Skip to main content Accessibility help
×
Hostname: page-component-7479d7b7d-jwnkl Total loading time: 0 Render date: 2024-07-11T11:33:57.007Z Has data issue: false hasContentIssue false

30 - Cell Death Regulation in the Hematopoietic System

from Part II - Cell Death in Tissues and Organs

Published online by Cambridge University Press:  07 September 2011

Douglas R. Green
Affiliation:
St. Jude Children's Research Hospital, Memphis, Tennessee
Paul A. Ney
Affiliation:
St. Jude Children’s Research Hospital
John C. Reed
Affiliation:
Sanford-Burnham Medical Research Institute, La Jolla, California
Get access

Summary

Introduction

The hematopoietic system is a dynamic multilineage organ, from which we can gain insights into the physiologic roles of cell death pathways. The role of hematopoiesis is to produce blood cells under normal and stress conditions throughout the lifespan of an organism. In humans, effective function of the hematopoietic system entails the steady and regulated production of erythrocytes, which carry oxygen; platelets, which prevent bleeding; and granulocytes, monocytes, and lymphocytes, which are required for host defense. Remarkably, these varied cell types are derived from a common ancestor, the hematopoietic stem cell (HSC). HSCs are known for certain key properties: they are long-lived and self-renewing and give rise to a steady supply of multipotential progenitor cells (MPPs) that are committed to undergo differentiation. MPPs in turn give rise to progenitors with a progressively restricted developmental potential; these undergo regulated expansion and, eventually, commitment to terminal differentiation. The terminal differentiation program is characterized by decreased proliferation or mitotic arrest, global changes in gene expression, and cellular remodeling. Finally, cells that have fulfilled their purpose, or senescent cells, are cleared from the body to make room for new cells, or after a stress response to re-establish homeostasis.

Type
Chapter
Information
Apoptosis
Physiology and Pathology
, pp. 350 - 362
Publisher: Cambridge University Press
Print publication year: 2011

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

Testa, U. Apoptotic mechanisms in the control of erythropoiesis. Leukemia. 2004;18:1176–99.
Secchiero, P, Zauli, G. Tumor-necrosis-factor-related apoptosis-inducing ligand and the regulation of hematopoiesis. Curr Opin Hematol. 2008;15:42–8.
Ware, CF. The TNF superfamily. Cytokine Growth Factor Rev. 2003;14:181–4.
Wolf, BB, Green, DR. Suicidal tendencies: apoptotic cell death by caspase family proteinases. J Biol Chem. 1999;274:20049–52.
Youle, RJ, Strasser, A. The BCL-2 protein family: opposing activities that mediate cell death. Nat Rev Mol Cell Biol. 2008;9:47–59.
Danial, NN, Korsmeyer, SJ. Cell death: critical control points. Cell. 2004;116:205–19.
Certo, M, Del Gaizo Moore, V, Nishino, M et al. Mitochondria primed by death signals determine cellular addiction to antiapoptotic BCL-2 family members. Cancer Cell. 2006;9:351–65.
Willis, SN, Fletcher, JI, Kaufmann, T et al. Apoptosis initiated when BH3 ligands engage multiple Bcl-2 homologs, not Bax or Bak. Science. 2007;315:856–9.
Jurgensmeier, JM, Xie, Z, Deveraux, Q et al. Bax directly induces release of cytochrome c from isolated mitochondria. Proc Natl Acad Sci U S A. 1998;95:4997–5002.
Rosse, T, Olivier, R, Monney, L et al. Bcl-2 prolongs cell survival after Bax-induced release of cytochrome c. Nature. 1998;391:496–9.
Li, H, Zhu, H, Xu, CJ, Yuan, J. Cleavage of BID by caspase 8 mediates the mitochondrial damage in the Fas pathway of apoptosis. Cell. 1998;94:491–501.
Luo, X, Budihardjo, I, Zou, H, Slaughter, C, Wang, X. Bid, a Bcl2 interacting protein, mediates cytochrome c release from mitochondria in response to activation of cell surface death receptors. Cell. 1998;94:481–90.
Cheshier, SH, Morrison, SJ, Liao, X, Weissman, IL. In vivo proliferation and cell cycle kinetics of long-term self-renewing hematopoietic stem cells. Proc Natl Acad Sci U S A. 1999;96:3120–5.
Nijnik, A, Woodbine, L, Marchetti, C et al. DNA repair is limiting for haematopoietic stem cells during ageing. Nature. 2007;447:686–90.
