of the practice of therapeutic bloodletting (which, fortunately, was performed infrequently on children) through the mid nineteenth century as a way to rid the body of the imbalance of humors believed to cause a wide variety of diseases.

Type
Chapter
Information
Neonatal Hematology
Pathogenesis, Diagnosis, and Management of Hematologic Problems
, pp. 1 - 42
Publisher: Cambridge University Press
Print publication year: 2021

Access options

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

References

References

Wintrobe, MM (ed.). Blood, Pure and Eloquent (New York: McGraw-Hill, 1980), pp. 131.Google Scholar
Leeuwenhoek, A. Microscopical observations concerning blood, milk, bones, the brain, cuticula and spittle. Philos Trans (London) 1674;9:121–8.Google Scholar
Hewson, W. On the figure and configuration of the red particles of the blood, commonly called the red globules. Philos Trans 1963;63(part 2):303–23.Google Scholar
Bizzozero, G. Uber einen neuen Formbestandtheil des Blutes und dessen Rolle bei der Thrombose und der Blutgerinnung. Virch Archiv Pathol Anat Physiol 1882; 90:261332.Google Scholar
Osler, W. An account of certain organisms occurring in the liquor sanguinis. Proc R Soc Lond 1874;22:391–8.Google Scholar
Ehrlich, P. Beitrag zur Kenntnis der Amilinfarbungen und ihrer Verwendung in der mikroscopischen Technik. Arch Mikr Anat 1877;13:263–77.Google Scholar
Wintrobe, MM. A simple and accurate hematocrit. J Lab Clin Med 1929;15:287–9.Google Scholar
Wintrobe, MM. Anemia: Classification and treatment on the basis of differences in the average volume and hemoglobin content of the red corpuscles. Arch Intern Med 1934;54:256–80.CrossRefGoogle Scholar
Dewees, WP. A Treatise on the Physical and Medical Treatment of Children (Philadelphia, PA: B.C. Carey & L. Lea, 1825).Google Scholar
Smith, JL. A Treatise on the Diseases of Infancy and Childhood (Philadelphia PA: H.C. Lea, 1869).Google Scholar
Holt, LE. The Diseases of Infancy and Childhood (New York: D. Appleton and Co, 1897).CrossRefGoogle Scholar
Lucas, WP, Dearing, BF, Hoobler, HR, Cox, A, Smythe, F. Blood studies in the new-born: Morphological chemical coagulation, urobilin and bilirubin. Am J Dis Child 1921;22:524–58.Google Scholar
Lippman, HS. A morphologic and quantitative study of the blood corpuscles in the new-born period. Am J Dis Child 1924;27:473526.Google Scholar
Mattoth, Y, Zaizov, R, Varsano, L. Postnatal changes in some red cell parameters. Acta Paediatr 1971;60:317–23.Google Scholar
Korber, E. Inaugural dissertation; Dorpet, 1866. Cited by Bischoff H. Untersuchungen uberdie Resistenz des Hamoglobins des menschenblutes mit besonderer beruchsichtigung des Sauglingalters. Z Gesante Exp Med 1926;48:472–89.Google Scholar
Singer, K, Chernoff, AI, Singer, L. Studies on abnormal hemoglobins 1. Their demonstration in sickle cell anemia and other hematologic disorders by means of alkali denaturation. Blood 1951;6:413–28.Google Scholar
Apt, L, Downey, WS. Melena neonatorum: the swallowed blood syndrome; a simple test for the differentiation of adult and fetal hemoglobin in bloody stools. J Pediatr 1955;47:612.Google Scholar
Kleihauer, E, Braun, H, Betke, K. Demonstration von fetalen Haemoglobin in den Erythrocyten eines Blutaustrichs. Klin Wochenschr 1957;35:637–8.CrossRefGoogle Scholar
Rhinesmith, HS, Schroeder, WA, Pauling, LA. A quantitative study of hydrolysis of human dinitrophenyl (DNP) globin: the number and kind of polypeptide chains in normal adult human hemoglobin. J Am Chem Soc 1957;79:4682–6.Google Scholar
Schroeder, WA. N-terminal residues of human fetal hemoglobin. J Am Chem Soc 1958;80:1521.Google Scholar
Bensch, R, Bensch, RE, Yu, CI. Reciprocal binding of oxygen and diphosphoglycerate by human hemoglobins. Proc Natl Acad Sci USA 1968;59:526–32.Google Scholar
Scott, RB. Screening for sickle cell in newborn infants. Am J Dis Child 1948;75:842–6.Google Scholar
Pearson, HA, O’Brien, RT, McIntosh, S, Aspenes, GT, Yang, MM. Routine screening of umbilical cord blood for sickle cell diseases. J Am Med Assoc 1974;227:420–2.Google Scholar
Benz, EJ, Forget, BG. The molecular genetics of the thalassemia syndromes. Prog Hematol 1975;129:107–55.Google Scholar
Singer, ST, Styles, L, Bojanski, J, Vinchinsky, E. Changing outcomes of homozygous alpha-thalassemia: cautious optimism. J Pediatr Hematol Oncol 2000;22:539–42.Google Scholar
Oski, FA, Naiman, JL. Hematologic Problems in the Newborn (Philadelphia, PA: W.B. Saunders Co., 1966).Google Scholar
Diamond, LK, Forward. In Miller, D, Pearson, HA, McMillan, CW, eds. Smith’s Blood Diseases of Infancy and Childhood (St. Louis, MO: C.V. Mosby Co., 1978).Google Scholar
Diamond, LK, Blackfan, ED, Baty, JM. Erythroblastosis fetalis, and its association with universal edema of the fetus, icterus gravis neonatorum and anemia of the newborn. J Pediatr 1932;1:269309.CrossRefGoogle Scholar
Darrow, RR. Icterus gravis neonatorum: an examination of etiologic considerations. Arch Pathol 1938;25:378417.Google Scholar
Landsteiner, K, Weiner, P. An agglutinable factor in human blood recognized by human sera for rhesus blood. Proc Soc Exp Biol Med 1940;43:223.Google Scholar
Landsteiner, K. Uber Agglutinationsercheiunungen normalen menschlichen Blutes. Wein Klin Wocheschr 1901;14:1132–4.Google Scholar
