Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-nr4z6 Total loading time: 0 Render date: 2024-06-07T19:07:27.391Z Has data issue: false hasContentIssue false

Chapter 17 - Functional Phagocyte Disorders in the Neonate

from Section V - Leucocyte Disorders

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

In addition to quantitative neutrophil abnormalities, innate immunity, and thus risk of infection in a neonate, may be negatively impacted by qualitative phagocyte defects. The term phagocyte stems from the Greek “phagein” meaning “to eat or devour” and “cyte” meaning “cell” and refers to hematopoietic derived cells, namely monocytes, macrophages and neutrophils capable of engulfing and digesting microorganisms, foreign particles, and cellular debris. Neutrophils are also classified as granulocytes, given the characteristic presence of granules in their cytoplasm that play a key role in neutrophil function.

Type
Chapter
Information
Neonatal Hematology
Pathogenesis, Diagnosis, and Management of Hematologic Problems
, pp. 279 - 292
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

Orkin, SH, Fisher, DE, Ginsburg, D, et al. eds.Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. 8th ed. (Philadelphia PA: Saunders/Elsevier, 2015).Google Scholar
Urlichs, F, Speer, CP. Neutrophil function in preterm and term infants. NeoReviews 2004 ;5(10): e417e430.Google Scholar
Lawrence, SM, Corriden, R, Nizet, V. Age-appropriate functions and dysfunctions of the neonatal neutrophil. Front Pediatr 2017;5:23.CrossRefGoogle ScholarPubMed
Melvan, JN, Bagby, GJ, Welsh, DA, Nelson, S, Zhang, P. Neonatal sepsis and neutrophil insufficiencies. Int Rev Immunol 2010;29(3):315–48.CrossRefGoogle ScholarPubMed
Levy, O. Innate immunity of the newborn: Basic mechanisms and clinical correlates. Nat Rev Immunol 2007;7(5):379–90.CrossRefGoogle ScholarPubMed
Carr, R. Neutrophil production and function in newborn infants. BJHaem 2000;110(1):1828.Google Scholar
Chandra, S, Haines, H, Michie, C, Maheshwari, A. Developmental defects in neutrophils from preterm infants. NeoReviews 2007;8(9):e368–76.Google Scholar
Lipp, P, Ruhnau, J, Lange, A, et al. Less neutrophil extracellular trap formation in term newborns than in adults. Neonatology 2017;111(2):182–8.Google Scholar
Chaudhuri, J, Mitra, S, Mukhopadhyay, D, Chakraborty, S, Chatterjee, S. Granulocyte colony-stimulating factor for preterms with sepsis and neutropenia: A randomized controlled trial. J Clin Neonatol 2012;1(4):202–6.Google Scholar
Aktaş, D, Demirel, B, Gürsoy, T, Ovalı, F. A randomized case-controlled study of recombinant human granulocyte colony stimulating factor for the treatment of sepsis in preterm neutropenic infants. Pediatr Neonatol 2015;56(3):171–5.Google Scholar
Carr, R, Modi, N, Doré, C. G-CSF and GM-CSF for treating or preventing neonatal infections. Cochrane Database Syst Rev 2003;3: CD003066.Google Scholar
Carr, R, Brocklehurst, P, Doré, CJ, Modi, N. Granulocyte-macrophage colony stimulating factor administered as prophylaxis for reduction of sepsis in extremely preterm, small for gestational age neonates (the PROGRAMS trial): a single-blind, multicentre, randomised controlled trial. Lancet 2009;373(9659):226–33.CrossRefGoogle Scholar
Capasso, L, Borrelli, A, Cerullo, J, et al. Role of immunoglobulins in neonatal sepsis. Transl Med UniSa 2015;11:2833.Google ScholarPubMed
Franco, ACBF, Torrico, AC, Moreira, FT, et al. Adjuvant use of intravenous immunoglobulin in the treatment of neonatal sepsis: A systematic review with a meta-analysis. J Pediatr (Rio J) 2012;88(5):377–83.Google Scholar