Rossi, DJ, Bryder, D, Seita, J et al. Deficiencies in DNA damage repair limit the function of haematopoietic stem cells with age. Nature. 2007;447:725–9.
Rossi, DJ, Seita, J, Czechowicz, A et al. Hematopoietic stem cell quiescence attenuates DNA damage response and permits DNA damage accumulation during aging. Cell Cycle. 2007;6:2371–6.
Ivanova, NB, Dimos, JT, Schaniel, C et al. A stem cell molecular signature. Science. 2002;298:6014.
Ramalho-Santos, M, Yoon, S, Matsuzaki, Y, Mulligan, RC, Melton, DA. “Stemness”: transcriptional profiling of embryonic and adult stem cells. Science. 2002;298:597–600.
Bruno, L, Hoffmann, R, McBlane, F et al. Molecular signatures of self-renewal, differentiation, and lineage choice in multipotential hemopoietic progenitor cells in vitro. Mol Cell Biol. 2004;24:741–56.
Lessard, J, Sauvageau, G. Bmi-1 determines the proliferative capacity of normal and leukaemic stem cells. Nature. 2003;423:255–60.
Park, IK, Qian, D, Kiel, M et al. Bmi-1 is required for maintenance of adult self- renewing haematopoietic stem cells. Nature. 2003;423:302–5.
Galan-Caridad, JM, Harel, S, Arenzana, TL et al. Zfx controls the self-renewal of embryonic and hematopoietic stem cells. Cell. 2007;129:345–57.
Yoshida, T, Hazan, I, Zhang, J et al. The role of the chromatin remodeler Mi-2beta in hematopoietic stem cell self-renewal and multilineage differentiation. Genes Dev. 2008;22:1174–89.
Wilson, A, Trumpp, A. Bone-marrow haematopoietic-stem-cell niches. Nat Rev Immunol. 2006;6:93–106.
Zhang, CC, Lodish, HF. Cytokines regulating hematopoietic stem cell function. Curr Opin Hematol. 2008;15:307–11.
Opferman, JT, Iwasaki, H, Ong, CC et al. Obligate role of anti-apoptotic MCL-1 in the survival of hematopoietic stem cells. Science. 2005;307:1101–4.
Kikushige, Y, Yoshimoto, G, Miyamoto, T et al. Human Flt3 is expressed at the hematopoietic stem cell and the granulocyte/macrophage progenitor stages to maintain cell survival. J Immunol. 2008;180:7358–67.
Matsuzaki, Y, Nakayama, K, Nakayama, K et al. Role of bcl-2 in the development of lymphoid cells from the hematopoietic stem cell. Blood. 1997;89:853–62.
Motoyama, N, Wang, F, Roth, KA et al. Massive cell death of immature hematopoietic cells and neurons in Bcl-x-deficient mice. Science 1995;267:1506–10.
Domen, J, Cheshier, SH, Weissman, IL. The role of apoptosis in the regulation of hematopoietic stem cells: overexpression of Bcl-2 increases both their number and repopulation potential. J Exp Med. 2000;191:253–64.
Zhang, J, Grindley, JC, Yin, T et al. PTEN maintains haematopoietic stem cells and acts in lineage choice and leukaemia prevention. Nature. 2006;441:518–522.
Tothova, Z, Kollipara, R, Huntly, BJ et al. FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell. 2007;128:325–39.
Necas, E, Sefc, L, Sulc, K, Barthel, E, Seidel, HJ. Estimation of extent of cell death in different stages of normal murine hematopoiesis. Stem Cells. 1998;16:107–11.
Ogilvy, S, Metcalf, D, Print, CG et al. Constitutive Bcl-2 expression throughout the hematopoietic compartment affects multiple lineages and enhances progenitor cell survival. Proc Natl Acad Sci U S A. 1999;96:14943–8.
Bouillet, P, Metcalf, D, Huang, DC et al. Proapoptotic Bcl-2 relative Bim required for certain apoptotic responses, leukocyte homeostasis, and to preclude autoimmunity. Science. 1999;286:1735–8.
Lindsten, T, Ross, AJ, King, A et al. The combined functions of proapoptotic Bcl-2 family members bak and bax are essential for normal development of multiple tissues. Mol Cell. 2000;6:1389–99.
Schneider, E, Moreau, G, Arnould, A et al. Increased fetal and extramedullary hematopoiesis in Fas-deficient C57BL/6-lpr/lpr mice. Blood. 1999;94:2613–21.