Levine, P, Katzin, EM, Burnham, L. Isoimmunization in pregnancy; its possible bearing on the etiology of erythroblastosis fetalis. J Am Med Assoc 1941;116:825–8.CrossRefGoogle Scholar
Levine, P, Burnham, L. The role of isoimmunization in the pathogenesis of erythroblastosis fetalis. Am J Obstet Gyneocol 1941;42:825–7.Google Scholar
Hart, AP. Familial icterus gravis of the newborn and its treatment. Can Med Assoc J 1925;15:1008–19.Google Scholar
Wallerstein, H. Treatment of severe erythroblastosis by simultaneous removal and replacement of the blood of the new born infant. Science 1946;103:583–4.Google Scholar
Weiner, AS, Wexler, IB. The use of heparin in performing exchange transfusions in newborn infants. J Lab Clin Med 1946;31:1016–19.Google Scholar
Diamond, LK. Replacement transfusion as a treatment of erythoblastosis fetalis. Pediatrics 1948;2:520–4.Google Scholar
Allen, FH, Diamond, LK. Erythroblastosis Fetalis (Boston, MA: Little, Brown Co., 1957).Google Scholar
Weiner, AS. Diagnosis and treatment of anemia of the newborn caused by occult placental hemorrhage. Am J Obstet Gynecol 1948;56:717–22.Google Scholar
Chown, B. Anemia from bleeding of the fetus into the mother’s circulation: proof of bleeding. Lancet 1954;1:1213–15.Google Scholar
Pearson, HA, Diamond, LK. Fetomaternal transfusion. Am J Dis Child 1959;97:267–73.Google Scholar
Liley, AW. Liquor amni analysis in the management of the pregnancy complicated by rhesus sensitization. Am J Obst Gynecol 1961;82:1359–70.Google Scholar
Liley, AW. Intrauterine transfusion of foetus in haemolytic disease. Br Med J 1963;2:1107–11.CrossRefGoogle ScholarPubMed
Clarke, CA. Prevention of Rh hemolytic disease. Br Med J 1967;4:712.Google Scholar
Freda, VJ, Gorman, JG, Pollack, WI. Prevention of Rh immunization. J Am Med Assoc 1967;199:390–4.Google Scholar
Ross, ME, Waldron, PE, Cashore, WJ, de Alarcón, PA. Hemolytic disease of the fetus and newborn. In de Alarcón, PA, Werner, EJ, Christensen, RD eds. Neonatal Hematology 2nd ed. (Cambridge: Cambridge University Press, 2013), pp. 8081.Google Scholar
Garrison, EH. An Introduction to the History of Medicine, 4th ed. (Philadelphia, PA: Saunders, 1929).Google Scholar
Quick, AJ. The Hemorrhagic Diseases and the Physiology of Hemostasis (Springfield: IL: C.C. Thomas, 1942).Google Scholar
Townsend, CW. The haemorrhagic disease of the newborn. Arch Pediatr 1894;11:559–65.Google Scholar
Whipple, GH. Hemorrhagic disease. Arch Intern Med 1912;9:363–99.Google Scholar
Brinkhaus, KM, Smith, HP, Warner, ED. Plasma protein level in normal infancy and in hemorrhagic disease of the newborn. Am J Med Sci 1937;193:475–80.Google Scholar
Lambert, SW. Melena neonatorum with report of a case cured by transfusion. Med Record 1908;73:885–7.Google Scholar
Sidbury, JB. Transfusion through the umbilical vein in hemorrhage of the newborn. Am J Dis Child 1923;25:290–6.CrossRefGoogle Scholar
Zuelzer, WW. Pediatric hematology in historical perspective. In Nathan, DG, Orkin, SH, eds. Nathan and Oski’s Hematology of Infancy and Childhood, 5th ed. (Philadelphia, PA: W.R. Saunders, 1998).Google Scholar
Dam, H, Dyggve, H, Larsen, H. Cholesterolstoffwechsel in Huhneriern und Huhnchen. Biochem J 1929;215:475–92.Google Scholar
Dam, H, Dyggve, H, Larsen, H, Schonheyder, F, Tage-Hansen, E. Studies on the mode of action of Vitamin K. Biochem J 1936;30:1275–9.CrossRefGoogle ScholarPubMed
Brinkhaus, KM, Smith, HP, Warner, ED. Plasma protein level in normal infancy and in hemorrhagic disease of the newborn. Am J Med Sci 1937;193:475–80.Google Scholar
Dam, H, Dyggve, H, Larsen, H. The relationship of vitamin K deficiency to hemorrhagic disease of the newborn. Adv Pediatr 1952;5:129–53.Google Scholar
Waddell, WW, Jr., Guerry, DP III, Bray, WE, Waddell Kelley, OR. Possible effects of vitamin K on prothrombin and clotting time in the newly born infant. Proc Soc Exp Biol Med 1937;40:432–4.Google Scholar
Almquist, HJ, Klose, AA. Synthetic and natural antihemorrhagic compounds. Am J Chem Soc 1939;61:2557–8.Google Scholar
Committee on Nutrition of the American Academy of Pediatrics. Vitamin K in the newborn. Pediatrics 1961;28:501–7.Google Scholar
Dam, H, Glavind, J, Larsen, H, Plum, P. Investigations into the cause of physiological hypoprothrombinemia in newborn children. IV. The vitamin K content of women’s milk and cow’s milk. Acta Med Scand 1942;112:210–16.Google Scholar
Sutherland, JM, Glueck, HL, Gleser, G. Hemorrhagic disease of the newborn: breast feeding as a necessary factor in the pathogenesis. Am J Dis Child 1967;113:524–33.Google Scholar
Stentlo, J, Fernlund, P, Egan, W, Raestorff, P. Vitamin K dependent modifications of glutamic acid residues in prothrombin. Proc Natl Acad Sci USA 1974;71:2730–3.Google Scholar
Pearson, HA. Life span of the fetal red blood cell. J Pediatr 1967;70:166171.Google Scholar
O’Brien, RT, Pearson, HA. Physiological anemia of the new-born infant. J. Pediatr 1971;79:132–8.Google Scholar
Gallagher, PG, Ehrenkranz, RA. Erythropoietin therapy for anemia of prematurity. Clin Perinatol 1993;20:16991.Google Scholar
Von Kohorn, I, Ehrenkranz, RA. Anemia in the preterm infant: Erythropoietin versus erythrocyte transfusion – It’s not that simple. Clin Perinatol 2009;36:111–23.Google Scholar