INIS Collaborative Group, Brocklehurst, P, Farrell, B, et al. Treatment of neonatal sepsis with intravenous immune globulin. N Engl J Med 2011;365(13):1201–11.Google Scholar
Jenson, HB, Pollock, BH. Meta-analyses of the effectiveness of intravenous immune globulin for prevention and treatment of neonatal sepsis. Pediatrics 1997;99(2):E2.Google Scholar
Jenson, HB, Pollock, BH. The role of intravenous immunoglobulin for the prevention and treatment of neonatal sepsis. Semin Perinatol 1998;22(1):5063.Google Scholar
Ohlsson, A, Lacy, JB. Intravenous immunoglobulin for preventing infection in preterm and/or low birth weight infants. Cochrane Database Syst Rev 2013;2(7):CD000361.Google Scholar
Glocker, E-O, Hennigs, A, Nabavi, M, et al. A homozygous CARD9 mutation in a family with susceptibility to fungal infections. N Engl J Med 2009;361(18):1727–35.Google Scholar
Netea, MG, Maródi, L. Innate immune mechanisms for recognition and uptake of Candida species. Trends Immunol 2010;31(9):346–53.Google Scholar
Clark, RA, Kimball, HR. Defective granulocyte chemotaxis in the Chediak–Higashi syndrome. J Clin Invest 1971;50(12):2645–52.CrossRefGoogle ScholarPubMed
Kaplan, J, De Domenico, I, Ward, DM. Chediak–Higashi syndrome. Curr Opin Hematol 2008;15(1):22–9.Google Scholar
Gallin, JI, Elin, RJ, Hubert, RT, et al. Efficacy of ascorbic acid in Chediak–Higashi syndrome (CHS): Studies in humans and mice. Blood. 1979;53(2):226–34.Google Scholar
Boxer, LA, Watanabe, AM, Rister, M, et al. Correction of leukocyte function in Chediak–Higashi syndrome by ascorbate. N Engl J Med 1976;295(19):1041–5.Google Scholar
Boxer, LA, Albertini, DF, Baehner, RL, Oliver, JM. Impaired microtubule assembly and polymorphonuclear leucocyte function in the Chediak–Higashi syndrome correctable by ascorbic acid. Br J Haematol 1979;43(2):207–13.Google Scholar
Winkelstein, JA, Marino, MC, Johnston, RB, et al. Chronic granulomatous disease. Report on a national registry of 368 patients. Medicine (Baltimore) 2000;79(3):155–69.Google Scholar
Marciano, BE, Spalding, C, Fitzgerald, A, et al. Common severe infections in chronic granulomatous disease. Clin Infect Dis 201515;60(8):1176–83.Google Scholar
Marciano, BE, Rosenzweig, SD, Kleiner, DE, et al. Gastrointestinal involvement in chronic granulomatous disease. Pediatrics 2004;114(2):462–8.CrossRefGoogle ScholarPubMed
Kuhns, DB, Alvord, WG, Heller, T, et al. Residual NADPH oxidase and survival in chronic granulomatous disease. N Engl J Med 2010;363(27):2600–10.Google Scholar
Gallin, JI, Alling, DW, Malech, HL, et al. Itraconazole to prevent fungal infections in chronic granulomatous disease. N Engl J Med 2003;348(24):2416–22.Google Scholar
The International Chronic Granulomatous Disease Cooperative Study Group. A controlled trial of interferon gamma to prevent infection in chronic granulomatous disease. N Engl J Med 1991;324(8):509–16.Google Scholar
Güngör, T, Teira, P, Slatter, M, et al. Reduced-intensity conditioning and HLA-matched haemopoietic stem-cell transplantation in patients with chronic granulomatous disease: a prospective multicentre study. Lancet 2014;383(9915):436–48.CrossRefGoogle ScholarPubMed
Visser, G, Rake, JP, Fernandes, J, et al. Neutropenia, neutrophil dysfunction, and inflammatory bowel disease in glycogen storage disease type Ib: Results of the European study on glycogen storage disease type I. J Pediatr 2000;137(2):187–91.CrossRefGoogle Scholar
Melis, D, Fulceri, R, Parenti, G, et al. Genotype/phenotype correlation in glycogen storage disease type 1b: A multicentre study and review of the literature. Eur J Pediatr 2005;164(8):501–8.Google Scholar