Suda, T, Suda, J, Ogawa, M. Single-cell origin of mouse hemopoietic colonies expressing multiple lineages in variable combinations. Proc Natl Acad Sci U S A. 1983;80:6689–93.
Leary, AG, Strauss, LC, Civin, CI, Ogawa, M. Disparate differentiation in hemopoietic colonies derived from human paired progenitors. Blood. 1985;66:327–32.
Akashi, K, He, X, Chen, J et al. Transcriptional accessibility for genes of multiple tissues and hematopoietic lineages is hierarchically controlled during early hematopoiesis. Blood. 2003;101:383–9.
Terskikh, AV, Miyamoto, T, Chang, C, Diatchenko, L, Weissman, IL. Gene expression analysis of purified hematopoietic stem cells and committed progenitors. Blood. 2003;102:94–101.
Lagasse, E, Weissman, IL. Enforced expression of Bcl-2 in monocytes rescues macrophages and partially reverses osteopetrosis in op/op mice. Cell. 1997;89:1021–31.
Akashi, K, Kondo, M, von Freeden-Jeffry, U, Murray, R, Weissman, IL. Bcl-2 rescues T lymphopoiesis in interleukin-7 receptor-deficient mice. Cell. 1997;89:1033–41.
Kondo, M, Scherer, DC, Miyamoto, T et al. Cell-fate conversion of lymphoid- committed progenitors by instructive actions of cytokines. Nature. 2000;407:383–6.
Huang, S, Guo, YP, May, G, Enver, T. Bifurcation dynamics in lineage-commitment in bipotent progenitor cells. Dev Biol. 2007;305:695–713.
Sawada, K, Krantz, SB, Dai, CH et al. Purification of human blood burst-forming units-erythroid and demonstration of the evolution of erythropoietin receptors. J Cell Physiol. 1990;142:219–30.
Wickrema, A, Krantz, SB, Winkelmann, JC, Bondurant, MC. Differentiation and erythropoietin receptor gene expression in human erythroid progenitor cells. Blood. 1992;80:1940–9.
Wu, H, Liu, X, Jaenisch, R, Lodish, HF. Generation of committed erythroid BFU-E and CFU-E progenitors does not require erythropoietin or the erythropoietin receptor. Cell. 1995;83:59–67.
Witthuhn, BA, Quelle, FW, Silvennoinen, O et al. Jak2 associates with the erythropoietin receptor and is tyrosine-phosphorylated and activated following stimulation with erythropoietin. Cell. 1993;74:227–36.
Parganas, E, Wang, D, Stravopodis, D et al. Jak2 is essential for signaling through a variety of cytokine receptors. Cell. 1998;93:385–95.
Gregoli, PA, Bondurant, MC. The roles of Bcl-X(L) and apopain in the control of erythropoiesis by erythropoietin. Blood. 1997;90:630–40.
Silva, M, Grillot, D, Benito, A et al. Erythropoietin can promote erythroid progenitor survival by repressing apoptosis through Bcl-XL and Bcl-2. Blood. 1996;88:1576–82.
Socolovsky, M, Fallon, AE, Wang, S, Brugnara, C, Lodish, HF. Fetal anemia and apoptosis of red cell progenitors in Stat5a-/-5b-/- mice: a direct role for Stat5 in Bcl-X(L) induction. Cell. 1999;98:181–91.
Dolznig, H, Habermann, B, Stangl, K et al. Apoptosis protection by the Epo target Bcl-X(L) allows factor-independent differentiation of primary erythroblasts. Curr Biol. 2002;12:1076–85.
Rhodes, MM, Kopsombut, P, Bondurant, MC, Price, JO, Koury, MJ. Bcl-x(L) prevents apoptosis of late-stage erythroblasts but does not mediate the antiapoptotic effect of erythropoietin. Blood. 2005;106:1857–63.
Motoyama, N, Kimura, T, Takahashi, T, Watanabe, T, Nakano, T. Bcl-X prevents apoptotic cell death of both primitive and definitive erythrocytes at the end of maturation. J Exp Med. 1999;189:1691–8.
Koury, MJ, Bondurant, MC. Erythropoietin retards DNA breakdown and prevents programmed death in erythroid progenitor cells. Science. 1990;248:378–81.
Gregoli, PA, Bondurant, MC. Function of caspases in regulating apoptosis caused by erythropoietin deprivation in erythroid progenitors. J Cell Physiol. 1999;178:133–43.