References

Purton, LE, Scadden, DT. Limiting factors in murine hematopoietic stem cell assays. Cell Stem Cell 2007;1:263–70.Google Scholar
Wintrobe, M. Milestones on the path of progress. In Wintrobe, M. ed. Blood, Pure, and Eloquent (New York: McGraw-Hill, 1980), p. 1.Google Scholar
Donne, A. De I-oringe des gloubles du sang, de leur mode de formation et de leur fin. C R Seances Acad Sci 1842;366–8.Google Scholar
Bizzozero, G. Uber einen Formbestandteil des Blutes und dessen Rolle bei der Thrombose und der Blutgerinnung. Virch Archiv Pathol Anat Physiol 1882; 90:261332.CrossRefGoogle Scholar
Jacobson, LO, Marks, EK, et al. The role of the spleen in radiation injury. Proc Soc Exp Biol Med 1949;70:740–2.Google Scholar
Ford, CE, Hamerton, JL, Barnes, DW, Loutit, JF. Cytological identification of radiation-chimaeras. Nature 1956;177:452–4.Google Scholar
Becker, AJ, McCulloch, EA, Till, JE. Cytological demonstration of the clonal nature of spleen colonies derived from transplanted mouse marrow cells. Nature 1963;197:452–4.Google Scholar
Pluznik, DH, Sachs, L. The cloning of normal “mast” cells in tissue culture. J Cell Physiol 1965;66:319–24.Google ScholarPubMed
Bradley, TR, Metcalf, D. The growth of mouse bone marrow cells in vitro. Aust J Exp Biol Med Sci 1966;44:287–99.Google Scholar
Stephenson, JR, Axelrad, AA, McLeod, DL, Shreeve, MM. Induction of colonies of hemoglobin-synthesizing cells by erythropoietin in vitro. Proc Natl Acad Sci USA 1971;68:1542–6.Google Scholar
Spangrude, GJ. Biological and clinical aspects of hematopoietic stem cells. Annu Rev Med 1994;45:93104.CrossRefGoogle ScholarPubMed
Harrison, . Competitive repopulation: a new assay for long-term stem cell functional capacity. Blood 1980;55:7781.Google Scholar
Sutherland, HJ, Eaves, CJ, Eaves, AC, Dragowska, W, Lansdorp, PM. Characterization and partial purification of human marrow cells capable of initiating long-term hematopoiesis in vitro. Blood 1989;74:1563–70.CrossRefGoogle ScholarPubMed
Orlic, D, Bodine, DM. What defines a pluripotent hematopoietic stem cell (PHSC): will the real PHSC please stand up! Blood 1994;84:3991–4.CrossRefGoogle ScholarPubMed
Dexter, TM, Allen, TD, Lajtha, LG. Conditions controlling the proliferation of haemopoietic stem cells in vitro. J Cell Physiol 1977;91:335–44.Google Scholar
Ploemacher, RE, van der Sluijs, JP, Voerman, JS, Brons, NH. An in vitro limiting-dilution assay of long-term repopulating hematopoietic stem cells in the mouse. Blood 1989;74:2755–63.Google Scholar
Verfaillie, CM, Miller, JS. A novel single-cell proliferation assay shows that long-term culture-initiating cell (LTC-IC) maintenance over time results from the extensive proliferation of a small fraction of LTC-IC. Blood 1995;86:2137–45.Google Scholar
Zanjani, ED. Pallavicini, MG, Ascensao, JL, et al. Engraftment and long-term expression of human fetal hemopoietic stem cells in sheep following transplantation in utero. J Clin Invest 1992:89;1178–88.Google Scholar
Shultz, LD. Schweitzer, PA, Christianson, SW, et al. Multiple defects in innate and adaptive immunologic function in NOD/LtSz-scid mice. J Immunol 1995;154:180–91.Google Scholar
Mezquita, P, Beard, BC, Kiem, H-P. NOD/SCID repopulating cells contribute only to short-term repopulation in the baboon. Gene Ther 2008;15:1460–2.Google Scholar
Park, YS, Lee, CH, Kim, JW, Shin, S, Park, CM. Differentiation of hepatocellular carcinoma from its various mimickers in liver magnetic resonance imaging: What are the tips when using hepatocyte-specific agents? World J Gastroenterol 2016;22:284–99.Google Scholar
Okada, S, Harada, H, Ito, T, Saito, T, Suzu, S. Early development of human hematopoietic and acquired immune systems in new born NOD/Scid/Jak3null mice intrahepatic engrafted with cord blood-derived CD34+ cells. Int J Hematol 2008;88:476–82.Google Scholar
Traggiai, E. Chicha, L, Mazzucchelli, L, et al. Development of a human adaptive immune system in cord blood cell-transplanted mice. Science 2004;304:104–7.Google Scholar
Ishikawa, F, Yasukawa, M, Lyons, B, et al. Development of functional human blood and immune systems in NOD/SCID/IL2 receptor {gamma} chain(null) mice. Blood 2005;106:1565–73.Google Scholar
Bradford, GB, Williams, B, Rossi, R, Bertoncello, I. Quiescence, cycling, and turnover in the primitive hematopoietic stem cell compartment. Exp Hematol 1997;25:445–53.Google Scholar
Foudi, A, Hochedlinger, K, van Buren, D, et al. Analysis of histone 2B-GFP retention reveals slowly cycling hematopoietic stem cells. Nat Biotechnol 2009;27:8490.Google Scholar
Ogawa, M. Differentiation and proliferation of hematopoietic stem cells. Blood 1993;81:2844–53.Google Scholar
Watowich, SS, Wu, H, Socolovsky, M, et al. Cytokine receptor signal transduction and the control of hematopoietic cell development. Annu Rev Cell Dev Biol 1996;12:91128.Google Scholar
Enver, T, Pera, M, Peterson, C, Andrews, PW. Stem cell states, fates, and the rules of attraction. Cell Stem Cell 2009;4:387–97.Google Scholar
Golan, K, Kumari, A, Kollet, O, et al. Daily onset of light and darkness differentially controls hematopoietic stem cell differentiation and maintenance. Cell Stem Cell 2018;23:572–85, doi:10.1016/j.stem.2018.08.002.Google Scholar
Wang, C, Cheng, L. Gankyrin as a potential therapeutic target for cancer. Invest New Drugs 2017;35 :655–6, doi:10.1007/s10637-017-0474-8.Google Scholar
Okawa, S, Nicklas, S, Zickenrott, S, Schwamborn, JC, Del Sol, AA. Generalized gene-regulatory network model of stem cell differentiation for predicting lineage specifiers. Stem Cell Rep 2016;7:307–15.Google Scholar
Osawa, M, Hanada, K, Hamada, H, Nakauchi, H. Long-term lymphohematopoietic reconstitution by a single CD34-low/negative hematopoietic stem cell. Science 1996;273:242–5.Google Scholar
Kent, DG, Copley, MR, Benz, C, et al. Prospective isolation and molecular characterization of hematopoietic stem cells with durable self-renewal potential. Blood 2009;113:6342–50.Google Scholar
Fraser, CC, Eaves, CJ, Szilvassy, SJ, Humphries, RK. Expansion in vitro of retrovirally marked totipotent hematopoietic stem cells. Blood 1990;76:1071–6.Google Scholar
Miller, CL, Eaves, CJ. Expansion in vitro of adult murine hematopoietic stem cells with transplantable lympho-myeloid reconstituting ability. Proc Natl Acad Sci USA 1997;94:13648–53.Google Scholar
Wineman, J, Moore, K, Lemischka, I, Müller-Sieburg, C. Functional heterogeneity of the hematopoietic microenvironment: rare stromal elements maintain long-term repopulating stem cells. Blood 1996;87:4082–90.Google Scholar
Bowie, MB, Kent, DG, Copley, MR, Eaves, CJ. Steel factor responsiveness regulates the high self-renewal phenotype of fetal hematopoietic stem cells. Blood 2007;109:5043–8.CrossRefGoogle ScholarPubMed