Sowerwine, KJ, Holland, SM, Freeman, AF. Hyper-IgE syndrome update. Ann N Y Acad Sci 2012;1250:2532.CrossRefGoogle ScholarPubMed
Hill, HR, Ochs, HD, Quie, PG, et al. Defect in neutrophil granulocyte chemotaxis in Job’s syndrome of recurrent “cold” staphylococcal abscesses. Lancet 1974;2(7881):617–19.Google Scholar
Dinauer, MC. Disorders of neutrophil function: An overview. Methods Mol Biol 2007;412:489504.Google Scholar
Szczawinska-Poplonyk, A, Kycler, Z, Pietrucha, B, et al. The hyperimmunoglobulin E syndrome: Clinical manifestation diversity in primary immune deficiency. Orphanet J Rare Dis 2011;6(1):76.CrossRefGoogle ScholarPubMed
Sassi, A, Lazaroski, S, Wu, G, et al. Hypomorphic homozygous mutations in phosphoglucomutase 3 (PGM3) impair immunity and increase serum IgE levels. J Allergy Clin Immunol. 2014;133(5):1410–19.E13.Google Scholar
Zhang, Y, Yu, X, Ichikawa, M, et al. Autosomal recessive phosphoglucomutase 3 (PGM3) mutations link glycosylation defects to atopy, immune deficiency, autoimmunity, and neurocognitive impairment. J Allergy Clin Immunol 2014;133(5):1400–9, e14095.CrossRefGoogle ScholarPubMed
Mogensen, TH. STAT3 and the Hyper-IgE syndrome: Clinical presentation, genetic origin, pathogenesis, novel findings and remaining uncertainties. JAKSTAT. 2013;2(2):e23435.Google Scholar
Davies, EG, Thrasher, AJ. Update on the hyper immunoglobulin M syndromes. Br J Haematol 2010;149(2):167–80.Google Scholar
Picard, C, von Bernuth, H, Ghandil, P, et al. Clinical Features and Outcome of Patients With IRAK-4 and MyD88 Deficiency. Medicine (Baltimore) 2010;89(6):403–25.Google Scholar
Bouma, G, Doffinger, R, Patel, SY, et al. Impaired neutrophil migration and phagocytosis in IRAK-4 deficiency. Br J Haematol 2009;147(1):153–6.Google Scholar
Jarchum, I, Liu, M, Shi, C, Equinda, M, Pamer, EG. Critical role for MyD88-mediated neutrophil recruitment during Clostridium difficile colitis. Infect Immun 2012;80(9):2989–96.Google Scholar
Harris, ES, Weyrich, AS, Zimmerman, GA. Lessons from rare maladies: Leukocyte adhesion deficiency syndromes. Curr Opin Hematol 2013;20(1):1625.Google Scholar
Rotrosen, D, Gallin, JI. Disorders of phagocyte function. Annu Rev Immunol 1987;5:127–50.Google Scholar
Moutsopoulos, NM, Konkel, J, Sarmadi, M, et al. Defective neutrophil recruitment in leukocyte adhesion deficiency type I disease causes local IL-17-driven inflammatory bone loss. Sci Transl Med 2014;6(229):229ra40.Google Scholar
Bustamante, J, Boisson-Dupuis, S, Abel, L, Casanova, J-L. Mendelian susceptibility to mycobacterial disease: Genetic, immunological, and clinical features of inborn errors of IFN-γ immunity. Semin Immunol 2014;26(6):454–70.Google Scholar
Bigley, V, Maisuria, S, Cytlak, U, et al. Biallelic interferon regulatory factor 8 mutation: A complex immunodeficiency syndrome with dendritic cell deficiency, monocytopenia, and immune dysregulation. Journal of Allergy and Clinical Immunology 2018;141(6):2234–48.CrossRefGoogle ScholarPubMed
Klebanoff, SJ, Kettle, AJ, Rosen, H, Winterbourn, CC, Nauseef, WM. Myeloperoxidase: A front-line defender against phagocytosed microorganisms. J Leukoc Biol 2013;93(2):185–98.CrossRefGoogle ScholarPubMed
Chiang, AK, Chan, GC, Ma, SK, et al. Disseminated fungal infection associated with myeloperoxidase deficiency in a premature neonate. Pediatr Infect Dis J 2000;19(10):1027–9.Google Scholar
Ludviksson, BR, Thorarensen, O, Gudnason, T, Halldorsson, S. Candida albicans meningitis in a child with myeloperoxidase deficiency. Pediatr Infect Dis J 1993;12(2):162–4.Google Scholar
Uzel, G. The range of defects associated with nuclear factor kappaB essential modulator. Curr Opin Allergy Clin Immunol 2005;5(6):513–18.Google Scholar