De Maria, R, Testa, U, Luchetti, L et al. Apoptotic role of Fas/Fas ligand system in the regulation of erythropoiesis. Blood. 1999;93:796–803.
Mohandas, N, Prenant, M. Three-dimensional model of bone marrow. Blood. 1978;51:633–43.
Liu, Y, Pop, R, Sadegh, C, et al. Suppression of Fas-FasL coexpression by erythropoietin mediates erythroblast expansion during the erythropoietic stress response in vivo. Blood. 2006;108:123–133.
De Maria, R., Zeuner, A, Eramo, A et al. Negative regulation of erythropoiesis by caspase-mediated cleavage of GATA-1. Nature. 1999;401:489–93.
Varfolomeev, EE, Schuchmann, M, Luria, V et al. Targeted disruption of the mouse Caspase 8 gene ablates cell death induction by the TNF receptors, Fas/Apo1, and DR3 and is lethal prenatally. Immunity. 1998;9:267–76.
Yeh, WC, Pompa, JL, McCurrach, ME et al. FADD: essential for embryo development and signaling from some, but not all, inducers of apoptosis. Science. 1998;279:1954–8.
Dolznig, H, Bartunek, P, Nasmyth, K, Mullner, EW, Beug, H. Terminal differentiation of normal chicken erythroid progenitors: shortening of G1 correlates with loss of D-cyclin/cdk4 expression and altered cell size control. Cell Growth Differ. 1995;6:1341–52.
Kelley, LL, Green, WF, Hicks, GG et al. Apoptosis in erythroid progenitors deprived of erythropoietin occurs during the G1 and S phases of the cell cycle without growth arrest or stabilization of wild-type p53. Mol Cell Biol. 1994;14:4183–92.
Dolznig, H, Boulme, F, Stangl, K et al. Establishment of normal, terminally differentiating mouse erythroid progenitors: molecular characterization by cDNA arrays. FASEB J. 2001;15:1442–4.
Hou, VC, Conboy, JG. Regulation of alternative pre-mRNA splicing during erythroid differentiation. Curr Opin Hematol. 2001;8:74–9.
Groudine, M, Weintraub, H. Activation of globin genes during chicken development. Cell. 1981;24:393–401.
Zermati, Y, Garrido, C, Amsellem, S et al. Caspase activation is required for terminal erythroid differentiation. J Exp Med. 2001;193:247–54.
Carlile, GW, Smith, DH, Wiedmann, M. Caspase-3 has a nonapoptotic function in erythroid maturation. Blood. 2004;103:4310–16.
Krauss, SW, Lo, AJ, Short, SA et al. Nuclear substructure reorganization during late-stage erythropoiesis is selective and does not involve caspase cleavage of major nuclear substructural proteins. Blood. 2005;106:2200–205.
Tarbutt, RG. Cell population kinetics of the erythroid system in the rat. The response to protracted anaemia and to continuous gamma-irradiation. Br J Haematol. 1969;16:9–24.
Ganzoni, A, Hillman, RS, Finch, CA. Maturation of the macroreticulocyte. Br J Haematol. 1969;16:119–35.
Mel, HC, Prenant, M, Mohandas, N. Reticulocyte motility and form: studies on maturation and classification. Blood. 1977;49:1001–9.
Waugh, RE, McKenney, JB, Bauserman, RG et al. Surface area and volume changes during maturation of reticulocytes in the circulation of the baboon. J Lab Clin Med. 1997;129:527–35.
Chasis, JA, Prenant, M, Leung, A, Mohandas, N. Membrane assembly and remodeling during reticulocyte maturation. Blood. 1989;74:1112–20.
Gronowicz, G, Swift, H, Steck, TL. Maturation of the reticulocyte in vitro. J Cell Sci. 1984;71:177–97.
Koury, MJ, Koury, ST, Kopsombut, P, Bondurant, MC. In vitro maturation of nascent reticulocytes to erythrocytes. Blood. 2005;105:2168–74.
Yorimitsu, T, Klionsky, DJ. Autophagy: molecular machinery for self-eating. Cell Death Differ. 2005;12 Suppl 2:1542–52.
Heynen, MJ, Tricot, G, Verwilghen, RL. Autophagy of mitochondria in rat bone marrow erythroid cells. Relation to nuclear extrusion. Cell Tissue Res. 1985;239:235–9.
Kundu, M, Lindsten, T, Yang, C et al. Ulk1 plays a critical role in the autophagic clearance of mitochondria and ribosomes during reticulocyte maturation. Blood. 2008;112:1493–502.