Jollie, WP. Ultrastructural studies of protein transfer across rodent yolk sac. Placenta 1986;7:263–81.CrossRefGoogle ScholarPubMed
Zon, LI. Developmental biology of hematopoiesis. Blood 1995;86:2876–91.Google Scholar
Shalaby, F, Ho, J, Stanford, WL, et al. A requirement for Flk1 in primitive and definitive hematopoiesis and vasculogenesis. Cell 1997;89:981–90.Google Scholar
Choi, K, Kennedy, M, Kazarov, A, Papadimitriou, JC, Keller, G. A common precursor for hematopoietic and endothelial cells. Development 1998;125:725–32.Google Scholar
Kabrun, N, Buhring, HJ, Choi, K, et al. Flk-1 expression defines a population of early embryonic hematopoietic precursors. Development 1997;124:2039–48.Google Scholar
Huber, TL, Kouskoff, V, Fehling, HJ, Palis, J Keller, G. Haemangioblast commitment is initiated in the primitive streak of the mouse embryo. Nature 2004;432:625–30.Google Scholar
Teixeira, V, Arede, N, Gardner, R, Rodríguez-León, J, Tavares, AT. Targeting the hemangioblast with a novel cell type-specific enhancer. BMC Dev Biol 2011;11:76.Google Scholar
Serrado Marques, J, Teixeira, V, Jacinto, A Tavares, AT. Identification of novel hemangioblast genes in the early chick embryo. Cells 2018;7:9.Google Scholar
Shivdasani, RA, Orkin, SH. The transcriptional control of hematopoiesis. Blood 1996;87:4025–39.Google Scholar
Maximow, A. Relation of blood cells to connective tissues and endothelium. Physiol Rev 1924;4:533–63.Google Scholar
Rhodes, KE, Gekas, C, Wang, Y, et al. The emergence of hematopoietic stem cells is initiated in the placental vasculature in the absence of circulation. Cell Stem Cell 2008;2:252–63.Google Scholar
Moore, MA, Metcalf, D. Ontogeny of the haemopoietic system: yolk sac origin of in vivo and in vitro colony forming cells in the developing mouse embryo. Br J Haematol 1970;18:279–96.Google Scholar
Samokhvalov, IM, Samokhvalova, NI, Nishikawa, S. Cell tracing shows the contribution of the yolk sac to adult haematopoiesis. Nature 2007;446:1056–61.CrossRefGoogle ScholarPubMed
Mikkola, HKA, Gekas, C, Orkin, SH, Dieterlen-Lievre, F. Placenta as a site for hematopoietic stem cell development. Exp Hematol 2005;33:1048–54.CrossRefGoogle ScholarPubMed
Gekas, C, Dieterlen-Lièvre, F, Orkin, SH, Mikkola, HKA. The placenta is a niche for hematopoietic stem cells. Dev Cell 2005;8:365–75.Google Scholar
Müller, AM, Medvinsky, A, Strouboulis, J, Grosveld, F, Dzierzak, E. Development of hematopoietic stem cell activity in the mouse embryo. Immunity 1994;1:291301.CrossRefGoogle ScholarPubMed
Medvinsky, A, Dzierzak, E. Definitive hematopoiesis is autonomously initiated by the AGM region. Cell 1996;86:897906.Google Scholar
Cumano, A, Dieterlen-Lievre, F, Godin, I. Lymphoid potential, probed before circulation in mouse, is restricted to caudal intraembryonic splanchnopleura. Cell 1996;86:907–16.Google Scholar
Chen, MJ, Yokomizo, T, Zeigler, BM, Dzierzak, E, Speck, NA. Runx1 is required for the endothelial to haematopoietic cell transition but not thereafter. Nature 2009;457:887–91.Google Scholar
Bertrand, JY, Giroux, S, Golub, R, et al. Characterization of purified intraembryonic hematopoietic stem cells as a tool to define their site of origin. Proc Natl Acad Sci USA 2005;102:134–9.Google Scholar
Lis, R, Karrasch, CC, Poulos, MG, et al. Conversion of adult endothelium to immunocompetent haematopoietic stem cells. Nature 2017;545:439–45.Google Scholar
Zovein, AC, Hofmann, JJ, Lynch, M, et al. Fate tracing reveals the endothelial origin of hematopoietic stem cells. Cell Stem Cell 2008;3:625–36.Google Scholar
Adamo, L, Naveiras, O, Wenzel, PL, et al. Biomechanical forces promote embryonic haematopoiesis. Nature 2009;459:1131–5.Google Scholar
North, TE, Goessling, W, Peeters, M, et al. Hematopoietic stem cell development is dependent on blood flow. Cell 2009;137:736–48.Google Scholar
Diaz, MF, Li, N, Lee, HY, et al. Biomechanical forces promote blood development through prostaglandin E2 and the cAMP-PKA signaling axis. J Exp Med 2015;212:665–80.Google Scholar
Tavassoli, M. Embryonic and fetal hemopoiesis: an overview. Blood Cells 1991;17:269–81; discussion 282–6.Google Scholar
Rich, IN. The developmental biology of hemopoiesis: effect of growth factors on the colony formation by embryonic cells. Exp Hematol 1992;20:368–70.Google Scholar
Lux, CT, Yoshimoto, M, McGrath, K, et al. All primitive and definitive hematopoietic progenitor cells emerging before E10 in the mouse embryo are products of the yolk sac. Blood 2008;111:3435–8.Google Scholar
Enders, A, King, B. Development of the human yolk sac. In Nogales, F, ed. The Human Yolk Sac and Yolk Sac Tumors (Berlin: Springer-Verlag, 1993) pp. 3347.Google Scholar
Brent, RL, Beckman, DA, Jensen, M, Koszalka, TR. Experimental yolk sac dysfunction as a model for studying nutritional disturbances in the embryo during early organogenesis. Teratology 1990;41:405–13.Google Scholar
Pereda, J, Niimi, G. Embryonic erythropoiesis in human yolk sac: two different compartments for two different processes. Microsc Res Tech 2008;71:856–62.Google Scholar
Bremer, J. The earliest blood vessels in man. Am J Anat 1914;16:447–76.Google Scholar
Pereda, J, Monge, JI, Niimi, G. Two different pathways for the transport of primitive and definitive blood cells from the yolk sac to the embryo in humans. Microsc Res Tech 2010;73:803–9.Google Scholar
Freyer, C, Renfree, MB. The mammalian yolk sac placenta. J Exp Zoolog B Mol Dev Evol 2009;312:545–54.Google Scholar
Gilmour, J. Normal hematopoiesis in intra-uterine and neonatal life. J Path 1941;52:2555.Google Scholar
Thomas, DB, Yoffey, JM. Human foetal haemopoiesis. I. The cellular composition of foetal blood. Br J Haematol 1962;8:290–5.Google Scholar
Thomas, DB, Yoffey, JM. Human foetal haematopoiesis. Ii. Hepatic haematopoiesis in the human foetus. Br J Haematol 1964;10:1937.Google Scholar
Kelemen, E, Calvo, W, Fliedner, TM. Atlas of Human Hemopoietic Development (New York: Springer-Verlag, 1979).Google Scholar
Charbord, P, Tavian, M, Humeau, L, Péault, B. Early ontogeny of the human marrow from long bones: an immunohistochemical study of hematopoiesis and its microenvironment. Blood 1996;87:4109–19.CrossRefGoogle ScholarPubMed
Tavassoli, M. Ontogeny of hempoiesis. In Meisami, E, Timiras, P, eds. Handbook of Human Growth and Developmental Biology (Boca Raton:CRC Press, 1988), pp. 101–12.Google Scholar
Kelemen, E, Gulya, E, Vass, K. Ontogeny of human neutrophil granulocyte alkaline phosphatase. J Cell Physiol 1978;95:353–4.Google Scholar