Kawai, T, Nishikomori, R, Heike, T. Diagnosis and treatment in anhidrotic ectodermal dysplasia with immunodeficiency. Allergol Int 2012;61(2):207–17.Google Scholar
Choi, M, Rolle, S, Wellner, M, et al. Inhibition of NF-kappaB by a TAT-NEMO-binding domain peptide accelerates constitutive apoptosis and abrogates LPS-delayed neutrophil apoptosis. Blood 2003;102(6):2259–67.Google Scholar
Singh, A, Zarember, KA, Kuhns, DB, Gallin, JI. Impaired priming and activation of the neutrophil NADPH oxidase in patients with IRAK4 or NEMO deficiency. J Immunol 2009;182(10):6410–17.Google Scholar
Pai, S-Y, Levy, O, Jabara, HH, et al. Allogeneic transplantation successfully corrects immune defects, but not susceptibility to colitis, in a patient with nuclear factor-kappaB essential modulator deficiency. J Allergy Clin Immunol 2008;122(6):1113–1118.e1.Google Scholar
Dupuis-Girod, S, Cancrini, C, Le Deist, F, et al. Successful allogeneic hemopoietic stem cell transplantation in a child who had anhidrotic ectodermal dysplasia with immunodeficiency. Pediatrics 2006;118(1):e205211.Google Scholar
Gallin, JI, Fletcher, MP, Seligmann, BE, et al. Human neutrophil-specific granule deficiency: a model to assess the role of neutrophil-specific granules in the evolution of the inflammatory response. Blood 1982;59(6):1317–29.Google Scholar
McIlwaine, L, Parker, A, Sandilands, G, Gallipoli, P, Leach, M. Neutrophil-specific granule deficiency. Br J Haematol 2013;160(6):735.Google Scholar
Wynn, RF, Sood, M, Theilgaard-Mönch, K, et al. Intractable diarrhoea of infancy caused by neutrophil specific granule deficiency and cured by stem cell transplantation. Gut 2006;55(2):292–3.CrossRefGoogle ScholarPubMed
Dhanraj, S, Matveev, A, Li, H, et al. Biallelic mutations in DNAJC21 cause Shwachman–Diamond syndrome. Blood 2017;129(11):1557–62.Google Scholar
Stepensky, P, Chacón-Flores, M, Kim, KH, et al. Mutations in EFL1, an SBDS partner, are associated with infantile pancytopenia, exocrine pancreatic insufficiency and skeletal anomalies in a Shwachman-Diamond like syndrome. J Med Genet 2017 54(8):558–66.Google Scholar
Carapito, R, Konantz, M, Paillard, C, et al. Mutations in signal recognition particle SRP54 cause syndromic neutropenia with Shwachman–Diamond-like features. J Clin Invest 2017;127(11):4090–103.Google Scholar
Bezzerri, V, Cipolli, M. Shwachman–Diamond syndrome: Molecular mechanisms and current perspectives. Mol Diagn Ther 2019;23(2):281–90.Google Scholar
Rochowski, A, Sun, C, Glogauer, M, Alter, BP. Neutrophil functions in patients with inherited bone marrow failure syndromes. Pediatr Blood Cancer 2011;57(2):306–9.Google Scholar
Nelson, AS, Myers, KC. Diagnosis, treatment, and molecular pathology of Shwachman–Diamond syndrome. Hematol Oncol Clin North Am 2018;32(4):687700.Google Scholar
Thrasher, AJ, Burns, SO. WASP: a key immunological multitasker. Nat Rev Immunol 2010;10(3):182–92.Google Scholar
Ochs, HD, Slichter, SJ, Harker, LA, et al. The Wiskott–Aldrich syndrome: studies of lymphocytes, granulocytes, and platelets. Blood 1980;55(2):243–52.Google Scholar
Massaad, MJ, Ramesh, N, Geha, RS. Wiskott–Aldrich syndrome: A comprehensive review. Ann N Y Acad Sci 2013;1285:2643. CrossRefGoogle ScholarPubMed

Save book to Kindle

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

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

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

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

Available formats
×

Save book to Google Drive

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

Available formats
×