Schweers, RL, Zhang, J, Randall, MS et al. NIX is required for programmed mitochondrial clearance during reticulocyte maturation. Proc Natl Acad Sci U S A. 2007;104:19500–5.
Sandoval, H, Thiagarajan, P, Dasgupta, SK et al. Essential role for Nix in autophagic maturation of erythroid cells. Nature. 2008;454:232–5.
Aerbajinai, W, Giattina, M, Lee, YT, Raffeld, M, Miller, JL. The proapoptotic factor Nix is coexpressed with Bcl-xL during terminal erythroid differentiation. Blood. 2003;102:712–17.
Diwan, A, Koesters, AG, Odley, AM et al. Unrestrained erythroblast development in Nix−/− mice reveals a mechanism for apoptotic modulation of erythropoiesis. Proc Natl Acad Sci U S A. 2007;104:6794–9.
Maiuri, MC, Le, TG, Criollo, A et al. Functional and physical interaction between Bcl-X(L) and a BH3-like domain in Beclin-1. EMBO J. 2007;26:2527–39.
Bratosin, D, Estaquier, J, Petit, F et al. Programmed cell death in mature erythrocytes: a model for investigating death effector pathways operating in the absence of mitochondria. Cell Death Differ. 2001;8:1143–56.
Yoshida, H, Kawane, K, Koike, M et al. Phosphatidylserine-dependent engulfment by macrophages of nuclei from erythroid precursor cells. Nature. 2005;437:754–8.
Berg, CP, Engels, IH, Rothbart, A et al. Human mature red blood cells express caspase-3 and caspase-8, but are devoid of mitochondrial regulators of apoptosis. Cell Death Differ. 2001;8:1197–206.
Patel, SR, Hartwig, JH, ItalianoJE, Jr JE, Jr. The biogenesis of platelets from megakaryocyte proplatelets. J Clin Invest. 2005;115:3348–54.
Gurney, AL, Carver-Moore, K, de Sauvage, FJ, Moore, MW. Thrombocytopenia in c-mpl-deficient mice. Science. 1994;265:1445–7.
De, BS, Sabri, S, Daugas, E et al. Platelet formation is the consequence of caspase activation within megakaryocytes. Blood. 2002;100:1310–17.
Clarke, MC, Savill, J, Jones, DB, Noble, BS, Brown, SB. Compartmentalized megakaryocyte death generates functional platelets committed to caspase- independent death. J Cell Biol. 2003;160:577–87.
Zauli, G, Vitale, M, Falcieri, E et al. In vitro senescence and apoptotic cell death of human megakaryocytes. Blood. 1997;90:2234–43.
Kaluzhny, Y, Yu, G, Sun, S et al. BclxL overexpression in megakaryocytes leads to impaired platelet fragmentation. Blood. 2002;100:1670–8.
Wolf, BB, Goldstein, JC, Stennicke, HR et al. Calpain functions in a caspase- independent manner to promote apoptosis-like events during platelet activation. Blood. 1999;94:1683–92.
Sanz, C, Benet, I, Richard, C et al. Antiapoptotic protein Bcl-x(L) is up-regulated during megakaryocytic differentiation of CD34(+) progenitors but is absent from senescent megakaryocytes. Exp Hematol. 2001;29:728–35.
Mason, KD, Carpinelli, MR, Fletcher, JI et al. Programmed anuclear cell death delimits platelet life span. Cell. 2007;128:1173–86.
Richards, MK, Liu, F, Iwasaki, H, Akashi, K, Link, DC. Pivotal role of granulocyte colony-stimulating factor in the development of progenitors in the common myeloid pathway. Blood. 2003;102:3562–8.
Basu, S, Hodgson, G, Katz, M, Dunn, AR. Evaluation of role of G-CSF in the production, survival, and release of neutrophils from bone marrow into circulation. Blood. 2002;100:854–61.
Zhou, P, Qian, L, Bieszczad, CK et al. Mcl-1 in transgenic mice promotes survival in a spectrum of hematopoietic cell types and immortalization in the myeloid lineage. Blood. 1998;92:3226–39.
Dzhagalov, I, St, JA, He, YW. The antiapoptotic protein Mcl-1 is essential for the survival of neutrophils but not macrophages. Blood. 2007;109:1620–6.
Steimer, DA, Boyd, K, Takeuchi, O et al. Selective roles for antiapoptotic MCL-1 during granulocyte development and macrophage effector function. Blood. 2009;113:2805–15.