Wolf, BC, Luevano, E, Neiman, RS. Evidence to suggest that the human fetal spleen is not a hematopoietic organ. Am J Clin Pathol 1983;80:140–4.CrossRefGoogle Scholar
Calhoun, DA, Li, Y, Braylan, RC, Christensen, RD. Assessment of the contribution of the spleen to granulocytopoiesis and erythropoiesis of the mid-gestation human fetus. Early Hum Dev 1996;46:217–27.CrossRefGoogle ScholarPubMed
Forestier, F, Daffos, F, Galacteros, F, et al. Hematological values of 163 normal fetuses between 18 and 30 weeks of gestation. Pediatr Res 1986;20:342–6.Google Scholar
Migliaccio, G, Migliaccio, AR, Petti, S, et al. Human embryonic hemopoiesis. Kinetics of progenitors and precursors underlying the yolk sac–liver transition. J Clin Invest 1986;78:5160.Google Scholar
Porcellini, A, Manna, A, Manna, M. Ontogeny of granulocyte-macrophage progenitor cells in the human fetus. Int J Cell Cloning 1983;1:92104.Google Scholar
Peschle, C, Migliaccio, AR, Migliaccio, G, et al. Embryonic–fetal Hb switch in humans: studies on erythroid bursts generated by embryonic progenitors from yolk sac and liver. Proc Natl Acad Sci USA 1984;81:2416–20.Google Scholar
Park, CY, Majeti, R, Weissman, IL. In vivo evaluation of human hematopoiesis through xenotransplantation of purified hematopoietic stem cells from umbilical cord blood. Nat Protoc 2008;3:1932–40.Google Scholar
Barclay, A Birkeland, M, Brown, M, et al. The Leucocyte Antigen Facts Book (London: Academic Press, 1993).Google Scholar
Krause, DS, Fackler, MJ, Civin, CI, May, WS. CD34: structure, biology, and clinical utility. Blood 1996;87:113.Google Scholar
Gilles, JM, Divon, MY, Bentolila, E, et al. Immunophenotypic characterization of human fetal liver hematopoietic stem cells during the midtrimester of gestation. Am J Obstet Gynecol 1997;177:619–25.CrossRefGoogle ScholarPubMed
Muench, MO, Roncarolo, MG, Namikawa, R. Phenotypic and functional evidence for the expression of CD4 by hematopoietic stem cells isolated from human fetal liver. Blood 1997;89:1364–75.Google Scholar
Emerson, SG. The regulation of hematopoiesis in the human fetal liver. Prog Clin Biol Res 1990;352:21–8.Google Scholar
Tavian, M, Coulombel, L, Luton, D, et al. Aorta-associated CD34+ hematopoietic cells in the early human embryo. Blood 1996;87:6772.Google Scholar
Ivanovs, A, Rybtsov, S, Welch, L, et al. Highly potent human hematopoietic stem cells first emerge in the intraembryonic aorta-gonad-mesonephros region. J Exp Med 2011;208:2417–27.Google Scholar
Oberlin, E, Tavian, M, Blazsek, I, Péault, B. Blood-forming potential of vascular endothelium in the human embryo. Development 2002;129:4147–57.Google Scholar
Zambidis, ET, Oberlin, E, Tavian, M, Péault, B. Blood-forming endothelium in human ontogeny: lessons from in utero development and embryonic stem cell culture. Trends Cardiovasc Med 2006;16:95101.CrossRefGoogle ScholarPubMed
Huyhn, A, Dommergues, M, Izac, B, et al. Characterization of hematopoietic progenitors from human yolk sacs and embryos. Blood 1995;86:4474–85.Google Scholar
Nicolini, FE, Holyoake, TL, Cashman, JD, et al. Unique differentiation programs of human fetal liver stem cells shown both in vitro and in vivo in NOD/SCID mice. Blood 1999;94:2686–95.Google Scholar
Zanjani, ED, Flake, AW, Rice, H, Hedrick, M, Tavassoli, M. Long-term repopulating ability of xenogeneic transplanted human fetal liver hematopoietic stem cells in sheep. J Clin Invest 1994;93:1051–5.Google Scholar
Touraine, J. Transplantation of fetal hematopoietic and lymphopoietic cells in humans, with special reference to in utero transplantation. In Edwards, R, ed. Fetal Tissue Transplants in Medicine (Cambridge: Cambridge Univeristy Press, 1992), pp. 15576.Google Scholar
Kochupillai, V, Sharma, S, Sundaram, KR, Ahuja, RK. Hemopoietic improvement following fetal liver infusion in aplastic anemia. Eur J Haematol 1991;47:319–25.Google Scholar
Turner, CW, Yeager, AM, Waller, EK, Wingard, JR, Fleming, WH. Engraftment potential of different sources of human hematopoietic progenitor cells in BNX Mice. Blood 1996;87:3237–44.Google Scholar
Kumar, P, Defor, TE, Brunstein, C, et al. Allogeneic hematopoietic stem cell transplantation in adult acute lymphocytic leukemia: impact of donor source on survival. Biol Blood Marrow Transplant 2008;14:13941400.Google Scholar
Atsuta, Y, Suzuki, R, Nagamura-Inoue, T, et al. Disease-specific analyses of unrelated cord blood transplantation compared with unrelated bone marrow transplantation in adult patients with acute leukemia. Blood 2009;113:1631–8.Google Scholar
Sauter, C, Barker, JN. Unrelated donor umbilical cord blood transplantation for the treatment of hematologic malignancies. Curr Opin Hematol 2008;15:568–75.Google Scholar
Brunstein, CG. Umbilical cord blood transplantation for the treatment of hematologic malignancies. Cancer Control J Moffitt Cancer Cent 2011;18:222–36.Google Scholar
Chen, Y, Xu, LP, Liu, DH, et al. Comparative outcomes between cord blood transplantation and bone marrow or peripheral blood stem cell transplantation from unrelated donors in patients with hematologic malignancies: a single-institute analysis. Chin Med J (Engl ) 2013;126:2499–503.Google Scholar
McKinney-Freeman, SL, Daley, GQ. Towards hematopoietic reconstitution from embryonic stem cells: a sanguine future. Curr Opin Hematol 2007;14:343–7.Google Scholar
Vodyanik, MA, Slukvin, II. Hematoendothelial differentiation of human embryonic stem cells. Curr Protoc Cell Biol 2007; Chapter 23:Unit 23.6.Google Scholar
Tian, X, Woll, PS, Morris, JK, Linehan, JL, Kaufman, DS. Hematopoietic engraftment of human embryonic stem cell-derived cells is regulated by recipient innate immunity. Stem Cells 2006;24:1370–80.Google Scholar
Li, Z, Han, Z, Wu, JC. Transplantation of human embryonic stem cell-derived endothelial cells for vascular diseases. J Cell Biochem 2009;106:194–9.Google Scholar
Gomes, KMS, Costa, IC, Santos, JF, et al. Induced pluripotent stem cells reprogramming: Epigenetics and applications in the regenerative medicine. Rev Assoc Med Bras(1992) 2017;63:180–9.Google Scholar
van den Hurk, M, Kenis, G, Bardy, C, et al. Transcriptional and epigenetic mechanisms of cellular reprogramming to induced pluripotency. Epigenomics 2016;8:1131–49.Google Scholar
Hu, C, Li, L. Current reprogramming systems in regenerative medicine: from somatic cells to induced pluripotent stem cells. Regen Med 2016;11:105–32.Google Scholar
Sugimura, R, Jha, DK, Han, A, et al. Haematopoietic stem and progenitor cells from human pluripotent stem cells. Nature 2017;545:432–8.Google Scholar
Barcia Durán, JG, Lis, R, Lu, TM, Rafii, S. In vitro conversion of adult murine endothelial cells to hematopoietic stem cells. Nat Protoc 2018;13:2758–80.Google Scholar

References

Collins, A, Weitkamp, JH, Wynn, JL. Why are preterm newborns at increased risk of infection? Arch Dis Child Fetal Neonatal Ed 2018;103:F391F394.Google Scholar
Simon, AK, Hollander, GA, McMichael, A. Evolution of the immune system in humans from infancy to old age. Proc Biol Sci 2015;282:20143085.Google Scholar
Meissner, HC, Reef, SE, Cochi, S. Elimination of rubella from the United States: a milestone on the road to global elimination. Pediatrics 2006;117:933–5.Google Scholar
May, M, Relich, RF. A comprehensive systems biology approach to studying zika virus. PLoS One 2016;11:e0161355.Google Scholar
Mehta, R, Soares, CN, Medialdea-Carrera, R, et al. The spectrum of neurological disease associated with Zika and chikungunya viruses in adults in Rio de Janeiro, Brazil: A case series. PLoS Negl Trop Dis 2018;12:e0006212.CrossRefGoogle Scholar
Narendran, V, Visscher, MO, Abril, I, et al. Biomarkers of epidermal innate immunity in premature and full-term infants. Pediatr Res 2010;67:382–6.Google Scholar
Whitsett, JA. Review: The intersection of surfactant homeostasis and innate host defense of the lung: lessons from newborn infants. Innate Immun 2010;16:138–42.Google Scholar
Collado, MC, Cernada, M, Neu, J, et al. Factors influencing gastrointestinal tract and microbiota immune interaction in preterm infants. Pediatr Res 2015;77:726–31.Google Scholar
Tamburini, S, Shen, N, Wu, HC, et al. The microbiome in early life: implications for health outcomes. Nat Med 2016;22:713–22.Google Scholar
Cefalo, RC, Berghmans, RL, Hall, SP. The bioethics of human fetal tissue research and therapy: moral decision making of professionals. Am J Obstet Gynecol 1994;170:1219.Google Scholar
Reardon, S. Trump administration launches sweeping review of fetal-tissue research. Nature 2018;562:1617.Google Scholar
Dudley, DJ, Wiedmeier, S. The ontogeny of the immune response: perinatal perspectives. Semin Perinatol 1991;15:184–95.Google Scholar
Haddad, R, Guimiot, F, Six, E, et al. Dynamics of thymus-colonizing cells during human development. Immunity 2006;24:217–30.Google Scholar
Zlotoff, DA, Schwarz, BA, Bhandoola, A. The long road to the thymus: The generation, mobilization, and circulation of T-cell progenitors in mouse and man. Semin Immunopathol 2008;30:371–82.Google Scholar
Westra, ER, Swarts, DC, Staals, RH, et al. The CRISPRs, they are a-changin’: How prokaryotes generate adaptive immunity. Annu Rev Genet 2012;46:311–39.Google Scholar
Dishaw, LJ, Cannon, JP, Litman, GW, et al. Immune-directed support of rich microbial communities in the gut has ancient roots. Dev Comp Immunol 2014;47:3651.Google Scholar
Rohlfing, K, Stuhlmann, F, Docker, MF, et al. Convergent evolution of hemoglobin switching in jawed and jawless vertebrates. BMC Evol Biol 2016; 16:30.Google Scholar
Buchmann, K. Evolution of innate immunity: Clues from invertebrates via fish to mammals. Front Immunol 2014;5:459.Google Scholar
Beck, G, Habicht, GS. Immunity and the invertebrates. Sci Am 1996;275:60–3, 66.Google Scholar
Orkin, SH. Molecular genetics of chronic granulomatous disease. Annu Rev Immunol 1989;7:277307.Google Scholar
Segal, AW, Jones, OT, Webster, D, et al. Absence of a newly described cytochrome b from neutrophils of patients with chronic granulomatous disease. Lancet 1978;2:446–9.Google Scholar
Yoshida, LS, Saruta, F, Yoshikawa, K, et al. Mutation at histidine 338 of gp91(phox) depletes FAD and affects expression of cytochrome b558 of the human NADPH oxidase. J Biol Chem 1998;273:27879–86.Google Scholar
Sharp, JG, Crouse, DA, Purtilo, DT. Ontogeny and regulation of the immune system. Arch Pathol Lab Med 1987;111:1106–13.Google Scholar
Dishaw, LJ, Litman, GW. Invertebrate allorecognition: The origins of histocompatibility. Curr Biol 2009;19:R286–8.Google Scholar
Fagan, MB, Weissman, IL. Sequence and characterization of two HSP70 genes in the colonial protochordate Botryllus schlosseri. Immunogenetics 1996;44:134–42.Google Scholar
Fagan, MB, Weissman, IL. Linkage analysis of HSP70 genes and historecognition locus in botryllus schlosseri. Immunogenetics 1998;47:468–76.Google Scholar
Parrinello, N. Cytotoxic activity of tunicate hemocytes. Prog Mol Subcell Biol 1996;15:190217.Google Scholar
Hirano, M, Guo, P, McCurley, N, et al. Evolutionary implications of a third lymphocyte lineage in lampreys. Nature 2013;501:435–8.Google Scholar
Potts, KS, Sargeant, TJ, Markham, JF, et al. A lineage of diploid platelet-forming cells precedes polyploid megakaryocyte formation in the mouse embryo. Blood 2014; 124:2725–9.Google Scholar
Tober, J, Maijenburg, MW, Speck, NA. Taking the leap: runx1 in the formation of blood from endothelium. Curr Top Dev Biol 2016;118:113–62.Google Scholar
Bockamp, EO, McLaughlin, F, Gottgens, B, et al. Distinct mechanisms direct SCL/tal-1 expression in erythroid cells and CD34 positive primitive myeloid cells. J Biol Chem 1997;272:8781–90.Google Scholar
Endoh, M, Ogawa, M, Orkin, S, et al. SCL/tal-1-dependent process determines a competence to select the definitive hematopoietic lineage prior to endothelial differentiation. EMBO J 2002;21:6700–8.Google Scholar
Robertson, SM, Kennedy, M, Shannon, JM, et al. A transitional stage in the commitment of mesoderm to hematopoiesis requiring the transcription factor SCL/tal-1. Development 2000;127:2447–59.Google Scholar
Stanulovic, VS, Cauchy, P, Assi, SA, et al. LMO2 is required for TAL1 DNA binding activity and initiation of definitive haematopoiesis at the haemangioblast stage. Nucleic Acids Res 2017;45:9874–88.Google Scholar
Zhu, H, Traver, D, Davidson, AJ, et al. Regulation of the lmo2 promoter during hematopoietic and vascular development in zebrafish. Dev Biol 2005;281:256–69.Google Scholar
Kingsley, PD, Malik, J, Fantauzzo, KA, et al. Yolk sac-derived primitive erythroblasts enucleate during mammalian embryogenesis. Blood 2004;104:1925.Google Scholar
Palis, J. Primitive and definitive erythropoiesis in mammals. Front Physiol 2014;5:3.Google Scholar
Enzan, H. Electron microscopic studies of macrophages in early human yolk sacs. Acta Pathol Jpn 1986;36:4964.Google Scholar
Gomez Perdiguero, E, Klapproth, K, Schulz, C, et al. Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature 2015;518:547–51.Google Scholar
Beaudin, AE, Boyer, SW, Perez-Cunningham, J, et al. A transient developmental hematopoietic stem cell gives rise to innate-like B and T cells. Cell Stem Cell 2016;19:768–83.Google Scholar
Beaudin, AE, Forsberg, EC. To B1a or not to B1a: Do hematopoietic stem cells contribute to tissue-resident immune cells? Blood 2016;128:2765–9.Google Scholar
Dieterlen-Lievre, F. On the origin of haemopoietic stem cells in the avian embryo: an experimental approach. J Embryol Exp Morphol 1975;33:607–19.Google Scholar
Bertrand, JY, Giroux, S, Golub, R, et al. Characterization of purified intraembryonic hematopoietic stem cells as a tool to define their site of origin. Proc Natl Acad Sci USA 2005;102:134–9.Google Scholar
Choi, K, Kennedy, M, Kazarov, A, et al. A common precursor for hematopoietic and endothelial cells. Development 1998;125:725–32.Google Scholar
Cumano, A, Dieterlen-Lievre, F, Godin, I. Lymphoid potential, probed before circulation in mouse, is restricted to caudal intraembryonic splanchnopleura. Cell 1996;86:907–16.Google Scholar
Cumano, A, Godin, I. Ontogeny of the hematopoietic system. Annu Rev Immunol 2007; 25:745–85.Google Scholar
Tavian, M, Robin, C, Coulombel, L, et al. The human embryo, but not its yolk sac, generates lympho-myeloid stem cells: Mapping multipotent hematopoietic cell fate in intraembryonic mesoderm. Immunity 2001;15:487–95.Google Scholar