Cheers, C, Haigh, AM, Kelso, A et al. Production of colony-stimulating factors (CSFs) during infection: separate determinations of macrophage-, granulocyte-, granulocyte-macrophage-, and multi-CSFs. Infect Immun. 1988;56:247–51.
Tamura, M, Hattori, K, Nomura, H et al. Induction of neutrophilic granulocytosis in mice by administration of purified human native granulocyte colony-stimulating factor (G-CSF). Biochem Biophys Res Commun. 1987;142:454–60.
Cohen, AM, Zsebo, KM, Inoue, H et al. In vivo stimulation of granulopoiesis by recombinant human granulocyte colony-stimulating factor. Proc Natl Acad Sci U S A. 1987;84:2484–8.
Metcalf, D, Robb, L, Dunn, AR, Mifsud, S, Di, RL. Role of granulocyte-macrophage colony-stimulating factor and granulocyte colony-stimulating factor in the development of an acute neutrophil inflammatory response in mice. Blood. 1996;88:3755–64.
Gregory, AD, Hogue, LA, Ferkol, TW, Link, DC. Regulation of systemic and local neutrophil responses by G-CSF during pulmonary Pseudomonas aeruginosa infection. Blood. 2007;109:3235–43.
Lieschke, GJ, Grail, D, Hodgson, G et al. Mice lacking granulocyte colony-stimulating factor have chronic neutropenia, granulocyte and macrophage progenitor cell deficiency, and impaired neutrophil mobilization. Blood. 1994;84:1737–46.
Liu, F, Wu, HY, Wesselschmidt, R, Kornaga, T, Link, DC. Impaired production and increased apoptosis of neutrophils in granulocyte colony-stimulating factor receptor-deficient mice. Immunity. 1996;5:491–501.
Stanley, E, Lieschke, GJ, Grail, D et al. Granulocyte/macrophage colony-stimulating factor-deficient mice show no major perturbation of hematopoiesis but develop a characteristic pulmonary pathology. Proc Natl Acad Sci U S A. 1994;91:5592–6.
Yoshida, H, Hayashi, S, Kunisada, T et al. The murine mutation osteopetrosis is in the coding region of the macrophage colony stimulating factor gene. Nature. 1990;345:442–4.
Lieschke, GJ, Stanley, E, Grail, D et al. Mice lacking both macrophage- and granulocyte-macrophage colony-stimulating factor have macrophages and coexistent osteopetrosis and severe lung disease. Blood. 1994;84:27–35.
Cartwright, GE, Athens, JW, Wintrobe, MM. The kinetics of granulopoiesis in normal man. Blood. 1964;24:780–803.
Luo, HR, Loison, F. Constitutive neutrophil apoptosis: mechanisms and regulation. Am J Hematol. 2008;83:288–95.
Zhu, D, Hattori, H, Jo, H et al. Deactivation of phosphatidylinositol 3,4,5- trisphosphate/Akt signaling mediates neutrophil spontaneous death. Proc Natl Acad Sci U S A. 2006;103:14836–41.
Wu, R, Kausar, H, Johnson, P et al. Hsp27 regulates Akt activation and polymorphonuclear leukocyte apoptosis by scaffolding MK2 to Akt signal complex. J Biol Chem. 2007;282:21598–608.
Moulding, DA, Akgul, C, Derouet, M, White, MR, Edwards, SW. BCL-2 family expression in human neutrophils during delayed and accelerated apoptosis. J Leukoc Biol. 2001;70:783–92.
Derouet, M, Thomas, L, Cross, A, Moots, RJ, Edwards, SW. Granulocyte macrophage colony-stimulating factor signaling and proteasome inhibition delay neutrophil apoptosis by increasing the stability of Mcl-1. J Biol Chem. 2004;279:26915–21.
Hatakeyama, S, Hamasaki, A, Negishi, I et al. Multiple gene duplication and expression of mouse bcl-2-related genes, A1. Int Immunol. 1998;10:631–7.
Hamasaki, A, Sendo, F, Nakayama, K et al. Accelerated neutrophil apoptosis in mice lacking A1-a, a subtype of the bcl-2-related A1 gene. J Exp Med. 1998;188:1985–92.
Villunger, A, O’Reilly, LA, Holler, N, Adams, J, Strasser, A. Fas ligand, Bcl-2, granulocyte colony-stimulating factor, and p38 mitogen-activated protein kinase: Regulators of distinct cell death and survival pathways in granulocytes. J Exp Med. 2000;192:647–58.