de Bruijn, M, Dzierzak, E. Runx transcription factors in the development and function of the definitive hematopoietic system. Blood 2017;129:2061–9.Google Scholar
Draper, JE, Sroczynska, P, Tsoulaki, O, et al. RUNX1B expression is highly heterogeneous and distinguishes megakaryocytic and erythroid lineage fate in adult mouse hematopoiesis. PLoS Genet 2016;12:e1005814.Google Scholar
Kamikubo, Y. Genetic compensation of RUNX family transcription factors in leukemia. Cancer Sci 2018;109:2358–63.Google Scholar
Yoder, MC. Inducing definitive hematopoiesis in a dish. Nat Biotechnol 2014;32:539–41.Google Scholar
Boiers, C, Carrelha, J, Lutteropp, M, et al. Lymphomyeloid contribution of an immune-restricted progenitor emerging prior to definitive hematopoietic stem cells. Cell Stem Cell 2013;13:535–48.Google Scholar
Gekas, C, Dieterlen-Lievre, F, Orkin, SH, et al. The placenta is a niche for hematopoietic stem cells. Dev Cell 2005;8:365–75.Google Scholar
Gordon-Keylock, S, Sobiesiak, M, Rybtsov, S, et al. Mouse extraembryonic arterial vessels harbor precursors capable of maturing into definitive HSCs. Blood 2013;122:2338–45.Google Scholar
Lin, Y, Yoder, MC, Yoshimoto, M. Lymphoid progenitor emergence in the murine embryo and yolk sac precedes stem cell detection. Stem Cells Dev 2014;23:1168–77.Google Scholar
Medvinsky, A, Dzierzak, E. Definitive hematopoiesis is autonomously initiated by the AGM region. Cell 1996;86:897906.Google Scholar
Muller, AM, Medvinsky, A, Strouboulis, J, et al. Development of hematopoietic stem cell activity in the mouse embryo. Immunity 1994;1 291301.Google Scholar
Hadland, BK, Varnum-Finney, B, Mandal, PK, et al. A common origin for B-1a and B-2 lymphocytes in clonal pre-hematopoietic stem cells. Stem Cell Reports 2017;8:15631572.Google Scholar
Zhang, Y, Gao, S, Xia, J, et al. Hematopoietic hierarchy: An updated roadmap. Trends Cell Biol 2018;28:97686.Google Scholar
Ignatz, M, Sola-Visner, M, Rimsza, LM, et al. Umbilical cord blood produces small megakaryocytes after transplantation. Biol Blood Marrow Transplant 2007;13:145–50.Google Scholar
Slayton, WB, Wainman, DA, Li, XM, et al. Developmental differences in megakaryocyte maturation are determined by the microenvironment. Stem Cells 2005;23:1400–8.Google Scholar
Hiwarkar, P, Hubank, M, Qasim, W, et al. Cord blood transplantation recapitulates fetal ontogeny with a distinct molecular signature that supports CD4(+) T-cell reconstitution. Blood Adv 2017;1:2206–16.Google Scholar
Tavassoli, M. Embryonic and fetal hemopoiesis: an overview. Blood Cells 1991;17:269–81; discussion 282–6.Google Scholar
Fukuda, T. Fetal hemopoiesis. I. Electron microscopic studies on human yolk sac hemopoiesis. Virchows Arch B Cell Pathol 1973;14:197213.Google Scholar
Luckett, WP. Origin and differentiation of the yolk sac and extraembryonic mesoderm in presomite human and rhesus monkey embryos. Am J Anat 1978;152:5997.Google Scholar
Barcena, A, Kapidzic, M, Muench, MO, et al. The human placenta is a hematopoietic organ during the embryonic and fetal periods of development. Dev Biol 2009;327:2433.Google Scholar
Barcena, A, Muench, MO, Kapidzic, M, et al. A new role for the human placenta as a hematopoietic site throughout gestation. Reprod Sci 2009;16:178–87.Google Scholar
Kelemen, E, Janossa, M. Macrophages are the first differentiated blood cells formed in human embryonic liver. Exp Hematol 1980;8:9961000.Google Scholar
Fomin, ME, Beyer, AI, Muench, MO. Human fetal liver cultures support multiple cell lineages that can engraft immunodeficient mice. Open Biol 2017;7:170108.Google Scholar
Yurasov, S, Kollmann, TR, Kim, A, et al. Severe combined immunodeficiency mice engrafted with human T cells, B cells, and myeloid cells after transplantation with human fetal bone marrow or liver cells and implanted with human fetal thymus: a model for studying human gene therapy. Blood 1997;89:1800–10.Google Scholar
Charbord, P, Tavian, M, Humeau, L, et al. Early ontogeny of the human marrow from long bones: an immunohistochemical study of hematopoiesis and its microenvironment. Blood 1996;87:4109–19.Google Scholar
Kelemen, E, Calvo, W, Fliedner, TM. Atlas of Human Hematopoietic Development (New York: Springer-Verlag, 1979).Google Scholar
Slayton, WB, Juul, SE, Calhoun, DA, et al. Hematopoiesis in the liver and marrow of human fetuses at 5 to 16 weeks post-conception: quantitative assessment of macrophage and neutrophil populations. Pediatr Res 1998;43:774–82.Google Scholar
Slayton, WB, Li, Y, Calhoun, DA, et al. The first-appearance of neutrophils in the human fetal bone marrow cavity. Early Hum Dev 1998;53:129–44.Google Scholar
Haynes, BF, Denning, SM, Singer, KH, et al. Ontogeny of T-cell precursors: A model for the initial stages of human T-cell development. Immunol Today 1989;10:8791.Google Scholar
Haynes, BF, Martin, ME, Kay, HH, et al. Early events in human T cell ontogeny. Phenotypic characterization and immunohistologic localization of T cell precursors in early human fetal tissues. J Exp Med 1988;168:1061–80.Google Scholar
MacDonald, TT, Spencer, J. Ontogeny of the gut-associated lymphoid system in man. Acta Paediatr Suppl 1994;83:35.Google Scholar
Hinchliffe, D. Development of the Vertebrate Limb (Oxford: Clarendon Press, 1980).Google Scholar
Hofman, FM, Danilovs, J, Husmann, L, et al. Ontogeny of B cell markers in the human fetal liver. J Immunol 1984;133:1197–201.Google Scholar
Naito, K, Takahashi, H, Kojima, M. Ontogenic Development of Kupffer Cells (Amsterdam: Elsevier Biomedical Press, 1982).Google Scholar
Guilliams, M, De Kleer, I, Henri, S, et al. Alveolar macrophages develop from fetal monocytes that differentiate into long-lived cells in the first week of life via GM-CSF. J Exp Med 2013;210:1977–92.Google Scholar
Schulz, C, Gomez Perdiguero, E, Chorro, L, et al. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science 2012;336:8690.Google Scholar
Hoeffel, G, Chen, J, Lavin, Y, et al. C-Myb(+) erythro-myeloid progenitor-derived fetal monocytes give rise to adult tissue-resident macrophages. Immunity 2015;42:665–78.Google Scholar
Orlikowsky, TW, Spring, B, Dannecker, GE, et al. Expression and regulation of B7 family molecules on macrophages (MPhi) in preterm and term neonatal cord blood and peripheral blood of adults. Cytometry B Clin Cytom 2003;53:40–7.Google Scholar
Marodi, L. Deficient interferon-gamma receptor-mediated signaling in neonatal macrophages. Acta Paediatr Suppl 2002;91:117–9.Google Scholar
Kraft, JD, Horzempa, J, Davis, C, et al. Neonatal macrophages express elevated levels of interleukin-27 that oppose immune responses. Immunology 2013;139:484–93.Google Scholar
Cuenca, AG, Joiner, DN, Gentile, LF, et al. TRIF-dependent innate immune activation is critical for survival to neonatal gram-negative sepsis. J Immunol 2015;194:1169–77.Google Scholar
Chelvarajan, RL, Collins, SM, Doubinskaia, IE, et al. Defective macrophage function in neonates and its impact on unresponsiveness of neonates to polysaccharide antigens. J Leukoc Biol 2004;75:982–94.Google Scholar
Olin, A, Henckel, E, Chen, Y, et al. Stereotypic immune system development in newborn children. Cell 2018;174:1277–92 e14.Google Scholar
Christensen, RD, Hill, HR. Rothstein G: granulocytic stem cell (CFUc) proliferation in experimental group B streptococcal sepsis. Pediatr Res 1983;17:278–80.Google Scholar
Christensen, RD. Rothstein G: Pre- and postnatal development of granulocytic stem cells in the rat. Pediatr Res 1984;18:599602.Google Scholar
Makoni, M, Eckert, J, Anne Pereira, H, et al. Alterations in neonatal neutrophil function attributable to increased immature forms. Early Hum Dev 2016;103:17.Google Scholar
Prosser, A, Hibbert, J, Strunk, T, et al. Phagocytosis of neonatal pathogens by peripheral blood neutrophils and monocytes from newborn preterm and term infants. Pediatr Res 2013;74:503–10.Google Scholar
Henkart, P, Yue CC: The role of cytoplasmic granules in lymphocyte cytotoxicity. Prog Allergy 1988;40:82110.Google ScholarPubMed
Strauss-Albee, DM, Liang, EC, Ranganath, T, et al. The newborn human NK cell repertoire is phenotypically formed but functionally reduced. Cytometry B Clin Cytom 2017;92:3341.Google Scholar
Lopez, C. Immunology and Pathogenesis of Persistent Virus Infections (Washington, DC: American Society of Microbiology, 1988).Google Scholar
Yu, JC, Khodadadi, H, Malik, A, et al. Innate immunity of neonates and infants. Front Immunol 2018;9:1759.Google Scholar
Eberl, G, Colonna, M, Di Santo, JP, et al. Innate lymphoid cells. Innate lymphoid cells: a new paradigm in immunology. Science 2015;348:aaa6566.Google Scholar
Bernink, JH, Peters, CP, Munneke, M, et al. Human type 1 innate lymphoid cells accumulate in inflamed mucosal tissues. Nat Immunol 2013;14:221–9.Google Scholar
Fuchs, A, Vermi, W, Lee, JS, et al. Intraepithelial type 1 innate lymphoid cells are a unique subset of IL-12- and IL-15-responsive IFN-gamma-producing cells. Immunity 2013;38:769–81.Google Scholar
Moro, K, Yamada, T, Tanabe, M, et al. Innate production of T(H)2 cytokines by adipose tissue-associated c-Kit(+)Sca-1(+) lymphoid cells. Nature 2010;463:540–4.Google Scholar
Neill, DR, Wong, SH, Bellosi, A, et al. Nuocytes represent a new innate effector leukocyte that mediates type-2 immunity. Nature 2010;464:1367–70.Google Scholar
Cella, M, Fuchs, A, Vermi, W, et al. A human natural killer cell subset provides an innate source of IL-22 for mucosal immunity. Nature 2009;457:722–5.Google Scholar
Luci, C, Reynders, A, Ivanov, II, et al. Influence of the transcription factor RORgammat on the development of NKp46+ cell populations in gut and skin. Nat Immunol 2009;10:7582.Google Scholar
Sanos, SL, Bui, VL, Mortha, A, et al. RORgammat and commensal microflora are required for the differentiation of mucosal interleukin 22-producing NKp46+ cells. Nat Immunol 2009;10:8391.Google Scholar
Satoh-Takayama, N, Vosshenrich, CA, Lesjean-Pottier, S, et al. Microbial flora drives interleukin 22 production in intestinal NKp46+ cells that provide innate mucosal immune defense. Immunity 2008;29:958–70.Google Scholar
Miller, D, Motomura, K, Garcia-Flores, V, et al. Innate lymphoid cells in the maternal and fetal compartments. Front Immunol 2018;9:2396.Google Scholar
Bank, U, Deiser, K, Finke, D, et al. Cutting edge: Innate lymphoid cells suppress homeostatic T cell expansion in neonatal mice. J Immunol 2016;196:3532–6.Google Scholar
Colten, HR, Goldberger, G. Ontogeny of serum complement proteins. Pediatrics 1979;64:775–80.Google Scholar
Ballow, M, Fang, F, Good, RA, et al. Developmental aspects of complement components in the newborn. The presence of complement components and C3 proactivator (properdin factor B) in human colostrum. Clin Exp Immunol 1974;18:257–66.Google Scholar
Grumach, AS, Ceccon, ME, Rutz, R, et al. Complement profile in neonates of different gestational ages. Scand J Immunol 2014;79:276–81.Google Scholar
Adkins, B, Mueller, C, Okada, CY, et al. Early events in T-cell maturation. Annu Rev Immunol 1987;5:325–65.Google Scholar
Haynes, BF. The role of the thymic microenvironment in promotion of early stages of human T cell maturation. Clin Res 1986;34:422–31.Google Scholar
Germain, RN, Margulies, DH. The biochemistry and cell biology of antigen processing and presentation. Annu Rev Immunol 1993;11:403–50.Google Scholar
Kingsley, G, Pitzalis, C, Waugh, AP, et al. Correlation of immunoregulatory function with cell phenotype in cord blood lymphocytes. Clin Exp Immunol 1988;73:40–5.Google Scholar
Bruning, T, Daiminger, A, Enders, G. Diagnostic value of CD45RO expression on circulating T lymphocytes of fetuses and newborn infants with pre-, peri- or early post-natal infections. Clin Exp Immunol 1997;107:306–11.Google Scholar
Paganelli, R, Cherchi, M, Scala, E, et al. Activated and “memory” phenotype of circulating T lymphocytes in intrauterine life. Cell Immunol 1994;155:486–92.Google Scholar
Durandy, A, De Saint Basile, G, Lisowska-Grospierre, B, et al. Undetectable CD40 ligand expression on T cells and low B cell responses to CD40 binding agonists in human newborns. J Immunol 1995;154:1560–8.Google Scholar
Granberg, C, Hirvonen, T. Cell-mediated lympholysis by fetal and neonatal lymphocytes in sheep and man. Cell Immunol 1980;51:1322.Google Scholar
Granberg, C, Manninen, K, Toivanen P: Cell-mediated lympholysis by human neonatal lymphocytes. Clin Immunol Immunopathol 1976;6:256–63.Google Scholar
Hayakawa, S, Ohno, N, Okada, S, et al. Significant augmentation of regulatory T cell numbers occurs during the early neonatal period. Clin Exp Immunol 2017;190:26879.Google Scholar
Xu, L, Tanaka, S, Bonno, M, et al. Cord blood CD4(+)CD25(+) regulatory T cells fail to inhibit cord blood NK cell functions due to insufficient production and expression of TGF-beta1. Cell Immunol 2014;290:8995.Google Scholar
Prince, LR, Maxwell, NC, Gill, SK, et al. Macrophage phenotype is associated with disease severity in preterm infants with chronic lung disease. PLoS One 2014;9:e103059.Google Scholar
Charrier, E, Cordeiro, P, Cordeau, M, et al. Post-transcriptional down-regulation of Toll-like receptor signaling pathway in umbilical cord blood plasmacytoid dendritic cells. Cell Immunol 2012;276:114–21.Google Scholar
Wilson, CB, Penix, L, Melvin, A, et al. Lymphokine regulation and the role of abnormal regulation in immunodeficiency. Clin Immunol Immunopathol 1993;67:S2532.Google Scholar
Lawton, AR, Cooper, MD. B cell ontogeny: Immunoglobulin genes and their expression. Pediatrics 1979;64:750–7.Google Scholar
Gathings, WE, Lawton, AR, Cooper, MD. Immunofluorescent studies of the development of pre-B cells, B lymphocytes and immunoglobulin isotype diversity in humans. Eur J Immunol 1977;7:804–10.Google Scholar
Coffman, RL, Seymour, BW, Lebman, DA, et al. The role of helper T cell products in mouse B cell differentiation and isotype regulation. Immunol Rev 1988;102:528.Google Scholar
Waddick, KG, Uckun, FM. CD5 antigen-positive B lymphocytes in human B cell ontogeny during fetal development and after autologous bone marrow transplantation. Exp Hematol 1993;21:791–8.Google Scholar
Cooper, MD. Current concepts. B lymphocytes. Normal development and function. N Engl J Med 1987;317:1452–6.Google Scholar
Gathings, WE, Kubagawa, H, Cooper, MD. A distinctive pattern of B cell immaturity in perinatal humans. Immunol Rev 1981;57:107–26.Google Scholar
Lucey, DR, Clerici, M, Shearer, GM. Type 1 and type 2 cytokine dysregulation in human infectious, neoplastic, and inflammatory diseases. Clin Microbiol Rev 1996;9:532–62.Google Scholar
Ghilardi, N, Ouyang, W. Targeting the development and effector functions of TH17 cells. Semin Immunol 2007;19:383–93.Google Scholar
Nesin, M, Cunningham-Rundles, S. Cytokines and neonates. Am J Perinatol 2000;17:393404.Google Scholar
Malhotra, I, Ouma, J, Wamachi, A, et al. In utero exposure to helminth and mycobacterial antigens generates cytokine responses similar to that observed in adults. J Clin Invest 1997;99:1759–66.Google Scholar

Save book to Kindle

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

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

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

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

Available formats
×

Save book to Google Drive

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

Available formats
×