Villunger, A, Scott, C, Bouillet, P, Strasser, A. Essential role for the BH3-only protein Bim but redundant roles for Bax, Bcl-2, and Bcl-w in the control of granulocyte survival. Blood. 2003;101:2393–400.
Fossati, G, Moulding, DA, Spiller, DG et al. The mitochondrial network of human neutrophils: role in chemotaxis, phagocytosis, respiratory burst activation, and commitment to apoptosis. J Immunol. 2003;170:1964–72.
Harter, L, Keel, M, Hentze, H et al. Spontaneous in contrast to CD95-induced neutrophil apoptosis is independent of caspase activity. J Trauma. 2001;50:982–8.
Maianski, NA, Mul, FP, van Buul, JD, Roos, D, Kuijpers, TW. Granulocyte colony- stimulating factor inhibits the mitochondria-dependent activation of caspase-3 in neutrophils. Blood. 2002;99:672–9.
Fadeel, B, Ahlin, A, Henter, JI, Orrenius, S, Hampton, MB. Involvement of caspases in neutrophil apoptosis: regulation by reactive oxygen species. Blood. 1998;92:4808–18.
Iwai, K, Miyawaki, T, Takizawa, T et al. Differential expression of bcl-2 and susceptibility to anti-Fas-mediated cell death in peripheral blood lymphocytes, monocytes, and neutrophils. Blood. 1994;84:1201–8.
Liles, WC, Kiener, PA, Ledbetter, JA, Aruffo, A, Klebanoff, SJ. Differential expression of Fas (CD95) and Fas ligand on normal human phagocytes: implications for the regulation of apoptosis in neutrophils. J Exp Med. 1996;184:429–40.
Brown, SB, Savill, J. Phagocytosis triggers macrophage release of Fas ligand and induces apoptosis of bystander leukocytes. J Immunol. 1999;162:480–5.
Dale, DC, Boxer, L, Liles, WC. The phagocytes: neutrophils and monocytes. Blood. 2008;112:935–45.
Simon, HU. Neutrophil apoptosis pathways and their modifications in inflammation. Immunol Rev. 2003;193:101–10.
Klein, JB, Buridi, A, Coxon, PY et al. Role of extracellular signal-regulated kinase and phosphatidylinositol-3 kinase in chemoattractant and LPS delay of constitutive neutrophil apoptosis. Cell Signal. 2001;13:335–43.
Francois, S, El, BJ, Dang, PM et al. Inhibition of neutrophil apoptosis by TLR agonists in whole blood: involvement of the phosphoinositide 3-kinase/Akt and NF-kappaB signaling pathways, leading to increased levels of Mcl-1, A1, and phosphorylated Bad. J Immunol. 2005;174:3633–42.
Moulding, DA, Quayle, JA, Hart, CA, Edwards, SW. Mcl-1 expression in human neutrophils: regulation by cytokines and correlation with cell survival. Blood. 1998;92:2495–502.
Hannah, S, Mecklenburgh, K, Rahman, I et al. Hypoxia prolongs neutrophil survival in vitro. FEBS Lett. 1995;372:233–7.
Watson, RW, Rotstein, OD, Nathens, AB, Parodo, J, Marshall, JC. Neutrophil apoptosis is modulated by endothelial transmigration and adhesion molecule engagement. J Immunol. 1997;158:945–53.
Cowburn, AS, White, JF, Deighton, J, Walmsley, SR, Chilvers, ER. z-VAD-fmk augmentation of TNF alpha-stimulated neutrophil apoptosis is compound specific and does not involve the generation of reactive oxygen species. Blood. 2005;105:2970–2.
Beg, AA, Baltimore, D. An essential role for NF-kappaB in preventing TNF-alpha-induced cell death. Science. 1996;274:782–4.
Van Antwerp, DJ, Martin, SJ, Kafri, T, Green, DR, Verma, IM. Suppression of TNF-alpha-induced apoptosis by NF-kappaB. Science. 1996;274:787–9.
Watson, RW, Redmond, HP, Wang, JH, Condron, C, Bouchier-Hayes, D. Neutrophils undergo apoptosis following ingestion of Escherichia coli. J Immunol. 1996;156:3986–92.
Babior, BM, Curnutte, JT, McMurrich, BJ. The particulate superoxide-forming system from human neutrophils. Properties of the system and further evidence supporting its participation in the respiratory burst. J Clin Invest 1976;58:989–96.
Klebanoff, SJ. Myeloperoxidase: friend and foe. J Leukoc Biol. 2005;77:598–625.
Karnovsky, ML. The metabolism of leukocytes. Semin Hematol. 1968;5:156–65.
Nunoi, H, Rotrosen, D, Gallin, JI, Malech, HL. Two forms of autosomal chronic granulomatous disease lack distinct neutrophil cytosol factors. Science. 1988;242:1298–301.
Volpp, BD, Nauseef, WM, Clark, RA. Two cytosolic neutrophil oxidase components absent in autosomal chronic granulomatous disease. Science. 1988;242:1295–7.
Kasahara, Y, Iwai, K, Yachie, A et al. Involvement of reactive oxygen intermediates in spontaneous and CD95 (Fas/APO-1)-mediated apoptosis of neutrophils. Blood. 1997;89:1748–53.
Baran, J, Guzik, K, Hryniewicz, W et al. Apoptosis of monocytes and prolonged survival of granulocytes as a result of phagocytosis of bacteria. Infect Immun. 1996;64:4242–8.
Lagasse, E, Weissman, IL. bcl-2 inhibits apoptosis of neutrophils but not their engulfment by macrophages. J Exp Med. 1994;179:1047–52.
Van, FR, Cohn, ZA. The origin and kinetics of mononuclear phagocytes. J Exp Med. 1968;128:415–35.
Perlman, H, Pagliari, LJ, Georganas, C et al. FLICE-inhibitory protein expression during macrophage differentiation confers resistance to fas-mediated apoptosis. J Exp Med. 1999;190:1679–88.
Liu, H, Perlman, H, Pagliari, LJ, Pope, RM. Constitutively activated Akt-1 is vital for the survival of human monocyte-differentiated macrophages. Role of Mcl-1, independent of nuclear factor (NF)-kappaB, Bad, or caspase activation. J Exp Med. 2001;194:113–26.
Hacker, H, Furmann, C, Wagner, H, Hacker, G. Caspase-9/-3 activation and apoptosis are induced in mouse macrophages upon ingestion and digestion of Escherichia coli bacteria. J Immunol. 2002;169:3172–9.
Marriott, HM, Bingle, CD, Read, RC et al. Dynamic changes in Mcl-1 expression regulate macrophage viability or commitment to apoptosis during bacterial clearance. J Clin Invest. 2005;115:359–68.
Kirschnek, S, Ying, S, Fischer, SF et al. Phagocytosis-induced apoptosis in macrophages is mediated by up-regulation and activation of the Bcl-2 homology domain 3-only protein Bim. J Immunol. 2005;174:671–9.
Albee, L, Shi, B, Perlman, H. Aspartic protease and caspase 3/7 activation are central for macrophage apoptosis following infection with Escherichia coli. J Leukoc Biol. 2007;81:229–37.
Yrlid, U, Wick, MJ. Salmonella-induced apoptosis of infected macrophages results in presentation of a bacteria-encoded antigen after uptake by bystander dendritic cells. J Exp Med. 2000;191:613–24.
Schaible, UE, Winau, F, Sieling, PA et al. Apoptosis facilitates antigen presentation to T lymphocytes through MHC-I and CD1 in tuberculosis. Nat Med. 2003;9:1039–46.
Labbe, K, Saleh, M. Cell death in the host response to infection. Cell Death Differ. 2008;15:1339–49.
Sly, LM, Hingley-Wilson, SM, Reiner, NE, McMaster, WR. Survival of Mycobacterium tuberculosis in host macrophages involves resistance to apoptosis dependent upon induction of antiapoptotic Bcl-2 family member Mcl-1. J Immunol. 2003;170:430–7.
Armstrong, JA, Hart, PD. Response of cultured macrophages to Mycobacterium tuberculosis, with observations on fusion of lysosomes with phagosomes. J Exp Med. 1971;134:713–40.
Orvedahl, A, Levine, B. Eating the enemy within: autophagy in infectious diseases. Cell Death Differ. 2008;16:57–69.
Gutierrez, MG, Master, SS, Singh, SB et al. Autophagy is a defense mechanism inhibiting BCG and Mycobacterium tuberculosis survival in infected macrophages. Cell. 2004;119:753–66.
Blander, JM, Medzhitov, R. Regulation of phagosome maturation by signals from toll-like receptors. Science. 2004;304:1014–18.
Sanjuan, MA, Dillon, CP, Tait, SW et al. Toll-like receptor signalling in macrophages links the autophagy pathway to phagocytosis. Nature. 2007;450:1253–7.

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
×